US20210172020A1 - Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof - Google Patents

Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof Download PDF

Info

Publication number
US20210172020A1
US20210172020A1 US16/986,943 US202016986943A US2021172020A1 US 20210172020 A1 US20210172020 A1 US 20210172020A1 US 202016986943 A US202016986943 A US 202016986943A US 2021172020 A1 US2021172020 A1 US 2021172020A1
Authority
US
United States
Prior art keywords
cells
car
expressing cell
cell
responder
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US16/986,943
Inventor
Felipe Bedoya
Hans Bitter
Jennifer Brogdon
Corin Dorfmeier
Abhishek Garg
David Jonathan Glass
Joan Mannick
Jan J. Melenhorst
Michael C. Milone
Leon Murphy
Elena Orlando
Nicholas Wilcox
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis AG
University of Pennsylvania Penn
Original Assignee
Novartis AG
University of Pennsylvania Penn
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis AG, University of Pennsylvania Penn filed Critical Novartis AG
Priority to US16/986,943 priority Critical patent/US20210172020A1/en
Publication of US20210172020A1 publication Critical patent/US20210172020A1/en
Assigned to NOVARTIS AG, THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA
Assigned to NOVARTIS AG, THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS AG
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4635Cytokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4636Immune checkpoint inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57426Specifically defined cancers leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/54Determining the risk of relapse

Definitions

  • the invention relates to cancer biomarkers and uses thereof.
  • CAR chimeric antigen receptor
  • CART modified autologous T cell
  • CTL019 the clinical results of a CART that binds to CD19 (i.e., “CTL019”) have shown promise in establishing complete remissions in patients suffering with chronic lymphocytic leukemia (CLL), as well as in childhood acute lymphocytic leukemia (ALL) (see, e.g., Kalos et al., SCI TRANSL MED 3:95ra73 (2011), Porter et al., NEJM 365:725-733 (2011), Grupp et al., NEJM 368:1509-1518 (2013)).
  • CLL chronic lymphocytic leukemia
  • ALL childhood acute lymphocytic leukemia
  • a successful therapeutic T cell therapy needs to have the ability to proliferate, to persist over time, and to further monitor for leukemic cell escapees.
  • the variable phenotypic state of T cells whether it is in a state of anergy, suppression or exhaustion, will have effects on CAR-transformed T cells' efficacy.
  • CAR transformed patient T cells need to persist and maintain the ability to proliferate in response to the CAR's antigen.
  • CAR-expressing cell e.g., T cell, NK cell
  • the present disclosure relates to the identification and use of analytes, analyte profiles, or markers (e.g., gene expression, flow cytometry and/or protein expression profiles) with clinical relevance to cancer (e.g., a hematological cancer such as chronic lymphocytic leukemia (CLL) and acute lymphocytic leukemia (ALL)).
  • cancer e.g., a hematological cancer such as chronic lymphocytic leukemia (CLL) and acute lymphocytic leukemia (ALL)).
  • CLL chronic lymphocytic leukemia
  • ALL acute lymphocytic leukemia
  • the disclosure provides the identity of genes, whose expression, at the transcriptional and protein levels, are correlated with CLL and ALL progression, e.g., as a way of predicting a response to a Chimeric Antigen Receptor (CAR)-expressing cell therapy (e.g., a therapy comprising a cell (e.g., an immune effector cell or population of cells) that expresses a CAR that binds to CD19 (also referred to herein as a “CAR19” or “CD19 CAR”-expressing cell).
  • CAR Chimeric Antigen Receptor
  • one or more of a CD19 CAR-expressing cell gene set signature a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g., a na ⁇ ve T cell (T N ), e.g.
  • a memory T cell e.g., a CD8+ memory T cell, e.g
  • a memory stem cell e.g. a central memory T cell (T CM ), e.g. an effector memory T cell (T EM )) gene set signature, and combinations thereof
  • T SCM memory stem cell
  • T CM central memory T cell
  • T EM effector memory T cell
  • These gene expression profiles may be applied to the diagnosis and/or prognosis of a cancer, e.g., a hematological cancer such as CLL and ALL, and are particularly useful in predicting whether a subject will respond favorably to a CAR therapy (e.g., a CD19 CAR therapy as described here, e.g., a CTL019 therapy) in a subject diagnosed with a cancer, e.g., a hematological cancer such as CLL or ALL.
  • a CAR therapy e.g., a CD19 CAR therapy as described here, e.g., a CTL019 therapy
  • the expression profiles of the genes disclosed herein constitute a more robust signature of hematological cancer progression (e.g., CLL and ALL progression) and provide a more reliable, non-subjective basis for the selection of appropriate therapeutic regimens.
  • the present disclosure provides novel gene signatures, e.g., at the transcriptional and protein levels, and methods of use thereof, that predict subject response to a cell expressing a CAR, e.g., a CD19 CAR (e.g., a CD19 CAR-expressing cell, e.g., T cell, NK cell, described herein such as, e.g., CTL019) therapy in a cancer, e.g., a hematological cancer such as CLL and ALL.
  • a CAR e.g., a CD19 CAR (e.g., a CD19 CAR-expressing cell, e.g., T cell, NK cell, described herein such as, e.g., CTL019) therapy in a cancer, e.g., a hematological cancer such as CLL and ALL.
  • a CAR e.g., a CD19 CAR (e.g., a CD19 CAR-expressing cell, e.g
  • the present disclosure demonstrates, at least in part, that expression profiles and gene signatures, e.g., at the transcriptional and protein levels, are useful to distinguish among a responder, a partial responder, a non-responder, a relapser or a non-relapser to a therapy comprising a CAR-expressing cell (e.g., a CAR-expressing immune effector cell, e.g., a T cell, or an NK cell), (also referred to herein as a “CAR-expressing cell therapy”), in a cancer (e.g., a hematological cancer such as CLL and ALL).
  • a CAR-expressing cell e.g., a CAR-expressing immune effector cell, e.g., a T cell, or an NK cell
  • a cancer e.g., a hematological cancer such as CLL and ALL.
  • the CAR-expressing cell is a CD19 CAR-expressing cell.
  • the therapy is a CTL019 therapy.
  • the expression profiles and gene signatures disclosed herein distinguish among a CAR (or CD19 CAR)-expressing cell responder, a CAR (or CD19 CAR)-expressing cell partial responder, or a CAR (or CD19 CAR)-expressing cell non-responder (e.g., a CTL019-responder, a CTL019-partial responder, and a CTL019-non-responder); or a CAR (or CD19 CAR)-expressing cell relapser, or a CAR (or CD19 CAR)-expressing cell non-relapser (e.g., a CTL019-relapser, or a CTL019-relapser), in a cancer (e.g., a hematological cancer such as CLL and ALL).
  • the present disclosure encompasses the identification of novel gene signatures predictive of subject response to a CAR-expressing cell therapy, e.g., a CAR-
  • Exemplary cancers include, but are not limited to, B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome,
  • B-ALL B-cell acute lymphocytic leukemia
  • T-ALL T-cell acute lymphocy
  • the invention features a method of evaluating a subject having a cancer, e.g., a hematological cancer.
  • the method includes acquiring a value of responder or relapser status (e.g., a value of responder or relapser status as described herein) to a therapy comprising a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells) for the subject, wherein said value is indicative of the subject's responsiveness or relapsing status to the CAR-expressing cell therapy.
  • a value of responder or relapser status e.g., a value of responder or relapser status as described herein
  • a therapy comprising a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells) for the subject, wherein said value is
  • the invention features a method of evaluating or monitoring the effectiveness of a CAR-expressing cell therapy in a subject having a cancer, e.g., a hematological cancer.
  • the method includes acquiring a value of responder or relapser status (e.g., a value of responder or relapser status as described herein) to a therapy comprising a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells) for the subject, wherein said value is indicative of the effectiveness of the CAR-expressing cell therapy, thereby evaluating the effectiveness of the CAR-expressing cell therapy in the subject.
  • a value of responder or relapser status e.g., a value of responder or relapser status as described herein
  • a therapy comprising a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.
  • the invention features a method for treating a subject having a cancer, e.g., a hematological cancer.
  • the method includes administering to the subject a therapeutically effective dose of a CAR-expressing cell therapy, if the subject is identified as being responsive (e.g., identified as a complete responder, partial responder or a non-relapser) to a therapy comprising a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells), wherein said identifying comprises a value of responder or relapser status (e.g., a value of responder or relapser status as described herein).
  • a CAR-expressing cell therapy e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells
  • said identifying comprises a value of responder or relapser status (e.g.,
  • the invention features a method of treating a cancer, e.g., a hematological cancer, in a subject.
  • the method includes acquiring a value of responder or relapser status (e.g., a value of responder or relapser status as described herein) to a therapy comprising a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells) for the subject; and responsive to said value, treating the cancer.
  • a value of responder or relapser status e.g., a value of responder or relapser status as described herein
  • a therapy comprising a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells) for the subject; and responsive to said value, treating the cancer.
  • the value of responder or relapser status comprises a measure of one, two, three, four, five, six, seven or more (all) of the following:
  • CD27 and/or CD45RO ⁇ e.g., CD27+CD45RO ⁇
  • immune effector cells e.g., in a CD4+ or a CD8+ T cell population, in a subject, e.g., a sample from the subject (e.g., an apheresis sample or a CAR-expressing cell product sample);
  • a subject e.g., a sample from the subject (e.g., an apheresis sample or a CAR-expressing cell product sample);
  • a subject e.g., a sample from the subject (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • an immune cell exhaustion marker e.g., one, two or more immune checkpoint inhibitors (e.g., PD-1, TIM-3 and/or LAG-3) in a subject, e.g., a sample from the subject (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • an immune cell exhaustion marker e.g., one, two or more immune checkpoint inhibitors (e.g., PD-1, TIM-3 and/or LAG-3)
  • a subject e.g., a sample from the subject (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • a cytokine level or activity e.g., quality of cytokine reportoire
  • a CAR-expressing cell product sample e.g., CAR19-expressing cell product sample (e.g., CTL019)
  • the cytokine is chosen from one, two, three, four, five or more (or all) of the cytokines listed in Table 16;
  • a quantity of CD27+PD-1 ⁇ cells in a subject e.g., a sample from the subject (e.g., an apheresis sample or a CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019)), e.g., a quantity greater than or equal to 1 ⁇ 10 7 cells.
  • the invention provides a CAR expressing cell therapy (e.g., CD19 CART cell, e.g., CTL019 cell) for use in the treatment of a subject, wherein the CAR expressing cell has been assayed according to a method herein, e.g., before or after transduction or transfection with a CAR nucleic acid.
  • a CAR expressing cell therapy e.g., CD19 CART cell, e.g., CTL019 cell
  • the invention provides a CAR expressing cell therapy (e.g., CD19 CART cell, e.g., CTL019 cell), for use in the treatment of a subject that has been identified as being responsive (e.g., identified as a complete responder, partial responder or a non-relapser) to a therapy comprising a CAR-expressing cell population (e.g., a CAR19-expressing cell population).
  • a CAR expressing cell therapy e.g., CD19 CART cell, e.g., CTL019 cell
  • a therapy comprising a CAR-expressing cell population (e.g., a CAR19-expressing cell population).
  • the composition for use can comprise a measure of one, two, three, four, five, six, seven, or more (all) of (i)-(viii) described herein.
  • identifying the subject as a complete responder, partial responder or non-responder, or a relapser or a non-relapser;
  • administering e.g., to a responder or a non-relapser, a CAR-expressing cell therapy
  • an additional agent in combination with a CAR-expressing cell therapy e.g., a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein;
  • modifying a process, e.g., a manufacturing process, of a CAR-expressing cell therapy e.g., enriching for younger T cells prior to introducing a nucleic acid encoding a CAR, or increasing the transduction efficiency, e.g., for a subject identified as a non-responder or a partial responder;
  • administering an alternative therapy e.g., for a non-responder or partial responder or relapser, e.g., a standard of care for a particular cancer type; or
  • T REG cell population and/or T REG gene signature e.g., by one or more of CD25 depletion, or administration of cyclophosphamide, anti-GITR antibody, an mTOR inhibitor, or a combination thereof.
  • the subject is pre-treated with an anti-GITR antibody. In certain embodiment, the subject is treated with an anti-GITR antibody prior to infusion or re-infusion. In some embodiments, the subject is a patient with CLL.
  • the invention features a method of, or assay for, identifying a subject having a cancer as having an increased or decreased likelihood to respond to a treatment that comprises a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells).
  • a CAR-expressing cell e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells).
  • the method includes:
  • a difference e.g., a statistically significant difference, between the determined level compared to a reference level is predictive of the subject's responsiveness to the CAR-expressing cell therapy
  • the invention features a method for treating a subject having a cancer comprising:
  • determining if the subject has an increased likelihood to respond, or a decreased likelihood to relapse, to a CAR-expressing cell therapy e.g., a CAR19-expressing therapy, e.g., CTL019
  • a CAR-expressing cell therapy e.g., a CAR19-expressing therapy, e.g., CTL019
  • determining the level or activity of one or more biomarkers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 e.g., CCL20, IL-17a and/or IL-6
  • the invention features a method for treating a subject having cancer comprising:
  • determining if the subject has an increased likelihood to relapse to a CAR-expressing cell therapy by acquiring a value for the level or activity of one or more markers in a Table herein, e.g., Table 17 in a sample from the subject (e.g., an apheresis sample or a manufactured CAR-expressing product sample), wherein a difference, e.g., a statistically significant difference, in the level or activity of one or more biomarker genes relative to the reference level is indicative of an increased likelihood of relapse to a CAR-expressing cell therapy; and
  • an immune cell has an exhausted phenotype, e.g., co-expresses at least two exhaustion markers, e.g., co-expresses PD-1 and TIM-3. In other embodiments, an immune cell has an exhausted phenotype, e.g., co-expresses at least two exhaustion markers, e.g., co-expresses PD-1 and LAG-3.
  • the CAR-expressing cell therapy comprises a plurality (e.g., a population) of CAR-expressing immune effector cells, e.g., a plurality (e.g., a population) of T cells or NK cells, or a combination thereof.
  • the CAR-expressing cell therapy is a CAR19 therapy (e.g., CTL019 therapy).
  • the CAR-expressing cell therapy comprises or consists of CTL019.
  • the CAR-expressing cell is a CTL019 product.
  • the CAR-expressing cell is a T cell.
  • the CAR-expressing cell is a NK cell.
  • the measure of one or more of (i)-(viii) is obtained from an apheresis sample acquired from the subject.
  • the apheresis sample can be evaluated prior to infusion or re-infusion.
  • the measure of one or more of (i)-(viii) is obtained from a manufactured CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019).
  • the manufactured CAR-expressing cell product can be evaluated prior to infusion or re-infusion.
  • the subject is evaluated prior to receiving, during, or after receiving, the CAR-expressing cell therapy.
  • the hematological cancer is an ALL or a CLL.
  • the subject can be a human patient.
  • the cell e.g., the population of immune effector cells (e.g., cells expressing a CAR molecule described herein) is administered in combination with an inhibitor of an immune checkpoint molecule chosen from one or more of PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, TGFR (e.g., TGFR beta), or a combination thereof.
  • an immune checkpoint molecule chosen from one or more of PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEA
  • the subject receives concurrent treatment with an agent, e.g., an mTOR inhibitor, and/or a checkpoint inhibitor.
  • the subject receives treatment with an agent, e.g., an mTOR inhibitor, and/or a checkpoint inhibitor, post-CAR-expressing cell therapy.
  • the subject receives a pre-treatment of with an agent, e.g., an mTOR inhibitor, and/or a checkpoint inhibitor, prior to the initiation of a CAR-expressing cell therapy.
  • T REG cell population and/or T REG gene signature is decreased prior to collection of cells for manufacturing. In some embodiments, the T REG cell population and/or T REG gene signature is decreased prior to CAR-expressing cell (e.g., T cell, NK cell) therapy. In some embodiments, the T REG cell population and/or T REG gene signature is decreased by administration of cyclophosphamide, anti-GITR antibody, an mTOR inhibitor, or a combination thereof.
  • the value of responder or relapser status comprises a measure of a combination of a gene signature and a biomarker.
  • the value of the responder or relapser status comprises a measure of a CD19 CAR-expressing cell gene set signature and a combination of one or more of: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, or KLRG1.
  • the method further comprises identifying the subject as a responder (e.g., a complete or partial responder), a non-responder, a relapser or a non-relapser, based on a measure of one or more of (i)-(viii).
  • a responder e.g., a complete or partial responder
  • a non-responder e.g., a relapser or a non-relapser
  • the measure of one or more of (i)-(viii) evaluates a profile for one or more of gene expression, flow cytometry or protein expression.
  • the expression profile includes one or more gene signatures based on mRNA expression levels of selected genes obtained from the apheresis sample or a manufactured CD19 CAR-expressing cell product (e.g., CTL019).
  • the expression profile includes one, two, three, four, five, ten, twenty or more of a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 16, Table 17, Table 18, Table 20, FIG. 2B , PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature.
  • the level or activity of a CD8+ T cell is evaluated using a profile or signature indicative of the percentage of CD8+ T cell in the sample.
  • the level or activity of CD27+CD45RO ⁇ immune effector cells is evaluated using a profile or signature indicative of the percentage of CD27+CD45RO ⁇ immune effector cells in the sample.
  • the level or activity e.g., in (i), (ii), or (v) is evaluated using a profile or gene signature according to one, two, three, four, five, ten, twenty, fifty, sixty, seventy, one hundred or more of a biomarker or gene set listed in Tables 1A, 1B, 3, 4, 5, 6, or FIG. 2B .
  • the level or activity one, two or more immune checkpoint inhibitors is evaluated, e.g., using flow cytometry, as an indicator of the percentage of PD-1+/LAG-3+ cells in the CAR-expressing cell population (e.g., a CAR19+ cell population).
  • the level or activity one, two or more immune checkpoint inhibitors is evaluated, e.g., using flow cytometry, as an indicator of the percentage of PD-1+/TIM-3+ cells in the CAR-expressing cell population (e.g., a CAR19+ cell population).
  • the level or activity of one, two, three, four, five, ten, twenty, fifty, sixty, seventy, one hundred or more of a biomarker or gene set listed in Table 7A, Table 7B, Table 8 and FIG. 2B predicts a subject's response to a CAR19+ cell product (e.g., CTL019).
  • the value of responder or relapser status comprises a measure of the level or activity of one, two, three, four, five, ten, twenty or more (e.g., all) of the biomarkers having a given FDR p-value, listed herein, e.g., in a Table herein.
  • the FDR p-value is below 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001.
  • the FDR p-value is below 0.1 or 0.01.
  • the biomarkers are biomarkers listed in Table 1A, Table 1B, Table 16, Table 17, Table 18, or Table 20, or a combination thereof.
  • the measure comprises a measure of all of the biomarkers in Table 1A that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001. In some embodiments, the measure comprises a measure of all of the biomarkers in Table 1B that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001. In some embodiments, the measure comprises a measure of all of the biomarkers in Table 16 that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001.
  • the measure comprises a measure of all of the biomarkers in Table 17 that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001. In some embodiments, the measure comprises a measure of all of the biomarkers in Table 18 that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001. In some embodiments, the measure comprises a measure of all of the biomarkers in Table 20 that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001.
  • the measure comprises a measure of all of the biomarkers having a p-value below the threshold. In some embodiments, the measure comprises a measure of one, two, three, four, five, ten, twenty, fifty, or one hundred biomarkers having a p-value below the threshold. In some embodiments, the measure comprises a measure of at least one, two, three, four, five, ten, twenty, fifty, or one hundred biomarkers having a p-value below the threshold. In some embodiments, the measure comprises a measure of 1-5, 5-10, 10-20, 20-50, or 50-100 biomarkers having a p-value below the threshold.
  • biomarkers of Table 7B that are designated “CR” in the table are upregulated in complete responders compared to non-responders. In some embodiments, biomarkers of Table 7B that are designated “NR” in the table are upregulated in non-responders compared to complete responders.
  • the biomarker is a secreted or a cell surface biomarker listed in Table 8.
  • the biomarker can be measured by flow cytometry.
  • a responder e.g., a complete responder
  • a non-responder has, or is identified as having, a greater level or activity of one, two, three, four, five, six, seven, or more (e.g., all) of IL22, IL-2RA, IL-21, IRF8, IL8, CCL17, CCL22, effector T cells, or regulatory T cells, as compared to a responder.
  • a relapser is a patient having, or who is identified as having, an increased level of expression of one or more of (e.g., 2, 3, 4, or all of) the following genes, compared to non relapsers: MIR199A1, MIR1203, uc021ovp, ITM2C, and HLA-DQB1 and/or a decreased levels of expression of one or more of (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or all of) the following genes, compared to non relapsers: PPIAL4D, TTTY10, TXLNG2P, MIR4650-1, KDMSD, USP9Y, PRKY, RPS4Y2, RPS4Y1, NCRNA00185, SULT1E1, and EIF1AY.
  • a complete responder has, or is identified as having, a greater, e.g., a statistically significant greater, percentage of CD8+ T cells compared to a reference value, e.g., a non-responder percentage of CD8+ T cells.
  • a complete responder has, or is identified as having, a greater percentage (e.g., 5%, 6%, 7%, 10%, 15%, 20%, 25%, 27%, 30%, 35%, or 40% or greater number) of CD27+CD45RO ⁇ immune effector cells, e.g., in the CD8+ population, compared to a reference value, e.g., a non-responder number of CD27+CD45RO ⁇ immune effector cells.
  • a reference value e.g., a non-responder number of CD27+CD45RO ⁇ immune effector cells.
  • a complete responder or a partial responder has, or is identified as having, a greater, e.g., a statistically significant greater, percentage of CD4+ T cells compared to a reference value, e.g., a non-responder percentage of CD4+ T cells.
  • a complete responder has, or is identified as having, a greater percentage of one, two, three, or more (e.g., all) of resting T EFF cells, resting T REG cells, younger T cells (e.g., younger CD4 or CD8 cells, or gamma/delta T cells), or early memory T cells, or a combination thereof, compared to a reference value, e.g., a non-responder number of resting T EFF cells, resting T REG cells, younger T cells (e.g., younger CD4 or CD8 cells), or early memory T cells.
  • a reference value e.g., a non-responder number of resting T EFF cells, resting T REG cells, younger T cells (e.g., younger CD4 or CD8 cells), or early memory T cells.
  • a non-responder has, or is identified as having, a greater percentage of one, two, three, or more (e.g., all) of activated T EFF cells, activated T REG cells, older T cells (e.g., older CD4 or CD8 cells), or late memory T cells, or a combination thereof, compared to a reference value, e.g., a responder number of activated T EFF cells, activated T REG cells, older T cells (e.g., older CD4 or CD8 cells), or late memory T cells.
  • a reference value e.g., a responder number of activated T EFF cells, activated T REG cells, older T cells (e.g., older CD4 or CD8 cells), or late memory T cells.
  • a non-responder has, or is identified as having, a greater percentage of an immune cell exhaustion marker, e.g., one, two or more immune checkpoint inhibitors (e.g., PD-1, TIM-3 and/or LAG-3).
  • an immune cell exhaustion marker e.g., one, two or more immune checkpoint inhibitors (e.g., PD-1, TIM-3 and/or LAG-3).
  • a non-responder has, or is identified as having, a greater percentage of PD-1 or LAG-3 expressing immune effector cells (e.g., CD4+ T cells and/or CD8+ T cells) (e.g., CAR-expressing CD4+ cells and/or CD8+ T cells) compared to the percentage of PD-1 or LAG-3 expressing immune effector cells from a responder.
  • immune effector cells e.g., CD4+ T cells and/or CD8+ T cells
  • CAR-expressing CD4+ cells and/or CD8+ T cells
  • a non-responder has, or is identified as having, a greater percentage of immune cells having an exhausted phenotype, e.g., immune cells that co-express at least two exhaustion markers, e.g., co-expresses PD-1 and TIM-3. In other embodiments, a non-responder has, or is identified as having, a greater percentage of immune cells having an exhausted phenotype, e.g., immune cells that co-express at least two exhaustion markers, e.g., co-expresses PD-1 and LAG-3.
  • a non-responder has, or is identified as having, a greater percentage of PD-1+/LAG-3+ cells in the CAR-expressing cell population (e.g., a CAR19+ cell population) compared to a responder (e.g., a complete responder) to the CAR-expressing cell therapy.
  • a partial responder has, or is identified as having, a higher percentages of PD-1+/LAG-3+ cells, than a responder, in the CAR-expressing cell population (e.g., a CAR19+ cell population).
  • a non-responder has, or is identified as having, an exhausted phenotype of PD1+CAR+ and co-expression of LAG3 in the CAR-expressing cell population (e.g., a CAR19+ cell population).
  • a non-responder has, or is identified as having, a greater percentage of PD-1+/TIM-3+ cells in the CAR-expressing cell population (e.g., a CAR19+ cell population) compared to the responder (e.g., a complete responder).
  • a partial responders has, or is identified as having, a higher percentage of PD-1+/TIM-3+ cells, than responders, in the CAR-expressing cell population (e.g., a CAR19+ cell population).
  • the presence of CD8+CD27+CD45RO ⁇ T cells in an apheresis sample is a positive predictor of the subject response to a CAR-expressing cell therapy (e.g., a CAR19 therapy (e.g., CTL019 therapy)).
  • a CAR-expressing cell therapy e.g., a CAR19 therapy (e.g., CTL019 therapy)
  • a high percentage of PD1+CAR+ and LAG3+ or TIM3+ T cells in an apheresis sample is a poor prognostic predictor of the subject response to a CAR-expressing cell therapy (e.g., a CAR19 therapy (e.g., CTL019 therapy)).
  • a CAR-expressing cell therapy e.g., a CAR19 therapy (e.g., CTL019 therapy
  • a responder e.g., a complete responder
  • a CAR19 therapy has, or is identified as having, the biomarker profile of Table 9.
  • a non-responder to a CAR19 therapy has, or is identified as having, the biomarker comprising one or more of PD-1+ immune effector cells, TIM-3+ immune effector cells, LAG-3+ immune effector cells, KLRG1+ immune effector cells, CD27-immune effector cells, activated T EFF cells, activated T REG cells, activated TH1, activated TH2 cells, stimulated memory cells, or late T memory cells, or a combination thereof,
  • a non-responder to a CAR19 therapy has, or is identified as having, the biomarker profile of Table 10.
  • expression of one, two, three, four or more (all) of KLRG1, CD57, CD27, CD122, or CD62L is predictive of patient response to CTL019 therapy.
  • a non-relapser is a patient with B-ALL, and has, or is identified as having, one or more expression profiles (e.g., protein or gene expression profiles) or gene signatures characteristic of resting T EFF cells or resting T REG cells.
  • expression profiles e.g., protein or gene expression profiles
  • gene signatures characteristic of resting T EFF cells or resting T REG cells.
  • a relapser is a patient with B-ALL, and has, or is identified as having, one or more expression profiles (e.g., protein or gene expression profiles) or gene signatures characteristic of activated T EFF cells or activated T REG cells.
  • expression profiles e.g., protein or gene expression profiles
  • a T REG cell (e.g., an activated T REG cell) has upregulated expression of one or more (e.g., at least 10, 20, 30, 40, 50, 60, 70, or all) of the following biomarkers: AIM2, ALAS1, BATF, C5orf32, CCL17, CD40LG, CHAC2, CSF1, CTSL1, EBNA1BP2, EDARADD, EMP1, EPAS1, FABP5, FAM40B, FKBP4, FOSL1, GCLM, GK, GPR56, HMOX1, HSPD1, HSPE1, IKBIP, IL10, IL13, IL15RA, IL1RN, IL2RA, IL3, IL4, IL5, IL9, KCNK5, LTA, MANF, MIR1182, MIR155, MIR155HG, MYOF, NDUFAF1, NLN, NME1, NME1-NME2, P
  • a T EFF cell (e.g., an activated T EFF cell) has upregulated expression of one or more (e.g., at least 10, 20, 30, 40, 50, 60, 70, or all) of the following biomarkers: AIM2, ALAS1, B4GALT5, BATF, C3orf26, C4orf43, CCL3, CCL4, CCT3, CCT7, CD40LG, CHAC2, CSF2, CTNNA1, EBNA1BP2, EDARADD, EEF1E1, EIF2B3, EIF2S1, FABP5, FAM40B, FKBP4, FOSL1, GFOD1, GLRX2, HSPD1, HSPE1, IFNG, IL15RA, IL21, IL2RA, IL3, KCNK5, KIAA0020, LARP4, LRP8, LTA, MANF, MIR1182, MIR155, MIR155HG, MTCH2, MYOF, KIAA0020, LARP4, LRP8, LTA, MANF, MIR
  • a relapser is a patient with B-ALL and has, or is identified as having, one or more protein or gene expression profiles comprising one, two, three, four, five, ten or more genes according to Table 7A, Table 7B or FIG. 2B or a combination thereof.
  • a relapser has, or is identified as having, an elevated level of one or more T REG cell biomarkers, or a combination thereof.
  • the relapser has, or is identified as having, upregulated expression of one or more (e.g., at least 10, 20, 30, 40, 50, 60, 70, or all) of the following genes: AIM2, ALAS1, BATF, C5orf32, CCL17, CD40LG, CHAC2, CSF1, CTSL1, EBNA1BP2, EDARADD, EMP1, EPAS1, FABP5, FAM40B, FKBP4, FOSL1, GCLM, GK, GPR56, HMOX1, HSPD1, HSPE1, IKBIP, IL10, IL13, IL15RA, IL1RN, IL2RA, IL3, IL4, IL5, IL9, KCNK5, LTA, MANF, MIR1182, MIR155, MIR155
  • the relapser has, or is identified as having, upregulated expression of one or more (e.g., at least 10, 20, 25, or all) of the following genes: C5orf32, CCL17, CSF1, CTSL1, EMP1, EPAS1, GCLM, GK, GPR56, HMOX1, IKBIP, IL10, IL13, IL1RN, IL4, IL5, IL9, MIR155, PANX2, PGAM4, PRKAR1B, TNFRSF11A, TNFRSF1B, TNFRSF8, VTRNA1-3, or ZNF282.
  • the upregulated expression may be, e.g., measured 16 hours after stimulation.
  • the upregulated expression may be determined, e.g., by measuring RNA levels for the indicated genes.
  • a subject has, or is identified as having, an elevated level of one or more T EFF cell biomarkers, or a combination thereof.
  • the subject has, or is identified as having, upregulated expression of one or more (e.g., at least 10, 20, 30, 40, 50, 60, 70, or all) of the following genes: AIM2, ALAS1, B4GALT5, BATF, C3orf26, C4orf43, CCL3, CCL4, CCT3, CCT7, CD40LG, CHAC2, CSF2, CTNNA1, EBNA1BP2, EDARADD, EEF1E1, EIF2B3, EIF2S1, FABP5, FAM40B, FKBP4, FOSL1, GFOD1, GLRX2, HSPD1, HSPE1, IFNG, IL15RA, IL21, IL2RA, IL3, KCNK5, KIAA0020, LARP4, LRP8, LTA, MANF, MIR
  • the subject has, or is identified as having, upregulated expression of one or more (e.g., at least 10, 20, 25, or all) of the following genes: B4GALT5, C3orf26, C4orf43, CCL3, CCL4, CCT3, CCT7, CSF2, CTNNA1, EEF1E1, EIF2B3, EIF2S1, GFOD1, GLRX2, IL21, IL2RA, IL3, KIAA0020, LARP4, LRP8, OTUD7B, PAM, PEA15, PFKM, PGAM1, PGAM4, PRSS23, PSMD1, PSMD11, PSMD14, PTRH2, RPF2, SORD, SPR, TMCC2, TMEM165, or TXNDC5.
  • the upregulated expression may be, e.g., measured 16 hours after stimulation.
  • the upregulated expression may be determined, e.g., by measuring RNA levels for the indicated genes.
  • the responder e.g., the complete or partial responder
  • the responder has one, two, three or more (or all) of the following profile:
  • (i) has a greater number of CD27+ immune effector cells compared to a reference value, e.g., a non-responder number of CD27+ immune effector cells;
  • (ii) has a greater number of CD8+ T cells compared to a reference value, e.g., a non-responder number of CD8+ T cells;
  • checkpoint inhibitors e.g., a checkpoint inhibitor chosen from PD-1, LAG-3, TIM-3, or KLRG-1, or a combination, compared to a reference value, e.g., a non-responder number of cells expressing one or more checkpoint inhibitors; or
  • (iv) has a greater number of one, two, three, four or more (all) of resting T EFF cells, resting T REG cells, younger cells, na ⁇ ve CD4 cells, unstimulated memory cells or early memory T cells, or a combination thereof, compared to a reference value, e.g., a non-responder number of resting T EFF cells, resting T REG cells, na ⁇ ve CD4 cells, unstimulated memory cells or early memory T cells.
  • a reference value e.g., a non-responder number of resting T EFF cells, resting T REG cells, na ⁇ ve CD4 cells, unstimulated memory cells or early memory T cells.
  • the cytokine level or activity is chosen from one, two, three, four, five, six, seven, eight, or more (or all) of cytokine CCL20/MIP3a, IL17A, IL6, GM-CSF, IFN ⁇ , IL10, IL13, IL2, IL21, IL4, IL5, IL9 or TNF ⁇ , or a combination thereof.
  • the cytokine can be chosen from one, two, three, four or more (all) of IL-17a, CCL20, IL2, IL6, or TNFa.
  • an increased level or activity of a cytokine is chosen from one or both of IL-17a and CCL20, is indicative of increased responsiveness or decreased relapse.
  • the cytokine level is measured after T cell activation.
  • a transduction efficiency of 15% or higher, e.g., in (vii), is indicative of increased responsiveness or decreased relapse.
  • a transduction efficiency of less than 15%, e.g., in (vii), is indicative of decreased responsiveness or increased relapse.
  • the invention features a method to identify a likely responder (e.g., a complete responder or a partial responder, a non-relapser) to a therapy comprising a CAR-expressing cell (e.g., a T cell, an NK cell) (e.g., a CD19 CAR-expressing cell therapy, e.g., described herein, e.g., a CTL019 therapy).
  • a responder status e.g.
  • a complete responder, a partial responder, a non-responder, a relapser or a non-relapser to a therapy comprising a CAR-expressing cell is determined by measuring one or more of a CD19 CAR-expressing cell gene set signature, a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T
  • a memory stem cell e.g. a central memory T cell (T CM ), e.g. an effector memory T cell (T EM )) gene set signature, and combinations thereof.
  • a complete responder has, e.g., two, three, four or more (e.g., all) of CD27 + , CD45RO ⁇ , PD-1 ⁇ , LAG-3 ⁇ , and TIM-3 ⁇ , as described in Table 9.
  • a non-responder has, e.g., two, three, or more of (e.g., all) of CD27 ⁇ CD45RO + , PD-1 + , LAG-3 + , and TIM-3 + , as described in Table 10.
  • the responder or relapser status (e.g. complete responder, partial responder, non-responder, relapser or non-relapser to a CAR-expressing cell therapy) is determined by evaluating, e.g., measuring, two, three, four, five, six, seven, eight, nine, ten, fifteen or more of a CD19 CAR-expressing cell gene set signature, a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1.
  • a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20,
  • any of the methods and compositions for use disclosed herein can be used prior to administration of a CAR-expressing cell therapy.
  • provided methods can be used before, at the same time, or during course of a CAR-expressing cell therapy.
  • any of the methods and compositions for use disclosed herein can be used to identify a subject having cancer, e.g., a hematological cancer such as, e.g., CLL or ALL, as having an increased or a decreased likelihood to respond to a treatment that comprises a CAR-expressing cell (e.g., T cell, NK cell) therapy, e.g., a CD19 CAR-expressing cell therapy.
  • a hematological cancer such as, e.g., CLL or ALL
  • a CAR-expressing cell e.g., T cell, NK cell
  • CD19 CAR-expressing cell therapy e.g., CD19 CAR-expressing cell therapy.
  • the method comprises: (1) acquiring a sample from the subject (e.g., an apheresis sample obtained from the blood of the subject; and/or e.g., a manufactured product sample, e.g., genetically engineered T cells); (2) determining a level (e.g., amount or activity) of one or more biomarkers (e.g., 2, 3, 4, 5, 10, 15 or more) listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 in the sample; and (3) (optionally) comparing the determined level of the one or more markers to a reference level; and (4) identifying the subject as a complete responder, partial responder, non-responder, a relapser or non-relapser to the C
  • provided methods comprise (1) acquiring a sample (e.g., an apheresis sample obtained from the subject; and/or e.g., a manufactured product sample, e.g., genetically engineered T cells, e.g., a manufactured CD19 CAR-expressing cell product); (2) acquiring, e.g., determining a gene signature of the sample; and (3) (optionally) comparing the gene signature to a reference gene signature; wherein a difference, e.g., a statistically significant difference, in expression level of one or more of the biomarkers in the determined gene signature is predictive of the subjects responsiveness to the CAR-expressing cell therapy.
  • a sample e.g., an apheresis sample obtained from the subject; and/or e.g., a manufactured product sample, e.g., genetically engineered T cells, e.g., a manufactured CD19 CAR-expressing cell product
  • acquiring e.g., determining a gene signature of the sample
  • the gene signature comprises one or more markers selected from Table 1A, Table 1B, Table 2, Table 3, Table 4, Table 5, Table 6, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and combinations thereof.
  • the sample is a biological sample selected from a blood, plasma, or a serum sample.
  • a biological sample is a blood sample.
  • the sample is an apheresis sample, e.g., immune effector cells (e.g., T cells) obtained from the blood of the subject.
  • the sample is a manufactured product sample, e.g. genetically engineered T cells, e.g., a manufactured CD19 CAR-expressing cell product.
  • methods for determining the responsiveness of a subject having cancer, e.g., a hematological cancer such as, e.g., CLL or ALL, to a treatment comprising a CAR-expressing cell (e.g., a T cell, an NK cell) therapy, e.g., a CD19 CAR-expressing cell therapy, e.g., described herein.
  • a CAR-expressing cell e.g., a T cell, an NK cell
  • CD19 CAR-expressing cell therapy e.g., described herein.
  • the method includes: determining a level or activity of one or more biomarkers (e.g., 2, 3, 4, 5, 10, 15 or more) listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 in a sample obtained prior to treatment; wherein a difference, e.g., a statistically significant difference, in a level (e.g., amount or activity) of one or more markers in the sample relative to a predetermined value is indicative of increased responsiveness to the CAR-expressing cell.
  • biomarkers e.g., 2, 3, 4, 5, 10, 15 or more listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a
  • the sample is a biological sample selected from a blood, plasma, or a serum sample.
  • a biological sample is a blood sample.
  • the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject.
  • the sample is a manufactured product sample, e.g. genetically engineered T cells, e.g., obtained from the blood of the subject, e.g., a manufactured CAR-expressing cell product, e.g., a manufactured CD19 CAR-expressing cell product.
  • methods for evaluating a subject having cancer, e.g., a hematological cancer such as, e.g., CLL or ALL, comprising acquiring a value of responder or relapser status for the subject that comprises a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4+ biomarker, a CD8+ biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell
  • T SCM memory stem cell
  • T CM central memory T cell
  • T EM effector memory T cell
  • CD19 CAR-expressing cell e.g., T cell, NK cell
  • methods comprise a measure of one or more of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature.
  • a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature.
  • methods comprise a measure of one or more of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature, for evaluating a subject having CLL.
  • a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature, for evaluating a subject having CLL.
  • methods comprise a measure of one or more of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature, for evaluating a subject having ALL.
  • a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature, for evaluating a subject having ALL.
  • the method comprises a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature in a biological sample selected from a blood, plasma, or a serum sample.
  • a biological sample is a blood sample.
  • the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject.
  • the sample is a manufactured product sample, e.g. genetically engineered T cells, e.g., obtained from the blood of the subject, e.g., a manufactured CAR-expressing cell product, e.g., a manufactured CD19 CAR-expressing cell product.
  • methods for evaluating or monitoring the effectiveness of a CAR-expressing cell (e.g., T cell, NK cell) therapy, e.g., a CD19 CAR-expressing cell therapy, in a subject having cancer comprising acquiring a value of responder or relapser status for the subject that comprises a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T
  • T SCM memory stem cell
  • T C M central memory T cell
  • T EM effector memory T cell
  • methods comprise a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature.
  • a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature.
  • methods comprise a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature, for evaluating or monitoring the effectiveness of a CAR-expressing cell therapy in a subject having CLL.
  • a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD
  • methods comprise a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature, for evaluating or monitoring the effectiveness of a CAR-expressing cell therapy in a subject having ALL.
  • a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19
  • the method comprises a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature in a biological sample selected from a blood, plasma, or a serum sample.
  • a biological sample is a blood sample.
  • the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject.
  • the sample is a manufactured product sample, e.g. genetically engineered T cells, e.g., obtained from the blood of the subject.
  • methods for providing a prediction for success rate of a CAR-expressing cell therapy, e.g., a CD19 CAR-expressing cell therapy, e.g., described herein, in a subject having cancer, said method comprising steps of providing a biological sample from the subject; determining the levels of expression of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) genes listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), and Table 17 to obtain a gene expression pattern for the sample; and based on the gene expression pattern obtained, providing a prognosis to the subject.
  • a CAR-expressing cell therapy e.g., a CD19 CAR-expressing cell therapy, e.g., described herein
  • said method comprising steps of providing a biological sample from the subject; determining the levels of expression of one or more (e.g., 2, 3, 4, 5, 10,
  • a biological sample includes, but is not limited to a blood, plasma, or a serum sample.
  • a biological sample is a blood sample.
  • the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject.
  • the subject has CLL.
  • the subject has ALL.
  • methods for treating a subject having cancer e.g., a hematological cancer.
  • methods for treating a subject having cancer determining if a subject has a difference, e.g., a statistically significant difference, in expression level of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) markers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6) and Table 17 relative to a reference level, and if there is a difference, e.g., a statistically significant difference, between the determined level and reference level, administering to the subject a therapeutically effective dose of a CAR-expressing cell (e.g., T cell, NK cell), thereby treating the subject.
  • a CAR-expressing cell e.g., T cell, NK cell
  • the method comprises modifying the CAR-expressing cell product prior to infusion into the subject. In an embodiment, wherein there is a difference, e.g., a statistically significant difference, between the determined level and the reference level, the method comprises modifying the manufacture of a CAR-expressing cell product prior to infusion into the subject. In an embodiment, if there is a difference, e.g., a statistically significant difference, between the determined level and reference level, the method comprises adjusting the CAR-expressing cell infusion dose to achieve an anti-cancer effect.
  • the methods of treatment described herein comprise determining if a subject has an increased likelihood to respond to a CAR-expressing cell (e.g., T cell, NK cell) therapy, e.g., a CD19 CAR-expressing cell therapy, e.g., a CD19 CAR-expressing cell therapy described herein, by comparing the level of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15 and Table 16 (e.g., CCL20, IL-17a and/or IL-6) in a sample from the subject relative to a reference level, wherein a difference, e.g., a statistically significant difference, in expression level of one or more maker genes relative to the reference level is indicative of an increased likelihood of response; and administering to the subject a therapeutically effective dose of a CAR-expressing cell, thereby treating the subject.
  • the sample is selected from a
  • the methods of treatment described herein further comprise obtaining a sample from a subject; determining a level of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1, in the sample; comparing the determined level of one or more markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1, to a reference level; and administering a therapeutically effective dose of a CAR-
  • the methods of treatment described herein comprise, or further comprise, acquiring a value of responder or relapser status for the subject that comprises a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g.,
  • a memory stem cell e.g. a central memory T cell (T CM ), e.g. an effector memory T cell (T EM )) gene set signature, and a CD19 CAR-expressing cell gene set signature, and responsive to a determination of responder or relapser status, performing one, two, three four or more of: identifying the subject as a complete responder, partial responder or non-responder; administering a CAR-expressing cell therapy; selecting or altering a dosing of a CAR-expressing cell therapy; selecting or altering the schedule or time course of a CAR-expressing cell therapy; administering, e.g., to a non-responder or a partial responder, an additional agent in combination with a CAR-expressing cell therapy, e.g., a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein, or a kinase inhibitor, e.g., a kinase inhibitor described herein; administering
  • kits for predicting subject response to CAR-expressing cell e.g., T cell, NK cell
  • CAR-expressing cell e.g., T cell, NK cell
  • a CD19 CAR-expressing cell therapy e.g., a CD19 CAR-expressing cell therapy described herein.
  • kits comprise at least one reagent that specifically detects the level or activity of a set of genes (e.g., 2, 3, 4, 5, 10, 15 or more of the genes) selected from Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and/or KLRG1; and instructions for using the kits, e.g., for predicting a subject's response to a CAR-expressing cell therapy.
  • a set of genes e.g., 2, 3, 4, 5, 10, 15 or more of the genes selected from Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3
  • said instructions for use provide that if one or more of the detected expression levels is different from, e.g., greater than a reference level, the subject is more likely to respond positively to a CAR-expressing cell therapy. In some embodiments, said instructions for use provide that if one or more of the detected expression levels is less than a reference level, the subject is more likely to respond positively to a CAR-expressing cell therapy. In some embodiments, if the level or activity of PD-1, LAG-3, or TIM-3, or any combination thereof, is less than a reference value, the subject is more likely to respond positively to the therapy.
  • At least one reagent that specifically detects expression levels of the set of genes comprises a nucleic acid probe complementary to mRNA expressed from the genes, for example a CDNA or an oligonucleotide.
  • the nucleic acid probe may be immobilized on a substrate surface or may be in solution.
  • the set of reagents may detect the expression of polypeptides, e.g., surface polypeptides, encoded by said set of genes.
  • the nucleic acid probe comprises a nucleic acid of about 10, 15, 20, 25, 30, 35, 40, 45, 50 or 100 nucleic acid residues complementary the nucleic acid sequence of a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and/or KLRG1.
  • the kits may further comprise one or more of: extraction buffer/reagents and protocol, amplification buffer/reagents and protocol, hybridization buffer/reagents and protocol, and labeling buffer/reagents and protocol.
  • kits further comprise at least one CD19 CAR-expressing cell (e.g., T cell, NK cell) gene set signature.
  • kit further comprises a reference standard.
  • the subject has CLL.
  • the subject has ALL.
  • the subject has B-cell ALL.
  • the disclosure features a reaction mixture comprising at least one reagent that specifically detects expression levels of a set of genes (e.g., 2, 3, 4, 5, 10, 15 or more of the genes) selected from Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and/or KLRG1; and a biological sample.
  • the sample is selected from a blood, plasma or a serum sample.
  • the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject.
  • the sample comprises CAR-expressing cells, e.g., CART-expressing cells, e.g., CART19 cells.
  • At least one reagent that specifically detects expression levels of the set of genes comprises a nucleic acid probe complementary to mRNA expressed from the genes, for example a CDNA or an oligonucleotide.
  • the nucleic acid probe may be immobilized on a substrate surface or may be in solution.
  • the set of reagents may detect the expression of polypeptides, e.g., surface polypeptides, encoded by said set of genes.
  • the nucleic acid probe comprises a nucleic acid of about 10, 15, 20, 25, 30, 35, 40, 45, 50 or 100 nucleic acid residues complementary the nucleic acid sequence of a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and/or KLRG1.
  • the reaction mixture may further comprise one or more of: extraction buffer/reagents, amplification buffer/reagents, hybridization buffer/reagents, and labeling buffer/reagents.
  • the present disclosure features a system for evaluating cancer in a subject.
  • the system comprises at least one processor operatively connected to a memory, the at least one processor when executing is configured to perform any one or more of the steps described herein.
  • the present disclosure features a system for evaluating cancer in a subject.
  • the system includes at least one processor operatively connected to a memory, the at least one processor when executing is configured to:
  • CD27 and/or CD45RO ⁇ e.g., CD27+CD45RO ⁇
  • immune effector cells e.g., in a CD4+ or a CD8+ T cell population in a sample (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • an immune cell exhaustion marker e.g., one, two or more immune checkpoint inhibitors (e.g., PD-1, TIM-3 and/or LAG-3) in a sample (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • an immune cell exhaustion marker e.g., one, two or more immune checkpoint inhibitors (e.g., PD-1, TIM-3 and/or LAG-3) in a sample (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • a cytokine level or activity e.g., quality of cytokine repertoire
  • a CAR-expressing cell product sample e.g., CAR19-expressing cell product sample (e.g., CTL019)
  • the cytokine is chosen from one, two, three, four, five or more (or all) of the cytokines listed in Table 16;
  • a quantity of CD27+PD-1 ⁇ cells in a subject e.g., a sample from the subject (e.g., an apheresis sample or a CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019)), e.g., a quantity greater than or equal to 1 ⁇ 10 7 cells.
  • a CAR-expressing cell therapy e.g., a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein;
  • a manufacturing process of a CAR-expressing cell therapy e.g., enrich for na ⁇ ve T cells prior to introducing a nucleic acid encoding a CAR, e.g., for a subject identified as a non-responder or a partial responder;
  • a selection of an alternative therapy e.g., for a non-responder or partial responder, e.g., a standard of care for a particular cancer type, or if the subject is, or is identified as, a non-responder or a relapser, recommend decreasing the T REG cell population and/or T REG gene signature, e.g., by CD25 depletion, administration of cyclophosphamide, anti-GITR antibody, mTOR inhibitor, or a combination thereof.
  • the value includes a measure of a CD19 CAR-expressing cell gene set signature and a combination of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, and a memory T cell (e.g., a CD8+ memory T cell, e.g., a na ⁇ ve T cell (T N)
  • T SCM memory stem cell
  • T CM central memory T cell
  • T EM effector memory T cell
  • the at least one processor when executing is configured to: acquire a value of responder status that comprises a measure of a CD19 CAR-expressing cell gene set signature and a combination of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1; and responsive to a determination of the value of responder status, perform one, two, three, four or more of: identify the subject as a complete responder, partial responder, or non-responder; recommend a CAR-expressing cell therapy; recommend a selection or alteration of a dosing of a CAR-expressing cell therapy; recommend an alternative therapy, e.g., a standard of care
  • the invention features a method of evaluating the potency of a CAR-expressing cell product, e.g., CAR19-expressing cell product sample (e.g., CTL019).
  • the method includes acquiring a value for one, two, three, four, five, six, seven, eight, or more (e.g., all) of:
  • CD27 and/or CD45RO ⁇ e.g., CD27+CD45RO ⁇
  • immune effector cells e.g., in a CD4+ or a CD8+ T cell population, in the CAR-expressing cell product
  • the invention features a method for optimizing manufacturing of a CAR-expressing cell product, e.g., CAR19-expressing cell product sample (e.g., CTL019).
  • the method includes:
  • CD27 and/or CD45RO ⁇ e.g., CD27+CD45RO ⁇
  • immune effector cells e.g., in a CD4+ or a CD8+ T cell population, in the CAR-expressing cell product
  • a quantity of CD27+PD-1 ⁇ cells in a subject e.g., a sample from the subject (e.g., an apheresis sample or a CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019)), e.g., a quantity greater than or equal to 1 ⁇ 10 7 cells; or
  • selecting the CAR-expressing cell product for administration to a subject optionally, selecting the CAR-expressing cell product for administration to a subject
  • any of the aforesaid manufacturing methods further comprise reducing the number (e.g., depleting) T REG cells, e.g., via CD25-depletion, GITR depletion, or mTOR inhibition.
  • the manufacturing methods further comprise contacting the sample, e.g., the apheresis sample, with an anti-GITR antibody.
  • any of the aforesaid manufacturing methods each of (i), (ii) or (iii), (iv), (v), (vi), (vii), (viii), (ix), or any combination thereof (e.g., all) are evaluated following activation in vitro.
  • the method further includes determining an expression level of one or more cytokines of Table 14, Table 15 or Table 16 (e.g., CCL20, IL-17a and/or IL-6) secreted by the CAR-expressing cell (e.g., T cell, NK cell) product.
  • secretion of one or more cytokines of Table 14, Table 15 or Table 16 (e.g., CCL20, IL-17a and/or IL-6) is in response to CAR-expressing cell product stimulation with one or more target tumor antigen(s).
  • the cytokine signature or level described herein predict subject response to a CAR-expressing cell product.
  • the cytokine signature or level described in Table 16 predict subject response to a CAR-expressing cell product.
  • the method further includes one or more of (e.g., one, two, three or all of): obtaining a blood sample, e.g., a population of T cells obtained from the blood of a subject; activating the population of T cells, e.g., by a method described herein; genetically engineering a cell from the population of T cells, e.g., transducing a cell from the population of T cells, with a vector comprising a nucleic acid encoding a CAR, e.g., a CAR described herein, e.g., a CD19 CAR described herein, e.g., CTL019; expanding a population of T cells that comprises a genetically engineered T cell, e.g., a cell transduced with a nucleic acid encoding a CAR, e.g., a CAR described herein, e.g., a CD19 CAR described herein, e.g., by a
  • the one or more gene signatures or expression profiles described herein are identified in a subject prior to CAR-expressing cell treatment (e.g., a CD19 CAR-expressing cell treatment, e.g., CTL019 therapy) that predict relapse to CAR-expressing cell treatment.
  • the one or more gene signatures or expression profiles described herein are identified in an apheresis sample.
  • the one or more gene signatures or expression profiles described herein are identified in a bone marrow sample.
  • the one or more gene signatures or expression profiles described herein are identified in a manufactured CAR-expressing cell product (e.g., a CD19 CAR-expressing cell product, e.g., CTL019) prior to infusion.
  • decreasing the T REG level or gene signature in a subject prior to apheresis or during manufacturing of a CAR-expressing cell product significantly reduces the risk of subject relapse.
  • a CAR-expressing cell manufacturing process is modified to deplete T REG cells prior to manufacturing of the CAR-expressing cell product (e.g., a CTL019 product).
  • CD25-depletion is used to deplete T REG cells prior to manufacturing of the CAR-expressing cell product (e.g., a CTL019 product).
  • the method further comprises:
  • the method further comprises expanding the population of cells after the nucleic acid molecule encoding a CAR has been introduced.
  • the population of cells is expanded in culture for 5 days, and the resulting cells exhibit higher proinflammatory IFN- ⁇ and/or GM-CSF levels, as compared to the same cells expanded in culture for 9 days under the same culture conditions.
  • the population of cells is expanded by culturing the cells in the presence of an agent that stimulates a CD3/TCR complex associated signal and/or a ligand that stimulates a costimulatory molecule on the surface of the cells.
  • the agent can be a bead conjugated with anti-CD3 antibody, or a fragment thereof, and/or anti-CD28 antibody, or a fragment thereof.
  • the method further includes cryopreserving the population of the cells after the appropriate expansion period.
  • the method of making disclosed herein further comprises contacting the population of immune effector cells with a nucleic acid encoding a telomerase subunit, e.g., hTERT.
  • the nucleic acid encoding the telomerase subunit can be DNA.
  • the method of making disclosed herein further comprises culturing the population of immune effector cells in serum comprising 2% hAB serum.
  • the CAR can comprise one of more of: a leader sequence, e.g., a leader sequence described herein, e.g., in Table 11; an anti-CD19 binding domain, e.g., an anti-CD19 binding domain described herein, e.g., in Table 12; a hinge region, e.g., a hinge region described herein, e.g., a hinge region described in Table 11; a transmembrane domain, e.g., a transmembrane domain described herein, e.g., in Table 11; and an intracellular signaling domain (e.g., a costimulatory domain and/or a primary signaling domain, e.g., a costimulatory domain described herein, e.g., in Table 11 and/or a primary signaling domain described herein, e.g., in Table 11).
  • the CD19 CAR-expressing cell e.g., T cell, NK cell
  • sequence accession numbers specified herein including in any Table herein, e.g., in any of Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 14, Table 17, Table 18, and Table 20, refer to the database entries current as of Oct. 8, 2014.
  • sequence accession numbers refer to the database entries current as of Oct. 8, 2014.
  • FIG. 1 depicts an exemplary model illustrating findings from a whole genome CTL019 RNAseq analysis performed on T cells at apheresis and after CTL019 manufacturing for 21 CLL and 7 ALL samples.
  • This model demonstrates that expression patterns of complete responders (CRs) have a younger T cell phenotype than non-responders (NRs).
  • Memory T cell subsets are differentially enriched between CRs versus NRs with CRs showing similarity to T memory stem cells.
  • FIG. 2B is an exemplary schematic illustrating an overview of the analysis. Briefly, for each gene set, a 3-group statistical model was applied to determine whether the meta-gene was statistically different between the CLL product CRs, PRs, and NRs. CRs are more like resting T EFF cells, whereas NR are more like activated T EFF cells. CTL019 NR samples are in a more activated state than CR samples.
  • FIG. 4 depicts an exemplary result illustrating meta-gene scores for the T SCM vs T CM upregulated gene set.
  • the y-axis is normalized meta-gene expression scores.
  • Gene sets enriched in CLL CRs e.g., CTL019 CRs
  • ALL and CLL CRs are enriched in T stem cell (T SCM ) subset specific genes
  • CLL PRs and NRs are enriched in T central memory (T CM ) subset genes.
  • T SCM T stem cell
  • T CM T central memory
  • FIG. 5A depicts an exemplary result from a Principle Component Analysis (PCA) of CTL019 samples.
  • PCA Principle Component Analysis
  • FIG. 5B depicts an exemplary result from a PCA of CTL019 and apheresis samples.
  • This exemplary PCA result illustrates that CRs, ALL and Normal samples cluster separately from PRs and NRs and from the apheresis cluster.
  • FIG. 6 depicts an exemplary schematic depicting immunophenotyping of apheresis and product samples.
  • FIG. 7A and FIG. 7B depict exemplary multi-color flow cytometry analysis results identifying correlates of response in product samples.
  • 36 manufactured CTL019 samples from CLL patients were analyzed. Samples included 5 CR, 8 PR, 19NR and 3 pending.
  • FIG. 7A depicts an exemplary result illustrating percent CD4+ cells and patient response.
  • FIG. 7B depicts an exemplary result illustrating percent CD8+ cells and patient response.
  • FIG. 10A and FIG. 10B depict an exemplary flow cytometry analysis of PD1, CAR 19, and TIM-3 expression on T cells from subjects that are complete responders (CR) or non-responders (NR) to CAR-expressing cell therapy.
  • FIG. 10C depicts exemplary results that show the distribution of PD1 and TIM-3 expression from groups of subjects with different responses to CAR-expressing cell therapy.
  • Non-responder (NR) products have higher percentages of CAR19+PD1+TIM3+ cells than CRs.
  • FIG. 11 depicts an exemplary result illustrating that CD27 levels in the CAR product correlate with patient response.
  • CRs CD8+ cells displayed a higher percentage of CD27+ cells as compared to PRs and NRs.
  • FIG. 13 depicts an exemplary multi-color flow cytometry analysis result illustrating a correlation between a younger T cell phenotype and response to CTL019 therapy.
  • FIG. 16 depicts an exemplary block diagram of a computer system on which various aspects and embodiments may be practiced.
  • FIG. 19A depicts an exemplary scatter plot showing log-normalized correlation of IL17A (y-axis) and CCL20 (x-axis) expression. Dashed lines represent the classification boundary for separating NRs from CRs/PRs. Each dot represents a CLL patient, and the cross-hatch (NR), black (PR) and white (CR) represent the clinical response.
  • the correlation coefficient is represented by “r” (e.g., a correlation coefficient of 0.928) and corresponding p-value for correlation using “p.value” (e.g., corresponding p-value of 1.36e-09).
  • r e.g., a correlation coefficient of 0.928
  • p.value e.g., corresponding p-value of 1.36e-09
  • FIG. 19B depicts an exemplary scatter plot showing correlation of CCL20 with percentage of CAR+ cells with a correlation coefficient of 0.395 and corresponding p-value of 0.0761.
  • Each dot represents a CLL patient, and the cross-hatch (NR), black (PR) and white (CR) represent the clinical response.
  • the correlation coefficient is represented by “r” and corresponding p-value for correlation using “p.value”.
  • FIG. 19C depicts an exemplary scatter plot showing correlation of IL17a with percentage of CAR+ cells with a correlation coefficient of 0.278 and corresponding p-value of 0.222.
  • Each dot represents a CLL patient, and the cross-hatch (NR), black (PR) and white (CR) represent the clinical response.
  • the correlation coefficient is represented by “r” and corresponding p-value for correlation using “p.value”.
  • the y-axis is normalized meta-gene expression scores.
  • FIG. 21 depicts an exemplary scatter plot showing percent of CAR+ cells (i.e., transduction rate) at pre-harvest for complete responders (CR) in white, partial responders (PR) in black and non-responders (NR) in hatching.
  • Transduction efficiencies were measured pre-harvest and correlated with subject response (e.g., CR, PR, or NR).
  • subject response e.g., CR, PR, or NR
  • the solid line represents a 15% transduction efficiency that separates the majority of non-responders from responders.
  • pre-harvest CAR transduction rate is a marker of response to CAR-expressing cell (e.g., T cell, NK cell) therapy in CLL.
  • FIG. 22 is a bar graph depicting the relationship between number of CD27+PD1 ⁇ CART cells infused and response to therapy.
  • FIG. 23 is a scatter plot depicting the relationship between number of CD27+PD1-CART cells infused and response to therapy.
  • an element means one element or more than one element.
  • Acquire or “acquiring” as the terms are used herein, refer to obtaining possession of a physical entity (e.g., a sample, a polypeptide, a nucleic acid, or a sequence), or a value, e.g., a numerical value, by “directly acquiring” or “indirectly acquiring” the physical entity or value.
  • Directly acquiring means performing a process (e.g., performing a synthetic or analytical method) to obtain the physical entity or value.
  • Indirectly acquiring refers to receiving the physical entity or value from another party or source (e.g., a third party laboratory that directly acquired the physical entity or value).
  • Directly acquiring a physical entity includes performing a process that includes a physical change in a physical substance, e.g., a starting material.
  • exemplary changes include making a physical entity from two or more starting materials, shearing or fragmenting a substance, separating or purifying a substance, combining two or more separate entities into a mixture, performing a chemical reaction that includes breaking or forming a covalent or non-covalent bond.
  • antibody refers to a protein, or polypeptide sequence derived from an immunoglobulin molecule which specifically binds with an antigen.
  • Antibodies can be polyclonal or monoclonal, multiple or single chain, or intact immunoglobulins, and may be derived from natural sources or from recombinant sources.
  • Antibodies can be tetramers of immunoglobulin molecules.
  • the alteration can be at least twice, at least twice three, at least twice four, at least twice five, or at least twice ten or more times greater than or less than the expression level of the biomarkers in a control sample (e.g., a sample from a healthy subject not having the associated disease), or the average expression level in several control samples.
  • a control sample e.g., a sample from a healthy subject not having the associated disease
  • An “altered level of expression” can be determined at the protein or nucleic acid (e.g., mRNA) level.
  • An antigen binding fragment can also be incorporated into single domain antibodies, maxibodies, minibodies, nanobodies, intrabodies, diabodies, triabodies, tetrabodies, v-NAR and bis-scFv (see, e.g., Hollinger and Hudson, Nature Biotechnology 23:1126-1136, 2005).
  • Antigen binding fragments can also be grafted into scaffolds based on polypeptides such as a fibronectin type III (Fn3)(see U.S. Pat. No. 6,703,199, which describes fibronectin polypeptide minibodies).
  • scFv refers to a fusion protein comprising at least one antibody fragment comprising a variable region of a light chain and at least one antibody fragment comprising a variable region of a heavy chain, wherein the light and heavy chain variable regions are contiguously linked via a short flexible polypeptide linker, and capable of being expressed as a single chain polypeptide, and wherein the scFv retains the specificity of the intact antibody from which it is derived.
  • an scFv may have the VL and VH variable regions in either order, e.g., with respect to the N-terminal and C-terminal ends of the polypeptide, the scFv may comprise VL-linker-VH or may comprise VH-linker-VL.
  • antibody heavy chain refers to the larger of the two types of polypeptide chains present in antibody molecules in their naturally occurring conformations, and which normally determines the class to which the antibody belongs.
  • antibody light chain refers to the smaller of the two types of polypeptide chains present in antibody molecules in their naturally occurring conformations. Kappa ( ⁇ ) and lambda ( ⁇ ) light chains refer to the two major antibody light chain isotypes.
  • the CDR amino acid residues in the heavy chain variable domain (VH) are numbered 31-35 (HCDR1), 50-65 (HCDR2), and 95-102 (HCDR3); and the CDR amino acid residues in the light chain variable domain (VL) are numbered 24-34 (LCDR1), 50-56 (LCDR2), and 89-97 (LCDR3).
  • the CDR amino acids in the VH are numbered 26-32 (HCDR1), 52-56 (HCDR2), and 95-102 (HCDR3); and the CDR amino acid residues in the VL are numbered 26-32 (LCDR1), 50-52 (LCDR2), and 91-96 (LCDR3).
  • the CDRs correspond to the amino acid residues that are part of a Kabat CDR, a Chothia CDR, or both.
  • the CDRs correspond to amino acid residues 26-35 (HCDR1), 50-65 (HCDR2), and 95-102 (HCDR3) in a VH, e.g., a mammalian VH, e.g., a human VH; and amino acid residues 24-34 (LCDR1), 50-56 (LCDR2), and 89-97 (LCDR3) in a VL, e.g., a mammalian VL, e.g., a human VL.
  • anti-cancer effect refers to a biological effect which can be manifested by various means, including but not limited to, e.g., a decrease in tumor volume, a decrease in the number of cancer cells, a decrease in the number of metastases, an increase in life expectancy, decrease in cancer cell proliferation, decrease in cancer cell survival, or amelioration of various physiological symptoms associated with the cancerous condition.
  • An “anti-cancer effect” can also be manifested by the ability of the peptides, polynucleotides, cells and antibodies in prevention of the occurrence of cancer in the first place.
  • anti-tumor effect refers to a biological effect which can be manifested by various means, including but not limited to, e.g., a decrease in tumor volume, a decrease in the number of tumor cells, a decrease in tumor cell proliferation, or a decrease in tumor cell survival.
  • allogeneic refers to any material derived from a different animal of the same species as the individual to whom the material is introduced. Two or more individuals are said to be allogeneic to one another when the genes at one or more loci are not identical. In some aspects, allogeneic material from individuals of the same species may be sufficiently unlike genetically to interact antigenically.
  • autologous refers to any material derived from the same individual to whom it is later to be re-introduced into the individual.
  • a marker of the invention which is predictive of responsiveness to therapeutics can have an altered expression level, protein level, or protein activity, in a biological sample obtained from a subject having, or suspected of having, cancer as compared to a biological sample obtained from a control subject.
  • cancer associated antigen or “tumor antigen” interchangeably refers to a molecule (typically protein, carbohydrate or lipid) that is preferentially expressed on the surface of a cancer cell, either entirely or as a fragment (e.g., MHC/peptide), in comparison to a normal cell, and which is useful for the preferential targeting of a pharmacological agent to the cancer cell.
  • a tumor antigen is a marker expressed by both normal cells and cancer cells, e.g., a lineage marker, e.g., CD19 on B cells.
  • CD19 includes proteins comprising mutations, e.g., point mutations, fragments, insertions, deletions and splice variants of full length wild-type CD19.
  • CD19 is expressed on most B lineage cancers, including, e.g., acute lymphoblastic leukemia, chronic lymphocyte leukemia and non-Hodgkin lymphoma. Other cells which express CD19 are provided below in the definition of “disease associated with expression of CD19.” It is also an early marker of B cell progenitors. See, e.g., Nicholson et al., M OL . I MMUN . 34 (16-17): 1157-1165 (1997).
  • the antigen-binding portion of the CAR-expressing cell recognizes and binds an antigen within the extracellular domain of the CD19 protein.
  • the CD19 protein is expressed on a cancer cell.
  • the CD19 has a wild-type sequence, e.g., a wild-type human sequence.
  • the CD19 has a mutant sequence, e.g., a mutant human sequence.
  • CAR Chimeric Antigen Receptor
  • a CAR refers to a set of polypeptides, typically two in the simplest embodiments, which when in an immune effector cell, provides the cell with specificity for a target cell, typically a cancer cell, and with intracellular signal generation.
  • a CAR comprises at least an extracellular antigen binding domain, a transmembrane domain and a cytoplasmic signaling domain (also referred to herein as “an intracellular signaling domain”) comprising a functional signaling domain derived from a stimulatory molecule and/or costimulatory molecule as defined below.
  • the set of polypeptides are in the same polypeptide chain (e.g., comprise a chimeric fusion protein). In some embodiments, the set of polypeptides are not contiguous with each other, e.g., are in different polypeptide chains. In some aspects, the set of polypeptides include a dimerization switch that, upon the presence of a dimerization molecule, can couple the polypeptides to one another, e.g., can couple an antigen binding domain to an intracellular signaling domain. In one aspect, the stimulatory molecule of the CAR is the zeta chain associated with the T cell receptor complex.
  • the cytoplasmic signaling domain comprises a primary signaling domain (e.g., a primary signaling domain of CD3-zeta). In one aspect, the cytoplasmic signaling domain further comprises one or more functional signaling domains derived from at least one costimulatory molecule as defined below. In one aspect, the costimulatory molecule is chosen from 4-1BB (i.e., CD137), CD27, ICOS, and/or CD28. In one aspect, the CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a stimulatory molecule.
  • 4-1BB i.e., CD137
  • CD27 CD27
  • ICOS ICOS
  • CD28 CD28
  • the CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from
  • the CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a co-stimulatory molecule and a functional signaling domain derived from a stimulatory molecule.
  • the CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising two functional signaling domains derived from one or more co-stimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule.
  • the CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising at least two functional signaling domains derived from one or more co-stimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule.
  • the CAR comprises an optional leader sequence at the amino-terminus (N-ter) of the CAR fusion protein.
  • the CAR further comprises a leader sequence at the N-terminus of the extracellular antigen recognition domain, wherein the leader sequence is optionally cleaved from the antigen recognition domain (e.g., a scFv) during cellular processing and localization of the CAR to the cellular membrane.
  • the CAR is CTL019.
  • the antigen binding domain of a CAR composition of the invention comprises an antibody fragment.
  • the CAR comprises an antibody fragment that comprises a scFv.
  • binding domain refers to a protein, e.g., an immunoglobulin chain or fragment thereof, comprising at least one immunoglobulin variable domain sequence.
  • binding domain or “antibody molecule” encompasses antibodies and antibody fragments.
  • an antibody molecule is a multispecific antibody molecule, e.g., it comprises a plurality of immunoglobulin variable domain sequences, wherein a first immunoglobulin variable domain sequence of the plurality has binding specificity for a first epitope and a second immunoglobulin variable domain sequence of the plurality has binding specificity for a second epitope.
  • a multispecific antibody molecule is a bispecific antibody molecule.
  • a bispecific antibody has specificity for no more than two antigens.
  • a bispecific antibody molecule is characterized by a first immunoglobulin variable domain sequence which has binding specificity for a first epitope and a second immunoglobulin variable domain sequence that has binding specificity for a second epitope.
  • disease associated with expression of CD19 includes, but is not limited to, a disease associated with expression of CD19 (e.g., wild type or mutant CD19) or condition associated with cells which express, or at any time expressed, CD19 including, e.g., proliferative diseases such as a cancer or malignancy or a precancerous condition such as a myelodysplasia, a myelodysplastic syndrome or a preleukemia; or a noncancer related indication associated with cells which express CD19.
  • proliferative diseases such as a cancer or malignancy or a precancerous condition such as a myelodysplasia, a myelodysplastic syndrome or a preleukemia
  • a noncancer related indication associated with cells which express CD19.
  • a disease associated with expression of CD19 may include a condition associated with cells which do not presently express CD19, e.g., because CD19 expression has been downregulated, e.g., due to treatment with a molecule targeting CD19, e.g., a CD19 CAR, but which at one time expressed CD19.
  • a cancer associated with expression of CD19 is a hematological cancer.
  • the hematological cancer is a leukemia or a lymphoma.
  • a cancer associated with expression of CD19 includes cancers and malignancies including, but not limited to, e.g., one or more acute leukemias including but not limited to, e.g., acute myeloid leukemia (AML), B-cell acute lymphocytic leukemia (“B-ALL”), T-cell acute lymphocytic leukemia (“T-ALL”), acute lymphocytic leukemia (ALL); one or more chronic leukemias including but not limited to, e.g., chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL).
  • one or more acute leukemias including but not limited to, e.g., acute myeloid leukemia (AML), B-cell acute lymphocytic leukemia (“B-ALL”), T-cell acute lymphocytic leukemia (“T-ALL”), acute lymphocytic leukemia (ALL); one or more chronic leukemias including but not limited to, e.g.
  • Additional cancers or hematologic conditions associated with expression of CD19 comprise, but are not limited to, e.g., B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, myeloproliferative neoplasm; a histiocytic disorder (e.g., a mast
  • Further diseases associated with expression of CD19 expression include, but not limited to, e.g., atypical and/or non-classical cancers, malignancies, precancerous conditions or proliferative diseases associated with expression of CD19.
  • Non-cancer related indications associated with expression of CD19 include, but are not limited to, e.g., autoimmune disease, (e.g., lupus), inflammatory disorders (allergy and asthma) and transplantation.
  • the tumor antigen-expressing cells express, or at any time expressed, mRNA encoding the tumor antigen.
  • the tumor antigen-expressing cells produce the tumor antigen protein (e.g., wild-type or mutant), and the tumor antigen protein may be present at normal levels or reduced levels.
  • the tumor antigen-expressing cells produced detectable levels of a tumor antigen protein at one point, and subsequently produced substantially no detectable tumor antigen protein.
  • the disease is a CD19-negative cancer, e.g., a CD19-negative relapsed cancer.
  • the tumor antigen (e.g., CD19)-expressing cell expresses, or at any time expressed, mRNA encoding the tumor antigen.
  • the tumor antigen (e.g., CD19)-expressing cell produces the tumor antigen protein (e.g., wild-type or mutant), and the tumor antigen protein may be present at normal levels or reduced levels.
  • the tumor antigen (e.g., CD19)-expressing cell produced detectable levels of a tumor antigen protein at one point, and subsequently produced substantially no detectable tumor antigen protein.
  • costimulatory molecule refers to the cognate binding partner on a T cell that specifically binds with a costimulatory ligand, thereby mediating a costimulatory response by the T cell, such as, but not limited to, proliferation.
  • Costimulatory molecules are cell surface molecules other than antigen receptors or their ligands that are required for an efficient immune response.
  • Costimulatory molecules include, but are not limited to MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signalling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM-1, ICOS (CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4,
  • a costimulatory intracellular signaling domain refers to an intracellular portion of a costimulatory molecule.
  • the intracellular signaling domain can comprise the entire intracellular portion, or the entire native intracellular signaling domain, of the molecule from which it is derived, or a functional fragment thereof.
  • a “value of responder or relapser status” includes a measure (e.g., level) predictive of responsiveness or relapse of a subject to a treatment (e.g., a treatment that comprises, or consists of, a CAR-expressing cell therapy as described herein).
  • the measure is qualitative or quantitative.
  • the value of responder or relapser status is complete responder, partial responder, non-responder, relapser or non-relapser.
  • the value of responder or relapser status is a probability of being a complete responder, a partial responder, a non-responder, a relapser or a non-relapser.
  • the value of responder or relapser status can be determined based on the measure of any of (i)-(viii) as described herein.
  • a subject responds to treatment if a parameter of a cancer (e.g., a hematological cancer, e.g., cancer cell growth, proliferation and/or survival) in the subject is retarded or reduced by a detectable amount, e.g., about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more as determined by any appropriate measure, e.g., by mass, cell count or volume.
  • a subject responds to treatment if the subject experiences a life expectancy extended by about 5%, 10%, 20%, 30%, 40%, 50% or more beyond the life expectancy predicted if no treatment is administered.
  • a subject responds to treatment, if the subject has an increased disease-free survival, overall survival or increased time to progression.
  • a complete response or complete responder may involve one or more of: ⁇ 5% BM blast, >1000 neutrophil/ANC (/ ⁇ L). >100,000 platelets (/ ⁇ L) with no circulating blasts or extramedullary disease (No lymphadenopathy, splenomegaly, skin/gum infiltration/testicular mass/CNS involvement), Trilineage hematopoiesis, and no recurrence for 4 weeks.
  • a partial responder may involve one or more of ⁇ 50% reduction in BM blast, >1000 neutrophil/ANC (/ ⁇ L). >100,000 platelets (/ ⁇ L).
  • a non-responder can show disease progression, e.g., >25% in BM blasts.
  • a “complete responder” as used herein refers to a subject having a disease, e.g., a cancer, who exhibits a complete response, e.g., a complete remission, to a treatment.
  • a complete response may be identified, e.g., using the NCCN Guidelines®, or Cheson et al, J Clin Oncol 17:1244 (1999) and Cheson et al., “Revised Response Criteria for Malignant Lymphoma”, J Clin Oncol 25:579-586 (2007) (both of which are incorporated by reference herein in their entireties), as described herein.
  • a “partial responder” as used herein refers to a subject having a disease, e.g., a cancer, who exhibits a partial response, e.g., a partial remission, to a treatment.
  • a partial response may be identified, e.g., using the NCCN Guidelines®, or Cheson criteria as described herein.
  • non-responder refers to a subject having a disease, e.g., a cancer, who does not exhibit a response to a treatment, e.g., the patient has stable disease or progressive disease.
  • a non-responder may be identified, e.g., using the NCCN Guidelines®, or Cheson criteria as described herein.
  • relapse refers to reappearance of a disease (e.g., cancer) after an initial period of responsiveness, e.g., after prior treatment with a therapy, e.g., cancer therapy (e.g., complete response or partial response).
  • the initial period of responsiveness may involve the level of cancer cells falling below a certain threshold, e.g., below 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%.
  • the reappearance may involve the level of cancer cells rising above a certain threshold, e.g., above 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%.
  • the reappearance may involve, e.g., a reappearance of blasts in the blood, bone marrow (>5%), or any extramedullary site, after a complete response.
  • a complete response in this context, may involve ⁇ 5% BM blast.
  • a response e.g., complete response or partial response
  • the initial period of responsiveness lasts at least 1, 2, 3, 4, 5, or 6 days; at least 1, 2, 3, or 4 weeks; at least 1, 2, 3, 4, 6, 8, 10, or 12 months; or at least 1, 2, 3, 4, or 5 years.
  • a therapy that includes a CD19 inhibitor may relapse or be refractory to treatment.
  • the relapse or resistance can be caused by CD19 loss (e.g., an antigen loss mutation) or other CD19 alteration that reduces the level of CD19 (e.g., caused by clonal selection of CD19-negative clones).
  • CD19 loss e.g., an antigen loss mutation
  • CD19 alteration that reduces the level of CD19 (e.g., caused by clonal selection of CD19-negative clones).
  • a cancer that harbors such CD19 loss or alteration is referred to herein as a “CD19-negative cancer” or a “CD19-negative relapsed cancer”).
  • a CD19-negative cancer need not have 100% loss of CD19, but a sufficient reduction to reduce the effectiveness of a CD19 therapy such that the cancer relapses or becomes refractory.
  • a CD19-negative cancer results from a CD19 CAR therapy.
  • a CD19-negative multiple myeloma can be treated with a CD19 CAR-expressing therapy, e.g., as described in PCT/US2015/024671, filed Apr. 7, 2015 (e.g., paragraphs 9 and 90, and Example 6 therein), which is incorporated by reference in its entirety.
  • a CD19-negative cancer can be treated with a CAR-expressing therapy, e.g., a CD123 CAR-expressing therapy, e.g., as described in PCT/US2015/045898 filed Aug. 19, 2015 (e.g., p. 26, p. 30, and Example 7 therein) which is incorporated by reference in its entirety.
  • a CAR-expressing therapy e.g., a CD123 CAR-expressing therapy, e.g., as described in PCT/US2015/045898 filed Aug. 19, 2015 (e.g., p. 26, p. 30, and Example 7 therein) which is incorporated by reference in its entirety.
  • endogenous refers to any material from or produced inside an organism, cell, tissue or system.
  • an effective amount or “therapeutically effective amount” are used interchangeably herein, and refer to an amount of a compound, formulation, material, or composition, as described herein effective to achieve a particular biological result.
  • exogenous refers to any material introduced from or produced outside an organism, cell, tissue or system.
  • expression refers to the transcription and/or translation of a particular nucleotide sequence driven by a promoter.
  • the term “flexible polypeptide linker” or “linker” as used in the context of a scFv refers to a peptide linker that consists of amino acids such as glycine and/or serine residues used alone or in combination, to link variable heavy and variable light chain regions together.
  • the flexible polypeptide linkers include, but are not limited to, (Gly 4 Ser) 4 (SEQ ID NO:29) or (Gly 4 Ser) 3 (SEQ ID NO:30).
  • the linkers include multiple repeats of (Gly 2 Ser), (GlySer) or (Gly 3 Ser) (SEQ ID NO:31). Also included within the scope of the invention are linkers described in WO2012/138475, incorporated herein by reference.
  • sequence similarity refers to sequence similarity between two polynucleotide sequences or between two polypeptide sequences, with identity being a more strict comparison.
  • percent identity or homology and “% identity or homology” refer to the percentage of sequence similarity found in a comparison of two or more polynucleotide sequences or two or more polypeptide sequences.
  • sequence similarity refers to the percent similarity in base pair sequence (as determined by any suitable method) between two or more polynucleotide sequences. Two or more sequences can be anywhere from 0-100% similar, or any integer value there between.
  • Identity or similarity can be determined by comparing a position in each sequence that can be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same nucleotide base or amino acid, then the molecules are identical at that position.
  • a degree of similarity or identity between polynucleotide sequences is a function of the number of identical or matching nucleotides at positions shared by the polynucleotide sequences.
  • a degree of identity of polypeptide sequences is a function of the number of identical amino acids at positions shared by the polypeptide sequences.
  • a degree of homology or similarity of polypeptide sequences is a function of the number of amino acids at positions shared by the polypeptide sequences.
  • the term “substantial homology,” as used herein, refers to homology of at least 50%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% or more.
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab, F(ab)2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies and antibody fragments thereof are human immunoglobulins (recipient antibody or antibody fragment) in which residues from a complementary-determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity.
  • Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • a humanized antibody/antibody fragment can comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications can further refine and optimize antibody or antibody fragment performance.
  • the humanized antibody or antibody fragment thereof will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or a significant portion of the FR regions are those of a human immunoglobulin sequence.
  • the humanized antibody or antibody fragment can also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Immuno effector cell refers to a cell that is involved in an immune response, e.g., in the promotion of an immune effector response.
  • immune effector cells include T cells, e.g., alpha/beta T cells and gamma/delta T cells, B cells, natural killer (NK) cells, natural killer T (NK-T) cells, mast cells, and myeloid-derived phagocytes.
  • Immuno effector function or immune effector response refers to function or response, e.g., of an immune effector cell, that enhances or promotes an immune attack of a target cell.
  • an immune effector function or response refers a property of a T or NK cell that promotes killing or the inhibition of growth or proliferation, of a target cell.
  • primary stimulation and co-stimulation are examples of immune effector function or response.
  • effector function refers to a specialized function of a cell. Effector function of a T cell, for example, may be cytolytic activity or helper activity including the secretion of cytokines.
  • 4-1BB refers to a member of the TNFR superfamily with an amino acid sequence provided as GenBank Acc. No. AAA62478.2, or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like; and a “4-1BB costimulatory domain” is defined as amino acid residues 214-255 of GenBank Acc No. AAA62478.2, or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like.
  • the “4-1BB costimulatory domain” is the sequence provided as SEQ ID NO:14 or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like.
  • intracellular signaling domain refers to an intracellular portion of a molecule.
  • the intracellular signaling domain can generate a signal that promotes an immune effector function of the CAR containing cell, e.g., a CAR-expressing cell, e.g., a T cell or an NK cell.
  • immune effector function e.g., in a CAR-expressing cell include, cytolytic activity and helper activity, including the secretion of cytokines.
  • the intracellular signal domain transduces the effector function signal and directs the cell to perform a specialized function. While the entire intracellular signaling domain can be employed, in many cases it is not necessary to use the entire chain.
  • intracellular signaling domain is thus meant to include any truncated portion of the intracellular signaling domain sufficient to transduce the effector function signal.
  • the intracellular signaling domain can comprise a primary intracellular signaling domain.
  • Exemplary primary intracellular signaling domains include those derived from the molecules responsible for primary stimulation, or antigen dependent simulation.
  • the intracellular signaling domain can comprise a costimulatory intracellular domain.
  • Exemplary costimulatory intracellular signaling domains include those derived from molecules responsible for costimulatory signals, or antigen independent stimulation.
  • a primary intracellular signaling domain can comprise a cytoplasmic sequence of a T cell receptor
  • a costimulatory intracellular signaling domain can comprise cytoplasmic sequence from co-receptor or costimulatory molecule.
  • a primary intracellular signaling domain can comprise a signaling motif which is known as an immunoreceptor tyrosine-based activation motif or ITAM.
  • ITAM containing primary cytoplasmic signaling sequences include, but are not limited to, those derived from CD3 zeta, FcR gamma, FcR beta, CD3 gamma, CD3 delta, CD3 epsilon, CD5, CD22, CD79a, CD79b, CD278 (“ICOS”), FccRI, CD66d, CD32, DAP10, and DAP12.
  • in vitro transcribed RNA refers to RNA, preferably mRNA, that has been synthesized in vitro.
  • the in vitro transcribed RNA is generated from an in vitro transcription vector.
  • the in vitro transcription vector comprises a template that is used to generate the in vitro transcribed RNA.
  • isolated means altered or removed from the natural state.
  • a nucleic acid or a peptide naturally present in a living animal is not “isolated,” but the same nucleic acid or peptide partially or completely separated from the coexisting materials of its natural state is “isolated.”
  • An isolated nucleic acid or protein can exist in substantially purified form, or can exist in a non-native environment such as, for example, a host cell.
  • lentivirus refers to a genus of the Retroviridae family. Lentiviruses are unique among the retroviruses in being able to infect non-dividing cells; they can deliver a significant amount of genetic information into the DNA of the host cell, so they are one of the most efficient methods of a gene delivery vector. HIV, SIV, and FIV are all examples of lentiviruses.
  • lentiviral vector refers to a vector derived from at least a portion of a lentivirus genome, including especially a self-inactivating lentiviral vector as provided in Milone et al., M OL . T HER . 17(8): 1453-1464 (2009).
  • Other examples of lentivirus vectors that may be used in the clinic include but are not limited to, e.g., the LENTIVECTOR® gene delivery technology from Oxford BioMedica, the LENTIMAXTM vector system from Lentigen and the like. Nonclinical types of lentiviral vectors are also available and would be known to one skilled in the art.
  • low, immune enhancing, dose when used in conjunction with an mTOR inhibitor, e.g., an allosteric mTOR inhibitor, e.g., RAD001 or rapamycin, or a catalytic mTOR inhibitor, refers to a dose of mTOR inhibitor that partially, but not fully, inhibits mTOR activity, e.g., as measured by the inhibition of P70 S6 kinase activity. Methods for evaluating mTOR activity, e.g., by inhibition of P70 S6 kinase, are discussed herein. The dose is insufficient to result in complete immune suppression but is sufficient to enhance the immune response.
  • an mTOR inhibitor e.g., an allosteric mTOR inhibitor, e.g., RAD001 or rapamycin, or a catalytic mTOR inhibitor
  • na ⁇ ve T cell refers to immune cells that comprise antigen-inexperienced cells, e.g., immune cells that are precursors of memory cells.
  • na ⁇ ve T cells may be differentiated, but have not yet encountered their cognate antigen, and therefore are activated T cells or memory T cells.
  • na ⁇ ve T cells may be characterized by expression of CD62L, CD27, CCR7, CD45RA, CD28, and CD127, and the absence of CD95, or CD45RO isoform.
  • a na ⁇ ve T cells is a type of younger T cell as described herein.
  • less exhausted or “less exhausted phenotype” refers to immune effector cells that have reduced (e.g., lack) expression of immune cell exhaustion markers, e.g. PD1, TIM3, and LAG3.
  • a less exhausted cell may be a younger T cell as described herein.
  • nucleic acid refers to deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), or a combination of a DNA or RNA thereof, and polymers thereof in either single- or double-stranded form.
  • nucleic acid includes a gene, CDNA or an mRNA.
  • the nucleic acid molecule is synthetic (e.g., chemically synthesized) or recombinant. Unless specifically limited, the term encompasses nucleic acids containing analogues or derivatives of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides.
  • nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated.
  • degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., N UCLEIC A CID R ES . 19:5081 (1991); Ohtsuka et al., J. B IOL . C HEM .
  • A refers to adenosine
  • C refers to cytosine
  • G refers to guanosine
  • T refers to thymidine
  • U refers to uridine.
  • a “marker protein” is a protein encoded by or corresponding to a marker of the invention.
  • a marker protein comprises the entire or a partial sequence of a protein encoded by any of the sequences set forth herein, or a fragment thereof.
  • the terms “protein” and “polypeptide” are used interchangeably herein.
  • an “overexpression” or “significantly higher level of expression” of the gene products refers to an expression level or copy number in a test sample that is greater than the standard error of the assay employed to assess the level of expression.
  • the overexpression can be at least two, at least three, at least four, at least five, or at least ten or more times the expression level of the gene in a control sample or the average expression level of gene products in several control samples.
  • peptide refers to a compound comprised of amino acid residues covalently linked by peptide bonds.
  • a protein or peptide must contain at least two amino acids, and no limitation is placed on the maximum number of amino acids that can comprise a protein's or peptide's sequence.
  • Polypeptides include any peptide or protein comprising two or more amino acids joined to each other by peptide bonds.
  • the term refers to both short chains, which also commonly are referred to in the art as peptides, oligopeptides and oligomers, for example, and to longer chains, which generally are referred to in the art as proteins, of which there are many types.
  • Polypeptides include, for example, biologically active fragments, substantially homologous polypeptides, oligopeptides, homodimers, heterodimers, variants of polypeptides, modified polypeptides, derivatives, analogs, fusion proteins, among others.
  • a polypeptide includes a natural peptide, a recombinant peptide, or a combination thereof.
  • a “poly(A)” is a series of adenosines attached by polyadenylation to the mRNA.
  • the polyA is between 50 and 5000 (SEQ ID NO: 34) (e.g., 2000; SEQ ID NO: 32), e.g., 64 (SEQ ID NO: 37), e.g., greater than 100 (e.g., 150, SEQ ID NO: 33), e.g., greater than 400 (SEQ ID NO: 38).
  • poly(A) sequences can be modified chemically or enzymatically to modulate mRNA functionality such as localization, stability or efficiency of translation.
  • promoter refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a polynucleotide sequence.
  • promoter/regulatory sequence refers to a nucleic acid sequence which is required for expression of a gene product operably linked to the promoter/regulatory sequence. In some instances, this sequence may be the core promoter sequence and in other instances, this sequence may also include an enhancer sequence and other regulatory elements which are required for expression of the gene product.
  • the promoter/regulatory sequence may, for example, be one which expresses the gene product in a tissue specific manner.
  • prophylaxis means the prevention of or protective treatment for a disease or disease state.
  • ranges throughout this disclosure, various aspects of the invention can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6.
  • a range such as 95-99% identity includes something with 95%, 96%, 97%, 98% or 99% identity, and includes subranges such as 96-99%, 96-98%, 96-97%, 97-99%, 97-98% and 98-99% identity. This applies regardless of the breadth of the range.
  • recombinant antibody refers to an antibody which is generated using recombinant DNA technology, such as, for example, an antibody expressed by a bacteriophage or yeast expression system.
  • the term should also be construed to mean an antibody which has been generated by the synthesis of a DNA molecule encoding the antibody and which DNA molecule expresses an antibody protein, or an amino acid sequence specifying the antibody, wherein the DNA or amino acid sequence has been obtained using recombinant DNA or amino acid sequence technology which is available and well known in the art.
  • Refractory refers to a disease, e.g., cancer, that does not respond to a treatment.
  • a refractory cancer can be resistant to a treatment before or at the beginning of the treatment.
  • the refractory cancer can become resistant during a treatment.
  • a refractory cancer is also called a resistant cancer.
  • a reference or control level or activity is the level and/or activity in a subject, e.g., a sample obtained from one or more of: a baseline or prior value for the subject (e.g., prior to treatment with a CAR-expressing cell); the subject at a different time interval; an average or median value for a cancer patient population; a healthy control; or a healthy subject population (e.g., a control).
  • tissue sample each refers to a biological sample obtained from a tissue or bodily fluid of a subject or patient.
  • the source of the tissue sample can be solid tissue as from a fresh, frozen and/or preserved organ, tissue sample, biopsy, or aspirate; blood or any blood constituents (e.g., serum, plasma); bodily fluids such as urine, cerebral spinal fluid, whole blood, plasma and serum.
  • the sample can include a non-cellular fraction (e.g., urine, plasma, serum, or other non-cellular body fluid).
  • the sample is a urine sample.
  • the body fluid from which the sample is obtained from an individual comprises blood (e.g., whole blood).
  • the sample is a whole blood sample obtained from the subject.
  • the blood can be further processed to obtain plasma or serum.
  • the sample is an apheresis sample obtained from the blood of the subject.
  • the sample is a manufactured product sample, e.g., genetically engineered T cells obtained from the blood of the subject, e.g., a manufactured CAR-expressing cell (e.g., T cell, NK cell) product, e.g., a manufactured CD19 CAR-expressing cell product.
  • the sample contains a tissue, cells (e.g., peripheral blood mononuclear cells (PBMC)).
  • PBMC peripheral blood mononuclear cells
  • the sample can be a fine needle biopsy sample, an archival sample (e.g., an archived sample with a known diagnosis and/or treatment history), a histological section (e.g., a frozen or formalin-fixed section, e.g., after long term storage), among others.
  • the term sample includes any material obtained and/or derived from a biological sample, including a polypeptide, and nucleic acid (e.g., genomic DNA, CDNA, RNA) purified or processed from the sample. Purification and/or processing of the sample can involve one or more of extraction, concentration, antibody isolation, sorting, concentration, fixation, addition of reagents and the like.
  • the sample can contain compounds that are not naturally intermixed with the tissue in nature such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics or the like.
  • a manufactured composition comprising a genetically engineered cell (e.g., an immune effector cell), e.g., a population of cells in which a plurality of cells are engineered to express a CAR, e.g., a CAR described herein.
  • a manufactured product can be any genetically engineered immune effector cell (e.g., T cell, NK cell), e.g., genetically engineered immune effector cells obtained from the blood of the subject, e.g., a manufactured CAR-expressing cell product, e.g., a manufactured CD19 CAR-expressing cell product.
  • a cell (e.g., an immune effector cell) engineered to express a CAR may be obtained from an activated cryopreserved expanded cell population (e.g., an expanded immune effector cell population).
  • signaling domain refers to the functional portion of a protein which acts by transmitting information within the cell to regulate cellular activity via defined signaling pathways by generating second messengers or functioning as effectors by responding to such messengers.
  • the amount of a biomarker e.g., expression of gene products (e.g., one or more the biomarkers described herein), in a subject is “significantly” higher or lower than the normal amount of a marker, if the amount of the marker is greater or less, respectively, than the normal level by an amount greater than the standard error of the assay employed to assess amount, or at least two, three, four, five, ten or more times that amount.
  • the amount of the marker in the subject can be considered “significantly” higher or lower than the normal amount if the amount is at least about 1.5, two, at least about three, at least about four, or at least about five times, higher or lower, respectively, than the normal amount of the marker.
  • the term “specifically binds,” refers to an antibody, or a ligand, which recognizes and binds with a cognate binding partner protein present in a sample, but which antibody or ligand does not substantially recognize or bind other molecules in the sample.
  • stimulation refers to a primary response induced by binding of a stimulatory molecule (e.g., a TCR/CD3 complex) with its cognate ligand thereby mediating a signal transduction event, such as, but not limited to, signal transduction via the TCR/CD3 complex.
  • a stimulatory molecule e.g., a TCR/CD3 complex
  • Stimulation can mediate altered expression of certain molecules, such as down regulation of TGF- ⁇ , and/or reorganization of cytoskeletal structures, and the like.
  • the primary signal is initiated by, for instance, binding of a TCR/CD3 complex with an MHC molecule loaded with peptide, and which leads to mediation of a T cell response, including, but not limited to, proliferation, activation, differentiation, and the like.
  • a primary cytoplasmic signaling sequence (also referred to as a “primary signaling domain”) that acts in a stimulatory manner may contain a signaling motif which is known as immunoreceptor tyrosine-based activation motif or ITAM.
  • an ITAM containing cytoplasmic signaling sequence that is of particular use in the invention includes, but is not limited to, those derived from CD3 zeta, common FcR gamma (FCER1G), Fc gamma RIIa, FcR beta (Fc Epsilon Rib), CD3 gamma, CD3 delta, CD3 epsilon, CD79a, CD79b, DAP10, and DAP12.
  • the intracellular signaling domain in any one or more CARS of the invention comprises an intracellular signaling sequence, e.g., a primary signaling sequence of CD3-zeta.
  • the primary signaling sequence of CD3-zeta is the sequence provided as SEQ ID NO:18 (mutant CD3 zeta), or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like.
  • the primary signaling sequence of CD3-zeta is the sequence as provided in SEQ ID NO:20 (wild-type human CD3 zeta), or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like.
  • subject is intended to include living organisms in which an immune response can be elicited (e.g., mammals, human).
  • a subject is a mammal.
  • a subject is a human.
  • a subject is a patient.
  • terapéutica as used herein means a treatment.
  • a therapeutic effect is obtained by reduction, suppression, remission, or eradication of a disease state.
  • transfected or “transformed” or “transduced” refers to a process by which exogenous nucleic acid is transferred or introduced into the host cell.
  • a “transfected” or “transformed” or “transduced” cell is one which has been transfected, transformed or transduced with exogenous nucleic acid.
  • the cell includes the primary subject cell and its progeny.
  • transient refers to expression of a non-integrated transgene for a period of hours, days or weeks, wherein the period of time of expression is less than the period of time for expression of the gene if integrated into the genome or contained within a stable plasmid replicon in the host cell.
  • transmembrane domain refers to a polypeptide that spans the plasma membrane. In an embodiment, it links an extracellular sequence, e.g., a switch domain, an extracellular recognition element, e.g., an antigen binding domain, an inhibitory counter ligand binding domain, or costimulatory ECD domain, to an intracellular sequence, e.g., to a switch domain or an intracellular signaling domain.
  • extracellular sequence e.g., a switch domain
  • an extracellular recognition element e.g., an antigen binding domain, an inhibitory counter ligand binding domain, or costimulatory ECD domain
  • a transmembrane domain can include one or more additional amino acids adjacent to the transmembrane region, e.g., one or more amino acid associated with the extracellular region of the protein from which the transmembrane was derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the extracellular region) and/or one or more additional amino acids associated with the intracellular region of the protein from which the transmembrane protein is derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the intracellular region). Examples of transmembrane domains are disclosed herein.
  • treat refers to the reduction or amelioration of the progression, severity and/or duration of a proliferative disorder, or the amelioration of one or more symptoms (e.g., one or more discernible symptoms) of a proliferative disorder resulting from the administration of one or more therapies (e.g., one or more therapeutic agents such as a CAR of the invention).
  • therapies e.g., one or more therapeutic agents such as a CAR of the invention.
  • the terms “treat”, “treatment” and “treating” refer to the amelioration of at least one measurable physical parameter of a proliferative disorder, such as growth of a tumor, not necessarily discernible by the patient.
  • the terms “treat”, “treatment” and “treating” refer to the inhibition of the progression of a proliferative disorder, either physically by, e.g., stabilization of a discernible symptom, physiologically by, e.g., stabilization of a physical parameter, or both.
  • the terms “treat”, “treatment” and “treating” refer to the reduction or stabilization of tumor size or cancerous cell count.
  • an “underexpression” or “significantly lower level of expression” of products refers to an expression level in a test sample that is greater than the standard error of the assay employed to assess expression, for example, at least 1.5, twice, at least three, at least four, at least five, or at least ten or more times less than the expression level of the gene in a control sample, or the average expression level of gene products in several control samples.
  • xenogeneic refers to a graft derived from an animal of a different species.
  • the term “young T cell” or “younger T cell”, refers to an immune effector cell that comprises a less differentiated phenotype, e.g., a younger cell, e.g., a young T cell.
  • a younger T cell may be a na ⁇ ve T cell (T N ).
  • a young T cell may be characterized by expression of CD62L, and the absence of CD25, CD44, or CD45RO isoform.
  • a younger T cell may be a memory stem cell (T SCM ).
  • a younger T cells may be a central memory T cell (T CM ).
  • T N Phenotypic markers associated with T N , T SCM and T CM are disclosed in, e.g., Maus, M. et al. (2014) Annu. Rev. Immunol. 32:189-225 (see for example, FIG. 3 ), incorporated by reference herein.
  • Exemplary phenotypes of T N include one or more (or all) of the following: CD45RA+, CD45RO ⁇ , CD62L high , CCR7 high , CD95 ⁇ , CD122 ⁇ , CD27 high , CD28+, CD57 ⁇ , KLRG-1 ⁇ , or long telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid T N markers).
  • Exemplary phenotypes of T SCM include one or more (or all) of the following: CD45RA+, CD45RO ⁇ , CD62L high , CCR7 high , CD95+, CD122+, CD27 high , CD28 high , CD57 ⁇ , KLRG-1 ⁇ , or long telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid T SCM markers).
  • Exemplary phenotypes of T CM include one or more (or all) of the following: CD45RA ⁇ , CD45RO high , CD62L high , CCR7+, CD95+, CD122 high , CD27+, CD28 high , CD57 ⁇ , KLRG-1 ⁇ /+, or long/intermediate telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid T CM markers).
  • Exemplary phenotypes of T EM include one or more (or all) of the following: CD45RA ⁇ /+, CD45RO high , CD62L ⁇ , CCR7 ⁇ , CD95 ⁇ , CD122 high , CD27 ⁇ /+, CD28 ⁇ /+, CD57 low , KLRG-1+, or intermediate telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid T EM markers).
  • Exemplary phenotypes of T EFF include one or more (or all) of the following: CD45RA ⁇ /+, CD45RO+, CD62L ⁇ , CCR7 ⁇ , CD95 high , CD122 ⁇ /+, CD27 ⁇ , CD28 ⁇ , CD57+, KLRG-1 high , or short/intermediate telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid T EFF markers).
  • Exemplary phenotypes of an exhausted T cell phenotype include one or more (or all) of the following: CD45RA ⁇ /+, CD45RO+, CD62L ⁇ , CCR7-, CD95 high , CD122 low , CD27 ⁇ , CD28 ⁇ , CD57 high , KLRG-1 high , or short telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid markers).
  • zeta or alternatively “zeta chain”, “CD3-zeta” or “TCR-zeta” is defined as the protein provided as GenBank Acc. No. BAG36664.1, or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like, and a “zeta stimulatory domain” or alternatively a “CD3-zeta stimulatory domain” or a “TCR-zeta stimulatory domain” is defined as the amino acid residues from the cytoplasmic domain of the zeta chain that are sufficient to functionally transmit an initial signal necessary for T cell activation.
  • the cytoplasmic domain of zeta comprises residues 52 through 164 of GenBank Acc. No. BAG36664.1 or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like, that are functional orthologs thereof.
  • the “zeta stimulatory domain” or a “CD3-zeta stimulatory domain” is the sequence provided as SEQ ID NO:18.
  • the “zeta stimulatory domain” or a “CD3-zeta stimulatory domain” is the sequence provided as SEQ ID NO:20.
  • Methods are provided for the diagnosis and monitoring of treatment of cancer (e.g., a hematological cancer such as ALL and CLL) based on detection of certain biomarkers in samples from patients who have, or are suspected of having, cancer. Further, expression of one or more such biomarkers can be used to distinguish subjects that respond favorably to a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., “complete responders” or “CR”) from subjects that don't respond to a CAR-expressing cell therapy (e.g., “non-responders” or “NR”) and from subjects that have a partial response to a CAR-expressing cell therapy (e.g., “partial responders” or “PR”).
  • a CAR-expressing cell e.g., T cell, NK cell
  • CR complete responders” or “CR”
  • NR non-responders
  • PR partial responders
  • 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more genes in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1 and/or a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene signature can be used with methods of the present disclosure to acquire a disease progression value.
  • the subject treated, or the subject from which the value is obtained is a subject having, or at risk of having, cancer at any stage of treatment.
  • exemplary cancers include, but are not limited to, B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myel
  • the subject has received a pretreatment of an additional therapy, e.g., a subject that has been identified as a partial responder or non-responder and subsequently has been pretreated with an additional therapy.
  • the subject receives pretreatment with an mTOR inhibitor.
  • the mTOR inhibitor is administered at a dose or dosing schedule described herein.
  • a low, immune enhancing dose of an mTOR inhibitor is given to the subject prior to treatment with a CAR-expressing cell (e.g., a T cell, an NK cell).
  • administering is initiated prior to administration of a CAR expressing cell described herein, e.g., T cells.
  • an mTOR inhibitor e.g., an allosteric inhibitor, e.g., RAD001, or a catalytic inhibitor
  • the CAR cells are administered after a sufficient time, or sufficient dosing, of an mTOR inhibitor, such that the level of PD1 negative immune effector cells, e.g., T cells, or the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells, has been, at least transiently, increased.
  • the subject has received a pretreatment with a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein.
  • inhibitory molecules e.g., checkpoint molecules include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR (e.g., TGFRbeta).
  • an inhibitory nucleic acid e.g., an inhibitory nucleic acid, e.g., a dsRNA, e.g., an siRNA or shRNA; or e.g., an inhibitory protein or system, e.g., a clustered regularly interspaced short palindromic repeats (CRISPR), a transcription-activator like effector nuclease (TALEN), or a zinc finger endonuclease (ZFN), e.g., as described herein, can be used to inhibit expression of a molecule that modulates or regulates, e.g., inhibits, T-cell function in the CAR-expressing cell.
  • CRISPR clustered regularly interspaced short palindromic repeats
  • TALEN transcription-activator like effector nuclease
  • ZFN zinc finger endonuclease
  • the agent is an shRNA, e.g., an shRNA described herein.
  • the inhibitor of checkpoint molecule can be, e.g., an antibody or antibody fragment that binds to a checkpoint molecule.
  • the agent can be an antibody or antibody fragment that binds to PD1, PD-L1, PD-L2 (e.g., as described herein) or CTLA4 (e.g., ipilimumab (also referred to as MDX-010 and MDX-101, and marketed as Yervoy®; Bristol-Myers Squibb; Tremelimumab (IgG2 monoclonal antibody available from Pfizer, formerly known as ticilimumab, CP-675,206)).
  • CTLA4 e.g., ipilimumab (also referred to as MDX-010 and MDX-101, and marketed as Yervoy®; Bristol-Myers Squibb; Tremelimumab (IgG
  • the subject receives an additional therapy in combination with CAR-expressing cell (e.g., a T cell, an NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019).
  • CAR-expressing cell e.g., a T cell, an NK cell
  • the subject receives an mTOR inhibitor, e.g., an mTOR inhibitor described herein, in combination with CAR-expressing cell therapy.
  • the mTOR inhibitor is administered at a dose and/or dosing schedule described herein.
  • the subject receives a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein, in combination with CAR-expressing cell therapy.
  • the subject has been identified as a non-responder and the subject receives a therapy other than a CAR-expressing cell therapy, e.g., a standard of care therapy for the particular cancer type.
  • the subject receives one or more of an anti-CD20 antibody, or functional fragment thereof (e.g., ofatumumab, rituximab, obinutuzumab), an anti-CD52 antibody or functional fragment thereof (e.g., alemtuzumab), an alkylating agent (e.g., a nitrogen mustard alkylating agent such as, e.g., bendamustine HCl, chlorambucel, cyclophosphamide), a kinase inhibitor (e.g., a kinase inhibitor described herein such as, e.g., a BTK inhibitor described herein or a phosphonositide-3 kinase inhibitor described herein).
  • the subject receives a stem cell transplant
  • the present invention provides methods for evaluation of expression level of one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, twenty-five, thirty, thirty-five, forty, forty-five, fifty, one hundred, or more genes from Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 that comprise a CD19 CAR-expressing cell gene signature.
  • Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 e.g., CCL20, IL-17a and/or IL-6
  • Table 17, Table 18, Table 20 PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 that comprise a CD19
  • methods of the present disclosure can be used to determine the responsiveness of a subject to treatment with a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy described herein such as, e.g., CTL019), wherein a statistically significant difference in the amount, e.g., expression, and/or activity of a marker disclosed herein relative to a reference, e.g., a median value for a cancer patient population (e.g., a hematological cancer such as CLL and ALL), a median value for a population of healthy, cancer-free subjects, a median value for a population of non-responders or partial responders, in a subjects sample, then the more likely the disease is to respond to CAR-expressing cell therapy.
  • a CAR-expressing cell therapy e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy described herein such as, e.g.
  • the disclosure provides a method of, or assay for, identifying a subject having a cancer including, but not limited to, B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphom
  • a CAR-expressing cell therapy comprises a CAR-expressing cell therapy described herein, e.g., CTL019.
  • a CAR-expressing cell therapy consists of a CAR-expressing cell therapy described herein, e.g., CTL019.
  • the disclosure provides a method of, or assay for, identifying a subject having cancer (e.g., a hematological cancer such as CLL and ALL) as having an increased or decreased likelihood to respond to a treatment that comprises a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019), the method comprising:
  • Table 16 e.g., CCL20, IL-17a and/or IL-6
  • a statistically significant difference in expression level of one or more markers in the sample relative to a predetermined value is indicative of increased responsiveness to the CAR-expressing cell.
  • the methods provided herein are particularly useful for identifying subjects that are likely to respond to CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019) prior to initiation of such treatment (e.g., pre-therapy) or early in the therapeutic regimen.
  • CAR-expressing cell e.g., T cell, NK cell
  • expression or activity of biomarkers is measured in a subject at least 2 weeks, at least 1 month, at least 3 months, at least 6 months, or at least 1 year prior to initiation of therapy.
  • expression or activity of biomarkers is measured less than 6 months prior to the initiation of therapy.
  • expression or activity of biomarkers is measured within 6 months, 5 months, 4 months, 3 months, 2 months, 1 month, 2 weeks, 1 week, 6 days, 5 days, 4 days, 3 days, 2 days, or 1 day prior to the initiation of therapy.
  • the expression or activity of biomarkers is determined after initiation of therapy (e.g., 1 month, 2 months, 3 months, 3.5 months, 4 months, 4.5 months, 5 months, 5.5 months, 6 months).
  • the invention provides a method of evaluating a subject having cancer (e.g., a hematological cancer such as CLL and ALL) comprising:
  • acquiring a value of responder status for the subject that comprises a measure of one or more of the following:
  • a memory T cell e.g.,
  • T SCM memory stem cell
  • T CM central memory T cell
  • T EM effector memory T cell
  • the disclosure provides a method of evaluating a subject having cancer (e.g., a hematological cancer such as CLL and ALL) comprising acquiring a value of responder status for the subject that comprises a measure of one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature, thereby evaluating the subject.
  • the disclosure provides a method of evaluating or monitoring the effectiveness of a CAR-expressing cell therapy in a subject having cancer comprising:
  • acquiring a value of responder status for the subject that comprises a measure of one or more of the following:
  • a memory T cell e.g., a CD8+ memory T cell, e.g., a na ⁇ ve T cell (T N ),
  • T SCM memory stem cell
  • T CM central memory T cell
  • T EM effector memory T cell
  • the disclosure provides a method of evaluating or monitoring the effectiveness of a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019) in a subject having cancer (e.g., a hematological cancer such as CLL and ALL) comprising: acquiring a value of responder status for the subject that comprises a measure of one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell
  • the value of responder status comprises a measure of a combination of a gene signature and a biomarker.
  • the value of the responder status comprises a measure of a CD19 CAR-expressing cell gene set signature and a combination of one or more of: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, and a memory T cell (e.g., a CD8+ memory T cell, e.g., a na ⁇ ve T cell (T N ), e.g. a memory stem
  • the value of the responder status comprises a measure of a CD19 CAR-expressing cell gene set signature and a combination of one or more of: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1.
  • the value of the responder status comprises a measure of one or more biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B and a combination of one or more of: a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g., a na ⁇ ve T cell (T N ), e.g.
  • a memory T cell e.g., a CD8+ memory T cell, e.g., a na ⁇ ve T cell (T N ), e.g.
  • T SCM memory stem cell
  • T CM central memory T cell
  • T EM effector memory T cell
  • the value of the responder status comprises a measure of one or more biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B and a combination of one or more of: CD57, CD27, CD122, CD62L, and KLRG1.
  • the CD19 CAR-expressing cell gene signature comprises a value for expression of at least 5, 6, 7, 8, 9 or 10 genes comprising a CD19 CAR-expressing cell gene signature.
  • the value for expression of the gene comprises a value for a transcriptional parameter, e.g., the level of an mRNA encoded by the gene.
  • the value for expression of the protein comprises a value for a translational parameter, e.g., the level of a protein.
  • provided methods further comprise obtaining a sample from the subject, wherein the sample comprises a cellular or tissue fraction.
  • the cellular fraction comprises blood.
  • the measure of biomarker and/or gene signature is acquired before, at the same time, or during course of a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019).
  • a CAR-expressing cell therapy e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019
  • the measure of biomarker and/or gene signature is acquired less than 6 months, 5 months, 4 months, 3 months, 2 months, 1 month, 2 weeks, 1 week, 6 days, 5 days, 4 days, 3 days, 2 days prior to the initiation of a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019).
  • a CAR-expressing cell therapy e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019
  • the methods described herein can also be used to monitor a positive response of a subject to CAR-expressing cell (e.g., T cell, NK cell) treatment (e.g., a CD19 CAR-expressing cell treatment described herein such as, e.g., CTL019).
  • CAR-expressing cell e.g., T cell, NK cell
  • Such methods are useful for early detection of tolerance to therapy or to predict whether disease in a subject will progress.
  • the expression or activity of biomarkers is determined e.g., at least every week, at least every 2 weeks, at least every month, at least every 2 months, at least every 3 months, at least every 4 months, at least every 5 months, at least every 6 months, at least every 7 months, at least every 8 months, at least every 9 months, at least every 10 months, at least every 11 months, at least every year, at least every 18 months, at least every 2 years, at least every 3 years, at least every 5 years or more. It is also contemplated that expression or activity of the biomarkers is at irregular intervals e.g., biomarkers can be detected in a subject at 3 months of treatment, at 6 months of treatment, and at 7 months of treatment. Thus, in some embodiments, the expression or activity of the biomarkers is determined when deemed necessary by the skilled physician monitoring treatment of the subject.
  • the methods described herein can be used in treating any subject having cancer (e.g., a hematological cancer such as CLL and ALL).
  • the invention pertains to methods of treating cancer (e.g., a hematological cancer such as CLL and ALL) in a subject.
  • the disclosure provides methods for treating cancer including, but not limited to, B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymph
  • provided methods comprise administering to the subject a cell expressing a CAR, e.g. a CAR T cell, e.g. a CD19 CAR T cell, e.g., a CTL019 product, if the subject is identified as having a difference, e.g., statistically significant difference in expression level of one or more markers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 relative to a reference level, such that the cancer (e.g., a hematological cancer such as ALL and CLL) is treated in the subject.
  • a CAR e.g. a CAR T cell, e.g. a CD19 CAR T cell, e.g., a CTL
  • an example of a cancer that is treatable by disclosed methods is a cancer associated with expression of CD19.
  • the cancer associated with expression of CD19 is a hematological cancer.
  • the hematological cancer is a leukemia or a lymphoma.
  • a cancer associated with expression of CD19 includes cancers and malignancies including, but not limited to, e.g., one or more acute leukemias including but not limited to, e.g., B-cell acute lymphoid leukemia (“B-ALL”), T-cell acute lymphoid leukemia (“T-ALL”), acute lymphoid leukemia (ALL); one or more chronic leukemias including but not limited to, e.g., chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL).
  • B-ALL B-cell acute lymphoid leukemia
  • T-ALL T-cell acute lymphoid leukemia
  • ALL acute lymphoid leukemia
  • chronic leukemias including but not limited to, e.g., chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL).
  • CML chronic myelogenous leukemia
  • CLL chronic lymphocytic leukemia
  • Additional cancers or hematologic conditions associated with expression of CD19 include, but are not limited to, e.g., B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, and “preleukemia” which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of
  • the disclosure provides a method for treating a subject having cancer (e.g., a hematological cancer such as ALL and CLL) comprising:
  • determining if the subject has an increased likelihood to respond to a CAR-expressing cell (e.g., T cell, NK cell) therapy e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019) by comparing the level of one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more) markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 in a sample from the subject relative to a reference level, wherein a statistically significant difference in expression level of one or more maker genes relative to the reference level is indicative of an increased likelihood of response; and
  • a CAR-expressing cell e.g., T cell
  • the disclosure provides a method for treating a subject having cancer (e.g., a hematological cancer such as ALL and CLL), comprising:
  • Table 16 e.g., CCL20, IL-17a and/or IL-6
  • Table 16 e.g., CCL20, IL-17a and/or IL-6
  • Table 17 Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 to a reference level;
  • a CAR-expressing cell e.g., T cell, NK cell
  • a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019
  • the subject is identified as having a statistically significant difference in the determined level of one or more markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 to a reference level, in the sample.
  • a CAR-expressing cell e.g., T cell, NK cell
  • a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019
  • the disclosure provides a method of treating cancer (e.g., a hematological cancer such as ALL and CLL) in a subject, comprising:
  • acquiring a value of responder status for the subject that comprises a measure of one or more of the following:
  • a memory T cell e.g., a CD8+ memory T cell, e.g., a na ⁇ ve T cell (T N ),
  • T SCM memory stem cell
  • T CM central memory T cell
  • T EM effector memory T cell
  • a CAR-expressing cell therapy e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019;
  • an additional agent in combination with a CAR-expressing cell therapy e.g., a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein;
  • modifying a manufacturing process of a CAR-expressing cell therapy e.g., enrich for na ⁇ ve T cells prior to introducing a nucleic acid encoding a CAR, e.g., for a subject identified as a non-responder or a partial responder; or selecting an alternative therapy, e.g., for a non-responder or partial responder; or
  • an alternative therapy e.g., an alternative therapy described herein, e.g., a standard of care therapy for the cancer; thereby treating cancer in a subject.
  • the disclosure provides a method of treating cancer (e.g., a hematological cancer such as ALL and CLL) in a subject, comprising: acquiring a value of responder status for the subject that comprises a measure of one or more of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature, and responsive to a determination of responder status, performing one, two, three four or more of: identifying the subject as a complete responder, partial responder or non-responder; administering a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019
  • the amount of the biomarker determined in a sample from a subject is quantified as an absolute measurement (e.g., ng/mL).
  • Absolute measurements can easily be compared to a reference value or cut-off value.
  • a cut-off value can be determined that represents a disease progressing status; any absolute values falling either above (i.e., for biomarkers that increase expression with progression of a cancer, e.g., a hematological cancer such as ALL and CLL) or falling below (i.e., for biomarkers with decreased expression with progression of a cancer, e.g., a hematological cancer such as ALL and CLL) the cut-off value are likely to be disease progressing.
  • the present disclosure also pertains to the field of predictive medicine in which diagnostic assays, pharmacogenomics, and monitoring clinical trials are used for predictive purposes to thereby treat an individual prophylactically. Accordingly, one aspect of the present disclosure relates to assays for determining the amount, structure, and/or activity of polypeptides or nucleic acids corresponding to one or more markers described herein, in order to determine whether an individual having cancer (e.g., a hematological cancer such as CLL and ALL) or at risk of developing cancer (e.g., a hematological cancer such as CLL and ALL) will be more likely to respond to CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019).
  • cancer e.g., a hematological cancer such as CLL and ALL
  • CAR-expressing cell therapy e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL
  • the disclosure provides a method for determining whether a subject with cancer (e.g., a hematological cancer such as CLL and ALL) is likely to respond to a cell expressing a CAR, e.g., a CD19 CAR-expressing cell described herein, such as CTL019.
  • a subject with cancer e.g., a hematological cancer such as CLL and ALL
  • the disclosure is drawn to a method for predicting a time course of disease.
  • the method is drawn to a method for predicting a probability of a significant event in the time course of the disease (e.g., reoccurrence or remission).
  • the method comprises detecting a combination of biomarkers associated with responsiveness to treatment as described herein and determining whether the subject is likely to respond to treatment.
  • the disclosure provides a method for providing a prognosis for success rate of a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019) in a subject having cancer (e.g., a hematological cancer such as ALL and CLL), said method comprising steps of:
  • one or more genes listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 to obtain a gene expression pattern for the sample; and
  • the step of determining the levels of expression of the set of genes further comprises detecting the expression of mRNA expressed from said genes.
  • provided methods further comprise a step wherein determining the expression of mRNA comprises exposing said mRNA to a nucleic acid probe complementary to said mRNA.
  • the step of determining the levels of expression of the set of genes further comprises detecting the expression of a polypeptide encoded by said genes.
  • provided methods comprise selecting a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019) for the subject, based on the prognosis provided.
  • a CAR-expressing cell e.g., T cell, NK cell
  • a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019
  • the one or more alterations, e.g., alterations in biomarker expression are assessed at pre-determined intervals, e.g., a first point in time and at least at a subsequent point in time.
  • a time course is measured by determining the time between significant events in the course of a subject disease, wherein the measurement is predictive of whether a subject has a long time course.
  • the significant event is the progression from diagnosis to death. In another embodiment, the significant event is the progression from diagnosis to worsening disease.
  • Biomarker expression level can also be assayed. Expression of a marker described herein can be assessed by any of a wide variety of known methods for detecting expression of a transcribed molecule or protein. Non-limiting examples of such methods include immunological methods for detection of secreted, cell-surface, cytoplasmic, or nuclear proteins, protein purification methods, protein function or activity assays, nucleic acid hybridization methods, nucleic acid reverse transcription methods, and nucleic acid amplification methods.
  • activity of a particular gene is characterized by a measure of gene transcript (e.g., mRNA), by a measure of the quantity of translated protein, or by a measure of gene product activity.
  • Marker expression can be monitored in a variety of ways, including by detecting mRNA levels, protein levels, or protein activity, any of which can be measured using standard techniques. Detection can involve quantification of the level of gene expression (e.g., genomic DNA, CDNA, mRNA, protein, or enzyme activity), or, alternatively, can be a qualitative assessment of the level of gene expression, in particular in comparison with a control level. The type of level being detected will be clear from the context.
  • mRNA or CDNA made therefrom Methods of detecting and/or quantifying the gene transcript (mRNA or CDNA made therefrom) using nucleic acid hybridization techniques are known to those of skill in the art (see e.g., Sambrook et al. supra).
  • one method for evaluating the presence, absence, or quantity of CDNA involves a Southern transfer as described above. Briefly, the mRNA is isolated (e.g., using an acid guanidinium-phenol-chloroform extraction method, Sambrook et al. supra.) and reverse transcribed to produce CDNA. The CDNA is then optionally digested and run on a gel in buffer and transferred to membranes. Hybridization is then carried out using the nucleic acid probes specific for the target CDNA.
  • one method to conduct such an assay would involve anchoring the marker or probe onto a solid phase support, also referred to as a substrate, and detecting target marker/probe complexes anchored on the solid phase at the end of the reaction.
  • a sample from a subject which is to be assayed for presence and/or concentration of marker, can be anchored onto a carrier or solid phase support.
  • the reverse situation is possible, in which the probe can be anchored to a solid phase and a sample from a subject can be allowed to react as an unanchored component of the assay.
  • the non-immobilized component is added to the solid phase upon which the second component is anchored.
  • uncomplexed components can be removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized upon the solid phase.
  • the detection of marker/probe complexes anchored to the solid phase can be accomplished in a number of methods outlined herein.
  • the probe when it is the unanchored assay component, can be labeled for the purpose of detection and readout of the assay, either directly or indirectly, with detectable labels discussed herein and which are well-known to one skilled in the art.
  • marker/probe complex formation without further manipulation or labeling of either component (marker or probe), for example by utilizing the technique of fluorescence energy transfer (see, for example, Lakowicz et al., U.S. Pat. No. 5,631,169; Stavrianopoulos, et al., U.S. Pat. No. 4,868,103).
  • a fluorophore label on the first, ‘donor’ molecule is selected such that, upon excitation with incident light of appropriate wavelength, its emitted fluorescent energy will be absorbed by a fluorescent label on a second ‘acceptor’ molecule, which in turn is able to fluoresce due to the absorbed energy.
  • the ‘donor’ protein molecule can simply utilize the natural fluorescent energy of tryptophan residues. Labels are chosen that emit different wavelengths of light, such that the ‘acceptor’ molecule label can be differentiated from that of the ‘donor’. Since the efficiency of energy transfer between the labels is related to the distance separating the molecules, spatial relationships between the molecules can be assessed. In a situation in which binding occurs between the molecules, the fluorescent emission of the ‘acceptor’ molecule label in the assay should be maximal. An FET binding event can be conveniently measured through standard fluorometric detection means well known in the art (e.g., using a fluorimeter).
  • determination of the ability of a probe to recognize a marker can be accomplished without labeling either assay component (probe or marker) by utilizing a technology such as real-time Biomolecular Interaction Analysis (BIA) (see, e.g., Sjolander, S. and Urbaniczky, C., 1991, A NAL . C HEM . 63:2338-2345 and Szabo et al., 1995, C URR . O PIN . S TRUCT . B IOL . 5:699-705).
  • BIOA Biomolecular Interaction Analysis
  • surface plasmon resonance is a technology for studying biospecific interactions in real time, without labeling any of the interactants (e.g., BIAcore).
  • analogous diagnostic and prognostic assays can be conducted with marker and probe as solutes in a liquid phase.
  • the complexed marker and probe are separated from uncomplexed components by any of a number of standard techniques, including but not limited to: differential centrifugation, chromatography, electrophoresis and immunoprecipitation.
  • differential centrifugation marker/probe complexes can be separated from uncomplexed assay components through a series of centrifugal steps, due to the different sedimentation equilibria of complexes based on their different sizes and densities (see, for example, Rivas, G., and Minton, A. P., 1993 , Trends Biochem Sci.
  • Standard chromatographic techniques can also be utilized to separate complexed molecules from uncomplexed ones.
  • gel filtration chromatography separates molecules based on size, and through the utilization of an appropriate gel filtration resin in a column format, for example, the relatively larger complex can be separated from the relatively smaller uncomplexed components.
  • the relatively different charge properties of the marker/probe complex as compared to the uncomplexed components can be exploited to differentiate the complex from uncomplexed components, for example, through the utilization of ion-exchange chromatography resins.
  • Such resins and chromatographic techniques are well known to one skilled in the art (see, e.g., Heegaard, N. H., 1998, J. M OL .
  • Gel electrophoresis can also be employed to separate complexed assay components from unbound components (see, e.g., Ausubel et al., ed., C URRENT P ROTOCOLS IN M OLECULAR B IOLOGY , John Wiley & Sons, New York, 1987-1999).
  • protein or nucleic acid complexes are separated based on size or charge, for example.
  • non-denaturing gel matrix materials and conditions in the absence of reducing agent are typical. Appropriate conditions to the particular assay and components thereof will be well known to one skilled in the art.
  • the level of mRNA corresponding to the marker can be determined both by in situ and by in vitro formats in a biological sample using methods known in the art.
  • biological sample is intended to include tissues, cells, biological fluids and isolates thereof, isolated from a subject, as well as tissues, cells and fluids present within a subject.
  • Many expression detection methods use isolated RNA.
  • any RNA isolation technique that does not select against the isolation of mRNA can be utilized for the purification of RNA from cells (see, e.g., Ausubel et al., ed., C URRENT P ROTOCOLS IN M OLECULAR B IOLOGY , John Wiley & Sons, New York 1987-1999).
  • large numbers of tissue samples can readily be processed using techniques well known to those of skill in the art, such as, for example, the single-step RNA isolation process of Chomczynski (1989, U.S. Pat. No. 4,843,155).
  • the isolated nucleic acid can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, polymerase chain reaction analyses and probe arrays.
  • One diagnostic method for the detection of mRNA levels involves contacting the isolated mRNA with a nucleic acid molecule (probe) that can hybridize to the mRNA encoded by the gene being detected.
  • the nucleic acid probe can be, for example, a full-length CDNA, or a portion thereof, such as an oligonucleotide of at least 7, 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to a mRNA or genomic DNA encoding a marker of the present invention.
  • Other suitable probes for use in the diagnostic assays are described herein. Hybridization of an mRNA with the probe indicates that the marker in question is being expressed.
  • the mRNA is immobilized on a solid surface and contacted with a probe, for example by running the isolated mRNA on an agarose gel and transferring the mRNA from the gel to a membrane, such as nitrocellulose.
  • the probe(s) are immobilized on a solid surface and the mRNA is contacted with the probe(s), for example, in an Affymetrix gene chip array.
  • a skilled artisan can readily adapt known mRNA detection methods for use in detecting the level of mRNA encoded by the markers described herein.
  • the probes can be full length or less than the full length of the nucleic acid sequence encoding the protein. Shorter probes are empirically tested for specificity. Exemplary nucleic acid probes are 20 bases or longer in length (See, e.g., Sambrook et al. for methods of selecting nucleic acid probe sequences for use in nucleic acid hybridization). Visualization of the hybridized portions allows the qualitative determination of the presence or absence of CDNA.
  • Fluorogenic rtPCR can also be used in the methods of the invention. In fluorogenic rtPCR, quantitation is based on amount of fluorescence signals, e.g., TaqMan and sybr green.
  • amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5′ or 3′ regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between.
  • amplification primers are from about 10 to 30 nucleotides in length and flank a region from about 50 to 200 nucleotides in length. Under appropriate conditions and with appropriate reagents, such primers permit the amplification of a nucleic acid molecule comprising the nucleotide sequence flanked by the primers.
  • mRNA does not need to be isolated from the cells prior to detection.
  • a cell or tissue sample is prepared/processed using known histological methods. The sample is then immobilized on a support, typically a glass slide, and then contacted with a probe that can hybridize to mRNA that encodes the marker.
  • the expression level can be provided as a relative expression level.
  • the level of expression of the marker can be determined for 10 or more samples of normal versus cancer isolates, or even 50 or more samples, prior to the determination of the expression level for the sample in question.
  • the mean expression level of each of the genes assayed in the larger number of samples can be determined and this can be used as a baseline expression level for the marker.
  • the expression level of the marker determined for the test sample (absolute level of expression) then can be divided by the mean expression value obtained for that marker. This provides a relative expression level.
  • the samples used in the baseline determination will be from samples derived from a subject having cancer (e.g., a hematological cancer such as ALL and CLL) versus samples from a healthy subject of the same tissue type.
  • a subject having cancer e.g., a hematological cancer such as ALL and CLL
  • the choice of the cell source is dependent on the use of the relative expression level.
  • Using expression found in normal tissues as a mean expression score aids in validating whether the marker assayed is specific to the tissue from which the cell was derived (versus normal cells).
  • the mean expression value can be revised, providing improved relative expression values based on accumulated data. Expression data from normal cells provides a means for grading the severity of the cancer disease state.
  • expression of a marker is assessed by preparing genomic DNA or mRNA/CDNA (i.e., a transcribed polynucleotide) from cells in a subject sample, and by hybridizing the genomic DNA or mRNA/CDNA with a reference polynucleotide which is a complement of a polynucleotide comprising the marker, and fragments thereof.
  • CDNA can, optionally, be amplified using any of a variety of polymerase chain reaction methods prior to hybridization with the reference polynucleotide.
  • Expression of one or more markers can likewise be detected using quantitative PCR (QPCR) to assess the level of expression of the marker(s).
  • a mixture of transcribed polynucleotides obtained from the sample is contacted with a substrate having fixed thereto a polynucleotide complementary to or homologous with at least a portion (e.g., at least 7, at least 10, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, at least 100, at least 500, or more nucleotide residues) of a marker described herein.
  • a portion e.g., at least 7, at least 10, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, at least 100, at least 500, or more nucleotide residues
  • polynucleotides complementary to, or homologous with, a marker described herein are differentially detectable on the substrate (e.g., detectable using different chromophores or fluorophores, or fixed to different selected positions), then the levels of expression of a plurality of markers can be assessed simultaneously using a single substrate (e.g., a “gene chip” microarray of polynucleotides fixed at selected positions).
  • a method of assessing marker expression which involves hybridization of one nucleic acid with another, the hybridization can be performed under stringent hybridization conditions.
  • a combination of methods to assess the expression of a marker is utilized.
  • the level of expression of the marker is significantly greater than the minimum detection limit of the method used to assess expression in at least one of a biological sample from a subject with cancer (e.g., a hematological cancer such as ALL and CLL) or a reference (e.g., a biological sample from a healthy subject, e.g., a subject without cancer).
  • a biological sample from a subject with cancer e.g., a hematological cancer such as ALL and CLL
  • a reference e.g., a biological sample from a healthy subject, e.g., a subject without cancer.
  • nucleic acid molecules that correspond to one or markers described herein, including nucleic acids which encode a polypeptide corresponding to one or more markers described herein or a portion of such a polypeptide.
  • the nucleic acid molecules include those nucleic acid molecules which reside in genomic regions identified herein.
  • Isolated nucleic acid molecules also include nucleic acid molecules sufficient for use as hybridization probes to identify nucleic acid molecules that correspond to a marker described herein, including nucleic acid molecules which encode a polypeptide corresponding to a marker described herein, and fragments of such nucleic acid molecules, e.g., those suitable for use as PCR primers for the amplification or mutation of nucleic acid molecules.
  • Probes based on the sequence of a nucleic acid molecule of the invention can be used to detect transcripts (e.g., mRNA) or genomic sequences corresponding to one or more markers described herein.
  • the probe comprises a label group attached thereto, e.g., a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.
  • Such probes can be used as part of a diagnostic test kit for identifying cells or tissues which mis-express the protein, such as by measuring levels of a nucleic acid molecule encoding the protein in a sample of cells from a subject, e.g., detecting mRNA levels or determining whether a gene encoding the protein has been mutated or deleted.
  • the activity or level of a marker protein can also be detected and/or quantified by detecting or quantifying the expressed polypeptide.
  • the polypeptide can be detected and quantified by any of a number of means well known to those of skill in the art. These can include analytic biochemical methods such as electrophoresis, capillary electrophoresis, high performance liquid chromatography (HPLC), thin layer chromatography (TLC), hyperdiffusion chromatography, and the like, or various immunological methods such as fluid or gel precipitin reactions, immunodiffusion (single or double), immunoelectrophoresis, radioimmunoassay (RIA), enzyme-linked immunosorbent assays (ELISAs), immunofluorescent assays, Western blotting, immunohistochemistry and the like.
  • HPLC high performance liquid chromatography
  • TLC thin layer chromatography
  • ELISAs enzyme-linked immunosorbent assays
  • immunofluorescent assays Western blotting, immunohistochemistry and the like.
  • Another agent for detecting a polypeptide is an antibody capable of binding to a polypeptide corresponding to a marker described herein, e.g., an antibody with a detectable label.
  • Antibodies can be polyclonal or monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab) 2 ) can be used.
  • the term “labeled”, with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently labeled streptavidin.
  • the antibody is labeled, e.g., a radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody.
  • an antibody derivative e.g., an antibody conjugated with a substrate or with the protein or ligand of a protein-ligand pair (e.g., biotin-streptavidin)
  • an antibody fragment e.g., a single-chain antibody, an isolated antibody hypervariable domain, etc.
  • a protein corresponding to the marker such as the protein encoded by the open reading frame corresponding to the marker or such a protein which has undergone all or a portion of its normal post-translational modification, is used.
  • Proteins from cells can be isolated using techniques that are well known to those of skill in the art.
  • the protein isolation methods employed can, for example, be such as those described in Harlow and Lane (Harlow and Lane, 1988 , Antibodies: A Laboratory Manual , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.).
  • antibodies, or antibody fragments can be used in methods such as Western blots or immunofluorescence techniques to detect the expressed proteins.
  • Suitable solid phase supports or carriers include any support capable of binding an antigen or an antibody.
  • Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.
  • the polypeptide is detected and/or quantified using Luminex® assay technology.
  • the Luminex® assay separates tiny color-coded beads into e.g., distinct sets that are each coated with a reagent for a particular bioassay, allowing the capture and detection of specific analytes from a sample in a multiplex manner.
  • the Luminex® assay technology can be compared to a multiplex ELISA assay using bead-based fluorescence cytometry to detect analytes such as biomarkers.
  • a kit can be used to assess the disease progression of a cancer including, but not limited to, B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma, marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, Hod
  • the disclosure provides a kit for assessing and characterizing responder status (e.g., compete responder, partial responder or non-responder) of a subject having a hematological cancer to a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy as described herein, such as e.g., CTL019).
  • a CAR-expressing cell e.g., T cell, NK cell
  • a CD19 CAR-expressing cell therapy as described herein, such as e.g., CTL019
  • Suitable reagents for binding with a polypeptide corresponding to a marker described herein include antibodies, antibody derivatives, antibody fragments, and the like.
  • Suitable reagents for binding with a nucleic acid include complementary nucleic acids.
  • the nucleic acid reagents can include oligonucleotides (labeled or non-labeled) fixed to a substrate, labeled oligonucleotides not bound with a substrate, pairs of PCR primers, molecular beacon probes, and the like.
  • the kit can optionally comprise additional components useful for performing the methods described herein.
  • the kit can comprise fluids (e.g., SSC buffer) suitable for annealing complementary nucleic acids or for binding an antibody with a protein with which it specifically binds, one or more sample compartments, an instructional material which describes performance of a method of the invention, a reference sample for comparison of expression levels of the biomarkers described herein, and the like.
  • kits of the invention can comprise a reagent useful for determining protein level or protein activity of a marker.
  • a set of reagents that specifically detects expression levels of one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 50 or more) genes listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 and a CD19 CAR-expressing cell gene set signature; and
  • one or more e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 50 or more genes listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27
  • said instructions for use provide that if one or more of the detected expression levels is greater than a reference level, the subject is more likely to respond positively to a CAR-expressing cell therapy.
  • the set of reagents detects the expression of polypeptides encoded by said set of genes.
  • the methods described herein can be used to assess a responder status to a cell expressing a CAR.
  • the cell expresses a CAR molecule comprising an antigen binding domain (e.g., an antibody or antibody fragment that specifically binds to a tumor antigen), a transmembrane domain, and an intracellular signaling domain (e.g., an intracellular signaling domain comprising a costimulatory domain and/or a primary signaling domain).
  • the antigen binding domain comprises any antibody, or a fragment thereof, e.g., an scFv, known in the art that targets or specifically binds to any of the tumor antigens described herein.
  • the tumor antigen is BCMA (also known as TNFRSF17, Tumor Necrosis Factor Receptor Superfamily, Member 17, or B Cell Maturation Antigen), CD33, CLL-1 (also known as C-type Lectin-Like domain family 1, or CLECL1) or claudin-6 (CLDN6).
  • BCMA also known as TNFRSF17, Tumor Necrosis Factor Receptor Superfamily, Member 17, or B Cell Maturation Antigen
  • CD33 also known as TNFRSF17, Tumor Necrosis Factor Receptor Superfamily, Member 17, or B Cell Maturation Antigen
  • CLL-1 also known as C-type Lectin-Like domain family 1, or CLECL1
  • CLDN6 claudin-6
  • the CAR comprises an antibody or antibody fragment which includes an anti-CD19 binding domain described herein (e.g., a murine or humanized antibody or antibody fragment that specifically binds to CD19 as described herein), a transmembrane domain described herein, and an intracellular signaling domain described herein (e.g., an intracellular signaling domain comprising a costimulatory domain and/or a primary signaling domain described herein).
  • an anti-CD19 binding domain described herein e.g., a murine or humanized antibody or antibody fragment that specifically binds to CD19 as described herein
  • a transmembrane domain described herein e.g., a transmembrane domain described herein
  • an intracellular signaling domain described herein e.g., an intracellular signaling domain comprising a costimulatory domain and/or a primary signaling domain described herein.
  • the CAR of the invention comprises a target-specific binding element otherwise referred to as an antigen binding domain.
  • an antigen binding domain The choice of moiety depends upon the type and number of ligands that define the surface of a target cell.
  • the antigen binding domain may be chosen to recognize a ligand that acts as a cell surface marker on target cells associated with a particular disease state.
  • examples of cell surface markers that may act as ligands for the antigen binding domain in a CAR of the invention include those associated with viral, bacterial and parasitic infections, autoimmune disease and cancer cells.
  • the CAR-mediated T-cell response can be directed to an antigen of interest by way of engineering an antigen binding domain that specifically binds a desired antigen into the CAR.
  • the portion of the CAR comprising the antigen binding domain comprises an antigen binding domain that targets a tumor antigen, e.g., a tumor antigen described herein.
  • the antigen binding domain can be any domain that binds to the antigen including but not limited to a monoclonal antibody, a polyclonal antibody, a recombinant antibody, a human antibody, a humanized antibody, and a functional fragment thereof, including but not limited to a single-domain antibody such as a heavy chain variable domain (VH), a light chain variable domain (VL) and a variable domain (VHH) of camelid derived nanobody, and to an alternative scaffold known in the art to function as antigen binding domain, such as a recombinant fibronectin domain, a T cell receptor (TCR), or a fragment there of, e.g., single chain TCR, and the like.
  • VH heavy chain variable domain
  • VL light chain variable domain
  • VHH variable domain of camelid derived nanobody
  • an alternative scaffold known in the art to function as antigen binding domain such as a recombinant fibronectin domain, a T cell receptor (TCR), or a fragment there of,
  • the antigen binding domain it is beneficial for the antigen binding domain to be derived from the same species in which the CAR will ultimately be used in.
  • the antigen binding domain of the CAR it may be beneficial for the antigen binding domain of the CAR to comprise human or humanized residues for the antigen binding domain of an antibody or antibody fragment.
  • any known CD19 CAR e.g., the CD19 antigen binding domain of any known CD19 CAR, in the art can be used in accordance with the instant invention.
  • CD19 CAR described in the U.S. Pat. Nos. 8,399,645; 7,446,190; Xu et al., Leuk Lymphoma.
  • target antigens that can be targeted using the CAR-expressing cells, include, but are not limited to, CD19, CD123, EGFRvIII, mesothelin, among others, as described in, for example, WO 2014/130635, WO 2014/130657, and WO 2015/090230, each of which is herein incorporated by reference in its entirety.
  • the CAR T cell that specifically binds to CD19 has the USAN designation TISAGENLECLEUCEL-T.
  • CTL019 is made by a gene modification of T cells is mediated by stable insertion via transduction with a self-inactivating, replication deficient Lentiviral (LV) vector containing the CTL019 transgene under the control of the EF-1 alpha promoter.
  • LV replication deficient Lentiviral
  • CTL019 can be a mixture of transgene positive and negative T cells that are delivered to the subject on the basis of percent transgene positive T cells.
  • the CAR-expressing cells can specifically bind to human CD19, e.g., can include a CAR molecule, or an antigen binding domain (e.g., a humanized antigen binding domain) according to Table 3 of WO2014/153270, incorporated herein by reference.
  • an antigen binding domain e.g., a humanized antigen binding domain
  • the CAR-expressing cells can specifically bind to CD123, e.g., can include a CAR molecule (e.g., any of the CAR1-CAR8), or an antigen binding domain according to Tables 1-2 of WO 2014/130635, incorporated herein by reference.
  • a CAR molecule e.g., any of the CAR1-CAR8
  • an antigen binding domain according to Tables 1-2 of WO 2014/130635, incorporated herein by reference.
  • the CAR-expressing cells can specifically bind to EGFRvIII, e.g., can include a CAR molecule, or an antigen binding domain according to Table 2 or SEQ ID NO:11 of WO 2014/130657, incorporated herein by reference.
  • the CAR-expressing cells can specifically bind to mesothelin, e.g., can include a CAR molecule, or an antigen binding domain according to Tables 2-3 of WO 2015/090230, incorporated herein by reference.
  • the antigen binding domain comprises one, two three (e.g., all three) heavy chain CDRs, HC CDR1, HC CDR2 and HC CDR3, from an antibody listed above, and/or one, two, three (e.g., all three) light chain CDRs, LC CDR1, LC CDR2 and LC CDR3, from an antibody listed above.
  • the antigen binding domain comprises a heavy chain variable region and/or a variable light chain region of an antibody listed or described above.
  • the tumor antigen is a tumor antigen described in International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • the tumor antigen is chosen from one or more of: CD19; CD123; CD22; CD30; CD171; CS-1 (also referred to as CD2 subset 1, CRACC, SLAMF7, CD319, and 19A24); C-type lectin-like molecule-1 (CLL-1 or CLECL1); CD33; epidermal growth factor receptor variant III (EGFRvIII); ganglioside G2 (GD2); ganglioside GD3 (aNeu5 Ac(2-8)aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer); TNF receptor family member B cell maturation (BCMA); Tn antigen ((Tn Ag) or (GalNAc ⁇ -Ser/Thr)); prostate-specific membrane antigen (PSMA); Receptor ty
  • Murine CD19 CAR constructs are described in PCT publication WO 2012/079000, incorporated herein by reference, and the amino acid sequence of the murine CD19 CAR and scFv constructs are shown in Table 2 below.
  • CD19 CAR constructs containing humanized anti-CD19 scFv domains are described in PCT publication WO 2014/153270, incorporated herein by reference.
  • the antigen binding domain comprises an anti-CD19 antibody, or fragment thereof, e.g., an scFv.
  • the antigen binding domain comprises a variable heavy chain and a variable light chain listed in Table 12.
  • the linker sequence joining the variable heavy and variable light chains can be, e.g., any of the linker sequences described herein, or alternatively, can be GSTSGSGKPGSGEGSTKG (SEQ ID NO:45).
  • CD19 CAR e.g., the CD19 antigen binding domain of any known CD19 CAR
  • any known CD19 CAR e.g., the CD19 antigen binding domain of any known CD19 CAR, in the art can be used in accordance with the present disclosure.
  • the antigen binding domain comprises one, two three (e.g., all three) heavy chain CDRs, HC CDR1, HC CDR2 and HC CDR3, from an antibody listed above, and/or one, two, three (e.g., all three) light chain CDRs, LC CDR1, LC CDR2 and LC CDR3, from an antibody listed above.
  • the antigen binding domain comprises a heavy chain variable region and/or a variable light chain region of an antibody listed or described above.
  • an antigen binding domain against CD22 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Haso et al., B LOOD , 121(7): 1165-1174 (2013); Wayne et al., C LIN C ANCER R ES 16(6): 1894-1903 (2010); Kato et al., L EUK R ES 37(1):83-88 (2013); Creative BioMart (creativebiomart.net): MOM-18047-S(P).
  • CDRs antigen binding portion
  • an antigen binding domain against CD20 is an antigen binding portion, e.g., CDRs, of the anitbody Rituximab, Ofatumumab, Ocrelizumab, Veltuzumab, or GA101.
  • an antigen binding domain against ROR1 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Hudecek et al., C LIN C ANCER R ES 19(12):3153-3164 (2013); WO 2011159847; and US20130101607.
  • a multispecific antibody molecule is a bispecific antibody molecule.
  • a bispecific antibody has specificity for no more than two antigens.
  • a bispecific antibody molecule is characterized by a first immunoglobulin variable domain sequence which has binding specificity for a first epitope and a second immunoglobulin variable domain sequence that has binding specificity for a second epitope.
  • the first and second epitopes are on the same antigen, e.g., the same protein (or subunit of a multimeric protein).
  • the first and second epitopes overlap.
  • the first and second epitopes do not overlap.
  • first and second epitopes are on different antigens, e.g., different proteins (or different subunits of a multimeric protein).
  • a bispecific antibody molecule comprises a heavy chain variable domain sequence and a light chain variable domain sequence which have binding specificity for a first epitope and a heavy chain variable domain sequence and a light chain variable domain sequence which have binding specificity for a second epitope.
  • a bispecific antibody molecule comprises a half antibody having binding specificity for a first epitope and a half antibody having binding specificity for a second epitope.
  • a bispecific antibody molecule comprises a half antibody, or fragment thereof, having binding specificity for a first epitope and a half antibody, or fragment thereof, having binding specificity for a second epitope.
  • a bispecific antibody molecule comprises a scFv, or fragment thereof, have binding specificity for a first epitope and a scFv, or fragment thereof, have binding specificity for a second epitope.
  • the antibody molecule is a multi-specific (e.g., a bispecific or a trispecific) antibody molecule.
  • a multi-specific antibody molecule e.g., a bispecific or a trispecific
  • Protocols for generating bispecific or heterodimeric antibody molecules, and various configurations for bispecific antibody molecules, are described in, e.g., paragraphs 455-458 of WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.
  • the bispecific antibody molecule is characterized by a first immunoglobulin variable domain sequence, e.g., a scFv, which has binding specificity for CD19, e.g., comprises a scFv as described herein, or comprises the light chain CDRs and/or heavy chain CDRs from a scFv described herein, and a second immunoglobulin variable domain sequence that has binding specificity for a second epitope on a different antigen.
  • a first immunoglobulin variable domain sequence e.g., a scFv
  • CD19 e.g., comprises a scFv as described herein, or comprises the light chain CDRs and/or heavy chain CDRs from a scFv described herein
  • a second immunoglobulin variable domain sequence that has binding specificity for a second epitope on a different antigen.
  • the antibodies and antibody fragments of the present invention can be grafted to one or more constant domain of a T cell receptor (“TCR”) chain, for example, a TCR alpha or TCR beta chain, to create a chimeric TCR.
  • TCR T cell receptor
  • an scFv as disclosed herein can be grafted to the constant domain, e.g., at least a portion of the extracellular constant domain, the transmembrane domain and the cytoplasmic domain, of a TCR chain, for example, the TCR alpha chain and/or the TCR beta chain.
  • an antibody fragment for example a VL domain as described herein, can be grafted to the constant domain of a TCR alpha chain
  • an antibody fragment for example a VH domain as described herein, can be grafted to the constant domain of a TCR beta chain
  • a VL domain may be grafted to the constant domain of the TCR beta chain
  • a VH domain may be grafted to a TCR alpha chain
  • the CDRs of an antibody or antibody fragment may be grafted into a TCR alpha and/or beta chain to create a chimeric TCR.
  • the LCDRs disclosed herein may be grafted into the variable domain of a TCR alpha chain and the HCDRs disclosed herein may be grafted to the variable domain of a TCR beta chain, or vice versa.
  • Such chimeric TCRs may be produced, e.g., by methods known in the art (For example, Willemsen R A et al, Gene Therapy 2000; 7: 1369-1377; Zhang T et al, Cancer Gene Ther 2004; 11: 487-496; Aggen et al, Gene Ther. 2012 April; 19(4):365-74).
  • the antigen binding domain comprises a non-antibody scaffold, e.g., a fibronectin, ankyrin, domain antibody, lipocalin, small modular immuno-pharmaceutical, maxybody, Protein A, or affilin.
  • the non-antibody scaffold has the ability to bind to target antigen on a cell.
  • the antigen binding domain is a polypeptide or fragment thereof of a naturally occurring protein expressed on a cell.
  • the antigen binding domain comprises a non-antibody scaffold.
  • a wide variety of non-antibody scaffolds can be employed so long as the resulting polypeptide includes at least one binding region which specifically binds to the target antigen on a target cell.
  • Non-antibody scaffolds include: fibronectin (Novartis, MA), ankyrin (Molecular Partners AG, Zurich, Switzerland), domain antibodies (Domantis, Ltd., Cambridge, Mass., and Ablynx nv, Zwijnaarde, Belgium), lipocalin ( Pieris Proteolab AG, Freising, Germany), small modular immuno-pharmaceuticals (Trubion Pharmaceuticals Inc., Seattle, Wash.), maxybodies (Avidia, Inc., Mountain View, Calif.), Protein A (Affibody AG, Sweden), and affilin (gamma-crystallin or ubiquitin) (Scil Proteins GmbH, Halle, Germany).
  • fibronectin Novartis, MA
  • ankyrin Molecular Partners AG, Zurich, Switzerland
  • domain antibodies Domantis, Ltd., Cambridge, Mass., and Ablynx nv, Zwijnaarde, Belgium
  • lipocalin Pieris Proteolab AG, Freising, Germany
  • the antigen binding domain comprises the extracellular domain, or a counter-ligand binding fragment thereof, of molecule that binds a counterligand on the surface of a target cell.
  • a CAR described herein comprises a transmembrane domain that is fused to an extracellular sequence, e.g., an extracellular recognition element, which can comprise an antigen binding domain.
  • the transmembrane domain is one that naturally is associated with one of the domains in the CAR.
  • the transmembrane domain is one that is not naturally associated with one of the domains in the CAR.
  • a transmembrane domain can include one or more additional amino acids adjacent to the transmembrane region, e.g., one or more amino acid associated with the extracellular region of the protein from which the transmembrane was derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the extracellular region) and/or one or more additional amino acids associated with the intracellular region of the protein from which the transmembrane protein is derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the intracellular region).
  • one or more amino acid associated with the extracellular region of the protein from which the transmembrane was derived e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the extracellular region
  • additional amino acids associated with the intracellular region of the protein from which the transmembrane protein is derived e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the intracellular region
  • the transmembrane domain is one which minimizes interactions with other elements, e.g., other transmembrane domains. In some instances, the transmembrane domain minimizes binding of such domains to the transmembrane domains of the same or different surface membrane proteins, e.g., to minimize interactions with other members of the receptor complex. Suitable examples can be derived by selection or modification of amino acid substitution of a known transmembrane domain. In an embodiment, the transmembrane domain is capable of promoting homodimerization with another CAR on the cell surface.
  • Transmembrane regions suitable for use in molecules described herein may be derived from any one or more of e.g., the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154.
  • a transmembrane domain may include at least the transmembrane region(s) of, e.g., KIRDS2, OX40, CD2, CD27, LFA-1 (CD11a, CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD160, CD19, IL2R beta, IL2R gamma, IL7R a, ITGA1, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, DNAM1 (CD22
  • the transmembrane domain is derived from CD8. In an embodiment the transmembrane domain is derived from CD28. In one aspect, the transmembrane domain is a transmembrane domain from the sequence provided as SEQ ID NO: 12 or SEQ ID NO: 42.
  • a sequence e.g., a hinge or spacer sequence
  • a sequence can be disposed between a transmembrane domain and another sequence or domain to which it is fused.
  • a variety of human hinges aka “spacers” can be employed as well, e.g., including but not limited to the human Ig (immunoglobulin) hinge.
  • a short oligo- or polypeptide linker between 2 and 10 amino acids in length may form the linkage between the transmembrane domain and another domain, e.g., an intracellular signaling domain or costimulatory domain, of a CAR.
  • a glycine-serine doublet provides a particularly suitable linker.
  • the hinge or spacer is the amino acid sequence provided as SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 8.
  • the hinge or spacer comprises a KIR2DS2 hinge.
  • the transmembrane domain may be a non-naturally occurring sequence, in which case can comprise predominantly hydrophobic residues such as leucine and valine.
  • a triplet of phenylalanine, tryptophan and valine will be found at each end of a transmembrane domain.
  • a short oligo- or polypeptide linker may form the linkage between the transmembrane domain and the cytoplasmic region of the CAR.
  • a glycine-serine doublet provides a particularly suitable linker.
  • the linker comprises the amino acid sequence of GGGGSGGGGS (SEQ ID NO:10).
  • the linker is encoded by a nucleotide sequence of GGTGGCGGAGGTTCTGGAGGTGGAGGTTCC (SEQ ID NO:11).
  • the cytoplasmic domain or region of the CAR includes an intracellular signaling domain.
  • An intracellular signaling domain is generally responsible for activation of at least one of the normal effector functions of the immune cell in which the CAR has been introduced.
  • intracellular signaling domains for use in the CAR of the invention include the cytoplasmic sequences of the T cell receptor (TCR) and co-receptors that act in concert to initiate signal transduction following antigen receptor engagement, as well as any derivative or variant of these sequences and any recombinant sequence that has the same functional capability.
  • TCR T cell receptor
  • T cell activation can be said to be mediated by two distinct classes of cytoplasmic signaling sequences: those that initiate antigen-dependent primary activation through the TCR (primary intracellular signaling domains) and those that act in an antigen-independent manner to provide a secondary or costimulatory signal (secondary cytoplasmic domain, e.g., a costimulatory domain).
  • a primary signaling domain regulates primary activation of the TCR complex either in a stimulatory way, or in an inhibitory way.
  • Primary intracellular signaling domains that act in a stimulatory manner may contain signaling motifs which are known as immunoreceptor tyrosine-based activation motifs or ITAMs.
  • a CAR of the invention comprises an intracellular signaling domain, e.g., a primary signaling domain of CD3-zeta, e.g., a CD3-zeta sequence described herein.
  • a primary signaling domain comprises a modified ITAM domain, e.g., a mutated ITAM domain which has altered (e.g., increased or decreased) activity as compared to the native ITAM domain.
  • a primary signaling domain comprises a modified ITAM-containing primary intracellular signaling domain, e.g., an optimized and/or truncated ITAM-containing primary intracellular signaling domain.
  • a primary signaling domain comprises one, two, three, four or more ITAM motifs. Further examples of molecules containing a primary intracellular signaling domain that are of particular use in the invention include those of DAP10, DAP12, and CD32.
  • a primary intracellular signaling domain comprises a functional fragment, or analog, of a primary stimulatory molecule (e.g., CD3 zeta-GenBank Acc. No. BAG36664.1).
  • the primary intracellular signaling domain can comprise the entire intracellular region or a fragment of the intracellular region which is sufficient for generation of an intracellular signal when an antigen binding domain to which it is fused binds cognate antigen.
  • the primary intracellular signaling domain has at least 70, 75, 80, 85, 90, 95, 98, or 99% sequence identity with the entire intracellular region, or a fragment of the intracellular region which is sufficient for generation of an intracellular signal, of a naturally occurring primary stimulatory molecule, e.g., a human (GenBank Acc No.
  • BAG36664.1 or other mammalian, e.g., a nonhuman species, e.g., rodent, monkey, ape or murine intracellular primary stimulatory molecule.
  • the primary intracellular signaling domain has at least 70, 75, 80, 85, 90, 95, 98, or 99% sequence identity with SEQ ID NO: 18 or SEQ ID NO: 20.
  • the primary intracellular signaling domain has at least 70, 75, 80, 85, 90, 95, 96, 97, 98, or 99% identity with, or differs by no more than 30, 25, 20, 15, 10, 5, 4, 3, 2, or 1 amino acid residues from the corresponding residues of the entire intracellular region, or a fragment of the intracellular region which is sufficient for generation of an intracellular signal, of a naturally occurring human primary stimulatory molecule, e.g., a naturally occurring human primary stimulatory molecule disclosed herein.
  • the intracellular signalling domain of the CAR can comprise the CD3-zeta signalling domain by itself or it can be combined with any other desired intracellular signalling domain(s) useful in the context of a CAR of the invention.
  • the intracellular signalling domain of the CAR can comprise a CD3 zeta chain portion and a costimulatory signaling domain.
  • the costimulatory signaling domain refers to a portion of the CAR comprising the intracellular domain of a costimulatory molecule.
  • the intracellular domain is designed to comprise the signaling domain of CD3-zeta and the signaling domain of CD28.
  • the intracellular domain is designed to comprise the signaling domain of CD3-zeta and the signaling domain of ICOS.
  • a costimulatory molecule is a cell surface molecule other than an antigen receptor or its ligands that is required for an efficient response of lymphocytes to an antigen.
  • examples of such molecules include MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM-1, ICOS (CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD19, CD4, CD8alpha, CD8beta,
  • CD27 costimulation has been demonstrated to enhance expansion, effector function, and survival of human CAR-expressing cell (e.g., T cell, NK cell) cells in vitro and augments human T cell persistence and antitumor activity in vivo (Song et al. BLOOD. 2012; 119(3):696-706).
  • human CAR-expressing cell e.g., T cell, NK cell
  • costimulatory molecules include CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD
  • the intracellular signaling sequences within the cytoplasmic portion of the CAR of the invention may be linked to each other in a random or specified order.
  • a short oligo- or polypeptide linker for example, between 2 and 10 amino acids (e.g., 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) in length may form the linkage between intracellular signaling sequence.
  • a glycine-serine doublet can be used as a suitable linker.
  • a single amino acid e.g., an alanine, a glycine, can be used as a suitable linker.
  • the intracellular signaling domain is designed to comprise two or more, e.g., 2, 3, 4, 5, or more, costimulatory signaling domains.
  • the two or more, e.g., 2, 3, 4, 5, or more, costimulatory signaling domains are separated by a linker molecule, e.g., a linker molecule described herein.
  • the intracellular signaling domain comprises two costimulatory signaling domains.
  • the linker molecule is a glycine residue. In some embodiments, the linker is an alanine residue.
  • a costimulatory domain comprises a functional fragment, or analog, of a costimulatory molecule (e.g., ICOS, CD28, or 4-1BB). It can comprise the entire intracellular region or a fragment of the intracellular region which is sufficient for generation of an intracellular signal, e.g., when an antigen binding domain to which it is fused binds cognate antigen.
  • a costimulatory molecule e.g., ICOS, CD28, or 4-1BB.
  • the costimulatory domain has at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity with the entire intracellular region, or a fragment of the intracellular region which is sufficient for generation of an intracellular signal, of a naturally occurring costimulatory molecule as described herein, e.g., a human, or other mammalian, e.g., a nonhuman species, e.g., rodent, monkey, ape or murine intracellular costimulatory molecule.
  • a naturally occurring costimulatory molecule as described herein, e.g., a human, or other mammalian, e.g., a nonhuman species, e.g., rodent, monkey, ape or murine intracellular costimulatory molecule.
  • the costimulatory domain has at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity with SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 40, or SEQ ID NO: 44.
  • the costimulatory signaling domain has at least 70, 75, 80, 85, 90, 95, 96, 97, 98, or 99% identity with, or differs by no more than 30, 25, 20, 15, 10, 5, 4, 3, 2, or 1 amino acid residues from the corresponding residues of the entire intracellular region, or a fragment of the intracellular region which is sufficient for generation of an intracellular signal, of, a naturally occurring human costimulatory molecule, e.g., a naturally occurring human costimulatory molecule disclosed herein.
  • CARs described herein can include one or more of the components listed in Table 11.
  • the CAR-expressing cell described herein can further comprise a second CAR, e.g., a second CAR that includes a different antigen binding domain, e.g., to the same target or a different target (e.g., a target other than a cancer associated antigen described herein or a different cancer associated antigen described herein, e.g., CD19, CD33, CLL-1, CD34, FLT3, or folate receptor beta).
  • the second CAR includes an antigen binding domain to a target expressed the same cancer cell type as the cancer associated antigen.
  • the CAR-expressing cell comprises a first CAR that targets a first antigen and includes an intracellular signaling domain having a costimulatory signaling domain but not a primary signaling domain, and a second CAR that targets a second, different, antigen and includes an intracellular signaling domain having a primary signaling domain but not a costimulatory signaling domain.
  • a costimulatory signaling domain e.g., 4-1BB, CD28, ICOS, CD27 or OX-40
  • the CAR expressing cell comprises a first cancer associated antigen CAR that includes an antigen binding domain that binds a target antigen described herein, a transmembrane domain and a costimulatory domain and a second CAR that targets a different target antigen (e.g., an antigen expressed on that same cancer cell type as the first target antigen) and includes an antigen binding domain, a transmembrane domain and a primary signaling domain.
  • a target antigen e.g., an antigen expressed on that same cancer cell type as the first target antigen
  • the CAR expressing cell comprises a first CAR that includes an antigen binding domain that binds a target antigen described herein, a transmembrane domain and a primary signaling domain and a second CAR that targets an antigen other than the first target antigen (e.g., an antigen expressed on the same cancer cell type as the first target antigen) and includes an antigen binding domain to the antigen, a transmembrane domain and a costimulatory signaling domain.
  • a first CAR that includes an antigen binding domain that binds a target antigen described herein, a transmembrane domain and a primary signaling domain
  • a second CAR that targets an antigen other than the first target antigen e.g., an antigen expressed on the same cancer cell type as the first target antigen
  • the CAR-expressing cell comprises a CAR described herein (e.g., a CD19 CAR) and an inhibitory CAR.
  • the inhibitory CAR comprises an antigen binding domain that binds an antigen found on normal cells but not cancer cells, e.g., normal cells that also express CLL.
  • the inhibitory CAR comprises the antigen binding domain, a transmembrane domain and an intracellular domain of an inhibitory molecule.
  • the intracellular domain of the inhibitory CAR can be an intracellular domain of PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR (e.g., TGFRbeta).
  • CEACAM e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5
  • LAG3, VISTA e.g., VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (
  • the antigen binding domains of the different CARs can be such that the antigen binding domains do not interact with one another.
  • a cell expressing a first and second CAR can have an antigen binding domain of the first CAR, e.g., as a fragment, e.g., an scFv, that does not form an association with the antigen binding domain of the second CAR, e.g., the antigen binding domain of the second CAR is a VHH.
  • binding of the antigen binding domain of the first CAR to its cognate antigen is not substantially reduced by the presence of the second CAR. In some embodiments, binding of the antigen binding domain of the first CAR to its cognate antigen in the presence of the second CAR is 85%, 90%, 95%, 96%, 97%, 98% or 99% of binding of the antigen binding domain of the first CAR to its cognate antigen in the absence of the second CAR.
  • the antigen binding domains of the first CAR said second CAR when present on the surface of a cell, associate with one another less than if both were scFv antigen binding domains. In some embodiments, the antigen binding domains of the first CAR and the second CAR, associate with one another 85%, 90%, 95%, 96%, 97%, 98% or 99% less than if both were scFv antigen binding domains.
  • the CARs described herein are expressed on cells, e.g., immune effector cells, e.g., T cells.
  • a nucleic acid construct of a CAR described herein is transduced to a T cell.
  • the cells expressing the CARs described herein are an in vitro transcribed RNA CAR T cell.
  • a source of cells e.g., immune effector cells, e.g., T cells or NK cells
  • T cells can be obtained from a number of sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors.
  • the cells obtained as described in this section are subjected to an assay described herein, e.g., one or more biomarkers are assayed.
  • immune effector cells e.g., T cells or NK cells
  • T cells or NK cells can be obtained from a unit of blood collected from a subject using any number of techniques known to the skilled artisan, such as FicollTM separation.
  • cells from the circulating blood of an individual are obtained by apheresis.
  • the apheresis product typically contains lymphocytes, including T cells, monocytes, granulocytes, B cells, other nucleated white blood cells, red blood cells, and platelets.
  • the cells collected by apheresis may be washed to remove the plasma fraction and, optionally, to place the cells in an appropriate buffer or media for subsequent processing steps.
  • the cells are washed with phosphate buffered saline (PBS).
  • the wash solution lacks calcium and may lack magnesium or may lack many if not all divalent cations.
  • a washing step may be accomplished by methods known to those in the art, such as by using a semi-automated “flow-through” centrifuge (for example, the Cobe 2991 cell processor, the Baxter CytoMate, or the Haemonetics Cell Saver 5) according to the manufacturer's instructions.
  • a semi-automated “flow-through” centrifuge for example, the Cobe 2991 cell processor, the Baxter CytoMate, or the Haemonetics Cell Saver 5
  • the cells may be resuspended in a variety of biocompatible buffers, such as, for example, Ca-free, Mg-free PBS, PlasmaLyte A, or other saline solution with or without buffer.
  • the undesirable components of the apheresis sample may be removed and the cells directly resuspended in culture media.
  • the methods of the application can utilize culture media conditions comprising 5% or less, for example 2%, human AB serum, and employ known culture media conditions and compositions, for example those described in Smith et al., “Ex vivo expansion of human T cells for adoptive immunotherapy using the novel Xeno-free CTS Immune Cell Serum Replacement” Clinical & Translational Immunology (2015) 4, e31; doi:10.1038/cti.2014.31.
  • T cells are isolated from peripheral blood lymphocytes by lysing the red blood cells and depleting the monocytes, for example, by centrifugation through a PERCOLLTM gradient or by counterflow centrifugal elutriation.
  • the methods described herein can include, e.g., selection of a specific subpopulation of immune effector cells, e.g., T cells, that are a T regulatory cell-depleted population, CD25+ depleted cells, using, e.g., a negative selection technique, e.g., described herein.
  • the population of T regulatory depleted cells contains less than 30%, 25%, 20%, 15%, 10%, 5%, 4%, 3%, 2%, 1% of CD25+ cells.
  • T regulatory cells e.g., CD25+ T cells
  • T regulatory cells are removed from the population using an anti-CD25 antibody, or fragment thereof, or a CD25-binding ligand, e.g., IL-2.
  • the anti-CD25 antibody, or fragment thereof, or CD25-binding ligand is conjugated to a substrate, e.g., a bead, or is otherwise coated on a substrate, e.g., a bead.
  • the anti-CD25 antibody, or fragment thereof is conjugated to a substrate as described herein.
  • the T regulatory cells are removed from the population using CD25 depletion reagent from MiltenyiTM.
  • the ratio of cells to CD25 depletion reagent is 1e7 cells to 20 uL, or 1e7 cells to15 uL, or 1e7 cells to 10 uL, or 1e7 cells to 5 uL, or 1e7 cells to 2.5 uL, or 1e7 cells to 1.25 uL.
  • greater than 500 million cells/ml is used.
  • a concentration of cells of 600, 700, 800, or 900 million cells/ml is used.
  • the population of immune effector cells to be depleted includes about 6 ⁇ 10 9 CD25+ T cells. In other aspects, the population of immune effector cells to be depleted include about 1 ⁇ 10 9 to 1 ⁇ 10 10 CD25+ T cell, and any integer value in between. In one embodiment, the resulting population T regulatory depleted cells has 2 ⁇ 10 9 T regulatory cells, e.g., CD25+ cells, or less (e.g., 1 ⁇ 10 9 , 5 ⁇ 10 8 , 1 ⁇ 10 8 , 5 ⁇ 10 7 , 1 ⁇ 10 7 , or less CD25+ cells).
  • the T regulatory cells e.g., CD25+ cells
  • a depletion tubing set such as, e.g., tubing 162-01.
  • the CliniMAC system is run on a depletion setting such as, e.g., DEPLETION2.1.
  • decreasing the level of negative regulators of immune cells e.g., decreasing the number of unwanted immune cells, e.g., T REG cells
  • a patient is pre-treated with one or more therapies that reduce T REG cells prior to collection of cells for CAR-expressing cell (e.g., T cell, NK cell) product manufacturing, thereby reducing the risk of patient relapse to CAR-expressing cell (e.g., T cell, NK cell) treatment (e.g., CTL019 treatment).
  • Methods of depleting T REG cells are known in the art. Methods of decreasing T REG cells include, but are not limited to, cyclophosphamide, anti-GITR antibody, CD25-depletion, and combinations thereof.
  • the manufacturing methods comprise reducing the number of (e.g., depleting) T REG cells prior to manufacturing of the CAR-expressing cell.
  • manufacturing methods comprise contacting the sample, e.g., the apheresis sample, with an anti-GITR antibody and/or an anti-CD25 antibody (or fragment thereof, or a CD25-binding ligand), e.g., to deplete T REG cells prior to manufacturing of the CAR-expressing cell (e.g., T cell, NK cell) product.
  • a patient is pre-treated with cyclophosphamide prior to collection of cells for CAR-expressing cell (e.g., T cell, NK cell) product manufacturing, thereby reducing the risk of patient relapse to CAR-expressing cell treatment (e.g., CTL019 treatment).
  • a patient is pre-treated with an anti-GITR antibody prior to collection of cells for CAR-expressing cell (e.g., T cell, NK cell) product manufacturing, thereby reducing the risk of patient relapse to CAR-expressing cell treatment (e.g., CTL019 treatment).
  • the CAR-expressing cell (e.g., T cell, NK cell) manufacturing process is modified to deplete T REG cells prior to manufacturing of the CAR-expressing cell (e.g., T cell, NK cell) product (e.g., a CTL019 product).
  • CD25-depletion is used to deplete T REG cells prior to manufacturing of the CAR-expressing cell (e.g., T cell, NK cell) product (e.g., a CTL019 product).
  • the methods described herein can include more than one selection step, e.g., more than one depletion step.
  • Enrichment of a T cell population by negative selection can be accomplished, e.g., with a combination of antibodies directed to surface markers unique to the negatively selected cells.
  • One method is cell sorting and/or selection via negative magnetic immunoadherence or flow cytometry that uses a cocktail of monoclonal antibodies directed to cell surface markers present on the cells negatively selected.
  • a monoclonal antibody cocktail can include antibodies to CD14, CD20, CD11b, CD16, HLA-DR, and CD8.
  • the methods described herein can further include removing cells from the population which express a tumor antigen, e.g., a tumor antigen that does not comprise CD25, e.g., CD19, CD30, CD38, CD123, CD20, CD14 or CD11b, to thereby provide a population of T regulatory depleted, e.g., CD25+ depleted, and tumor antigen depleted cells that are suitable for expression of a CAR, e.g., a CAR described herein.
  • tumor antigen expressing cells are removed simultaneously with the T regulatory, e.g., CD25+ cells.
  • an anti-CD25 antibody, or fragment thereof, and an anti-tumor antigen antibody, or fragment thereof can be attached to the same substrate, e.g., bead, which can be used to remove the cells or an anti-CD25 antibody, or fragment thereof, or the anti-tumor antigen antibody, or fragment thereof, can be attached to separate beads, a mixture of which can be used to remove the cells.
  • the removal of T regulatory cells, e.g., CD25+ cells, and the removal of the tumor antigen expressing cells is sequential, and can occur, e.g., in either order.
  • a check point inhibitor e.g., a check point inhibitor described herein, e.g., one or more of PD1+ cells, LAG3+ cells, and TIM3+ cells
  • check point inhibitors include B7-H1, B&-1, CD160, P1H, 2B4, PD1, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, TIGIT, CTLA-4, BTLA and LAIR1.
  • check point inhibitor expressing cells are removed simultaneously with the T regulatory, e.g., CD25+ cells.
  • an anti-CD25 antibody, or fragment thereof, and an anti-check point inhibitor antibody, or fragment thereof can be attached to the same bead which can be used to remove the cells, or an anti-CD25 antibody, or fragment thereof, and the anti-check point inhibitor antibody, or fragment there, can be attached to separate beads, a mixture of which can be used to remove the cells.
  • the removal of T regulatory cells, e.g., CD25+ cells, and the removal of the check point inhibitor expressing cells is sequential, and can occur, e.g., in either order.
  • T cells can isolated by incubation with anti-CD3/anti-CD28 (e.g., 3 ⁇ 28)-conjugated beads, such as DYNABEADS® M-450 CD3/CD28 T, for a time period sufficient for positive selection of the desired T cells.
  • the time period is about 30 minutes.
  • the time period ranges from 30 minutes to 36 hours or longer and all integer values there between.
  • the time period is at least 1, 2, 3, 4, 5, or 6 hours.
  • the time period is 10 to 24 hours, e.g., 24 hours.
  • TIL tumor infiltrating lymphocytes
  • use of longer incubation times can increase the efficiency of capture of CD8+ T cells.
  • T cells by simply shortening or lengthening the time T cells are allowed to bind to the CD3/CD28 beads and/or by increasing or decreasing the ratio of beads to T cells (as described further herein), subpopulations of T cells can be preferentially selected for or against at culture initiation or at other time points during the process.
  • subpopulations of T cells can be preferentially selected for or against at culture initiation or at other desired time points.
  • a T cell population can be selected that expresses one or more of IFN- ⁇ , TNF ⁇ , IL-17A, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-21, CCL20,GM-CSF, IL-10, IL-13, granzyme B, and perforin, or other appropriate molecules, e.g., other cytokines.
  • Methods for screening for cell expression can be determined, e.g., by the methods described in PCT Publication No.: WO 2013/126712.
  • the T cell population expresses cytokine CCL20, IL-17a, IL-6, and combinations thereof.
  • the concentration of cells and surface can be varied.
  • it may be desirable to significantly decrease the volume in which beads and cells are mixed together e.g., increase the concentration of cells, to ensure maximum contact of cells and beads.
  • a concentration of 10 billion cells/ml, 9 billion/ml, 8 billion/ml, 7 billion/ml, 6 billion/ml, or 5 billion/ml is used.
  • a concentration of 1 billion cells/ml is used.
  • a concentration of cells from 75, 80, 85, 90, 95, or 100 million cells/ml is used.
  • concentrations of 125 or 150 million cells/ml can be used.
  • Using high concentrations can result in increased cell yield, cell activation, and cell expansion. Further, use of high cell concentrations allows more efficient capture of cells that may weakly express target antigens of interest, such as CD28-negative T cells, or from samples where there are many tumor cells present (e.g., leukemic blood, tumor tissue, etc.). Such populations of cells may have therapeutic value and would be desirable to obtain. For example, using high concentration of cells allows more efficient selection of CD8+ T cells that normally have weaker CD28 expression.
  • the concentration of cells used is 5 ⁇ 10 6 /ml. In other aspects, the concentration used can be from about 1 ⁇ 10 5 /ml to 1 ⁇ 10 6 /ml, and any integer value in between.
  • the cells may be incubated on a rotator for varying lengths of time at varying speeds at either 2-10° C. or at room temperature.
  • T cells for stimulation can also be frozen after a washing step.
  • the freeze and subsequent thaw step provides a more uniform product by removing granulocytes and to some extent monocytes in the cell population.
  • the cells may be suspended in a freezing solution.
  • one method involves using PBS containing 20% DMSO and 8% human serum albumin, or culture media containing 10% Dextran 40 and 5% Dextrose, 20% Human Serum Albumin and 7.5% DMSO, or 31.25% Plasmalyte-A, 31.25% Dextrose 5%, 0.45% NaCl, 10% Dextran 40 and 5% Dextrose, 20% Human Serum Albumin, and 7.5% DMSO or other suitable cell freezing media containing for example, Hespan and PlasmaLyte A, the cells then are frozen to ⁇ 80° C. at a rate of 1° per minute and stored in the vapor phase of a liquid nitrogen storage tank. Other methods of controlled freezing may be used as well as uncontrolled freezing immediately at ⁇ 20° C. or in liquid nitrogen.
  • cryopreserved cells are thawed and washed as described herein and allowed to rest for one hour at room temperature prior to activation using the methods of the present invention.
  • a blood sample or an apheresis product is taken from a generally healthy subject.
  • a blood sample or an apheresis is taken from a generally healthy subject who is at risk of developing a disease, but who has not yet developed a disease, and the cells of interest are isolated and frozen for later use.
  • the T cells may be expanded, frozen, and used at a later time.
  • samples are collected from a patient shortly after diagnosis of a particular disease as described herein but prior to any treatments.
  • the cells are isolated from a blood sample or an apheresis from a subject prior to any number of relevant treatment modalities, including but not limited to treatment with agents such as natalizumab, efalizumab, antiviral agents, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies, cytoxan, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, and irradiation.
  • agents such as natalizumab, efalizumab, antiviral agents, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3
  • T cells are obtained from a patient directly following treatment that leaves the subject with functional T cells.
  • the quality of T cells obtained may be optimal or improved for their ability to expand ex vivo.
  • these cells may be in a preferred state for enhanced engraftment and in vivo expansion.
  • the immune effector cells expressing a CAR molecule are obtained from a subject that has received a low, immune enhancing dose of an mTOR inhibitor.
  • the population of immune effector cells, e.g., T cells, to be engineered to express a CAR are harvested after a sufficient time, or after sufficient dosing of the low, immune enhancing, dose of an mTOR inhibitor, such that the level of PD1 negative immune effector cells, e.g., T cells, or the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells, in the subject or harvested from the subject has been, at least transiently, increased.
  • population of immune effector cells e.g., T cells, which have, or will be engineered to express a CAR
  • population of immune effector cells can be treated ex vivo by contact with an amount of an mTOR inhibitor that increases the number of PD1 negative immune effector cells, e.g., T cells or increases the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells.
  • a T cell population is Ikaros-deficient.
  • Ikaros-deficient cells include cells that do not express Ikaros RNA or protein, or have reduced or inhibited Ikaros activity, Ikaros-deficient cells can be generated by genetic approaches, e.g., administering RNA-interfering agents, e.g., siRNA, shRNA, miRNA, to reduce or prevent Ikaros expression.
  • RNA-interfering agents e.g., siRNA, shRNA, miRNA
  • Ikaros-deficient cells can be generated by treatment with Ikaros inhibitors, e.g., lenalidomide.
  • a T cell population is DGK-deficient and Ikaros-deficient, e.g., does not express DGK and Ikaros, or has reduced or inhibited DGK and Ikaros activity.
  • DGK and Ikaros-deficient cells can be generated by any of the methods described herein.
  • the immune effector cell can be an allogeneic immune effector cell, e.g., T cell.
  • the cell can be an allogeneic T cell, e.g., an allogeneic T cell lacking expression of a functional T cell receptor (TCR) and/or human leukocyte antigen (HLA), e.g., HLA class I and/or HLA class II.
  • TCR T cell receptor
  • HLA human leukocyte antigen
  • a T cell lacking a functional TCR can be, e.g., engineered such that it does not express any functional TCR on its surface, engineered such that it does not express one or more subunits that comprise a functional TCR (e.g., engineered such that it does not express (or exhibits reduced expression) of TCR alpha, TCR beta, TCR gamma, TCR delta, TCR epsilon, and/or TCR zeta) or engineered such that it produces very little functional TCR on its surface.
  • the T cell can express a substantially impaired TCR, e.g., by expression of mutated or truncated forms of one or more of the subunits of the TCR.
  • substantially impaired TCR means that this TCR will not elicit an adverse immune reaction in a host.
  • a T cell described herein can be, e.g., engineered such that it does not express a functional HLA on its surface.
  • a T cell described herein can be engineered such that cell surface expression HLA, e.g., HLA class 1 and/or HLA class II, is downregulated.
  • HLA e.g., HLA class 1 and/or HLA class II
  • downregulation of HLA may be accomplished by reducing or eliminating expression of beta-2 microglobulin (B2M).
  • the T cell can lack a functional TCR and a functional HLA, e.g., HLA class I and/or HLA class II.
  • a functional TCR e.g., HLA class I and/or HLA class II.
  • Modified T cells that lack expression of a functional TCR and/or HLA can be obtained by any suitable means, including a knock out or knock down of one or more subunit of TCR or HLA.
  • the T cell can include a knock down of TCR and/or HLA using siRNA, shRNA, clustered regularly interspaced short palindromic repeats (CRISPR) transcription-activator like effector nuclease (TALEN), or zinc finger endonuclease (ZFN).
  • siRNA siRNA
  • shRNA clustered regularly interspaced short palindromic repeats
  • CRISPR clustered regularly interspaced short palindromic repeats
  • TALEN transcription-activator like effector nuclease
  • ZFN zinc finger endonuclease
  • the allogeneic cell can be a cell which does not expresses or expresses at low levels an inhibitory molecule, e.g. by any method described herein.
  • the cell can be a cell that does not express or expresses at low levels an inhibitory molecule, e.g., that can decrease the ability of a CAR-expressing cell to mount an immune effector response.
  • inhibitory molecules include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR (e.g., TGFRbeta).
  • TCR expression and/or HLA expression can be inhibited using siRNA or shRNA that targets a nucleic acid encoding a TCR and/or HLA, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, WIC class I, WIC class II, GAL9, adenosine, and TGFR beta), in a cell, e.g., T cell.
  • siRNA or shRNA that targets a nucleic acid encoding a TCR and/or HLA, and/or an inhibitory molecule described herein (
  • siRNA and shRNAs are described, e.g., in paragraphs 649 and 650 of International Application WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.
  • CRISPR or “CRISPR to TCR and/or HLA” or “CRISPR to inhibit TCR and/or HLA” as used herein refers to a set of clustered regularly interspaced short palindromic repeats, or a system comprising such a set of repeats. “Cas”, as used herein, refers to a CRISPR-associated protein.
  • CRISPR/Cas refers to a system derived from CRISPR and Cas which can be used to silence or mutate a TCR and/or HLA gene, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MEW class II, GAL9, adenosine, and TGFR beta), in a cell, e.g., T cell.
  • an inhibitory molecule described herein e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM
  • TALEN or “TALEN to HLA and/or TCR” or “TALEN to inhibit HLA and/or TCR” refers to a transcription activator-like effector nuclease, an artificial nuclease which can be used to edit the HLA and/or TCR gene, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, WIC class II, GAL9, adenosine, and TGFR beta), in a cell, e.g., T cell.
  • TALENs and uses thereof, are described, e.g., in paragraphs 659-665 of International Application WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.
  • ZFN Zinc Finger Nuclease or “ZFN to HLA and/or TCR” or “ZFN to inhibit HLA and/or TCR” refer to a zinc finger nuclease, an artificial nuclease which can be used to edit the HLA and/or TCR gene, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, WIC class I, MHC class II, GAL9, adenosine, and TGFR beta), in a cell, e.g., T cell.
  • a therapeutic T cell has short term persistence in a patient, due to shortened telomeres in the T cell; accordingly, transfection with a telomerase gene can lengthen the telomeres of the T cell and improve persistence of the T cell in the patient.
  • an immune effector cell e.g., a T cell
  • ectopically expresses a telomerase subunit, e.g., the catalytic subunit of telomerase, e.g., TERT, e.g., hTERT.
  • this disclosure provides a method of producing a CAR-expressing cell, comprising contacting a cell with a nucleic acid encoding a telomerase subunit, e.g., the catalytic subunit of telomerase, e.g., TERT, e.g., hTERT.
  • the cell may be contacted with the nucleic acid before, simultaneous with, or after being contacted with a construct encoding a CAR.
  • the disclosure features a method of making a population of immune effector cells (e.g., T cells, NK cells).
  • the method comprises: providing a population of immune effector cells (e.g., T cells or NK cells), contacting the population of immune effector cells with a nucleic acid encoding a CAR; and contacting the population of immune effector cells with a nucleic acid encoding a telomerase subunit, e.g., hTERT, under conditions that allow for CAR and telomerase expression.
  • the nucleic acid encoding the telomerase subunit is DNA. In an embodiment, the nucleic acid encoding the telomerase subunit comprises a promoter capable of driving expression of the telomerase subunit.
  • hTERT has the amino acid sequence of GenBank Protein ID AAC51724.1 (Meyerson et al., “hEST2, the Putative Human Telomerase Catalytic Subunit Gene, Is Up-Regulated in Tumor Cells and during Immortalization” Cell Volume 90, Issue 4, 22 Aug. 1997, Pages 785-795) as set out in SEQ ID NO: 82 herein.
  • the hTERT has a sequence at least 80%, 85%, 90%, 95%, 96 ⁇ circumflex over ( ) ⁇ , 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 82. In an embodiment, the hTERT has a sequence of SEQ ID NO: 82. In an embodiment, the hTERT comprises a deletion (e.g., of no more than 5, 10, 15, 20, or 30 amino acids) at the N-terminus, the C-terminus, or both. In an embodiment, the hTERT comprises a transgenic amino acid sequence (e.g., of no more than 5, 10, 15, 20, or 30 amino acids) at the N-terminus, the C-terminus, or both.
  • the hTERT is encoded by the nucleic acid sequence of GenBank Accession No. AF018167 (Meyerson et al., “hEST2, the Putative Human Telomerase Catalytic Subunit Gene, Is Up-Regulated in Tumor Cells and during Immortalization” Cell Volume 90, Issue 4, 22 Aug. 1997, Pages 785-795) as set out in SEQ ID NO: 83 herein.
  • the hTERT is encoded by a nucleic acid having a sequence at least 80%, 85%, 90%, 95%, 96, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 83. In an embodiment, the hTERT is encoded by a nucleic acid of SEQ ID NO: 83.
  • Immune effector cells such as T cells may be activated and expanded generally using methods as described, for example, in U.S. Pat. Nos. 6,352,694; 6,534,055; 6,905,680; 6,692,964; 5,858,358; 6,887,466; 6,905,681; 7,144,575; 7,067,318; 7,172,869; 7,232,566; 7,175,843; 5,883,223; 6,905,874; 6,797,514; 6,867,041; and U.S. Patent Application Publication No. 20060121005.
  • immune effector cells are subjected to an assay as described herein (e.g., one or more biomarkers are assayed) before, during, or after activation, or before, during, or after expansion.
  • a population of immune effector cells may be expanded by contact with a surface having attached thereto an agent that stimulates a CD3/TCR complex associated signal and a ligand that stimulates a costimulatory molecule on the surface of the T cells.
  • T cell populations may be stimulated as described herein, such as by contact with an anti-CD3 antibody, or antigen-binding fragment thereof, or an anti-CD2 antibody immobilized on a surface, or by contact with a protein kinase C activator (e.g., bryostatin) in conjunction with a calcium ionophore.
  • a ligand that binds the accessory molecule is used for co-stimulation of an accessory molecule on the surface of the T cells.
  • a population of T cells can be contacted with an anti-CD3 antibody and an anti-CD28 antibody, under conditions appropriate for stimulating proliferation of the T cells.
  • an anti-CD3 antibody and an anti-CD28 antibody can be used.
  • an anti-CD28 antibody include 9.3, B-T3, XR-CD28 (Diaclone, Besancon, France) can be used as can other methods commonly known in the art (Berg et al., Transplant Proc. 30(8):3975-3977, 1998; Haanen et al., J. Exp. Med. 190(9):13191328, 1999; Garland et al., J. Immunol Meth. 227(1-2):53-63, 1999).
  • the primary stimulatory signal and the costimulatory signal for the T cell may be provided by different protocols.
  • the agents providing each signal may be in solution or coupled to a surface. When coupled to a surface, the agents may be coupled to the same surface (i.e., in “cis” formation) or to separate surfaces (i.e., in “trans” formation).
  • one agent may be coupled to a surface and the other agent in solution.
  • the agent providing the costimulatory signal is bound to a cell surface and the agent providing the primary activation signal is in solution or coupled to a surface. In certain aspects, both agents can be in solution.
  • the agents may be in soluble form, and then cross-linked to a surface, such as a cell expressing Fc receptors or an antibody or other binding agent which will bind to the agents.
  • a surface such as a cell expressing Fc receptors or an antibody or other binding agent which will bind to the agents.
  • aAPCs artificial antigen presenting cells
  • the two agents are immobilized on beads, either on the same bead, i.e., “cis,” or to separate beads, i.e., “trans.”
  • the agent providing the primary activation signal is an anti-CD3 antibody or an antigen-binding fragment thereof and the agent providing the costimulatory signal is an anti-CD28 antibody or antigen-binding fragment thereof; and both agents are co-immobilized to the same bead in equivalent molecular amounts.
  • a 1:1 ratio of each antibody bound to the beads for CD4+ T cell expansion and T cell growth is used.
  • a ratio of anti CD3:CD28 antibodies bound to the beads is used such that an increase in T cell expansion is observed as compared to the expansion observed using a ratio of 1:1. In one particular aspect an increase of from about 1 to about 3 fold is observed as compared to the expansion observed using a ratio of 1:1.
  • the ratio of CD3:CD28 antibody bound to the beads ranges from 100:1 to 1:100 and all integer values there between. In one aspect, more anti-CD28 antibody is bound to the particles than anti-CD3 antibody, i.e., the ratio of CD3:CD28 is less than one. In certain aspects, the ratio of anti CD28 antibody to anti CD3 antibody bound to the beads is greater than 2:1.
  • a 1:100 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:75 CD3:CD28 ratio of antibody bound to beads is used. In a further aspect, a 1:50 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:30 CD3:CD28 ratio of antibody bound to beads is used. In one preferred aspect, a 1:10 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:3 CD3:CD28 ratio of antibody bound to the beads is used. In yet one aspect, a 3:1 CD3:CD28 ratio of antibody bound to the beads is used.
  • Ratios of particles to cells from 1:500 to 500:1 and any integer values in between may be used to stimulate T cells or other target cells.
  • the ratio of particles to cells may depend on particle size relative to the target cell. For example, small sized beads could only bind a few cells, while larger beads could bind many.
  • the ratio of cells to particles ranges from 1:100 to 100:1 and any integer values in-between and in further aspects the ratio comprises 1:9 to 9:1 and any integer values in between, can also be used to stimulate T cells.
  • the ratio of anti-CD3- and anti-CD28-coupled particles to T cells that result in T cell stimulation can vary as noted above, however certain suitable values include 1:100, 1:50, 1:40, 1:30, 1:20, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, and 15:1 with one suitable ratio being at least 1:1 particles per T cell.
  • a ratio of particles to cells of 1:1 or less is used.
  • a suitable particle: cell ratio is 1:5.
  • the ratio of particles to cells can be varied depending on the day of stimulation.
  • the ratio of particles to cells is from 1:1 to 10:1 on the first day and additional particles are added to the cells every day or every other day thereafter for up to 10 days, at final ratios of from 1:1 to 1:10 (based on cell counts on the day of addition).
  • the ratio of particles to cells is 1:1 on the first day of stimulation and adjusted to 1:5 on the third and fifth days of stimulation.
  • particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:5 on the third and fifth days of stimulation.
  • the ratio of particles to cells is 2:1 on the first day of stimulation and adjusted to 1:10 on the third and fifth days of stimulation.
  • particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:10 on the third and fifth days of stimulation.
  • ratios will vary depending on particle size and on cell size and type.
  • the most typical ratios for use are in the neighborhood of 1:1, 2:1 and 3:1 on the first day.
  • the cells such as T cells
  • the cells are combined with agent-coated beads, the beads and the cells are subsequently separated, and then the cells are cultured.
  • the agent-coated beads and cells prior to culture, are not separated but are cultured together.
  • the beads and cells are first concentrated by application of a force, such as a magnetic force, resulting in increased ligation of cell surface markers, thereby inducing cell stimulation.
  • cell surface proteins may be ligated by allowing paramagnetic beads to which anti-CD3 and anti-CD28 are attached (3 ⁇ 28 beads) to contact the T cells.
  • the cells for example, 10 4 to 10 9 T cells
  • beads for example, DYNABEADS® M-450 CD3/CD28 T paramagnetic beads at a ratio of 1:1
  • a buffer for example PBS (without divalent cations such as, calcium and magnesium).
  • the target cell may be very rare in the sample and comprise only 0.01% of the sample or the entire sample (i.e., 100%) may comprise the target cell of interest.
  • any cell number is within the context of the present invention.
  • it may be desirable to significantly decrease the volume in which particles and cells are mixed together i.e., increase the concentration of cells, to ensure maximum contact of cells and particles.
  • a concentration of about 10 billion cells/ml, 9 billion/ml, 8 billion/ml, 7 billion/ml, 6 billion/ml, 5 billion/ml, or 2 billion cells/ml is used.
  • greater than 100 million cells/ml is used.
  • a concentration of cells of 10, 15, 20, 25, 30, 35, 40, 45, or 50 million cells/ml is used.
  • a concentration of cells from 75, 80, 85, 90, 95, or 100 million cells/ml is used. In further aspects, concentrations of 125 or 150 million cells/ml can be used. Using high concentrations can result in increased cell yield, cell activation, and cell expansion. Further, use of high cell concentrations allows more efficient capture of cells that may weakly express target antigens of interest, such as CD28-negative T cells. Such populations of cells may have therapeutic value and would be desirable to obtain in certain aspects. For example, using high concentration of cells allows more efficient selection of CD8+ T cells that normally have weaker CD28 expression.
  • cells transduced with a nucleic acid encoding a CAR are expanded, e.g., by a method described herein.
  • the cells are expanded in culture for a period of several hours (e.g., about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 18, 21 hours) to about 14 days (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days).
  • the cells are expanded for a period of 4 to 9 days.
  • the cells are expanded for a period of 8 days or less, e.g., 7, 6 or 5 days.
  • the cells are expanded in culture for 5 days, and the resulting cells are more potent than the same cells expanded in culture for 9 days under the same culture conditions. Potency can be defined, e.g., by various T cell functions, e.g. proliferation, target cell killing, cytokine production, activation, migration, or combinations thereof.
  • the cells, e.g., a CD19 CAR cell described herein, expanded for 5 days show at least a one, two, three or four fold increase in cells doublings upon antigen stimulation as compared to the same cells expanded in culture for 9 days under the same culture conditions.
  • the cells e.g., the cells expressing a CD19 CAR described herein, are expanded in culture for 5 days, and the resulting cells exhibit higher proinflammatory cytokine production, e.g., IFN- ⁇ and/or GM-CSF levels, as compared to the same cells expanded in culture for 9 days under the same culture conditions.
  • proinflammatory cytokine production e.g., IFN- ⁇ and/or GM-CSF levels
  • the cells e.g., a CD19 CAR cell described herein, expanded for 5 days show at least a one, two, three, four, five, tenfold or more increase in pg/ml of proinflammatory cytokine production, e.g., IFN- ⁇ and/or GM-CSF levels, as compared to the same cells expanded in culture for 9 days under the same culture conditions.
  • proinflammatory cytokine production e.g., IFN- ⁇ and/or GM-CSF levels
  • T cell culture includes an appropriate media (e.g., Minimal Essential Media or RPMI Media 1640 or, X-vivo 15, (Lonza)) that may contain factors necessary for proliferation and viability, including serum (e.g., fetal bovine or human serum), interleukin-2 (IL-2), insulin, IFN- ⁇ , IL-4, IL-7, GM-CSF, IL-10, IL-12, IL-15, TGF ⁇ , and TNF- ⁇ or any other additives for the growth of cells known to the skilled artisan.
  • serum e.g., fetal bovine or human serum
  • IL-2 interleukin-2
  • insulin IFN- ⁇
  • IL-4 interleukin-7
  • GM-CSF interleukin-10
  • IL-12 interleukin-12
  • TGF ⁇ TGF ⁇
  • TNF- ⁇ any other additives for the growth of cells known to the skilled artisan.
  • additives for the growth of cells include, but are not limited to, surfactant, plasmanate, and reducing agents such as N-acetyl-cysteine and 2-mercaptoethanol.
  • Media can include RPMI 1640, AIM-V, DMEM, MEM, ⁇ -MEM, F-12, X-Vivo 15, and X-Vivo 20, Optimizer, with added amino acids, sodium pyruvate, and vitamins, either serum-free or supplemented with an appropriate amount of serum (or plasma) or a defined set of hormones, and/or an amount of cytokine(s) sufficient for the growth and expansion of T cells.
  • Antibiotics e.g., penicillin and streptomycin
  • the target cells are maintained under conditions necessary to support growth, for example, an appropriate temperature (e.g., 37° C.) and atmosphere (e.g., air plus 5% CO 2 ).
  • the cells are expanded in an appropriate media (e.g., media described herein) that includes one or more interleukin that result in at least a 200-fold (e.g., 200-fold, 250-fold, 300-fold, 350-fold) increase in cells over a 14 day expansion period, e.g., as measured by a method described herein such as flow cytometry.
  • the cells are expanded in the presence IL-15 and/or IL-7 (e.g., IL-15 and IL-7).
  • the methods further comprise contacting a cell population (e.g., a cell population in which T regulatory cells, such as CD25+ T cells, have been depleted; or a cell population that has previously contacted an anti-CD25 antibody, fragment thereof, or CD25-binding ligand) with IL-15 and/or IL-7.
  • a cell population e.g., a cell population in which T regulatory cells, such as CD25+ T cells, have been depleted; or a cell population that has previously contacted an anti-CD25 antibody, fragment thereof, or CD25-binding ligand
  • the cell population e.g., that has previously contacted an anti-CD25 antibody, fragment thereof, or CD25-binding ligand
  • a CAR-expressing cell described herein is contacted with a composition comprising a interleukin-15 (IL-15) polypeptide, a interleukin-15 receptor alpha (IL-15Ra) polypeptide, or a combination of both a IL-15 polypeptide and a IL-15Ra polypeptide e.g., hetIL-15, during the manufacturing of the CAR-expressing cell, e.g., ex vivo.
  • a CAR-expressing cell described herein is contacted with a composition comprising a IL-15 polypeptide during the manufacturing of the CAR-expressing cell, e.g., ex vivo.
  • a CAR-expressing cell described herein is contacted with a composition comprising a combination of both a IL-15 polypeptide and a IL-15 Ra polypeptide during the manufacturing of the CAR-expressing cell, e.g., ex vivo.
  • a CAR-expressing cell described herein is contacted with a composition comprising hetlL-15 during the manufacturing of the CAR-expressing cell, e.g., ex vivo.
  • the CAR-expressing cell described herein is contacted with a composition comprising hetIL-15 during ex vivo expansion. In an embodiment, the CAR-expressing cell described herein is contacted with a composition comprising an IL-15 polypeptide during ex vivo expansion. In an embodiment, the CAR-expressing cell described herein is contacted with a composition comprising both an IL-15 polypeptide and an IL-15Ra polypeptide during ex vivo expansion. In one embodiment the contacting results in the survival and proliferation of a lymphocyte subpopulation, e.g., CD8+ T cells.
  • a lymphocyte subpopulation e.g., CD8+ T cells.
  • the cells are cultured (e.g., expanded, simulated, and/or transduced) in media comprising serum.
  • the serum may be, e.g., human AB serum (hAB).
  • hAB serum is present at about 2%, about 5%, about 2-3%, about 3-4%, about 4-5%, or about 2-5%.
  • 2% and 5% serum are each suitable levels that allow for many fold expansion of T cells.
  • T cells that have been exposed to varied stimulation times may exhibit different characteristics.
  • typical blood or apheresed peripheral blood mononuclear cell products have a helper T cell population (TH, CD4+) that is greater than the cytotoxic or suppressor T cell population (TC, CD8+).
  • TH, CD4+ helper T cell population
  • TC cytotoxic or suppressor T cell population
  • Ex vivo expansion of T cells by stimulating CD3 and CD28 receptors produces a population of T cells that prior to about days 8-9 consists predominately of TH cells, while after about days 8-9, the population of T cells comprises an increasingly greater population of TC cells.
  • infusing a subject with a T cell population comprising predominately of TH cells may be advantageous.
  • an antigen-specific subset of TC cells has been isolated it may be beneficial to expand this subset to a greater degree.
  • CD4 and CD8 markers vary significantly, but in large part, reproducibly during the course of the cell expansion process. Thus, such reproducibility enables the ability to tailor an activated T cell product for specific purposes.
  • cells transduced with a nucleic acid encoding a CAR can be selected for administration based upon, e.g., protein expression levels of one or more of CCL20, GM-CSF, IFN ⁇ , IL-10, IL-13, IL-17a, IL-2, IL-21, IL-4, IL-5, IL-6, IL-9, TNF ⁇ and/or combinations thereof.
  • cells transduced with a nucleic acid encoding a CAR can be selected for administration based upon, e.g., protein expression levels of CCL20, IL-17a, IL-6 and combinations thereof.
  • a CAR described herein is constructed, various assays can be used to evaluate the activity of the molecule, such as but not limited to, the ability to expand T cells following antigen stimulation, sustain T cell expansion in the absence of re-stimulation, and anti-cancer activities in appropriate in vitro and animal models. Assays to evaluate the effects of a CAR are described in further detail below.
  • CAR + T cells following antigen stimulation can be measured by flow cytometry.
  • a mixture of CD4 + and CD8 + T cells are stimulated with ⁇ CD3/ ⁇ CD28 aAPCs followed by transduction with lentiviral vectors expressing GFP under the control of the promoters to be analyzed.
  • exemplary promoters include the CMV IE gene, EF-1 ⁇ , ubiquitin C, or phosphoglycerokinase (PGK) promoters.
  • GFP fluorescence is evaluated on day 6 of culture in the CD4 + and/or CD8 + T cell subsets by flow cytometry.
  • CD4 + and CD8 + T cells are stimulated with ⁇ CD3/ ⁇ CD28 coated magnetic beads on day 0, and transduced with CAR on day 1 using a bicistronic lentiviral vector expressing CAR along with eGFP using a 2A ribosomal skipping sequence.
  • Cultures are re-stimulated with either a cancer associate antigen as described herein + K562 cells (K562-a cancer associate antigen as described herein), wild-type K562 cells (K562 wild type) or K562 cells expressing hCD32 and 4-1BBL in the presence of antiCD3 and anti-CD28 antibody (K562-BBL-3/28) following washing.
  • Exogenous IL-2 is added to the cultures every other day at 100 IU/ml.
  • GFP + T cells are enumerated by flow cytometry using bead-based counting. See, e.g., Milone et al., M OLECULAR T HERAPY 17(8): 1453-1464 (2009).
  • Sustained CAR + T cell expansion in the absence of re-stimulation can also be measured. See, e.g., Milone et al., M OLECULAR T HERAPY 17(8): 1453-1464 (2009). Briefly, mean T cell volume (fl) is measured on day 8 of culture using a Coulter Multisizer III particle counter, a Nexcelom Cellometer Vision or Millipore Scepter, following stimulation with ⁇ CD3/ ⁇ CD28 coated magnetic beads on day 0, and transduction with the indicated CAR on day 1.
  • Animal models can also be used to measure a CAR-expressing cell (e.g., T cell, NK cell) activity, e.g., as described in paragraph 698 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • a CAR-expressing cell e.g., T cell, NK cell
  • Dose dependent CAR treatment response can be evaluated, e.g., as described in paragraph 699 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • potency of a cell (e.g., T cell, NK cell) population e.g. a CAR-expressing cell) product, e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) cell product, e.g., CTL019 cells
  • a cell population e.g. a CAR-expressing cell
  • a CD19 CAR-expressing cell e.g., T cell, NK cell
  • CTL019 cells e.g., CTL019 cells
  • Cell (e.g., T cell, NK cell) populations e.g, a manufactured CAR-expressing cell) cell product, e.g., a CD19 CAR-expressing cell product, e.g., CTL019 cells
  • CD19-expressing K562 (K562-19) cells which mimic CD19-expressing B cells in CLL.
  • cytokine expression profiles are measured in the co-cultured cell media and potency of activated cells (e.g., a CAR-expressing cell product, e.g., a CD19 CAR-expressing cell product, e.g., CTL019 cells) is correlated with expression of different cytokines including, but not limited to CCL-20/MIP-3a, GM-CSF, IFN ⁇ , IL-10, IL-13, IL-17a, IL-2, IL-21, IL-4, IL-5, IL-6, IL-9, TNF ⁇ and/or combinations thereof.
  • activated cells e.g., a CAR-expressing cell product, e.g., a CD19 CAR-expressing cell product, e.g., CTL019 cells
  • cytokines including, but not limited to CCL-20/MIP-3a, GM-CSF, IFN ⁇ , IL-10, IL-13, IL-17a, IL-2, IL-21, IL-4,
  • cytokine expression levels are informative with regards to the potency of a cell (e.g., T cell, NK cell) population (e.g., to kill tumor cells).
  • cytokine expression levels described herein are used to improve a cell (e.g., T cell, NK cell) population (e.g., a CAR-expressing cell product, e.g., a CD 19 CAR-expressing cell product, e.g., CTL019 cells) prior to infusion in patients.
  • cytokine expression levels described herein provide an endpoint during optimization of the manufacturing process.
  • Cytotoxicity can be assessed by a standard 51Cr-release assay, e.g., as described in paragraph 701 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • Imaging technologies can be used to evaluate specific trafficking and proliferation of CARs in tumor-bearing animal models, e.g., as described in paragraph 702 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • the CAR ligand is an antibody that binds to the CAR molecule, e.g., binds to the extracellular antigen binding domain of CAR (e.g., an antibody that binds to the antigen binding domain, e.g., an anti-idiotypic antibody; or an antibody that binds to a constant region of the extracellular binding domain).
  • the CAR ligand is a CAR antigen molecule (e.g., a CAR antigen molecule as described herein).
  • a method for detecting and/or quantifying CAR-expressing cells is disclosed.
  • the CAR ligand can be used to detect and/or quantify CAR-expressing cells in vitro or in vivo (e.g., clinical monitoring of CAR-expressing cells in a patient, or dosing a patient).
  • the method includes:
  • CAR ligand (optionally, a labelled CAR ligand, e.g., a CAR ligand that includes a tag, a bead, a radioactive or fluorescent label);
  • acquiring the CAR-expressing cell e.g., acquiring a sample containing CAR-expressing cells, such as a manufacturing sample or a clinical sample
  • binding of the CAR-expressing cell with the CAR ligand can be detected using standard techniques such as FACS, ELISA and the like.
  • a method of expanding and/or activating cells e.g., immune effector cells.
  • the method includes:
  • a CAR-expressing cell e.g., a first CAR-expressing cell or a transiently expressing CAR cell
  • a CAR ligand e.g., a CAR ligand as described herein
  • a CAR ligand e.g., a CAR ligand as described herein
  • the CAR ligand is present on (e.g., is immobilized or attached to a substrate, e.g., a non-naturally occurring substrate).
  • the substrate is a non-cellular substrate.
  • the non-cellular substrate can be a solid support chosen from, e.g., a plate (e.g., a microtiter plate), a membrane (e.g., a nitrocellulose membrane), a matrix, a chip or a bead.
  • the CAR ligand is present in the substrate (e.g., on the substrate surface).
  • the CAR ligand can be immobilized, attached, or associated covalently or non-covalently (e.g., cross-linked) to the substrate.
  • the CAR ligand is attached (e.g., covalently attached) to a bead.
  • the immune cell population can be expanded in vitro or ex vivo.
  • the method can further include culturing the population of immune cells in the presence of the ligand of the CAR molecule, e.g., using any of the methods described herein.
  • the method of expanding and/or activating the cells further comprises addition of a second stimulatory molecule, e.g., CD28.
  • a second stimulatory molecule e.g., CD28.
  • the CAR ligand and the second stimulatory molecule can be immobilized to a substrate, e.g., one or more beads, thereby providing increased cell expansion and/or activation.
  • a method for selecting or enriching for a CAR expressing cell includes contacting the CAR expressing cell with a CAR ligand as described herein; and selecting the cell on the basis of binding of the CAR ligand.
  • a method for depleting, reducing and/or killing a CAR expressing cell includes contacting the CAR expressing cell with a CAR ligand as described herein; and targeting the cell on the basis of binding of the CAR ligand, thereby reducing the number, and/or killing, the CAR-expressing cell.
  • the CAR ligand is coupled to a toxic agent (e.g., a toxin or a cell ablative drug).
  • the anti-idiotypic antibody can cause effector cell activity, e.g., ADCC or ADC activities.
  • anti-CAR antibodies that can be used in the methods disclosed herein are described, e.g., in WO 2014/190273 and by Jena et al., “Chimeric Antigen Receptor (CAR)-Specific Monoclonal Antibody to Detect CD19-Specific T cells in Clinical Trials”, PLOS March 2013 8:3 e57838, the contents of which are incorporated by reference.
  • CAR Chimeric Antigen Receptor
  • compositions and methods herein are optimized for a specific subset of T cells, e.g., as described in US Serial No. PCT/US2015/043219 filed Jul. 31, 2015, the contents of which are incorporated herein by reference in their entirety.
  • the optimized subsets of T cells display an enhanced persistence compared to a control T cell, e.g., a T cell of a different type (e.g., CD8 + or CD4 + ) expressing the same construct.
  • a CD4 + T cell comprises a CAR described herein, which CAR comprises an intracellular signaling domain suitable for (e.g., optimized for, e.g., leading to enhanced persistence in) a CD4 + T cell, e.g., an ICOS domain.
  • a CD8 + T cell comprises a CAR described herein, which CAR comprises an intracellular signaling domain suitable for (e.g., optimized for, e.g., leading to enhanced persistence of) a CD8 + T cell, e.g., a 4-1BB domain, a CD28 domain, or another costimulatory domain other than an ICOS domain.
  • the CAR described herein comprises an antigen binding domain described herein, e.g., a CAR comprising an antigen binding domain.
  • a method of treating a subject e.g., a subject having cancer.
  • the method includes administering to said subject, an effective amount of:
  • an antigen binding domain e.g., an antigen binding domain described herein;
  • an intracellular signaling domain e.g., a first costimulatory domain, e.g., an ICOS domain
  • a first costimulatory domain e.g., an ICOS domain
  • an antigen binding domain e.g., an antigen binding domain described herein;
  • an intracellular signaling domain e.g., a second costimulatory domain, e.g., a 4-1BB domain, a CD28 domain, or another costimulatory domain other than an ICOS domain;
  • a second costimulatory domain e.g., a 4-1BB domain, a CD28 domain, or another costimulatory domain other than an ICOS domain;
  • the method further includes administering:
  • an antigen binding domain e.g., an antigen binding domain described herein;
  • the second CAR CD8+ comprises an intracellular signaling domain, e.g., a costimulatory signaling domain, not present on the CAR CD8+ , and, optionally, does not comprise an ICOS signaling domain.
  • RNA CAR Disclosed herein are methods for producing an in vitro transcribed RNA CAR.
  • RNA CAR and methods of using the same are described, e.g., in paragraphs 553-570 of in International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • RNA can be introduced into target cells using any of a number of different methods, for instance, commercially available methods which include, but are not limited to, electroporation, the Gene Pulser II, Multiporator, cationic liposome mediated transfection using lipofection, polymer encapsulation, peptide mediated transfection, or biolistic particle delivery systems such as “gene guns”.
  • non-viral methods can be used to deliver a nucleic acid encoding a CAR described herein into a cell or tissue or a subject.
  • Suitable non-viral delivery methods include transposons (e.g., Sleeping Beauty, piggyBac, and pT2-based transposons). Exemplary non-viral delivery methods and methods of using the same are described, e.g., in paragraphs 571-579 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • methods of manufacturing a CAR-expressing cell according to the invention are disclosed herein (e.g., in “Source of Cells” and “Activation and Expansion of Cells”).
  • a method of manufacturing a CAR-expressing cell comprises:
  • provided methods comprise steps of providing a CAR-expressing cell (e.g., T cell, NK cell) preparation (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019);
  • a CAR-expressing cell e.g., T cell, NK cell
  • a CD19 CAR-expressing cell e.g., T cell, NK cell
  • CTL019 e.g., CTL019
  • provided methods comprise acquiring a value for the level (e.g., determining the expression level) of a cytokine, e.g., one or more cytokines listed in Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), and Table 17, Table 18, Table 20, secreted by CAR-expressing cells (e.g., T cell, NK cell) in response to antigen recognition.
  • a cytokine e.g., one or more cytokines listed in Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), and Table 17, Table 18, Table 20, secreted by CAR-expressing cells (e.g., T cell, NK cell) in response to antigen recognition.
  • provided methods comprise determining the expression levels of one or more cytokines CCL20/MIP3a, IL-17a, IL-6 and/or combinations thereof, secreted by CAR-expressing cells (e.g., T cell,
  • provided methods further comprise integration of cytokines secreted by CAR-expressing cells (e.g., T cells, NK cells), e.g., one or more cytokines listed in Table 14, Table 15 and Table 16, in a potency assay.
  • CAR-expressing cells e.g., T cells, NK cells
  • provided methods further comprise integration of cytokines CCL20/MIP3a, IL-17a, IL-6 and/or combinations thereof, in a potency assay.
  • provided methods comprise integration of cytokines secreted by CAR-expressing cells (e.g., T cells, NK cells), e.g., one or more cytokines listed in Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), and Table 17 in a potency assay, and determining whether a CAR-expressing cell (e.g., T cell, NK cell) preparation (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019) may have a clinical effect.
  • CAR-expressing cells e.g., T cells, NK cells
  • CCL20/MIP3a, IL-17a, IL-6 and/or combinations thereof are used in a potency assay to determine whether a CAR-expressing cell (e.g., T cell, NK cell) preparation (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019) may have a clinical effect.
  • a CAR-expressing cell e.g., T cell, NK cell
  • provided methods further comprise adjusting the CAR-expressing cell (e.g., T cell, NK cell) infusion dose to achieve clinical efficacy.
  • provided methods comprise a step of providing a blood sample, e.g., a T cell sample, from a subject having cancer.
  • provided methods further comprise a step of comparing the obtained gene expression pattern difference with that of a reference sample.
  • a reference sample is a CAR-expressing cell (e.g., T cell, NK cell) preparation (e.g., a CD19 CAR-expressing cell as described herein, such as, e.g., CTL019) from a different batch of cells producing the therapeutic CAR-expressing cell preparation.
  • a CAR-expressing cell e.g., T cell, NK cell
  • a reference sample is a healthy donor sample with a manufactured CAR-expressing cell (e.g., T cell, NK cell) product (e.g., a CD19 CAR-expressing cell as described herein, such as, e.g., CTL019).
  • a reference sample is a healthy donor sample with a manufactured CD19 CAR-expressing cell product, such as, e.g., CTL019 product.
  • the determined difference is compared with a historical record of the reference sample.
  • the CAR-expressing cell (e.g., T cell, NK cell) preparation is a CD19 CAR-expressing cell (e.g., CTL019) preparation.
  • the CAR-expressing cell (e.g., T cell, NK cell) preparation comprises a CD19 CAR-expressing cell (e.g., CTL019) preparation.
  • the CAR-expressing cell (e.g., T cell, NK cell) preparation consists of a CD19 CAR-expressing cell (e.g., CTL019) preparation.
  • a method comprising:
  • a blood sample e.g., a T cell sample
  • a subject having cancer e.g., a blood sample obtained from a subject having cancer
  • Table 14 e.g., CCL20, IL-17a and/or IL-6
  • Table 17 Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, or KLRG1 to obtain a gene expression pattern for the sample;
  • the methods disclosed herein further include administering a T cell depleting agent after treatment with the cell (e.g., an immune effector cell as described herein), thereby reducing (e.g., depleting) the CAR-expressing cells (e.g., the CD19CAR-expressing cells).
  • a T cell depleting agent after treatment with the cell (e.g., an immune effector cell as described herein), thereby reducing (e.g., depleting) the CAR-expressing cells (e.g., the CD19CAR-expressing cells).
  • T cell depleting agents can be used to effectively deplete CAR-expressing cells (e.g., CD19CAR-expressing cells) to mitigate toxicity.
  • the CAR-expressing cells were manufactured according to a method herein, e.g., assayed (e.g., before or after transfection or transduction) according to a method herein.
  • the T cell depleting agent is administered one, two, three, four, or five weeks after administration of the cell, e.g., the population of immune effector cells, described herein.
  • target proteins include, but are not limited to, EpCAM, VEGFR, integrins (e.g., integrins ⁇ v ⁇ 3, ⁇ 4, ⁇ I3/4 ⁇ 3, ⁇ 4 ⁇ 7, ⁇ 5 ⁇ 1, ⁇ v ⁇ 3, ⁇ v), members of the TNF receptor superfamily (e.g., TRAIL-R1, TRAIL-R2), PDGF Receptor, interferon receptor, folate receptor, GPNMB, ICAM-1, HLA-DR, CEA, CA-125, MUC1, TAG-72, IL-6 receptor, 5T4, GD2, GD3, CD2, CD3, CD4, CD5, CD11, CD11a/LFA-1, CD15, CD18/ITGB2, CD19, CD20, CD22, CD23/1gE Receptor, CD25, CD28, CD30, CD33, CD38, CD40, CD41, CD44, CD51, CD52, CD62L, CD74, CD80, CD125, CD147/basigin, CD152/CTLA
  • the T cell depleting agent is a CD52 inhibitor, e.g., an anti-CD52 antibody molecule, e.g., alemtuzumab.
  • the cell e.g., the population of immune effector cells, expresses a CAR molecule as described herein (e.g., CD19CAR) and the target protein recognized by the T cell depleting agent.
  • the target protein is CD20.
  • the T cell depleting agent is an anti-CD20 antibody, e.g., rituximab.
  • the methods further include transplanting a cell, e.g., a hematopoietic stem cell, or a bone marrow, into the mammal.
  • a cell e.g., a hematopoietic stem cell, or a bone marrow
  • the invention features a method of conditioning a mammal prior to cell transplantation.
  • the method includes administering to the mammal an effective amount of the cell comprising a CAR nucleic acid or polypeptide, e.g., a CD19 CAR nucleic acid or polypeptide.
  • the cell transplantation is a stem cell transplantation, e.g., a hematopoietic stem cell transplantation, or a bone marrow transplantation.
  • conditioning a subject prior to cell transplantation includes reducing the number of target-expressing cells in a subject, e.g., CD19-expressing normal cells or CD19-expressing cancer cells.
  • Nucleic acid molecules encoding one or more CAR constructs can be introduced into an immune effector cell (e.g., a T cell) as described herein.
  • the nucleic acid molecule is provided as a messenger RNA transcript.
  • the nucleic acid molecule is provided as a DNA construct.
  • a nucleic acid described herein is introduced into a cell that has been assayed by a method described herein, e.g., one or more biomarkers has been assayed.
  • a cell comprising a nucleic acid described herein is assayed by a method described herein, e.g., one or more biomarkers has been assayed.
  • the nucleic acid molecules described herein can be a DNA molecule, an RNA molecule, or a combination thereof.
  • the nucleic acid molecule is an mRNA encoding a CAR polypeptide as described herein.
  • the nucleic acid molecule is a vector that includes any of the aforesaid nucleic acid molecules.
  • Nucleic acid molecules can encode, e.g., a CAR molecule described herein, and can comprise, e.g., a nucleic acid sequence described herein, e.g., in Table 11, Table 12 or Table 13.
  • nucleic acid sequences coding for the desired molecules can be obtained using recombinant methods known in the art, such as, for example by screening libraries from cells expressing the gene, by deriving the gene from a vector known to include the same, or by isolating directly from cells and tissues containing the same, using standard techniques.
  • the gene of interest can be produced synthetically, rather than cloned.
  • vectors in which a nucleic acid of the present disclosure is inserted are suitable tools to achieve long-term gene transfer since they allow long-term, stable integration of a transgene and its propagation in daughter cells.
  • Lentiviral vectors have the added advantage over vectors derived from onco-retroviruses such as murine leukemia viruses in that they can transduce non-proliferating cells, such as hepatocytes. They also have the added advantage of low immunogenicity.
  • a retroviral vector may also be, e.g., a gammaretroviral vector.
  • a gammaretroviral vector may include, e.g., a promoter, a packaging signal (w), a primer binding site (PBS), one or more (e.g., two) long terminal repeats (LTR), and a transgene of interest, e.g., a gene encoding a CAR.
  • a gammaretroviral vector may lack viral structural gens such as gag, pol, and env.
  • Exemplary gammaretroviral vectors include Murine Leukemia Virus (MLV), Spleen-Focus Forming Virus (SFFV), and Myeloproliferative Sarcoma Virus (MPSV), and vectors derived therefrom.
  • gammaretroviral vectors are described, e.g., in Tobias Maetzig et al., “Gammaretroviral Vectors: Biology, Technology and Application” Viruses. 2011 June; 3(6): 677-713.
  • the vector comprising the nucleic acid encoding the desired CAR of the invention is an adenoviral vector (A5/35).
  • the expression of nucleic acids encoding CARs can be accomplished using of transposons such as sleeping beauty, CRISPR, CAS9, and zinc finger nucleases. See below June et al. 2009 NATURE REVIEWS IMMUNOLOGY 9.10: 704-716, is incorporated herein by reference.
  • the expression of natural or synthetic nucleic acids encoding CARs is typically achieved by operably linking a nucleic acid encoding the CAR polypeptide or portions thereof to a promoter, and incorporating the construct into an expression vector.
  • the vectors can be suitable for replication and integration eukaryotes.
  • Typical cloning vectors contain transcription and translation terminators, initiation sequences, and promoters useful for regulation of the expression of the desired nucleic acid sequence.
  • the expression constructs may also be used for nucleic acid immunization and gene therapy, using standard gene delivery protocols. Methods for gene delivery are known in the art. See, e.g., U.S. Pat. Nos. 5,399,346, 5,580,859, 5,589,466, incorporated by reference herein in their entireties.
  • the invention provides a gene therapy vector.
  • the nucleic acid can be cloned into a number of types of vectors.
  • the nucleic acid can be cloned into a vector including, but not limited to a plasmid, a phagemid, a phage derivative, an animal virus, and a cosmid.
  • Vectors of particular interest include expression vectors, replication vectors, probe generation vectors, and sequencing vectors.
  • the expression vector may be provided to a cell in the form of a viral vector.
  • Viral vector technology is well known in the art and is described, for example, in Sambrook et al., 2012, MOLECULAR CLONING: A LABORATORY MANUAL, volumes 1-4, Cold Spring Harbor Press, NY), and in other virology and molecular biology manuals.
  • Viruses, which are useful as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, and lentiviruses.
  • a suitable vector contains an origin of replication functional in at least one organism, a promoter sequence, convenient restriction endonuclease sites, and one or more selectable markers, (e.g., WO 01/96584; WO 01/29058; and U.S. Pat. No. 6,326,193).
  • retroviruses provide a convenient platform for gene delivery systems.
  • a selected gene can be inserted into a vector and packaged in retroviral particles using techniques known in the art.
  • the recombinant virus can then be isolated and delivered to cells of the subject either in vivo or ex vivo.
  • retroviral systems are known in the art.
  • adenovirus vectors are used.
  • a number of adenovirus vectors are known in the art.
  • lentivirus vectors are used.
  • promoter elements regulate the frequency of transcriptional initiation.
  • these are located in the region 30-110 bp upstream of the start site, although a number of promoters have been shown to contain functional elements downstream of the start site as well.
  • the spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another.
  • tk thymidine kinase
  • the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline.
  • individual elements can function either cooperatively or independently to activate transcription.
  • Exemplary promoters include the CMV IE gene, EF-1 ⁇ , ubiquitin C, or phosphoglycerokinase (PGK) promoters.
  • a promoter that is capable of expressing a CAR transgene in a mammalian T cell
  • the native EF1a promoter drives expression of the alpha subunit of the elongation factor-1 complex, which is responsible for the enzymatic delivery of aminoacyl tRNAs to the ribosome.
  • the EF1a promoter has been extensively used in mammalian expression plasmids and has been shown to be effective in driving CAR expression from transgenes cloned into a lentiviral vector. See, e.g., Milone et al., M OL . T HER . 17(8): 1453-1464 (2009).
  • the EF1a promoter comprises the sequence provided as SEQ ID NO:11.
  • CMV immediate early cytomegalovirus
  • This promoter sequence is a strong constitutive promoter sequence capable of driving high levels of expression of any polynucleotide sequence operatively linked thereto.
  • other constitutive promoter sequences may also be used, including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, an avian leukemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the elongation factor-1a promoter, the hemoglobin promoter, and the creatine kinase promoter.
  • SV40 simian virus 40
  • MMTV mouse mammary tumor virus
  • HSV human immunodeficiency virus
  • inducible promoters are also contemplated as part of the invention.
  • the use of an inducible promoter provides a molecular switch capable of turning on expression of the polynucleotide sequence which it is operatively linked when such expression is desired, or turning off the expression when expression is not desired.
  • inducible promoters include, but are not limited to a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline promoter.
  • a promoter is the phosphoglycerate kinase (PGK) promoter.
  • PGK phosphoglycerate kinase
  • a truncated PGK promoter e.g., a PGK promoter with one or more, e.g., 1, 2, 5, 10, 100, 200, 300, or 400, nucleotide deletions when compared to the wild-type PGK promoter sequence
  • PGK phosphoglycerate kinase
  • a vector may also include, e.g., a signal sequence to facilitate secretion, a polyadenylation signal and transcription terminator (e.g., from Bovine Growth Hormone (BGH) gene), an element allowing episomal replication and replication in prokaryotes (e.g. SV40 origin and ColE1 or others known in the art) and/or elements to allow selection (e.g., ampicillin resistance gene and/or zeocin marker).
  • BGH Bovine Growth Hormone
  • the expression vector to be introduced into a cell can also contain either a selectable marker gene or a reporter gene or both to facilitate identification and selection of expressing cells from the population of cells sought to be transfected or infected through viral vectors.
  • the selectable marker may be carried on a separate piece of DNA and used in a co-transfection procedure. Both selectable markers and reporter genes may be flanked with appropriate regulatory sequences to enable expression in the host cells.
  • Useful selectable markers include, for example, antibiotic-resistance genes, such as neo and the like.
  • Reporter genes are used for identifying potentially transfected cells and for evaluating the functionality of regulatory sequences. Reporter genes are described, e.g., in paragraph 599 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • the vector may comprise two or more nucleic acid sequences encoding a CAR, e.g., a CAR described herein, e.g., a CD19 CAR, and a second CAR, e.g., an inhibitory CAR or a CAR that specifically binds to an antigen other than CD19.
  • the two or more nucleic acid sequences encoding the CAR are encoded by a single nucleic molecule in the same frame and as a single polypeptide chain.
  • the two or more CARs can, e.g., be separated by one or more peptide cleavage sites. (e.g., an auto-cleavage site or a substrate for an intracellular protease). Examples of peptide cleavage sites include T2A, P2A, E2A, or F2A sites.
  • the vector can be readily introduced into a host cell, e.g., mammalian, bacterial, yeast, or insect cell by any method in the art.
  • the expression vector can be transferred into a host cell by physical, chemical, or biological means, e.g., those described in paragraphs 601-603 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • an exemplary delivery vehicle is a liposome.
  • lipid formulations is contemplated for the introduction of the nucleic acids into a host cell (in vitro, ex vivo or in vivo), and is described, e.g., in paragraphs 604-605 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • assays include, for example, “molecular biological” assays well known to those of skill in the art, such as Southern and Northern blotting, RT-PCR and PCR; “biochemical” assays, such as detecting the presence or absence of a particular peptide, e.g., by immunological means (ELISAs and Western blots) or by assays described herein to identify agents falling within the scope of the invention.
  • molecular biological assays well known to those of skill in the art, such as Southern and Northern blotting, RT-PCR and PCR
  • biochemical assays such as detecting the presence or absence of a particular peptide, e.g., by immunological means (ELISAs and Western blots) or by assays described herein to identify agents falling within the scope of the invention.
  • the disclosure provides methods for treating a disease associated with expression of a tumor antigen described herein.
  • immune effector cells are assayed by a method described herein, e.g., one or more biomarkers is assayed, and the cells are administered to a subject as part of a treatment described herein.
  • the immune effector cells can be administered as part of a combination therapy described herein.
  • the present disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CAR.
  • cancer e.g., a hematological cancer such as ALL and CLL
  • immune effector cells e.g., T cells, NK cells
  • the cancer to be treated is a B cell malignancy.
  • the cancer to be treated is ALL (acute lymphoblastic leukemia), CLL (chronic lymphocytic leukemia), DLBCL (diffuse large B-cell lymphoma), MCL (Mantle cell lymphoma, or MM (multiple myeloma).
  • the disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CD19 CAR, wherein the cancer cells express CD19.
  • cancer e.g., a hematological cancer such as ALL and CLL
  • immune effector cells e.g., T cells, NK cells
  • the cancer to be treated is a B cell malignancy.
  • the cancer to be treated is ALL (acute lymphoblastic leukemia), CLL (chronic lymphocytic leukemia), DLBCL (diffuse large B-cell lymphoma), MCL (Mantle cell lymphoma), Hodgkin lymphoma, or MM (multiple myeloma).
  • the present invention provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CD22 CAR, wherein the cancer cells express CD22.
  • cancer e.g., a hematological cancer such as ALL and CLL
  • immune effector cells e.g., T cells, NK cells
  • CD22 CAR e.g., CD22 CAR
  • the cancer to be treated is a B cell malignancy.
  • the cancer to be treated is ALL (acute lymphoblastic leukemia), CLL (chronic lymphocytic leukemia), DLBCL (diffuse large B-cell lymphoma), MCL (Mantle cell lymphoma), Hodgkin lymphoma, or MM (multiple myeloma).
  • the present invention provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CD20 CAR, wherein the cancer cells express CD20.
  • cancer e.g., a hematological cancer such as ALL and CLL
  • immune effector cells e.g., T cells, NK cells
  • CD20 CAR e.g., CD20 CAR
  • the cancer to be treated is a B cell malignancy.
  • the cancer to be treated is ALL (acute lymphoblastic leukemia), CLL (chronic lymphocytic leukemia), DLBCL (diffuse large B-cell lymphoma), MCL (Mantle cell lymphoma), Hodgkin lymphoma, or MM (multiple myeloma).
  • the present invention provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a ROR1 CAR, wherein the cancer cells express ROR1.
  • cancer e.g., a hematological cancer such as ALL and CLL
  • immune effector cells e.g., T cells, NK cells
  • the cancer to be treated is a B cell malignancy.
  • the cancer to be treated is ALL (acute lymphoblastic leukemia), CLL (chronic lymphocytic leukemia), DLBCL (diffuse large B-cell lymphoma), MCL (Mantle cell lymphoma), Hodgkin lymphoma, or MM (multiple myeloma).
  • the disclosure includes a type of cellular therapy where immune effector cells (e.g., T cells, NK cells) are genetically modified (e.g., via transduction of a lentiviral vector) to express a CAR and the CAR-expressing cell is infused to a recipient in need thereof.
  • the infused cell is able to kill tumor cells in the recipient.
  • CAR-modified immune effector cells e.g., T cells, NK cells
  • CAR-expressing cells e.g., T cells or NK cells
  • generated using lentiviral vectors will have stable CAR expression.
  • the immune effector cells e.g., T cells, NK cells
  • the immune effector cells persist in the patient for at least four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, twelve months, thirteen months, fourteen month, fifteen months, sixteen months, seventeen months, eighteen months, nineteen months, twenty months, twenty-one months, twenty-two months, twenty-three months, two years, three years, four years, or five years after administration of the T cell to the patient.
  • the invention also includes a type of cellular therapy where immune effector cells (e.g., T cells, NK cells) are modified, e.g., by in vitro transcribed RNA, to transiently express a CAR and the CAR-expressing cell is infused to a recipient in need thereof.
  • CAR-expressing cells e.g., T cells, NK cells
  • CAR RNA e.g., by transfection or electroporation
  • the infused cell is able to kill tumor cells in the recipient.
  • the immune effector cells (e.g., T cells, NK cells) administered to the patient is present for less than one month, e.g., three weeks, two weeks, one week, after administration of the T cell to the patient.
  • the present disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CAR, e.g., a CAR described herein.
  • cancer e.g., a hematological cancer such as ALL and CLL
  • immune effector cells e.g., T cells, NK cells
  • the present disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CAR that specifically targets or binds to a tumor antigen (or cancer associated antigen) described herein, wherein the subject has been identified as a responder or partial responder.
  • cancer e.g., a hematological cancer such as ALL and CLL
  • immune effector cells e.g., T cells, NK cells
  • the methods provide treating a cancer (e.g., a hematological cancer such as ALL and CLL) as a partial responder or non-responder by providing to the subject a cancer therapy other than a CAR therapy, e.g., providing the subject a treatment that is the standard of care for that particular type of cancer.
  • the method of treatment includes altering the manufacturing of a CAR-expressing cell to enrich for na ⁇ ve T cells, e.g., as described herein, for a subject identified as a partial responder or non-responder prior to administering a CAR-expressing cell, e.g., a CAR-expressing cell described herein.
  • the immune effector cells are engineered to express CD19 CAR, for treating a subject having cancer (e.g., a hematological cancer such as ALL and CLL), wherein the cancer cells express CD19.
  • a subject having cancer e.g., a hematological cancer such as ALL and CLL
  • the cancer to be treated is ALL or CLL.
  • the CD19 CAR molecules to be expressed in an immune effector cell can comprise any anti-CD19 antigen binding domain in the art (e.g., those provided in Table 12) in combination with any of the CAR domains described herein to generate a full CAR construct.
  • the full CAR construct is a CAR listed in Table 13.
  • Table 13 provides the exemplary full CD19 CAR constructs generated using the various CAR domains (e.g., transmembrane and intracellular signaling domains) listed in Table 12, and the anti-CD19 antigen binding domains listed in Table 12.
  • Amino acid sequences are designated (aa) and nucleic acid sequences are designated (nt).
  • the disclosure provides methods for treating cancer, e.g., a cancer associated with CD19 expression, with a CAR-expressing cell (e.g., T cell, NK cell) therapy.
  • exemplary cancers include, but are not limited to e.g., one or more acute leukemias including but not limited to, e.g., B-cell acute lymphocytic leukemia (“B-ALL”), T-cell acute lymphocytic leukemia (“T-ALL”), acute lymphocytic leukemia (ALL); one or more chronic leukemias including but not limited to, e.g., chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL).
  • B-ALL B-cell acute lymphocytic leukemia
  • T-ALL T-cell acute lymphocytic leukemia
  • ALL acute lymphocytic leukemia
  • chronic leukemias including but not limited to, e.g., chronic myelogenous leukemia (
  • Additional cancers or hematologic conditions associated with expression of CD19 include, but are not limited to, e.g., B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, and “preleukemia” which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of
  • the disclosure provides methods for treating CLL.
  • the disclosure provides methods for treating ALL.
  • the disclosure provides methods for treating B-cell ALL.
  • the disclosure provides methods of treating a responder (e.g., a complete responder and partial responder) having cancer (e.g., a hematological cancer such as ALL and CLL) with a CAR-expressing cell (e.g., T cell, NK cell) (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019).
  • a responder e.g., a complete responder and partial responder
  • cancer e.g., a hematological cancer such as ALL and CLL
  • a CAR-expressing cell e.g., T cell, NK cell
  • CD19 CAR-expressing cell e.g., T cell, NK cell
  • the disclosure provides methods of treating a responder (e.g., a complete responder and partial responder) with a CAR-expressing cell (e.g., T cell, NK cell) in combination with another therapeutic agent, e.g., another therapeutic agent described herein (e.g., another CAR, e.g., another CAR described herein, an inhibitory CAR, e.g., an inhibitory CAR described herein, a kinase inhibitor (e.g., a kinase inhibitor described herein, e.g., an mTOR inhibitor, a BTK inhibitor), a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein, a standard of care therapy, etc.).
  • another therapeutic agent described herein e.g., another CAR, e.g., another CAR described herein, an inhibitory CAR, e.g., an inhibitory CAR described herein, a kinase inhibitor (e.g., a
  • the combination can be, e.g., with any agent described herein.
  • a CAR-expressing cell e.g., T cell, NK cell
  • an initial CAR-expressing cell e.g., T cell, NK cell
  • a partial responder is tested by any one of the methods described herein, such as, e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene set signature, and if status has not changed and/or is down-graded to, e.g., a non-responder, then the subject is administered an alternative therapy, e.g., a standard of care for the particular cancer.
  • an alternative therapy e.g., a standard of care for the particular cancer.
  • the disclosure provides methods of treating a non-responder having cancer (e.g., a hematological cancer such as ALL and CLL) with a CAR-expressing cell (e.g., T cell, NK cell) (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019).
  • a non-responder having cancer e.g., a hematological cancer such as ALL and CLL
  • a CAR-expressing cell e.g., T cell, NK cell
  • a CD19 CAR-expressing cell e.g., T cell, NK cell
  • the disclosure provides methods of treating a non-responder with a CAR-expressing cell (e.g., T cell, NK cell) in combination with another therapeutic agent, e.g., another therapeutic agent described herein (e.g., another CAR, e.g., another CAR described herein, an inhibitory CAR, e.g., an inhibitory CAR described herein, a kinase inhibitor (e.g., a kinase inhibitor described herein, e.g., an mTOR inhibitor, a BTK inhibitor), a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein, a standard of care therapy, etc.).
  • another therapeutic agent described herein e.g., another CAR, e.g., another CAR described herein, an inhibitory CAR, e.g., an inhibitory CAR described herein, a kinase inhibitor (e.g., a kinase inhibitor described herein, e.g.
  • the combination can be, e.g., with any agent described herein.
  • a CAR-expressing cell e.g., T cell, NK cell
  • an initial CAR-expressing cell e.g., T cell, NK cell
  • a non-responder is tested by any one of the methods described herein, such as, e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene set signature, and if status has changed and/or is up-graded to, e.g., a partial-responder, e.g., a complete responder, then the subject is administered an alternative therapy described herein.
  • the disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL or CLL) comprising steps of: (1) identifying a partial responder subject and/or non-responder subject, (2) administering to the partial responder subject and/or non-responder subject an mTOR inhibitor described herein, such as, e.g., RAD001 and rapamycin, e.g., at a dose and/or dosing schedule described herein; and (3) administering a CAR (e.g., a CD19 CAR described herein, such as, e.g., CTL019), e.g., subsequent to the administration of the mTOR inhibitor, thus treating the cancer.
  • the method further includes administering the mTOR inhibitor and/or the CAR in combination with one or more checkpoint inhibitors described here, such as, e.g., a PD1inhibitor.
  • the disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL or CLL) comprising steps of: (1) identifying a partial responder subject (e.g., patient) and/or non-responder subject (e.g., patient), (2) enriching the T cell population of the partial responder subject and/or non-responder subject by selecting for a less exhausted and/or more na ⁇ ve T cell population, (3) introducing (e.g., by transforming, transducing, infecting, electroporating, etc.) a CAR (e.g., a CD19 CAR described herein, such as, e.g., CTL019) into said enriched T cell population thus transforming the subject's T cell population; and (4) administering the CAR-expressing T cell population into the partial responder subject and/or non-responder subject, thus treating the cancer.
  • a CAR e.g., a CD19 CAR described herein, such as, e.g., CTL019
  • the disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL or CLL) comprising steps of: (1) identifying a partial responder subject (e.g., patient) and/or non-responder subject (e.g., patient), (2) reevaluating a partial responder subject and/or non-responder subject (e.g., patient) at a later time period for na ⁇ ve T cells and/or less exhausted phenotype, and (3), e.g., if the subject has an increase in na ⁇ ve T cells and/or a less exhausted phenotype, administering a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019), thus treating the cancer.
  • a CAR-expressing cell e.g., T cell, NK cell
  • a later time period comprises at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 5 hours, at least 6 hours, at least 7 hours, at least 8 hours, at least 9 hours, at least 10 hours, at least 11 hours, at least 12 hours, at least 1 day, at least 2 days, at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least 1 week, at least 2 weeks, at least 3 weeks, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months at least 8 months, at least 9 months, at least 10 months, at least 11 months, at least 1 year, at least 2 years, at least 3 years, at least 4 years, at least 5 years, at least 6 years, at least 7 years, at least 8 years, at least 9 years, at least 10 years, or 11 years or more.
  • the disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL or CLL) comprising the steps of (1) identifying partial responders and/or non-responders; and (2) treating with an alternative therapy, e.g., a standard of care for the particular cancer (e.g., the standard of care for ALL or CLL).
  • a partial responder is treated only with the standard of care (e.g., the standard of care for a hematological cancer such as ALL or CLL) in the absence of treatment with a CAR.
  • a non-responder is treated only with the standard of care (e.g., the standard of care for a hematological cancer such as ALL or CLL) in the absence of treatment with a CAR.
  • standard of care for CLL includes, but is not limited to exemplary therapies described herein, e.g., described in Table A, and combinations thereof.
  • standard of care for CLL includes (1) radiation therapy, (2) chemotherapy, (3) surgery (e.g., removal of the spleen), (4) targeted therapy, (5) stem cell transplantation, and combinations thereof.
  • the standard of care comprises external radiation therapy.
  • the standard of care comprises internal radiation therapy (e.g., a radioactive substance sealed in needles, wires or catheters, for example, that are placed directly into or near the cancer).
  • standard of care for ALL includes, but is not limited to exemplary therapies described herein, e.g., described in Table B, and combinations thereof.
  • standard of care for ALL includes (1) chemotherapy, (2) radiation therapy, (3) stem cell transplantation, (4) biological therapy, (5) targeted therapy, and combinations thereof.
  • the standard of care includes, but is not limited to, fludarabine with cyclophosphamide (FC); fludarabine with rituximab (FR); fludarabine, cyclophosphamide, and rituximab (FCR); cyclophosphamide, doxorubicin, vincristine and prednisone (CHOP); and combinations thereof.
  • General chemotherapeutic agents considered for use include, but are not limited to anastrozole (Arimidex®), bicalutamide (Casodex®), bleomycin sulfate (Blenoxane®), busulfan (Myleran®), busulfan injection (Busulfex®), capecitabine (Xeloda®), N4-pentoxycarbonyl-5-deoxy-5-fluorocytidine, carboplatin (Paraplatin®), carmustine (BiCNU®), chlorambucil (Leukeran®), cisplatin (Platinol®), cladribine (Leustatin®), cyclophosphamide (Cytoxan® or Neosar®), cytarabine, cytosine arabinoside (Cytosar-U®), cytarabine liposome injection (DepoCyt®), dacarbazine (DTIC-Dome®), d
  • chemotherapy comprises an antimetabolite, including, but not limited to, folic acid antagonists (also referred to herein as antifolates), pyrimidine analogs, purine analogs and adenosine deaminase inhibitors): methotrexate (Rheumatrex®, Trexall®), 5-fluorouracil (Adrucil®, Efudex®, Fluoroplex®), floxuridine (FUDF®), cytarabine (Cytosar-U®, Tarabine PFS), 6-mercaptopurine (Puri-Nethol®)), 6-thioguanine (Thioguanine Tabloid®), fludarabine phosphate (Fludara®), pentostatin (Nipent®), pemetrexed (Alimta®), raltitrexed (Tomudex®), cladribine (Leustatin®), clofarabine (Clofarex®, Clolar®),
  • Suitable antimetabolites include, e.g., 5-fluorouracil (Adrucil®, Efudex®, Fluoroplex®), floxuridine (FUDF®), capecitabine (Xeloda®), pemetrexed (Alimta®), raltitrexed (Tomudex®) and gemcitabine (Gemzar®), and combinations thereof.
  • the purine analogue is fludarabine.
  • chemotherapy comprises an alkylating agent including, but not limited to nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes, uracil mustard (Aminouracil Mustard®, Chlorethaminacil®, Demethyldopan®, Desmethyldopan®, Haemanthamine®, Nordopan®, Uracil nitrogen Mustard®, Uracillost®, Uracilmostaza®, Uramustin®, Uramustine®), chlormethine (Mustargen®), cyclophosphamide (Cytoxan®, Neosar®, Clafen®, Endoxan®, Procytox®, RevimmuneTM), ifosfamide (Mitoxana®), melphalan (Alkeran®), Chlorambucil (Leukeran®), pipobroman (Amedel®, Vercyte®), triethylenemelamine (Hemel®
  • Additional exemplary alkylating agents include, without limitation, Oxaliplatin (Eloxatin®); Temozolomide (Temodar® and Temodal®); Dactinomycin (also known as actinomycin-D, Cosmegen®); Melphalan (also known as L-PAM, L-sarcolysin, and phenylalanine mustard, Alkeran®); Altretamine (also known as hexamethylmelamine (HMM), Hexalen®); Carmustine (BiCNU®); Bendamustine (Treanda®); Busulfan (Busulfex® and Myleran®); Carboplatin (Paraplatin®); Lomustine (also known as CCNU, CeeNU®); Cisplatin (also known as CDDP, Platinol® and Platinol®-AQ); Chlorambucil (Leukeran®); Cyclophosphamide (Cytoxan® and Neosar®); dacarbazine (also known
  • the chemotherapeutic agent is a kinase inhibitor, e.g., a tyrosine kinase inhibitor including, but not limited to, erlotinib hydrochloride (Tarceva®); linifanib (N-[4-(3-amino-1H-indazol-4-yl)phenyl]-N-2-fluoro-5-methylphenyl)urea, also known as ABT 869, available from Genentech); sunitinib malate (Sutent®); bosutinib (4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-methoxy-7-[3-(4-methylpiperazin-1-yl)propoxy]quinoline-3-carbonitrile, also known as SKI-606, and described in U.S.
  • a tyrosine kinase inhibitor including, but not limited to, erlotinib hydrochloride (Tarceva®); lini
  • the kinase inhibitor is a BTK inhibitor selected from ibrutinib (PCI-32765); GDC-0834; RN-486; CGI-560; CGI-1764; HM-71224; CC-292; ONO-4059; CNX-774; and LFM-A13.
  • the kinase inhibitor is a CDK4 inhibitor selected from aloisine A; flavopiridol or HMR-1275, 2-(2-chlorophenyl)-5,7-dihydroxy-8-[(3S,4R)-3-hydroxy-1-methyl-4-piperidinyl]-4-chromenone; crizotinib (PF-02341066; 2-(2-Chlorophenyl)-5,7-dihydroxy-8-[(2R,3S)-2-(hydroxymethyl)-1-methyl-3-pyrrolidinyl]-4H-1-benzopyran-4-one, hydrochloride (P276-00); 1-methyl-5-[[2-[5-(trifluoromethyl)-1H-imidazol-2-yl]-4-pyridinyl]oxy]-N-[4-(trifluoromethyl)phenyl]-1H-benzimidazol-2-amine (RAF265); indisulam (E7070); roscovitine (CYC12C
  • the kinase inhibitor is an MNK inhibitor selected from CGP052088; 4-amino-3-(p-fluorophenylamino)-pyrazolo [3,4-d] pyrimidine (CGP57380); cercosporamide; ETC-1780445-2; and 4-amino-5-(4-fluoroanilino)-pyrazolo [3,4-d] pyrimidine.
  • targeted therapy includes, but is not limited to an anti-CD20 antibody or functional fragment thereof, such as, e.g., rituximab (Riuxan® and MabThera®); tositumomab (Bexxar®); and ofatumumab (Arzerra®), and combinations thereof.
  • the targeted therapy includes, but is not limited to, an anti-CD52 antibody or functional fragment thereof such as, e.g., alemtuzumab (Campath®).
  • biologic therapy comprises immunotherapy.
  • exemplary anthracyclines include, without limitation, doxorubicin (Adriamycin® and Rubex®); bleomycin (Lenoxane®); daunorubicin (dauorubicin hydrochloride, daunomycin, and rubidomycin hydrochloride, Cerubidine®); daunorubicin liposomal (daunorubicin citrate liposome, DaunoXome®); mitoxantrone (DHAD, Novantrone®); epirubicin (EllenceTM); idarubicin (Idamycin®, Idamycin PFS®); mitomycin C (Mutamycin®); geldanamycin; herbimycin; ravidomycin; desacetylravidomycin and combinations thereof.
  • stem cell transplantation comprises an autogeneic stem cell transplant.
  • stem cell transplantation comprises an allogenic stem cell transplant.
  • stem cell transplantation comprises allogeneic bone marrow transplantation.
  • stem cell transplantation comprises a hematopoietic stem cell transplantation (HSCT).
  • hematopoietic stem cells are derived from various tissues including, but not limited to bone marrow, peripheral blood, umbilical cord blood, and combinations thereof.
  • the provided methods comprise determining if the subject is identified as having a statistically significant difference in expression level of one or more markers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15 and Table 16, or a PD-1 biomarker, LAG-3 biomarker, TIM-3 biomarker, CD57 biomarker CD27 biomarker, CD122 biomarker, CD62L biomarker and a KLRG1 biomarker, relative to a reference level, and administering to the subject a therapeutically effective dose of a CAR-expressing cell, e.g., a T cell or NK cell.
  • a CAR-expressing cell e.g., T cell, NK cell
  • a CD19 CAR-expressing cell e.g., T cell, NK cell described herein such as, e.g., CTL019.
  • provided methods comprise a vector comprising CD19 CAR operably linked to promoter for expression in mammalian cells (e.g., T cells or NK cells).
  • provided methods comprise a recombinant cell (e.g., T cell or NK cell) expressing a CD19 CAR for use in treating CD19-expressing tumors, wherein the recombinant T cell expressing the CD19 CAR is termed a CD19 CAR-expressing cell.
  • a CD19 CAR-expressing cell e.g., T cell, NK cell administered according to provided methods is capable of contacting a tumor cell with at least one CD19 CAR expressed on its surface such that the CAR-expressing cell targets the tumor cell and growth of the tumor is inhibited.
  • the disclosure features to a method of inhibiting growth of a CD19-expressing tumor cell, comprising contacting the tumor cell with a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein such that the CAR-expressing cell is activated in response to the antigen and targets the cancer cell, wherein the growth of the tumor is inhibited.
  • a CD19 CAR-expressing cell e.g., T cell, NK cell
  • the disclosure includes a type of cellular therapy where T cells are genetically modified to express a CAR and the CAR-expressing cell (e.g., T cell, NK cell) is infused to a recipient in need thereof.
  • the infused cell is able to kill tumor cells in the recipient.
  • CAR-modified cells e.g., T cells or NK cells
  • the cells administered to the patient, or their progeny persist in the patient for at least four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, twelve months, thirteen months, fourteen month, fifteen months, sixteen months, seventeen months, eighteen months, nineteen months, twenty months, twenty-one months, twenty-two months, twenty-three months, two years, three years, four years, or five years after administration of the cell to the patient.
  • the disclosure also includes a type of cellular therapy where cells (e.g., T cells, NK cells) are modified, e.g., by in vitro transcribed RNA, to transiently express a chimeric antigen receptor (CAR) and the CAR-expressing cell (e.g., T cell, NK cell) is infused to a recipient in need thereof.
  • the infused cell is able to kill tumor cells in the recipient.
  • the cells administered to the patient are present for less than one month, e.g., three weeks, two weeks, one week, after administration of the cell (e.g., T cell, NK cell) to the patient.
  • the anti-tumor immunity response elicited by the CAR-modified cells may be an active or a passive immune response, or alternatively may be due to a direct vs indirect immune response.
  • the CAR transduced T cells exhibit specific proinflammatory cytokine secretion and potent cytolytic activity in response to human cancer cells expressing the CD19, resist soluble CD19 inhibition, mediate bystander killing and mediate regression of an established human tumor.
  • antigen-less tumor cells within a heterogeneous field of CD19-expressing tumor may be susceptible to indirect destruction by CD19-redirected T cells that has previously reacted against adjacent antigen-positive cancer cells.
  • the fully-human CAR-modified cells e.g., T cells, NK cells
  • the fully-human CAR-modified cells may be a type of vaccine for ex vivo immunization and/or in vivo therapy in a mammal.
  • the mammal is a human.
  • cells are isolated from a subject (e.g., a human) and genetically modified (i.e., transduced or transfected in vitro) with a vector expressing a CAR disclosed herein.
  • the CAR-modified cell can be administered to a mammalian recipient to provide a therapeutic benefit.
  • the mammalian recipient may be a human and the CAR-modified cell can be autologous with respect to the recipient.
  • the cells can be allogeneic, syngeneic or xenogeneic with respect to the recipient.
  • Hematological cancer conditions are types of cancer such as leukemia and malignant lymphoproliferative conditions that affect blood, bone marrow and the lymphatic system.
  • Leukemia can be classified as acute leukemia and chronic leukemia.
  • Acute leukemia can be further classified as acute myelogenous leukemia (AML) and acute lymphoid leukemia (ALL).
  • Chronic leukemia includes chronic myelogenous leukemia (CML) and chronic lymphoid leukemia (CLL).
  • CML chronic myelogenous leukemia
  • CLL chronic lymphoid leukemia
  • Other related conditions include myelodysplastic syndromes (MDS, formerly known as “preleukemia”) which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells and risk of transformation to AML.
  • MDS myelodysplastic syndromes
  • Lymphoma is a group of blood cell tumors that develop from lymphocytes.
  • Exemplary lymphomas include non-Hodgkin lymphoma and Hodgkin lymphoma.
  • the cancer is a hematologic cancer including but is not limited to a leukemia or a lymphoma.
  • the CAR-expressing cells (e.g., T cells, NK cells) of the invention may be used to treat cancers and malignancies such as, but not limited to, e.g., acute leukemias including but not limited to, e.g., B-cell acute lymphoid leukemia (“B-ALL”), T-cell acute lymphoid leukemia (“T-ALL”), acute lymphoid leukemia (ALL); one or more chronic leukemias including but not limited to, e.g., chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL); additional hematologic cancers or hematologic conditions including, but not limited to, e.g., B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell
  • the present disclosure also provides methods for inhibiting the proliferation or reducing a CD19-expressing cell population, the methods comprising contacting a population of cells comprising a CD19-expressing cell with a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell.
  • a CD19 CAR-expressing cell e.g., T cell, NK cell
  • the disclosure provides methods for inhibiting the proliferation or reducing the population of cancer cells expressing CD19, the methods comprising contacting the CD19-expressing cancer cell population with a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell.
  • the present disclosure provides methods for inhibiting the proliferation or reducing the population of cancer cells expressing CD19, the methods comprising contacting the CD19-expressing cancer cell population with a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell.
  • a CD19 CAR-expressing cell e.g., T cell, NK cell
  • the anti-CD19 CAR-expressing cell reduces the quantity, number, amount or percentage of cells and/or cancer cells by at least 25%, at least 30%, at least 40%, at least 50%, at least 65%, at least 75%, at least 85%, at least 95%, or at least 99% in a subject with or animal model for myeloid leukemia or another cancer associated with CD19-expressing cells relative to a negative control.
  • the subject is a human.
  • the present disclosure also provides methods for preventing, treating and/or managing a disease associated with CD19-expressing cells (e.g., a hematologic cancer or atypical cancer expressing CD19), the methods comprising administering to a subject in need a CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell.
  • a CAR-expressing cell e.g., T cell, NK cell
  • the subject is a human.
  • disorders associated with CD19-expressing cells include autoimmune disorders (such as lupus), inflammatory disorders (such as allergies and asthma) and cancers (such as hematological cancers or atypical cancers expressing CD19).
  • the present disclosure also provides methods for preventing, treating and/or managing a disease associated with CD19-expressing cells, the methods comprising administering to a subject in need a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell.
  • a CD19 CAR-expressing cell e.g., T cell, NK cell
  • the subject is a human.
  • the present disclosure provides methods for preventing relapse of cancer associated with CD19-expressing cells (e.g., a hematological cancer such as ALL and CLL), the methods comprising administering to a subject in need thereof a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell.
  • the methods comprise administering to the subject in need thereof an effective amount of a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell in combination with an effective amount of another therapy.
  • any cancer therapy as described above and herein can be administered in combination with one or more additional therapies to treat and/or reduce the symptoms of cancer described herein.
  • the pharmaceutical compositions can be administered concurrently with, prior to, or subsequent to, one or more other additional therapies or therapeutic agents.
  • a CAR-expressing cell described herein may be used in combination with other known agents and therapies.
  • Administered “in combination”, as used herein, means that two (or more) different treatments are delivered to the subject during the course of the subject affliction with the disorder, e.g., the two or more treatments are delivered after the subjects has been diagnosed with the disorder and before the disorder has been cured or eliminated or treatment has ceased for other reasons.
  • the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap in terms of administration. This is sometimes referred to herein as “simultaneous” or “concurrent delivery”.
  • the delivery of one treatment ends before the delivery of the other treatment begins.
  • the treatment is more effective because of combined administration.
  • the second treatment is more effective, e.g., an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment or the analogous situation is seen with the first treatment.
  • delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other.
  • the effect of the two treatments can be partially additive, wholly additive, or greater than additive.
  • the delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered.
  • a CAR-expressing cell described herein and the at least one additional therapeutic agent can be administered simultaneously, in the same or in separate compositions, or sequentially.
  • the CAR-expressing cell described herein can be administered first, and the additional agent can be administered second, or the order of administration can be reversed.
  • the CAR therapy and/or other therapeutic agents, procedures or modalities can be administered during periods of active disorder, or during a period of remission or less active disease.
  • the CAR therapy can be administered before the other treatment, concurrently with the treatment, post-treatment, or during remission of the disorder.
  • the CAR therapy and the additional agent can be administered in an amount or dose that is higher, lower or the same than the amount or dosage of each agent used individually, e.g., as a monotherapy.
  • the administered amount or dosage of the CAR therapy, the additional agent (e.g., second or third agent), or all is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50%) than the amount or dosage of each agent used individually, e.g., as a monotherapy.
  • the amount or dosage of the CAR therapy, the additional agent (e.g., second or third agent), or all, that results in a desired effect is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50% lower) than the amount or dosage of each agent used individually, e.g., as a monotherapy, required to achieve the same therapeutic effect.
  • a CAR-expressing cell described herein may be used in a treatment regimen in combination with surgery, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies or other antibody therapies, cytoxin, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, cytokines, and irradiation peptide vaccine, such as that described in Izumoto et al. 2008 J NEUROSURG 108:963-971.
  • immunosuppressive agents such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies
  • immunoablative agents such as CAMPATH, anti-CD3 antibodies or other antibody therapies, cytoxin, fludarabine, cyclosporin, FK506, rap
  • a CAR-expressing cell described herein can be used in combination with a chemotherapeutic agent.
  • chemotherapeutic agents include those described in paragraphs 873-874 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.
  • Exemplary alkylating agents include, without limitation, those described in paragraph 875 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.
  • Exemplary mTOR inhibitors include, without limitation, RAD001, temsirolimus; ridaforolimus (formally known as deferolimus, (1R,2R,4S)-4-[(2R)-2 [(1R,9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28Z,30S,32S,35R)-1,18-dihydroxy-19,30-dimethoxy-15,17,21,23, 29,35-hexamethyl-2,3,10,14,20-pentaoxo-11,36-dioxa-4-azatricyclo[30.3.1.0 4,9 ] hexatriaconta-16,24,26,28-tetraen-12-yl]propyl]-2-methoxycyclohexyl dimethylphosphinate, also known as AP23573 and MK8669, and described in PCT Publication No.
  • WO 03/064383 everolimus (Afinitor® or RAD001); rapamycin (AY22989, Sirolimus®); simapimod (CAS 164301-51-3); emsirolimus, (5- ⁇ 2,4-Bis[(3S)-3-methylmorpholin-4-yl]pyrido[2,3-d]pyrimidin-7-yl ⁇ -2-methoxyphenyl)methanol (AZD8055); 2-Amino-8-[trans-4-(2-hydroxyethoxy)cyclohexyl]-6-(6-methoxy-3-pyridinyl)-4-methyl-pyrido[2,3-d]pyrimidin-7(8H)-one (PF04691502, CAS 1013101-36-4); and N 2 -[1,4-dioxo-4-[[4-(4-oxo-8-phenyl-4H-1-benzopyran-2-yl)morpholinium-4-yl]methoxy]butyl]-arg
  • Exemplary immunomodulators include, without limitation, those described in paragraph 882 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.
  • Exemplary anthracyclines include, without limitation, those described in paragraph 883 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.
  • Exemplary vinca alkaloids include, without limitation, those described in paragraph 884 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.
  • Exemplary proteosome inhibitors include, without limitation, those described in paragraph 884 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.

Abstract

Cancer biomarkers and methods of using them are disclosed.

Description

  • This application is a continuation of U.S. application Ser. No. 15/517,597, filed Apr. 7, 2017, now allowed, which is a U.S. national phase application under 35 U.S.C. § 371 of International Application No. PCT/US2015/054542, filed Oct. 7, 2015, which claims priority to U.S. Ser. No. 62/061,553 filed Oct. 8, 2014 and U.S. Ser. No. 62/144,682 filed Apr. 8, 2015, the contents of which are incorporated herein by reference in their entireties.
  • SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Oct. 7, 2015, is named N2067-7057WO_SL.txt and is 219,221 bytes in size.
  • FIELD OF THE INVENTION
  • The invention relates to cancer biomarkers and uses thereof.
  • BACKGROUND OF THE INVENTION
  • Many patients with B cell malignancies are incurable with standard therapy. In addition, traditional treatment options often have serious side effects. Attempts have been made in cancer immunotherapy, however, several obstacles render the goal of clinical effectiveness difficult to achieve. Although hundreds of so-called tumor antigens have been identified, these are generally derived from self and thus are poorly immunogenic. Furthermore, tumors use several mechanisms to render themselves hostile to the initiation and propagation of immune attack.
  • Recent developments using chimeric antigen receptor (CAR) modified autologous T cell (CART) therapy, which relies on redirecting T cells to a suitable cell-surface molecule on cancer cells such as B cell malignancies, show promising results in harnessing the power of the immune system to treat B cell malignancies and other cancers (see, e.g., Sadelain et al., CANCER DISCOVERY 3:388-398 (2013)). For example, the clinical results of a CART that binds to CD19 (i.e., “CTL019”) have shown promise in establishing complete remissions in patients suffering with chronic lymphocytic leukemia (CLL), as well as in childhood acute lymphocytic leukemia (ALL) (see, e.g., Kalos et al., SCI TRANSL MED 3:95ra73 (2011), Porter et al., NEJM 365:725-733 (2011), Grupp et al., NEJM 368:1509-1518 (2013)).
  • Besides the ability for the chimeric antigen receptor on the genetically modified T cells to recognize and destroy the targeted cells, a successful therapeutic T cell therapy needs to have the ability to proliferate, to persist over time, and to further monitor for leukemic cell escapees. The variable phenotypic state of T cells, whether it is in a state of anergy, suppression or exhaustion, will have effects on CAR-transformed T cells' efficacy. To be effective, CAR transformed patient T cells need to persist and maintain the ability to proliferate in response to the CAR's antigen.
  • A need, therefore, exists for a method of using biomarkers for use in connection with the differential diagnosis and treatment of cancer with CAR-expressing cell (e.g., T cell, NK cell) therapy. In particular, there is an unmet need for effective predictors of therapeutic response in subjects having a hematological cancer, such as CLL and ALL, to a CAR-expressing cell therapy, e.g., with CTL019 or other CD19 CAR-expressing cells.
  • SUMMARY OF THE INVENTION
  • The present disclosure relates to the identification and use of analytes, analyte profiles, or markers (e.g., gene expression, flow cytometry and/or protein expression profiles) with clinical relevance to cancer (e.g., a hematological cancer such as chronic lymphocytic leukemia (CLL) and acute lymphocytic leukemia (ALL)). In some embodiments, the disclosure provides the identity of genes, whose expression, at the transcriptional and protein levels, are correlated with CLL and ALL progression, e.g., as a way of predicting a response to a Chimeric Antigen Receptor (CAR)-expressing cell therapy (e.g., a therapy comprising a cell (e.g., an immune effector cell or population of cells) that expresses a CAR that binds to CD19 (also referred to herein as a “CAR19” or “CD19 CAR”-expressing cell). In certain embodiments, one or more of a CD19 CAR-expressing cell gene set signature, a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature, and combinations thereof) are evaluated. These gene expression profiles may be applied to the diagnosis and/or prognosis of a cancer, e.g., a hematological cancer such as CLL and ALL, and are particularly useful in predicting whether a subject will respond favorably to a CAR therapy (e.g., a CD19 CAR therapy as described here, e.g., a CTL019 therapy) in a subject diagnosed with a cancer, e.g., a hematological cancer such as CLL or ALL. Compared to clinical parameters or biochemical markers used in existing prognosis methods, the expression profiles of the genes disclosed herein constitute a more robust signature of hematological cancer progression (e.g., CLL and ALL progression) and provide a more reliable, non-subjective basis for the selection of appropriate therapeutic regimens.
  • Amongst other things, the present disclosure provides novel gene signatures, e.g., at the transcriptional and protein levels, and methods of use thereof, that predict subject response to a cell expressing a CAR, e.g., a CD19 CAR (e.g., a CD19 CAR-expressing cell, e.g., T cell, NK cell, described herein such as, e.g., CTL019) therapy in a cancer, e.g., a hematological cancer such as CLL and ALL.
  • The present disclosure demonstrates, at least in part, that expression profiles and gene signatures, e.g., at the transcriptional and protein levels, are useful to distinguish among a responder, a partial responder, a non-responder, a relapser or a non-relapser to a therapy comprising a CAR-expressing cell (e.g., a CAR-expressing immune effector cell, e.g., a T cell, or an NK cell), (also referred to herein as a “CAR-expressing cell therapy”), in a cancer (e.g., a hematological cancer such as CLL and ALL). In one embodiment, the CAR-expressing cell is a CD19 CAR-expressing cell. In one embodiment, the therapy is a CTL019 therapy. In embodiments, the expression profiles and gene signatures disclosed herein distinguish among a CAR (or CD19 CAR)-expressing cell responder, a CAR (or CD19 CAR)-expressing cell partial responder, or a CAR (or CD19 CAR)-expressing cell non-responder (e.g., a CTL019-responder, a CTL019-partial responder, and a CTL019-non-responder); or a CAR (or CD19 CAR)-expressing cell relapser, or a CAR (or CD19 CAR)-expressing cell non-relapser (e.g., a CTL019-relapser, or a CTL019-relapser), in a cancer (e.g., a hematological cancer such as CLL and ALL). The present disclosure encompasses the identification of novel gene signatures predictive of subject response to a CAR-expressing cell therapy, e.g., a CD19 CAR-expressing cell therapy such as CTL019.
  • Thus, disclosed herein are methods, systems, compositions, and kits for the identification, assessment and/or treatment of a subject having cancer. Exemplary cancers include, but are not limited to, B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt
    Figure US20210172020A1-20210610-P00001
    lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma (HL), plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, and Waldenstrom macroglobulinemia. In one embodiment, the cancer is ALL. In another embodiment, the cancer is CLL. In an embodiment, the cancer is associated with CD19 expression.
  • Accordingly, in one aspect, the invention features a method of evaluating a subject having a cancer, e.g., a hematological cancer. The method includes acquiring a value of responder or relapser status (e.g., a value of responder or relapser status as described herein) to a therapy comprising a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells) for the subject, wherein said value is indicative of the subject's responsiveness or relapsing status to the CAR-expressing cell therapy.
  • In a related aspect, the invention features a method of evaluating or monitoring the effectiveness of a CAR-expressing cell therapy in a subject having a cancer, e.g., a hematological cancer. The method includes acquiring a value of responder or relapser status (e.g., a value of responder or relapser status as described herein) to a therapy comprising a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells) for the subject, wherein said value is indicative of the effectiveness of the CAR-expressing cell therapy, thereby evaluating the effectiveness of the CAR-expressing cell therapy in the subject.
  • In another aspect, the invention features a method for treating a subject having a cancer, e.g., a hematological cancer. The method includes administering to the subject a therapeutically effective dose of a CAR-expressing cell therapy, if the subject is identified as being responsive (e.g., identified as a complete responder, partial responder or a non-relapser) to a therapy comprising a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells), wherein said identifying comprises a value of responder or relapser status (e.g., a value of responder or relapser status as described herein).
  • In a related aspect, the invention features a method of treating a cancer, e.g., a hematological cancer, in a subject. The method includes acquiring a value of responder or relapser status (e.g., a value of responder or relapser status as described herein) to a therapy comprising a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells) for the subject; and responsive to said value, treating the cancer.
  • In embodiments of any of the methods and compositions for use described herein, the value of responder or relapser status comprises a measure of one, two, three, four, five, six, seven or more (all) of the following:
  • (i) the level or activity of CD27 and/or CD45RO− (e.g., CD27+CD45RO−) immune effector cells, e.g., in a CD4+ or a CD8+ T cell population, in a subject, e.g., a sample from the subject (e.g., an apheresis sample or a CAR-expressing cell product sample);
  • (ii) the level or activity of one, two, three, or more (e.g., all) of resting TEFF cells, resting TREG cells, younger T cells (e.g., younger CD4 or CD8 cells, or gamma/delta T cells), or early memory T cells, or a combination thereof, in a subject, e.g., a sample from the subject (e.g., an apheresis sample or a CAR-expressing cell product sample);
  • (iii) the level or activity of one, two, three, or more (e.g., all) of activated TEFF cells, activated TREG cells, older T cells (e.g., older CD4 or CD8 cells), or late memory T cells, or a combination thereof, in a subject, e.g., a sample from the subject (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • (iv) the level or activity of an immune cell exhaustion marker, e.g., one, two or more immune checkpoint inhibitors (e.g., PD-1, TIM-3 and/or LAG-3) in a subject, e.g., a sample from the subject (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • (v) the level or activity of one, two, three, four, five, ten, twenty or more of the biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 16, Table 17, Table 18, Table 20, FIG. 2B, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature;
  • (vi) a cytokine level or activity (e.g., quality of cytokine reportoire) in a CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019), wherein the cytokine is chosen from one, two, three, four, five or more (or all) of the cytokines listed in Table 16;
  • (vii) a transduction efficiency of a CAR-expressing cell in a CAR-expressing cell product sample; or
  • (viii) a quantity of CD27+PD-1− cells in a subject, e.g., a sample from the subject (e.g., an apheresis sample or a CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019)), e.g., a quantity greater than or equal to 1×107 cells.
  • In an aspect, the invention provides a CAR expressing cell therapy (e.g., CD19 CART cell, e.g., CTL019 cell) for use in the treatment of a subject, wherein the CAR expressing cell has been assayed according to a method herein, e.g., before or after transduction or transfection with a CAR nucleic acid. In a related aspect, the invention provides a CAR expressing cell therapy (e.g., CD19 CART cell, e.g., CTL019 cell), for use in the treatment of a subject that has been identified as being responsive (e.g., identified as a complete responder, partial responder or a non-relapser) to a therapy comprising a CAR-expressing cell population (e.g., a CAR19-expressing cell population). The composition for use can comprise a measure of one, two, three, four, five, six, seven, or more (all) of (i)-(viii) described herein.
  • Alternatively, or in combination with the methods and compositions for use disclosed herein, responsive to said value, performing one, two, three, four, five, six, seven, or more (e.g., all) of:
  • identifying the subject as a complete responder, partial responder or non-responder, or a relapser or a non-relapser;
  • administering e.g., to a responder or a non-relapser, a CAR-expressing cell therapy;
  • administering an altered dosing of a CAR-expressing cell therapy;
  • altering the schedule or time course of a CAR-expressing cell therapy;
  • administering, e.g., to a non-responder or a partial responder, an additional agent in combination with a CAR-expressing cell therapy, e.g., a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein;
  • administering to a non-responder or partial responder a therapy that increases the number of younger T cells in the subject prior to treatment with a CAR-expressing cell therapy;
  • modifying a process, e.g., a manufacturing process, of a CAR-expressing cell therapy, e.g., enriching for younger T cells prior to introducing a nucleic acid encoding a CAR, or increasing the transduction efficiency, e.g., for a subject identified as a non-responder or a partial responder;
  • administering an alternative therapy, e.g., for a non-responder or partial responder or relapser, e.g., a standard of care for a particular cancer type; or
  • if the subject is, or is identified as, a non-responder or a relapser, decreasing the TREG cell population and/or TREG gene signature, e.g., by one or more of CD25 depletion, or administration of cyclophosphamide, anti-GITR antibody, an mTOR inhibitor, or a combination thereof.
  • In certain embodiments, the subject is pre-treated with an anti-GITR antibody. In certain embodiment, the subject is treated with an anti-GITR antibody prior to infusion or re-infusion. In some embodiments, the subject is a patient with CLL.
  • In another aspect, the invention features a method of, or assay for, identifying a subject having a cancer as having an increased or decreased likelihood to respond to a treatment that comprises a CAR-expressing cell (e.g., a plurality (e.g., a population) of CAR (e.g., CAR19-)-expressing cells). The method includes:
  • (1) providing, e.g., acquiring, a sample from the subject;
  • (2) determining a level or activity of one or more biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1 in the sample;
  • wherein a difference, e.g., a statistically significant difference, between the determined level compared to a reference level is predictive of the subject's responsiveness to the CAR-expressing cell therapy; and
  • (3) (optionally) identifying the subject as a complete responder, partial responder, non-responder, a relapser or a non-relapser, to the CAR-expressing cell therapy.
  • In yet another aspect, the invention features a method for treating a subject having a cancer comprising:
  • determining if the subject has an increased likelihood to respond, or a decreased likelihood to relapse, to a CAR-expressing cell therapy (e.g., a CAR19-expressing therapy, e.g., CTL019) by determining the level or activity of one or more biomarkers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), FIG. 2B, Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature in a sample from the subject, e.g., relative to a reference level; and administering to the subject a therapeutically effective dose of a CAR-expressing cell therapy.
  • In yet another aspect, the invention features a method for treating a subject having cancer comprising:
  • (1) determining if the subject has an increased likelihood to relapse to a CAR-expressing cell therapy by acquiring a value for the level or activity of one or more markers in a Table herein, e.g., Table 17 in a sample from the subject (e.g., an apheresis sample or a manufactured CAR-expressing product sample), wherein a difference, e.g., a statistically significant difference, in the level or activity of one or more biomarker genes relative to the reference level is indicative of an increased likelihood of relapse to a CAR-expressing cell therapy; and
  • (2) for a subject with an increased likelihood of relapse, decreasing the TREG cell population and/or decreasing TREG gene signature; and
  • (3) administering to the subject a therapeutically effective dose of a CAR-expressing cell therapy.
  • Additional features and embodiments of the present invention include one or more of the following:
  • In some embodiments of any of the methods and compositions for use disclosed herein, an immune cell has an exhausted phenotype, e.g., co-expresses at least two exhaustion markers, e.g., co-expresses PD-1 and TIM-3. In other embodiments, an immune cell has an exhausted phenotype, e.g., co-expresses at least two exhaustion markers, e.g., co-expresses PD-1 and LAG-3.
  • In some embodiments of any of the methods, systems, compositions for use, and kits disclosed herein, the CAR-expressing cell therapy comprises a plurality (e.g., a population) of CAR-expressing immune effector cells, e.g., a plurality (e.g., a population) of T cells or NK cells, or a combination thereof. In one embodiment, the CAR-expressing cell therapy is a CAR19 therapy (e.g., CTL019 therapy). In an embodiment, the CAR-expressing cell therapy comprises or consists of CTL019. In an embodiment, the CAR-expressing cell is a CTL019 product. In an embodiment, the CAR-expressing cell is a T cell. In an embodiment, the CAR-expressing cell is a NK cell.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the measure of one or more of (i)-(viii) is obtained from an apheresis sample acquired from the subject. The apheresis sample can be evaluated prior to infusion or re-infusion.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the measure of one or more of (i)-(viii) is obtained from a manufactured CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019). The manufactured CAR-expressing cell product can be evaluated prior to infusion or re-infusion.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the subject is evaluated prior to receiving, during, or after receiving, the CAR-expressing cell therapy.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the hematological cancer is an ALL or a CLL. The subject can be a human patient.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the cell, e.g., the population of immune effector cells (e.g., cells expressing a CAR molecule described herein) is administered in combination with an inhibitor of an immune checkpoint molecule chosen from one or more of PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, TGFR (e.g., TGFR beta), or a combination thereof.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the subject receives concurrent treatment with an agent, e.g., an mTOR inhibitor, and/or a checkpoint inhibitor. In some embodiments, the subject receives treatment with an agent, e.g., an mTOR inhibitor, and/or a checkpoint inhibitor, post-CAR-expressing cell therapy. In some embodiments, the subject receives a pre-treatment of with an agent, e.g., an mTOR inhibitor, and/or a checkpoint inhibitor, prior to the initiation of a CAR-expressing cell therapy.
  • In some embodiments of any of the methods and compositions for use disclosed herein, TREG cell population and/or TREG gene signature is decreased prior to collection of cells for manufacturing. In some embodiments, the TREG cell population and/or TREG gene signature is decreased prior to CAR-expressing cell (e.g., T cell, NK cell) therapy. In some embodiments, the TREG cell population and/or TREG gene signature is decreased by administration of cyclophosphamide, anti-GITR antibody, an mTOR inhibitor, or a combination thereof.
  • In some embodiments of any of the methods and compositions for use disclosed herein the value of responder or relapser status comprises a measure of a combination of a gene signature and a biomarker. In some embodiments, the value of the responder or relapser status comprises a measure of a CD19 CAR-expressing cell gene set signature and a combination of one or more of: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, or KLRG1.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the method further comprises identifying the subject as a responder (e.g., a complete or partial responder), a non-responder, a relapser or a non-relapser, based on a measure of one or more of (i)-(viii).
  • In some embodiments of any of the methods and compositions for use disclosed herein, the measure of one or more of (i)-(viii) evaluates a profile for one or more of gene expression, flow cytometry or protein expression.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the expression profile includes one or more gene signatures based on mRNA expression levels of selected genes obtained from the apheresis sample or a manufactured CD19 CAR-expressing cell product (e.g., CTL019). In one embodiment, the expression profile includes one, two, three, four, five, ten, twenty or more of a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 16, Table 17, Table 18, Table 20, FIG. 2B, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the level or activity of a CD8+ T cell is evaluated using a profile or signature indicative of the percentage of CD8+ T cell in the sample.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the level or activity of CD27+CD45RO− immune effector cells is evaluated using a profile or signature indicative of the percentage of CD27+CD45RO− immune effector cells in the sample.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the level or activity, e.g., in (i), (ii), or (v), is evaluated using a profile or gene signature according to one, two, three, four, five, ten, twenty, fifty, sixty, seventy, one hundred or more of a biomarker or gene set listed in Tables 1A, 1B, 3, 4, 5, 6, or FIG. 2B.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the level or activity one, two or more immune checkpoint inhibitors is evaluated, e.g., using flow cytometry, as an indicator of the percentage of PD-1+/LAG-3+ cells in the CAR-expressing cell population (e.g., a CAR19+ cell population).
  • In some embodiments of any of the methods and compositions for use disclosed herein, the level or activity one, two or more immune checkpoint inhibitors is evaluated, e.g., using flow cytometry, as an indicator of the percentage of PD-1+/TIM-3+ cells in the CAR-expressing cell population (e.g., a CAR19+ cell population).
  • In some embodiments of any of the methods and compositions for use disclosed herein, the level or activity of one, two, three, four, five, ten, twenty, fifty, sixty, seventy, one hundred or more of a biomarker or gene set listed in Table 7A, Table 7B, Table 8 and FIG. 2B predicts a subject's response to a CAR19+ cell product (e.g., CTL019).
  • In some embodiments of any of the methods and compositions for use disclosed herein, the value of responder or relapser status comprises a measure of the level or activity of one, two, three, four, five, ten, twenty or more (e.g., all) of the biomarkers having a given FDR p-value, listed herein, e.g., in a Table herein. In some embodiments, the FDR p-value is below 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001. In some embodiments, the FDR p-value is below 0.1 or 0.01. In some embodiments, the biomarkers are biomarkers listed in Table 1A, Table 1B, Table 16, Table 17, Table 18, or Table 20, or a combination thereof. In some embodiments, the measure comprises a measure of all of the biomarkers in Table 1A that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001. In some embodiments, the measure comprises a measure of all of the biomarkers in Table 1B that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001. In some embodiments, the measure comprises a measure of all of the biomarkers in Table 16 that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001. In some embodiments, the measure comprises a measure of all of the biomarkers in Table 17 that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001. In some embodiments, the measure comprises a measure of all of the biomarkers in Table 18 that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001. In some embodiments, the measure comprises a measure of all of the biomarkers in Table 20 that have a p-value below a threshold of 0.2, 0.1, 0.05, 0.02, 0.01, 0.005, 0.002, or 0.001. In some embodiments, the measure comprises a measure of all of the biomarkers having a p-value below the threshold. In some embodiments, the measure comprises a measure of one, two, three, four, five, ten, twenty, fifty, or one hundred biomarkers having a p-value below the threshold. In some embodiments, the measure comprises a measure of at least one, two, three, four, five, ten, twenty, fifty, or one hundred biomarkers having a p-value below the threshold. In some embodiments, the measure comprises a measure of 1-5, 5-10, 10-20, 20-50, or 50-100 biomarkers having a p-value below the threshold.
  • In some embodiments, biomarkers of Table 7B that are designated “CR” in the table are upregulated in complete responders compared to non-responders. In some embodiments, biomarkers of Table 7B that are designated “NR” in the table are upregulated in non-responders compared to complete responders.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the biomarker is a secreted or a cell surface biomarker listed in Table 8. For example the biomarker can be measured by flow cytometry.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a responder (e.g., a complete responder) has, or is identified as having, a greater level or activity of one, two, or more (all) of GZMK, PPF1BP2, or naïve T cells as compared to a non-responder. In some embodiments of any of the methods and compositions for use disclosed herein, a non-responder has, or is identified as having, a greater level or activity of one, two, three, four, five, six, seven, or more (e.g., all) of IL22, IL-2RA, IL-21, IRF8, IL8, CCL17, CCL22, effector T cells, or regulatory T cells, as compared to a responder.
  • In an embodiment of any of the methods and compositions for use disclosed herein, a relapser is a patient having, or who is identified as having, an increased level of expression of one or more of (e.g., 2, 3, 4, or all of) the following genes, compared to non relapsers: MIR199A1, MIR1203, uc021ovp, ITM2C, and HLA-DQB1 and/or a decreased levels of expression of one or more of (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or all of) the following genes, compared to non relapsers: PPIAL4D, TTTY10, TXLNG2P, MIR4650-1, KDMSD, USP9Y, PRKY, RPS4Y2, RPS4Y1, NCRNA00185, SULT1E1, and EIF1AY.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a complete responder has, or is identified as having, a greater, e.g., a statistically significant greater, percentage of CD8+ T cells compared to a reference value, e.g., a non-responder percentage of CD8+ T cells.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a complete responder has, or is identified as having, a greater percentage (e.g., 5%, 6%, 7%, 10%, 15%, 20%, 25%, 27%, 30%, 35%, or 40% or greater number) of CD27+CD45RO− immune effector cells, e.g., in the CD8+ population, compared to a reference value, e.g., a non-responder number of CD27+CD45RO− immune effector cells.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a complete responder or a partial responder has, or is identified as having, a greater, e.g., a statistically significant greater, percentage of CD4+ T cells compared to a reference value, e.g., a non-responder percentage of CD4+ T cells.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a complete responder has, or is identified as having, a greater percentage of one, two, three, or more (e.g., all) of resting TEFF cells, resting TREG cells, younger T cells (e.g., younger CD4 or CD8 cells, or gamma/delta T cells), or early memory T cells, or a combination thereof, compared to a reference value, e.g., a non-responder number of resting TEFF cells, resting TREG cells, younger T cells (e.g., younger CD4 or CD8 cells), or early memory T cells.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a non-responder has, or is identified as having, a greater percentage of one, two, three, or more (e.g., all) of activated TEFF cells, activated TREG cells, older T cells (e.g., older CD4 or CD8 cells), or late memory T cells, or a combination thereof, compared to a reference value, e.g., a responder number of activated TEFF cells, activated TREG cells, older T cells (e.g., older CD4 or CD8 cells), or late memory T cells.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a non-responder has, or is identified as having, a greater percentage of an immune cell exhaustion marker, e.g., one, two or more immune checkpoint inhibitors (e.g., PD-1, TIM-3 and/or LAG-3). In one embodiment, a non-responder has, or is identified as having, a greater percentage of PD-1 or LAG-3 expressing immune effector cells (e.g., CD4+ T cells and/or CD8+ T cells) (e.g., CAR-expressing CD4+ cells and/or CD8+ T cells) compared to the percentage of PD-1 or LAG-3 expressing immune effector cells from a responder.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a non-responder has, or is identified as having, a greater percentage of immune cells having an exhausted phenotype, e.g., immune cells that co-express at least two exhaustion markers, e.g., co-expresses PD-1 and TIM-3. In other embodiments, a non-responder has, or is identified as having, a greater percentage of immune cells having an exhausted phenotype, e.g., immune cells that co-express at least two exhaustion markers, e.g., co-expresses PD-1 and LAG-3.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a non-responder has, or is identified as having, a greater percentage of PD-1+/LAG-3+ cells in the CAR-expressing cell population (e.g., a CAR19+ cell population) compared to a responder (e.g., a complete responder) to the CAR-expressing cell therapy.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a partial responder has, or is identified as having, a higher percentages of PD-1+/LAG-3+ cells, than a responder, in the CAR-expressing cell population (e.g., a CAR19+ cell population).
  • In some embodiments of any of the methods and compositions for use disclosed herein, a non-responder has, or is identified as having, an exhausted phenotype of PD1+CAR+ and co-expression of LAG3 in the CAR-expressing cell population (e.g., a CAR19+ cell population).
  • In some embodiments of any of the methods and compositions for use disclosed herein, a non-responder has, or is identified as having, a greater percentage of PD-1+/TIM-3+ cells in the CAR-expressing cell population (e.g., a CAR19+ cell population) compared to the responder (e.g., a complete responder).
  • In some embodiments of any of the methods and compositions for use disclosed herein, a partial responders has, or is identified as having, a higher percentage of PD-1+/TIM-3+ cells, than responders, in the CAR-expressing cell population (e.g., a CAR19+ cell population).
  • In some embodiments of any of the methods and compositions for use disclosed herein, the presence of CD8+CD27+CD45RO− T cells in an apheresis sample is a positive predictor of the subject response to a CAR-expressing cell therapy (e.g., a CAR19 therapy (e.g., CTL019 therapy)).
  • In some embodiments of any of the methods and compositions for use disclosed herein, a high percentage of PD1+CAR+ and LAG3+ or TIM3+ T cells in an apheresis sample is a poor prognostic predictor of the subject response to a CAR-expressing cell therapy (e.g., a CAR19 therapy (e.g., CTL019 therapy)).
  • In some embodiments of any of the methods and compositions for use disclosed herein, a responder (e.g., a complete responder) to a CAR19 therapy has, or is identified as having, the biomarker profile of Table 9.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a non-responder to a CAR19 therapy has, or is identified as having, the biomarker comprising one or more of PD-1+ immune effector cells, TIM-3+ immune effector cells, LAG-3+ immune effector cells, KLRG1+ immune effector cells, CD27-immune effector cells, activated TEFF cells, activated TREG cells, activated TH1, activated TH2 cells, stimulated memory cells, or late T memory cells, or a combination thereof,
  • In some embodiments of any of the methods and compositions for use disclosed herein, a non-responder to a CAR19 therapy has, or is identified as having, the biomarker profile of Table 10.
  • In some embodiments of any of the methods and compositions for use disclosed herein, expression of one, two, three, four or more (all) of KLRG1, CD57, CD27, CD122, or CD62L is predictive of patient response to CTL019 therapy.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a non-relapser is a patient with B-ALL, and has, or is identified as having, one or more expression profiles (e.g., protein or gene expression profiles) or gene signatures characteristic of resting TEFF cells or resting TREG cells.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a relapser is a patient with B-ALL, and has, or is identified as having, one or more expression profiles (e.g., protein or gene expression profiles) or gene signatures characteristic of activated TEFF cells or activated TREG cells.
  • In some embodiments of any of the aforesaid methods and compositions for use, a TREG cell (e.g., an activated TREG cell) has upregulated expression of one or more (e.g., at least 10, 20, 30, 40, 50, 60, 70, or all) of the following biomarkers: AIM2, ALAS1, BATF, C5orf32, CCL17, CD40LG, CHAC2, CSF1, CTSL1, EBNA1BP2, EDARADD, EMP1, EPAS1, FABP5, FAM40B, FKBP4, FOSL1, GCLM, GK, GPR56, HMOX1, HSPD1, HSPE1, IKBIP, IL10, IL13, IL15RA, IL1RN, IL2RA, IL3, IL4, IL5, IL9, KCNK5, LTA, MANF, MIR1182, MIR155, MIR155HG, MYOF, NDUFAF1, NLN, NME1, NME1-NME2, PANX2, PDIA6, PGAM4, PPIL1, PPPDE2, PRDX4, PRKAR1B, PSMD1, PSMD11, PUS7, RBBP8, SLC27A2, SLC39A14, SLC43A3, SRXN1, STIP1, STT3A, TBX21, TNFRSF11A, TNFRSF1B, TNFRSF8, TNFRSF9, TXN, UCK2, VDR, VTRNA1-3, WDR12, YWHAG, ZDHHC16, or ZNF282. The upregulated expression may be, e.g., measured 16 hours after stimulation. The upregulated expression may be determined, e.g., by measuring RNA levels for the indicated genes.
  • In some embodiments of any of the aforesaid methods and compositions for use, a TEFF cell (e.g., an activated TEFF cell) has upregulated expression of one or more (e.g., at least 10, 20, 30, 40, 50, 60, 70, or all) of the following biomarkers: AIM2, ALAS1, B4GALT5, BATF, C3orf26, C4orf43, CCL3, CCL4, CCT3, CCT7, CD40LG, CHAC2, CSF2, CTNNA1, EBNA1BP2, EDARADD, EEF1E1, EIF2B3, EIF2S1, FABP5, FAM40B, FKBP4, FOSL1, GFOD1, GLRX2, HSPD1, HSPE1, IFNG, IL15RA, IL21, IL2RA, IL3, KCNK5, KIAA0020, LARP4, LRP8, LTA, MANF, MIR1182, MIR155, MIR155HG, MTCH2, MYOF, NDUFAF1, NLN, NME1, NME1-NME2, OTUD7B, PAM, PDIA6, PEA15, PFKM, PGAM1, PGAM4, PPIL1, PRDX4, PRSS23, PSMD1, PSMD11, PSMD14, PTRH2, PUS7, RBBP8, RPF2, RPP25, SFXN1, SLC27A2, SLC39A14, SLC43A3, SORD, SPR, SRXN1, STIP1, STT3A, TBX21, TMCC2, TMEM165, TNFRSF9, TXN, TXNDC5, UCK2, VDR, WDR12, YWHAG, or ZDHHC16. The upregulated expression may be, e.g., measured 16 hours after stimulation. The upregulated expression may be determined, e.g., by measuring RNA levels for the indicated genes.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a relapser is a patient with B-ALL and has, or is identified as having, one or more protein or gene expression profiles comprising one, two, three, four, five, ten or more genes according to Table 7A, Table 7B or FIG. 2B or a combination thereof.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a relapser has, or is identified as having, an elevated level of one or more TREG cell biomarkers, or a combination thereof. In some embodiments, the relapser has, or is identified as having, upregulated expression of one or more (e.g., at least 10, 20, 30, 40, 50, 60, 70, or all) of the following genes: AIM2, ALAS1, BATF, C5orf32, CCL17, CD40LG, CHAC2, CSF1, CTSL1, EBNA1BP2, EDARADD, EMP1, EPAS1, FABP5, FAM40B, FKBP4, FOSL1, GCLM, GK, GPR56, HMOX1, HSPD1, HSPE1, IKBIP, IL10, IL13, IL15RA, IL1RN, IL2RA, IL3, IL4, IL5, IL9, KCNK5, LTA, MANF, MIR1182, MIR155, MIR155HG, MYOF, NDUFAF1, NLN, NME1, NME1-NME2, PANX2, PDIA6, PGAM4, PPIL1, PPPDE2, PRDX4, PRKAR1B, PSMD1, PSMD11, PUS7, RBBP8, SLC27A2, SLC39A14, SLC43A3, SRXN1, STIP1, STT3A, TBX21, TNFRSF11A, TNFRSF1B, TNFRSF8, TNFRSF9, TXN, UCK2, VDR, VTRNA1-3, WDR12, YWHAG, ZDHHC16, or ZNF282. In certain embodiment, the relapser has, or is identified as having, upregulated expression of one or more (e.g., at least 10, 20, 25, or all) of the following genes: C5orf32, CCL17, CSF1, CTSL1, EMP1, EPAS1, GCLM, GK, GPR56, HMOX1, IKBIP, IL10, IL13, IL1RN, IL4, IL5, IL9, MIR155, PANX2, PGAM4, PRKAR1B, TNFRSF11A, TNFRSF1B, TNFRSF8, VTRNA1-3, or ZNF282. The upregulated expression may be, e.g., measured 16 hours after stimulation. The upregulated expression may be determined, e.g., by measuring RNA levels for the indicated genes.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a subject has, or is identified as having, an elevated level of one or more TEFF cell biomarkers, or a combination thereof. In some embodiments, the subject has, or is identified as having, upregulated expression of one or more (e.g., at least 10, 20, 30, 40, 50, 60, 70, or all) of the following genes: AIM2, ALAS1, B4GALT5, BATF, C3orf26, C4orf43, CCL3, CCL4, CCT3, CCT7, CD40LG, CHAC2, CSF2, CTNNA1, EBNA1BP2, EDARADD, EEF1E1, EIF2B3, EIF2S1, FABP5, FAM40B, FKBP4, FOSL1, GFOD1, GLRX2, HSPD1, HSPE1, IFNG, IL15RA, IL21, IL2RA, IL3, KCNK5, KIAA0020, LARP4, LRP8, LTA, MANF, MIR1182, MIR155, MIR155HG, MTCH2, MYOF, NDUFAF1, NLN, NME1, NME1-NME2, OTUD7B, PAM, PDIA6, PEA15, PFKM, PGAM1, PGAM4, PPIL1, PRDX4, PRSS23, PSMD1, PSMD11, PSMD14, PTRH2, PUS7, RBBP8, RPF2, RPP25, SFXN1, SLC27A2, SLC39A14, SLC43A3, SORD, SPR, SRXN1, STIP1, STT3A, TBX21, TMCC2, TMEM165, TNFRSF9, TXN, TXNDC5, UCK2, VDR, WDR12, YWHAG, or ZDHHC16. In certain embodiment, the subject has, or is identified as having, upregulated expression of one or more (e.g., at least 10, 20, 25, or all) of the following genes: B4GALT5, C3orf26, C4orf43, CCL3, CCL4, CCT3, CCT7, CSF2, CTNNA1, EEF1E1, EIF2B3, EIF2S1, GFOD1, GLRX2, IL21, IL2RA, IL3, KIAA0020, LARP4, LRP8, OTUD7B, PAM, PEA15, PFKM, PGAM1, PGAM4, PRSS23, PSMD1, PSMD11, PSMD14, PTRH2, RPF2, SORD, SPR, TMCC2, TMEM165, or TXNDC5. The upregulated expression may be, e.g., measured 16 hours after stimulation. The upregulated expression may be determined, e.g., by measuring RNA levels for the indicated genes.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the responder (e.g., the complete or partial responder) has one, two, three or more (or all) of the following profile:
  • (i) has a greater number of CD27+ immune effector cells compared to a reference value, e.g., a non-responder number of CD27+ immune effector cells;
  • (ii) has a greater number of CD8+ T cells compared to a reference value, e.g., a non-responder number of CD8+ T cells;
  • (iii) has a lower number of immune cells expressing one or more checkpoint inhibitors, e.g., a checkpoint inhibitor chosen from PD-1, LAG-3, TIM-3, or KLRG-1, or a combination, compared to a reference value, e.g., a non-responder number of cells expressing one or more checkpoint inhibitors; or
  • (iv) has a greater number of one, two, three, four or more (all) of resting TEFF cells, resting TREG cells, younger cells, naïve CD4 cells, unstimulated memory cells or early memory T cells, or a combination thereof, compared to a reference value, e.g., a non-responder number of resting TEFF cells, resting TREG cells, naïve CD4 cells, unstimulated memory cells or early memory T cells.
  • In some embodiments of any of the methods and compositions for use disclosed herein, the cytokine level or activity, e.g., of (vi), is chosen from one, two, three, four, five, six, seven, eight, or more (or all) of cytokine CCL20/MIP3a, IL17A, IL6, GM-CSF, IFNγ, IL10, IL13, IL2, IL21, IL4, IL5, IL9 or TNFα, or a combination thereof. The cytokine can be chosen from one, two, three, four or more (all) of IL-17a, CCL20, IL2, IL6, or TNFa. In one embodiment, an increased level or activity of a cytokine is chosen from one or both of IL-17a and CCL20, is indicative of increased responsiveness or decreased relapse. In some embodiments, the cytokine level is measured after T cell activation.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a transduction efficiency of 15% or higher, e.g., in (vii), is indicative of increased responsiveness or decreased relapse.
  • In some embodiments of any of the methods and compositions for use disclosed herein, a transduction efficiency of less than 15%, e.g., in (vii), is indicative of decreased responsiveness or increased relapse.
  • In another aspect, the invention features a method to identify a likely responder (e.g., a complete responder or a partial responder, a non-relapser) to a therapy comprising a CAR-expressing cell (e.g., a T cell, an NK cell) (e.g., a CD19 CAR-expressing cell therapy, e.g., described herein, e.g., a CTL019 therapy). In an embodiment, a responder status (e.g. a complete responder, a partial responder, a non-responder, a relapser or a non-relapser to a therapy comprising a CAR-expressing cell (e.g., a T cell, an NK cell)) is determined by measuring one or more of a CD19 CAR-expressing cell gene set signature, a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature, and combinations thereof. In one embodiment, a complete responder (CR) has, e.g., two, three, four or more (e.g., all) of CD27+, CD45RO, PD-1, LAG-3, and TIM-3, as described in Table 9. In one embodiment, a non-responder (NR) has, e.g., two, three, or more of (e.g., all) of CD27 CD45RO+, PD-1+, LAG-3+, and TIM-3+, as described in Table 10.
  • In an embodiment, the responder or relapser status (e.g. complete responder, partial responder, non-responder, relapser or non-relapser to a CAR-expressing cell therapy) is determined by evaluating, e.g., measuring, two, three, four, five, six, seven, eight, nine, ten, fifteen or more of a CD19 CAR-expressing cell gene set signature, a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1.
  • In an embodiment, any of the methods and compositions for use disclosed herein can be used prior to administration of a CAR-expressing cell therapy. In some embodiments, provided methods can be used before, at the same time, or during course of a CAR-expressing cell therapy.
  • In an embodiment, any of the methods and compositions for use disclosed herein can be used to identify a subject having cancer, e.g., a hematological cancer such as, e.g., CLL or ALL, as having an increased or a decreased likelihood to respond to a treatment that comprises a CAR-expressing cell (e.g., T cell, NK cell) therapy, e.g., a CD19 CAR-expressing cell therapy. The method comprises: (1) acquiring a sample from the subject (e.g., an apheresis sample obtained from the blood of the subject; and/or e.g., a manufactured product sample, e.g., genetically engineered T cells); (2) determining a level (e.g., amount or activity) of one or more biomarkers (e.g., 2, 3, 4, 5, 10, 15 or more) listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 in the sample; and (3) (optionally) comparing the determined level of the one or more markers to a reference level; and (4) identifying the subject as a complete responder, partial responder, non-responder, a relapser or non-relapser to the CAR-expressing cell therapy. In embodiments, a difference, e.g., a statistically significant difference, between the determined level compared to a reference level is predictive of the subjects responsiveness to the CAR-expressing cell therapy.
  • In an aspect, provided methods comprise (1) acquiring a sample (e.g., an apheresis sample obtained from the subject; and/or e.g., a manufactured product sample, e.g., genetically engineered T cells, e.g., a manufactured CD19 CAR-expressing cell product); (2) acquiring, e.g., determining a gene signature of the sample; and (3) (optionally) comparing the gene signature to a reference gene signature; wherein a difference, e.g., a statistically significant difference, in expression level of one or more of the biomarkers in the determined gene signature is predictive of the subjects responsiveness to the CAR-expressing cell therapy. In an embodiment, the gene signature comprises one or more markers selected from Table 1A, Table 1B, Table 2, Table 3, Table 4, Table 5, Table 6, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and combinations thereof. In one embodiment, the sample is a biological sample selected from a blood, plasma, or a serum sample. In a particular embodiment, a biological sample is a blood sample. In an embodiment, the sample is an apheresis sample, e.g., immune effector cells (e.g., T cells) obtained from the blood of the subject. In an embodiment, the sample is a manufactured product sample, e.g. genetically engineered T cells, e.g., a manufactured CD19 CAR-expressing cell product.
  • In an aspect, methods are provided for determining the responsiveness of a subject having cancer, e.g., a hematological cancer such as, e.g., CLL or ALL, to a treatment comprising a CAR-expressing cell (e.g., a T cell, an NK cell) therapy, e.g., a CD19 CAR-expressing cell therapy, e.g., described herein. The method includes: determining a level or activity of one or more biomarkers (e.g., 2, 3, 4, 5, 10, 15 or more) listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 in a sample obtained prior to treatment; wherein a difference, e.g., a statistically significant difference, in a level (e.g., amount or activity) of one or more markers in the sample relative to a predetermined value is indicative of increased responsiveness to the CAR-expressing cell. In one embodiment, the sample is a biological sample selected from a blood, plasma, or a serum sample. In a particular embodiment, a biological sample is a blood sample. In one embodiment, the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject. In an embodiment, the sample is a manufactured product sample, e.g. genetically engineered T cells, e.g., obtained from the blood of the subject, e.g., a manufactured CAR-expressing cell product, e.g., a manufactured CD19 CAR-expressing cell product.
  • In an embodiment, methods are provided for evaluating a subject having cancer, e.g., a hematological cancer such as, e.g., CLL or ALL, comprising acquiring a value of responder or relapser status for the subject that comprises a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4+ biomarker, a CD8+ biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature, and a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene set signature, thereby evaluating the subject. In an embodiment, methods comprise a measure of one or more of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature. In one embodiment, methods comprise a measure of one or more of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature, for evaluating a subject having CLL. In another embodiment, methods comprise a measure of one or more of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature, for evaluating a subject having ALL. In one embodiment, the method comprises a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature in a biological sample selected from a blood, plasma, or a serum sample. In a particular embodiment, a biological sample is a blood sample. In an embodiment, the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject. In an embodiment, the sample is a manufactured product sample, e.g. genetically engineered T cells, e.g., obtained from the blood of the subject, e.g., a manufactured CAR-expressing cell product, e.g., a manufactured CD19 CAR-expressing cell product.
  • In an embodiment, methods are provided for evaluating or monitoring the effectiveness of a CAR-expressing cell (e.g., T cell, NK cell) therapy, e.g., a CD19 CAR-expressing cell therapy, in a subject having cancer comprising acquiring a value of responder or relapser status for the subject that comprises a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature, and a CD19 CAR-expressing cell gene set signature, thereby evaluating or monitoring the effectiveness of the CAR-expressing cell therapy in the subject. In an embodiment, methods comprise a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature. In an embodiment, methods comprise a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature, for evaluating or monitoring the effectiveness of a CAR-expressing cell therapy in a subject having CLL. In another embodiment, methods comprise a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature, for evaluating or monitoring the effectiveness of a CAR-expressing cell therapy in a subject having ALL. In one embodiment, the method comprises a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature in a biological sample selected from a blood, plasma, or a serum sample. In one embodiment, a biological sample is a blood sample. In an embodiment, the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject. In an embodiment, the sample is a manufactured product sample, e.g. genetically engineered T cells, e.g., obtained from the blood of the subject.
  • In an embodiment, methods are provided for providing a prediction for success rate of a CAR-expressing cell therapy, e.g., a CD19 CAR-expressing cell therapy, e.g., described herein, in a subject having cancer, said method comprising steps of providing a biological sample from the subject; determining the levels of expression of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) genes listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), and Table 17 to obtain a gene expression pattern for the sample; and based on the gene expression pattern obtained, providing a prognosis to the subject. In an embodiment, a biological sample includes, but is not limited to a blood, plasma, or a serum sample. In a particular embodiment, a biological sample is a blood sample. In one embodiment, the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject. In an embodiment, the subject has CLL. In an embodiment, the subject has ALL.
  • In another aspect, methods for treating a subject having cancer, e.g., a hematological cancer, are provided. In an embodiment, methods are provided for treating a subject having cancer determining if a subject has a difference, e.g., a statistically significant difference, in expression level of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) markers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6) and Table 17 relative to a reference level, and if there is a difference, e.g., a statistically significant difference, between the determined level and reference level, administering to the subject a therapeutically effective dose of a CAR-expressing cell (e.g., T cell, NK cell), thereby treating the subject. In an embodiment, wherein there is a difference, e.g., a statistically significant difference, between the determined level and reference level, the method comprises modifying the CAR-expressing cell product prior to infusion into the subject. In an embodiment, wherein there is a difference, e.g., a statistically significant difference, between the determined level and the reference level, the method comprises modifying the manufacture of a CAR-expressing cell product prior to infusion into the subject. In an embodiment, if there is a difference, e.g., a statistically significant difference, between the determined level and reference level, the method comprises adjusting the CAR-expressing cell infusion dose to achieve an anti-cancer effect.
  • In an embodiment, the methods of treatment described herein comprise determining if a subject has an increased likelihood to respond to a CAR-expressing cell (e.g., T cell, NK cell) therapy, e.g., a CD19 CAR-expressing cell therapy, e.g., a CD19 CAR-expressing cell therapy described herein, by comparing the level of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15 and Table 16 (e.g., CCL20, IL-17a and/or IL-6) in a sample from the subject relative to a reference level, wherein a difference, e.g., a statistically significant difference, in expression level of one or more maker genes relative to the reference level is indicative of an increased likelihood of response; and administering to the subject a therapeutically effective dose of a CAR-expressing cell, thereby treating the subject. In one embodiment, the sample is selected from a blood, plasma or a serum sample. In one embodiment, the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject.
  • In an embodiment, the methods of treatment described herein further comprise obtaining a sample from a subject; determining a level of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1, in the sample; comparing the determined level of one or more markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1, to a reference level; and administering a therapeutically effective dose of a CAR-expressing cell (e.g., T cell, NK cell), if the subject is identified as having a difference, e.g., a statistically significant difference, in the determined level of one or more markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1, to a reference level in the sample.
  • In an embodiment, the methods of treatment described herein comprise, or further comprise, acquiring a value of responder or relapser status for the subject that comprises a measure of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature, and a CD19 CAR-expressing cell gene set signature, and responsive to a determination of responder or relapser status, performing one, two, three four or more of: identifying the subject as a complete responder, partial responder or non-responder; administering a CAR-expressing cell therapy; selecting or altering a dosing of a CAR-expressing cell therapy; selecting or altering the schedule or time course of a CAR-expressing cell therapy; administering, e.g., to a non-responder or a partial responder, an additional agent in combination with a CAR-expressing cell therapy, e.g., a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein, or a kinase inhibitor, e.g., a kinase inhibitor described herein; administering to a non-responder or partial responder a therapy that increases the number of naïve T cells in the subject prior to treatment with a CAR-expressing cell therapy; modifying a manufacturing process of a CAR-expressing cell therapy, e.g., enrich for naïve T cells prior to introducing a nucleic acid encoding a CAR, e.g., for a subject identified as a non-responder or a partial responder; or selecting an alternative therapy, e.g., a standard of care for a particular cancer (e.g., as described herein), e.g., for a non-responder, partial responder or relapser; thereby treating cancer in the subject.
  • Systems
  • In still another aspect, the present disclosure provides kits for predicting subject response to CAR-expressing cell (e.g., T cell, NK cell) therapy, e.g., a CD19 CAR-expressing cell therapy, e.g., a CD19 CAR-expressing cell therapy described herein. The kits comprise at least one reagent that specifically detects the level or activity of a set of genes (e.g., 2, 3, 4, 5, 10, 15 or more of the genes) selected from Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and/or KLRG1; and instructions for using the kits, e.g., for predicting a subject's response to a CAR-expressing cell therapy. In some embodiments, said instructions for use provide that if one or more of the detected expression levels is different from, e.g., greater than a reference level, the subject is more likely to respond positively to a CAR-expressing cell therapy. In some embodiments, said instructions for use provide that if one or more of the detected expression levels is less than a reference level, the subject is more likely to respond positively to a CAR-expressing cell therapy. In some embodiments, if the level or activity of PD-1, LAG-3, or TIM-3, or any combination thereof, is less than a reference value, the subject is more likely to respond positively to the therapy.
  • In certain embodiments, at least one reagent that specifically detects expression levels of the set of genes comprises a nucleic acid probe complementary to mRNA expressed from the genes, for example a CDNA or an oligonucleotide. The nucleic acid probe may be immobilized on a substrate surface or may be in solution. The set of reagents may detect the expression of polypeptides, e.g., surface polypeptides, encoded by said set of genes. In one embodiment, the nucleic acid probe comprises a nucleic acid of about 10, 15, 20, 25, 30, 35, 40, 45, 50 or 100 nucleic acid residues complementary the nucleic acid sequence of a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and/or KLRG1. The kits may further comprise one or more of: extraction buffer/reagents and protocol, amplification buffer/reagents and protocol, hybridization buffer/reagents and protocol, and labeling buffer/reagents and protocol.
  • In certain embodiments, the kits further comprise at least one CD19 CAR-expressing cell (e.g., T cell, NK cell) gene set signature. In one embodiment, the kit further comprises a reference standard.
  • In an embodiment, the subject has CLL.
  • In an embodiment, the subject has ALL.
  • In an embodiment, the subject has B-cell ALL.
  • In an aspect, the disclosure features a reaction mixture comprising at least one reagent that specifically detects expression levels of a set of genes (e.g., 2, 3, 4, 5, 10, 15 or more of the genes) selected from Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and/or KLRG1; and a biological sample. In one embodiment, the sample is selected from a blood, plasma or a serum sample. In one embodiment, the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject. In one embodiment, the sample comprises CAR-expressing cells, e.g., CART-expressing cells, e.g., CART19 cells.
  • In certain embodiments, at least one reagent that specifically detects expression levels of the set of genes comprises a nucleic acid probe complementary to mRNA expressed from the genes, for example a CDNA or an oligonucleotide. The nucleic acid probe may be immobilized on a substrate surface or may be in solution. The set of reagents may detect the expression of polypeptides, e.g., surface polypeptides, encoded by said set of genes. In one embodiment, the nucleic acid probe comprises a nucleic acid of about 10, 15, 20, 25, 30, 35, 40, 45, 50 or 100 nucleic acid residues complementary the nucleic acid sequence of a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and/or KLRG1. The reaction mixture may further comprise one or more of: extraction buffer/reagents, amplification buffer/reagents, hybridization buffer/reagents, and labeling buffer/reagents.
  • In another aspect, the present disclosure features a system for evaluating cancer in a subject. The system comprises at least one processor operatively connected to a memory, the at least one processor when executing is configured to perform any one or more of the steps described herein.
  • In still another aspect, the present disclosure features a system for evaluating cancer in a subject. The system includes at least one processor operatively connected to a memory, the at least one processor when executing is configured to:
      • acquire a value of responder or relapser status that comprises a measure of one, two, three, four, five, six, seven or more (all) of the following:
  • (i) the level or activity of CD27 and/or CD45RO− (e.g., CD27+CD45RO−) immune effector cells, e.g., in a CD4+ or a CD8+ T cell population in a sample (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • (ii) the level or activity of one, two, three, or more (e.g., all) of resting TEFF cells, resting TREG cells, younger T cells (e.g., younger CD4 or CD8 cells, or gamma/delta T cells), early memory T cells, or a combination thereof, in a sample (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • (iii) the level or activity of one, two, three, or more (e.g., all) of activated TEFF cells, activated TREG cells, older T cells (e.g., older CD4 or CD8 cells), or late memory T cells, or a combination thereof, in a sample (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • (iv) the level or activity of an immune cell exhaustion marker, e.g., one, two or more immune checkpoint inhibitors (e.g., PD-1, TIM-3 and/or LAG-3) in a sample (e.g., an apheresis sample or a manufactured CAR-expressing cell product sample);
  • (v) the level or activity of one, two, three, four, five, ten, twenty or more of the biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 16, Table 17, Table 18, Table 20, FIG. 2B, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature;
  • (vi) a cytokine level or activity (e.g., quality of cytokine repertoire) in a CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019), wherein the cytokine is chosen from one, two, three, four, five or more (or all) of the cytokines listed in Table 16;
  • (vii) a transduction efficiency of a CAR-expressing cell in a manufactured CAR-expressing cell product sample; or
  • (viii) a quantity of CD27+PD-1− cells in a subject, e.g., a sample from the subject (e.g., an apheresis sample or a CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019)), e.g., a quantity greater than or equal to 1×107 cells.
  • responsive to a determination of the value of responder status, perform one, two, three, four, five, six, seven, or more (e.g., all) of:
  • identify the subject as a complete responder, partial responder, non-responder, relapser or non-relapser;
  • recommend administering a CAR-expressing cell therapy;
  • recommend a selection or alteration of a dosing of a CAR-expressing cell therapy;
  • recommend a selection or alteration of a schedule or time course of a CAR-expressing cell therapy;
  • recommend administering, e.g., to a non-responder or a partial responder, an additional agent in combination with a CAR-expressing cell therapy, e.g., a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein;
  • recommend administering to a non-responder or partial responder a therapy that increases the number of naïve T cells in the subject prior to treatment with a CAR-expressing cell therapy;
  • recommend modifying a manufacturing process of a CAR-expressing cell therapy, e.g., enrich for naïve T cells prior to introducing a nucleic acid encoding a CAR, e.g., for a subject identified as a non-responder or a partial responder;
  • recommend modifying the CAR-expressing cell product prior to infusion into the patient;
  • recommend adjusting the CAR-expressing cell infusion dose to achieve clinical efficacy;
  • recommend administering an alternative therapy, e.g., for a non-responder or partial responder or relapser;
  • recommend a selection of an alternative therapy, e.g., for a non-responder or partial responder, e.g., a standard of care for a particular cancer type, or if the subject is, or is identified as, a non-responder or a relapser, recommend decreasing the TREG cell population and/or TREG gene signature, e.g., by CD25 depletion, administration of cyclophosphamide, anti-GITR antibody, mTOR inhibitor, or a combination thereof.
  • In one embodiment, the value includes a measure of a CD19 CAR-expressing cell gene set signature and a combination of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, and a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature; and responsive to a determination of the value of responder status, perform one, two, three, four or more of: identify the subject as a complete responder, partial responder, or non-responder; recommend a CAR-expressing cell therapy; recommend a selection or alteration of a dosing of a CAR-expressing cell therapy; recommend an alternative therapy, recommend a combination therapy, e.g., a combination with a CAR-expressing cell therapy, recommend or alter a manufacturing process of a CAR-expressing cell therapy.
  • In one embodiment, the at least one processor when executing is configured to: acquire a value of responder status that comprises a measure of a CD19 CAR-expressing cell gene set signature and a combination of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) of: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1; and responsive to a determination of the value of responder status, perform one, two, three, four or more of: identify the subject as a complete responder, partial responder, or non-responder; recommend a CAR-expressing cell therapy; recommend a selection or alteration of a dosing of a CAR-expressing cell therapy; recommend an alternative therapy, e.g., a standard of care for a particular cancer (e.g., as described herein); recommend a combination therapy, e.g., a combination with a CAR-expressing cell therapy, recommend or alter a manufacturing process of a CAR-expressing cell therapy, e.g., as described herein.
  • Manufacturing Methods
  • In another aspect, the invention features a method of evaluating the potency of a CAR-expressing cell product, e.g., CAR19-expressing cell product sample (e.g., CTL019). The method includes acquiring a value for one, two, three, four, five, six, seven, eight, or more (e.g., all) of:
  • (i) the level or activity of CD27 and/or CD45RO− (e.g., CD27+CD45RO−) immune effector cells, e.g., in a CD4+ or a CD8+ T cell population, in the CAR-expressing cell product;
  • (ii) the level or activity of one, two, three, or more (e.g., all) of resting TEFF cells, resting TREG cells, younger T cells (e.g., younger CD4 or CD8 cells, or gamma/delta T cells), or early memory T cells, or a combination thereof, in the CAR-expressing cell product;
  • (iii) the level or activity of one, two, three, or more (e.g., all) of activated TEFF cells, activated TREG cells, older T cells (e.g., older CD4 or CD8 cells), or late memory T cells, or a combination thereof, in the CAR-expressing cell product;
  • (iv) the level or activity of an immune cell exhaustion marker, e.g., one, two or more immune checkpoint inhibitors (e.g., PD-1, TIM-3 and/or LAG-3) in the CAR-expressing cell product;
  • (v) the level or activity of one, two, three, four, five, ten, twenty or more of the biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 16, Table 17, Table 18, Table 20, FIG. 2B, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature;
  • (vi) a cytokine level or activity in a CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019), wherein the cytokine is chosen from one, two, three, four, five or more (or all) of the cytokines listed in Table 16;
  • (vii) a transduction efficiency of CAR-expressing cells in the product;
  • (viii) a quantity of CD27+PD-1− cells in a subject, e.g., a sample from the subject (e.g., an apheresis sample or a CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019)), e.g., a quantity greater than or equal to 1×107 cells; or
  • (ix) the level or activity of a TREG cell or cell population,
  • wherein an increase in (i), (ii), (vi), (vii), (viii), or any combination thereof, is indicative of increased potency of the CAR-expressing cell product, and
  • wherein an increase in (iii), (iv), (ix), or any combination thereof is indicative of decreased potency of the CAR-expressing cell product.
  • In a related aspect, the invention features a method for optimizing manufacturing of a CAR-expressing cell product, e.g., CAR19-expressing cell product sample (e.g., CTL019). The method includes:
  • (1) acquiring a sample comprising CAR-expressing cell (e.g., a population of CAR-expressing immune effector cells);
  • (2) activating the CAR-expressing cell in vitro;
  • (3) evaluating the potency of the potency of the activated CAR-expressing cell by determining one, two, three, four, five, six, seven, eight, or more (e.g., all) of:
  • (i) the level or activity of CD27 and/or CD45RO− (e.g., CD27+CD45RO−) immune effector cells, e.g., in a CD4+ or a CD8+ T cell population, in the CAR-expressing cell product;
  • (ii) the level or activity of one, two, three, or more (e.g., all) of resting TEFF cells, resting TREG cells, younger T cells (e.g., younger CD4 or CD8 cells, or gamma/delta T cells), or early memory T cells, or a combination thereof, in the CAR-expressing cell product;
  • (iii) the level or activity of one, two, three, or more (e.g., all) of activated TEFF cells, activated TREG cells, older T cells (e.g., older CD4 or CD8 cells), or late memory T cells, or a combination thereof, in the CAR-expressing cell product;
  • (iv) the level or activity of an immune cell exhaustion marker, e.g., one, two or more immune checkpoint inhibitors (e.g., PD-1, TIM-3 and/or LAG-3) in the CAR-expressing cell product;
  • (v) the level or activity of one, two, three, four, five, ten, twenty or more of the biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 16, Table 17, Table 18, Table 20, FIG. 2B, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature;
  • (vi) a cytokine level or activity in a CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019), wherein the cytokine is chosen from one, two, three, four, five or more (or all) of the cytokines listed in Table 16;
  • (vii) a transduction efficiency of CAR-expressing cells in the product;
  • (viii) a quantity of CD27+PD-1− cells in a subject, e.g., a sample from the subject (e.g., an apheresis sample or a CAR-expressing cell product sample, e.g., CAR19-expressing cell product sample (e.g., CTL019)), e.g., a quantity greater than or equal to 1×107 cells; or
  • (ix) the level or activity of a TREG cell or cell population,
  • wherein an increase in (i), (ii), (vi), (vii), (viii), or any combination thereof is indicative of increased potency of the CAR-expressing cell product, and
  • wherein an increase in (iii), (iv), (ix), or any combination thereof, is indicative of decreased potency of the CAR-expressing cell product.
  • In a related aspect, disclosed herein is a manufacturing process of a CAR-expressing cell (e.g., T cell, NK cell) product (e.g., a CD19 CAR-expressing cell, e.g., a CD19 CAR-expressing cell described herein, e.g., CTL019) to determine the potency or efficacy of the product. In an embodiment, provided methods comprise steps of providing a biological sample from a subject, e.g., a blood, serum or plasma sample; determining the levels of expression of one or more (e.g., 2, 3, 4, 5, 10, 15 or more) genes listed in Table 1A, 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1, to obtain a gene expression pattern for the sample; (optionally) comparing the obtained gene expression pattern to a predetermined value; and determining a difference between the obtained and the predetermined value. In an embodiment, the determined difference is recorded in a quality control record.
  • In some embodiments, any of the methods disclosed herein further comprise a step of enriching for, e.g., isolating, cells having an increase in any of (i), (ii), (vi), (vii), (viii), or any combination thereof, or a decrease in any of (iii), (iv), (ix), or any combination thereof.
  • In another aspect, the invention features a method for manufacturing of a product sample, e.g., genetically engineered T cells, e.g., obtained from the blood of a subject, e.g., a manufactured CAR-expressing cell (e.g., T cell, NK cell) product, e.g., a CD 19 CAR-expressing cell product, e.g., a CD19 CAR-expressing cell product described herein, e.g., CTL019). In an embodiment, the method comprises:
  • providing a manufactured product sample, e.g., genetically engineered T cells, e.g., obtained from the blood of a subject, e.g., a manufactured CAR-expressing cell product, e.g., a CD 19 CAR-expressing cell product, e.g., a CD19 CAR-expressing cell product described herein, e.g., CTL019);
  • (i) acquiring a cytokine expression profile (e.g., of Table 14, Table 15 or Table 16 (e.g., CCL20, IL-6 and/or IL-17a) secreted from the CAR-expressing cell product; and/or
  • (ii) acquiring measure of a transduction efficiency of CAR-expressing cells in the product;
  • identifying the CAR-expressing cell product as suitable for administration based on the determined cytokine level or transduction efficiency (or both); and
  • optionally, selecting the CAR-expressing cell product for administration to a subject,
  • thereby manufacturing a CAR-expressing cell product.
  • In certain embodiments of the aforesaid manufacturing methods, the cytokine is chosen from one, two, three, four, five, six, seven, eight, or more (or all) of CCL20/MIP3a, IL17A, IL6, GM-CSF, IFNγ, IL10, IL13, IL2, IL21, IL4, IL5, IL9 or TNFα, or a combination thereof. In one embodiment, the cytokine is chosen from one, two, three, four or more (all) of IL-17a, CCL20, IL2, IL6, or TNFa. For example, the cytokine can be chosen from: one or both of IL-17a and CCL20; one or both of CCL20/MIP3a and IL17A; or one, two, or all of CCL20/MIP3a, IL17A and IL6. In one embodiment, the cytokine is CCL20/MIP3a. In another embodiment, the cytokine is IL17A. In yet another embodiment, the cytokine is IL6.
  • In certain embodiments of the aforesaid manufacturing methods, a transduction efficiency of 15% or higher is indicative of increased potency. In other embodiments, a transduction efficiency of less than 15% is indicative of decreased potency.
  • In certain embodiments, any of the aforesaid manufacturing methods further comprise reducing the number (e.g., depleting) TREG cells, e.g., via CD25-depletion, GITR depletion, or mTOR inhibition. Alternatively, or in combination, the manufacturing methods further comprise contacting the sample, e.g., the apheresis sample, with an anti-GITR antibody.
  • In certain embodiments, any of the aforesaid manufacturing methods each of (i), (ii) or (iii), (iv), (v), (vi), (vii), (viii), (ix), or any combination thereof (e.g., all) are evaluated following activation in vitro.
  • In an embodiment, the cytokine profile includes one or more (e.g., one, two, three, four, five, six or more) of CCL20 (also referred to as MIP3a), IL-17a, IL-6, GM-CSF, IFNγ, IL-10, IL-13, IL-2, IL-21, IL-4, IL-5, IL-9 and TNFα. In an embodiment, the cytokine profile includes CCL20. In an embodiment, the cytokine profile includes IL-17a. In an embodiment, the cytokine profile includes IL-6. In one embodiment, the cytokine profile includes two or more (e.g., all three) of CCL20, IL-17a and IL-6.
  • In one embodiment, the method further includes determining an expression level of one or more cytokines of Table 14, Table 15 or Table 16 (e.g., CCL20, IL-17a and/or IL-6) secreted by the CAR-expressing cell (e.g., T cell, NK cell) product. In an embodiment, secretion of one or more cytokines of Table 14, Table 15 or Table 16 (e.g., CCL20, IL-17a and/or IL-6) is in response to CAR-expressing cell product stimulation with one or more target tumor antigen(s).
  • In an embodiment, the cytokine signature or level described herein is indicative of the potency of a CAR-expressing cell product. In an embodiment, cytokine signatures described herein are markers of response to a CAR-expressing cell product in a hematological cancer (e.g., CLL and ALL).
  • In an embodiment, the cytokine signature or level described herein predict subject response to a CAR-expressing cell product.
  • In an embodiment, the cytokine signature or level described in Table 16 predict subject response to a CAR-expressing cell product.
  • In an embodiment, the IL-17a and CCL-20 expression level predict subject response to a CAR-expressing cell product.
  • In an embodiment, the method further includes one or more of (e.g., one, two, three or all of): obtaining a blood sample, e.g., a population of T cells obtained from the blood of a subject; activating the population of T cells, e.g., by a method described herein; genetically engineering a cell from the population of T cells, e.g., transducing a cell from the population of T cells, with a vector comprising a nucleic acid encoding a CAR, e.g., a CAR described herein, e.g., a CD19 CAR described herein, e.g., CTL019; expanding a population of T cells that comprises a genetically engineered T cell, e.g., a cell transduced with a nucleic acid encoding a CAR, e.g., a CAR described herein, e.g., a CD19 CAR described herein, e.g., by a method described herein. In an embodiment, the CAR transduction efficiency described herein is indicative of subject's response to a CAR-expressing cell therapy in a hematological cancer (e.g., CLL and ALL).
  • In an embodiment, a CAR transduction efficiency described herein is predictive of subject response to CAR-expressing cell therapy in a hematological disease (e.g., CLL and ALL).
  • In an embodiment, a cytokine signature or level described herein is used to improve and/or modify CAR-expressing cell product (e.g., a CD19 CAR-expressing cell product such as, e.g., CTL019) prior to infusion in patients.
  • In an embodiment, a cytokine signature or level described herein is used to assess manufactured CAR-expressing cell (e.g., T cell, NK cell) products. In an embodiment, the cytokine signature or level described herein provide an end point in manufacturing process optimization.
  • In an embodiment, any of the aforesaid manufacturing methods comprise a step of recording the result of the comparison in a quality control record for the CAR-expressing cell (e.g., T cell, NK cell) preparation (e.g., a CD19 CAR-expressing cell preparation as described herein such as, e.g., CTL019). In an embodiment, the method further comprises obtaining a preparation of T cells from a subject identified as a partial responder or non-responder and increasing the number of naïve T cells in the preparation. In one embodiment, the method further comprises introducing a nucleic acid encoding a CAR into the T cell preparation.
  • In an embodiment, the method further comprises obtaining a preparation of immune effector cells (e.g., T cells) from a subject identified as a partial responder or non-responder and who has been subsequently treated with an agent that increases the number of naïve T cells in the subject, e.g., the subject has been treated with a kinase inhibitor, e.g., an mTOR inhibitor, e.g., as described herein, and/or a checkpoint inhibitor, e.g., as described herein. In one embodiment, the method further comprises introducing a nucleic acid encoding a CAR into a plurality of the immune effector cells (e.g., T cells) of the preparation.
  • In an embodiment, the CAR-expressing cell (e.g., T cell, NK cell) product is a CD19 CAR-expressing cell, e.g., a CD19 CAR-expressing cell described herein, e.g., CTL019.
  • In still another aspect, the present disclosure provides one or more gene signatures or expression profiles that discriminate relapsers to CAR-expressing cell (e.g., T cell, NK cell) therapy from non-relapsers to CAR-expressing cell therapy in a cancer, e.g., a hematological cancer (e.g., ALL and CLL).
  • In an embodiment, the one or more gene signatures or expression profiles described herein enable manufactured product improvements, thereby reducing the likelihood of patient relapse. In an embodiment, gene signatures described herein are used to modify therapeutic application of manufactured product, thereby reducing the likelihood of patient relapse.
  • In an embodiment, the one or more gene signatures or expression profiles described herein are identified in a subject prior to CAR-expressing cell treatment (e.g., a CD19 CAR-expressing cell treatment, e.g., CTL019 therapy) that predict relapse to CAR-expressing cell treatment. In an embodiment, the one or more gene signatures or expression profiles described herein are identified in an apheresis sample. In an embodiment, the one or more gene signatures or expression profiles described herein are identified in a bone marrow sample. In an embodiment, the one or more gene signatures or expression profiles described herein are identified in a manufactured CAR-expressing cell product (e.g., a CD19 CAR-expressing cell product, e.g., CTL019) prior to infusion.
  • In an embodiment, decreasing the TREG level or gene signature in a subject prior to apheresis or during manufacturing of a CAR-expressing cell product significantly reduces the risk of subject relapse.
  • In an embodiment, a subject is pre-treated with one or more therapies that reduce TREG cells prior to collection of cells for CAR-expressing cell product manufacturing, thereby reducing the risk of subject relapse to CAR-expressing cell treatment (e.g., CTL019 treatment). In an embodiment, methods of decreasing TREG cells include, but are not limited to, administration to the subject of one or more of cyclophosphamide, anti-GITR antibody, CD25-depletion, mTOR inhibitor, or a combination thereof. Administration of one or more of cyclophosphamide, anti-GITR antibody, CD25-depletion, mTOR inhibitor, or a combination thereof, can occur before, during or after an infusion of the CAR-expressing cell product.
  • In an embodiment, a subject is pre-treated with cyclophosphamide prior to collection of cells for CAR-expressing cell product manufacturing, thereby reducing the risk of subject relapse to CAR-expressing cell treatment (e.g., CTL019 treatment).
  • In an embodiment, a subject is pre-treated with an anti-GITR antibody prior to collection of cells for CAR-expressing cell product manufacturing, thereby reducing the risk of subject relapse to CAR-expressing cell treatment (e.g., CTL019 treatment).
  • In an embodiment, a CAR-expressing cell manufacturing process is modified to deplete TREG cells prior to manufacturing of the CAR-expressing cell product (e.g., a CTL019 product). In an embodiment, CD25-depletion is used to deplete TREG cells prior to manufacturing of the CAR-expressing cell product (e.g., a CTL019 product). Accordingly, in some embodiments, the method further comprises:
  • a. providing a population of immune effector cells (e.g., T cells or NK cells); and
  • b. removing T regulatory cells from the population, thereby providing a population of T regulatory-depleted cells;
  • wherein steps a) and b) are performed prior to introducing the nucleic acid encoding the CAR to the population.
  • In embodiments of the methods, the T regulatory cells comprise CD25+ T cells, and are removed from the cell population using an anti-CD25 antibody, or fragment thereof. The anti-CD25 antibody, or fragment thereof, can be conjugated to a substrate, e.g., a bead.
  • In other embodiments, the population of T regulatory-depleted cells provided from step (b) contains less than 30%, 25%, 20%, 15%, 10%, 5%, 4%, 3%, 2%, 1% of CD25+ cells.
  • In yet other embodiments, the method further comprises removing cells from the population which express a tumor antigen that does not comprise CD25 to provide a population of T regulatory-depleted and tumor antigen depleted cells prior to introducing the nucleic acid encoding a CAR to the population. The tumor antigen can be selected from CD19, CD30, CD38, CD123, CD20, CD14 or CD11b, or a combination thereof.
  • In other embodiments, the method further comprises removing cells from the population which express a checkpoint inhibitor, to provide a population of T regulatory-depleted and inhibitory molecule depleted cells prior to introducing the nucleic acid encoding a CAR to the population. An inhibitory molecule, e.g., a checkpoint inhibitor, can be chosen from PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR (e.g., TGFRbeta), e.g., as described herein.
  • Further embodiments disclosed herein encompass providing a population of immune effector cells. The population of immune effector cells provided can be selected based upon the expression of one or more of CD3, CD28, CD4, CD8, CD45RA, and/or CD45RO. In certain embodiments, the population of immune effector cells provided are CD3+ and/or CD28+.
  • In certain embodiments of the method, the method further comprises expanding the population of cells after the nucleic acid molecule encoding a CAR has been introduced.
  • In embodiments, the population of cells is expanded for a period of 8 days or less.
  • In certain embodiments, the population of cells is expanded in culture for 5 days, and the resulting cells are more potent than the same cells expanded in culture for 9 days under the same culture conditions.
  • In other embodiments, the population of cells is expanded in culture for 5 days, and shows at least a one, two, three or four fold increase in cell doublings upon antigen stimulation as compared to the same cells expanded in culture for 9 days under the same culture conditions.
  • In yet other embodiments, the population of cells is expanded in culture for 5 days, and the resulting cells exhibit higher proinflammatory IFN-γ and/or GM-CSF levels, as compared to the same cells expanded in culture for 9 days under the same culture conditions.
  • In other embodiments, the population of cells is expanded by culturing the cells in the presence of an agent that stimulates a CD3/TCR complex associated signal and/or a ligand that stimulates a costimulatory molecule on the surface of the cells. The agent can be a bead conjugated with anti-CD3 antibody, or a fragment thereof, and/or anti-CD28 antibody, or a fragment thereof.
  • In other embodiments, the population of cells is expanded in an appropriate media that includes one or more interleukin that result in at least a 200-fold, 250-fold, 300-fold, or 350-fold increase in cells over a 14 day expansion period, as measured by flow cytometry.
  • In other embodiments, the population of cells is expanded in the presence IL-15 and/or IL-7.
  • In certain embodiments, the method further includes cryopreserving the population of the cells after the appropriate expansion period.
  • In yet other embodiments, the method of making disclosed herein further comprises contacting the population of immune effector cells with a nucleic acid encoding a telomerase subunit, e.g., hTERT. The nucleic acid encoding the telomerase subunit can be DNA.
  • In yet other embodiments, the method of making disclosed herein further comprises culturing the population of immune effector cells in serum comprising 2% hAB serum.
  • In any of the methods, systems and kits described herein, the CD19 CAR can comprise an anti-CD19 binding domain described in Table 12, or CDRs, e.g., one or more (e.g., all) of HC CDR1, HC CDR2, HC CDR3, LC CDR1, LC CDR2, and LC CDR3 of an anti-CD19 binding domain described in Table 12. In an embodiment, the CAR can comprise one of more of: a leader sequence, e.g., a leader sequence described herein, e.g., in Table 11; an anti-CD19 binding domain, e.g., an anti-CD19 binding domain described herein, e.g., in Table 12; a hinge region, e.g., a hinge region described herein, e.g., a hinge region described in Table 11; a transmembrane domain, e.g., a transmembrane domain described herein, e.g., in Table 11; and an intracellular signaling domain (e.g., a costimulatory domain and/or a primary signaling domain, e.g., a costimulatory domain described herein, e.g., in Table 11 and/or a primary signaling domain described herein, e.g., in Table 11). In an embodiment, the CD19 CAR-expressing cell (e.g., T cell, NK cell) is CTL019 or a CD19 CAR described in Table 13.
  • Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references (e.g., sequence database reference numbers) mentioned herein are incorporated by reference in their entirety. For example, all GenBank, Unigene, and Entrez sequences referred to herein, e.g., in any Table herein, e.g., in any of Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 14, Table 17, Table 18, and Table 20, are incorporated by reference. Unless otherwise specified, the sequence accession numbers specified herein, including in any Table herein, e.g., in any of Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 14, Table 17, Table 18, and Table 20, refer to the database entries current as of Oct. 8, 2014. When one gene or protein references a plurality of sequence accession numbers, all of the sequence variants are encompassed.
  • In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. Headings, sub-headings or numbered or lettered elements, e.g., (a), (b), (i) etc., are presented merely for ease of reading. The use of headings or numbered or lettered elements in this document does not require the steps or elements be performed in alphabetical order or that the steps or elements are necessarily discrete from one another. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts an exemplary model illustrating findings from a whole genome CTL019 RNAseq analysis performed on T cells at apheresis and after CTL019 manufacturing for 21 CLL and 7 ALL samples. This model demonstrates that expression patterns of complete responders (CRs) have a younger T cell phenotype than non-responders (NRs). Memory T cell subsets are differentially enriched between CRs versus NRs with CRs showing similarity to T memory stem cells.
  • FIG. 2A depicts an exemplary histogram of the number of samples analyzed in the whole genome CTL019 RNAseq analysis described herein. p=product; a=apheresis. FIG. 2B is an exemplary schematic illustrating an overview of the analysis. Briefly, for each gene set, a 3-group statistical model was applied to determine whether the meta-gene was statistically different between the CLL product CRs, PRs, and NRs. CRs are more like resting TEFF cells, whereas NR are more like activated TEFF cells. CTL019 NR samples are in a more activated state than CR samples.
  • FIG. 3 depicts an exemplary schematic of memory T cell precursors and subsets. Without wishing to be bound by a particular theory, the state of memory T cells in CTL019 samples is likely a major component of response.
  • FIG. 4 depicts an exemplary result illustrating meta-gene scores for the TSCM vs TCM upregulated gene set. The x-axis is samples by response group where a=apheresis and p=product. The y-axis is normalized meta-gene expression scores. Gene sets enriched in CLL CRs (e.g., CTL019 CRs) are also enriched in acute lymphoblastic leukemias (ALLs). ALL and CLL CRs are enriched in T stem cell (TSCM) subset specific genes, whereas CLL PRs and NRs are enriched in T central memory (TCM) subset genes. The same pattern is seen in apheresis as in product samples. ALL expression patters are most similar to CLL CRs and are even more extreme in the direction of resting/unstimulated/early memory T cells.
  • FIG. 5A depicts an exemplary result from a Principle Component Analysis (PCA) of CTL019 samples. This exemplary PCA result illustrates that CRs, ALL and Normal samples cluster separately from PRs and NRs. FIG. 5B depicts an exemplary result from a PCA of CTL019 and apheresis samples. This exemplary PCA result illustrates that CRs, ALL and Normal samples cluster separately from PRs and NRs and from the apheresis cluster.
  • FIG. 6 depicts an exemplary schematic depicting immunophenotyping of apheresis and product samples.
  • FIG. 7A and FIG. 7B depict exemplary multi-color flow cytometry analysis results identifying correlates of response in product samples. 36 manufactured CTL019 samples from CLL patients were analyzed. Samples included 5 CR, 8 PR, 19NR and 3 pending. FIG. 7A depicts an exemplary result illustrating percent CD4+ cells and patient response. FIG. 7B depicts an exemplary result illustrating percent CD8+ cells and patient response.
  • FIG. 8A, FIG. 8B, FIG. 8C and FIG. 8D depicts an exemplary flow cytometry analysis of PD1 and CAR19 expression on T cells. FIG. 8A and FIG. 8B are representative flow cytometry profiles demonstrating the distribution of PD-1 and CAR19 expression on CD4+ T cells from subjects that are complete responders (CR) or non-responders (NR) to CAR-expressing cell therapy. FIG. 8C is a graph showing the percent of PD1 cells in the CD4+ T cell population from groups of subjects with different responses to CAR-expressing cell therapy. FIG. 8D is a graph showing the percent of PD1 cells in the CD8+ T cell population from groups of subjects with different responses to CAR-expressing cell therapy.
  • FIG. 9A and FIG. 9B depict an exemplary flow cytometry analysis of PD1, CAR 19, and LAG-3 expression on T cells from subjects that are complete responders (CR) or non-responders (NR) to CAR-expressing cell therapy. FIG. 9C depicts exemplary results that show the distribution of PD1 and LAG-3 expression from groups of subjects with different responses to CAR-expressing cell therapy. Non-responder (NR) products have higher percentages of PD1+CAR19+LAG3+ T cells than CRs. These data demonstrate that NR products exhibit an exhausted phenotype of PD1+CAR+ and co-expression of LAG3.
  • FIG. 10A and FIG. 10B depict an exemplary flow cytometry analysis of PD1, CAR 19, and TIM-3 expression on T cells from subjects that are complete responders (CR) or non-responders (NR) to CAR-expressing cell therapy. FIG. 10C depicts exemplary results that show the distribution of PD1 and TIM-3 expression from groups of subjects with different responses to CAR-expressing cell therapy. Non-responder (NR) products have higher percentages of CAR19+PD1+TIM3+ cells than CRs. These data demonstrate that NR products exhibit an exhausted phenotype of PD1+CAR+ and co-expression of TIM3.
  • FIG. 11 depicts an exemplary result illustrating that CD27 levels in the CAR product correlate with patient response. CRs CD8+ cells displayed a higher percentage of CD27+ cells as compared to PRs and NRs.
  • FIG. 12 depicts an exemplary multi-color flow cytometry analysis result identifying correlates of response in apheresis samples. 26 apheresed samples from CLL patients were analyzed. Samples included 4 CR, 6 PR, 14NR and 1 patient was not infused.
  • FIG. 13 depicts an exemplary multi-color flow cytometry analysis result illustrating a correlation between a younger T cell phenotype and response to CTL019 therapy. These data demonstrate that the percentage of CD27+CD45RO− in CD8+ T cells is predictive of which CLL patients will undergo a complete response to CTL019.
  • FIG. 14 depicts an exemplary analysis of apheresis in a human patient prior to CTL019 therapy. Exemplary results illustrate that while patient 1000-00045 presented with very few T cells, 27% of the T cells were CD8+CD27+CD45RO−.
  • FIG. 15 depicts an exemplary result of a patient response (patient 1000-00045) to CTL019 therapy. CD8+CD27+CD45RO− T cells were a positive predictor of the patient response to CTL019 therapy. These exemplary results illustrate that a good prognostic phenotype in apheresis is a high percentage of CD8+CD27+CD45RO− T cells (naïve or TSCM phenotype). A poor prognostic phenotype in CTL019 product is a high percentage of PD1+CAR+ and LAG3+ or TIM3+ T cells (exhausted phenotype).
  • FIG. 16 depicts an exemplary block diagram of a computer system on which various aspects and embodiments may be practiced.
  • FIG. 17 depicts an exemplary heatmap showing bi-clustering of cytokine expression in stimulated CTL019 products and CLL patients. Two clusters (Cluster 1 and Cluster 3) were almost exclusively comprised of CRs and PRs, whereas the other two clusters (Cluster 2 and Cluster 4) contained predominantly NRs. On average, cytokine expression levels were higher in CRs/PRs versus NRs. The top panel shows the red channel of the heatmap image, the center panel shows the blue channel, and the bottom panel shows the green channel.
  • FIG. 18 depicts exemplary results of log-normalized expressions of statistically significant cytokines (e.g., CCL20/MIP3a, IL2, TNFα, IL17a and IL6) to distinguish CRs, PRs and NRs in CLL patients.
  • FIG. 19A depicts an exemplary scatter plot showing log-normalized correlation of IL17A (y-axis) and CCL20 (x-axis) expression. Dashed lines represent the classification boundary for separating NRs from CRs/PRs. Each dot represents a CLL patient, and the cross-hatch (NR), black (PR) and white (CR) represent the clinical response. The correlation coefficient is represented by “r” (e.g., a correlation coefficient of 0.928) and corresponding p-value for correlation using “p.value” (e.g., corresponding p-value of 1.36e-09). FIG. 19B depicts an exemplary scatter plot showing correlation of CCL20 with percentage of CAR+ cells with a correlation coefficient of 0.395 and corresponding p-value of 0.0761. Each dot represents a CLL patient, and the cross-hatch (NR), black (PR) and white (CR) represent the clinical response. The correlation coefficient is represented by “r” and corresponding p-value for correlation using “p.value”. FIG. 19C depicts an exemplary scatter plot showing correlation of IL17a with percentage of CAR+ cells with a correlation coefficient of 0.278 and corresponding p-value of 0.222. Each dot represents a CLL patient, and the cross-hatch (NR), black (PR) and white (CR) represent the clinical response. The correlation coefficient is represented by “r” and corresponding p-value for correlation using “p.value”.
  • FIG. 20 depicts exemplary results (p=0.000215) illustrating that TREG genes have high expression levels in relapsers (R) compared to non-relapser, complete responders (CR). The x-axis is samples by response group where CR=complete responder and R=relapser. The y-axis is normalized meta-gene expression scores.
  • FIG. 21 depicts an exemplary scatter plot showing percent of CAR+ cells (i.e., transduction rate) at pre-harvest for complete responders (CR) in white, partial responders (PR) in black and non-responders (NR) in hatching. Transduction efficiencies were measured pre-harvest and correlated with subject response (e.g., CR, PR, or NR). The solid line represents a 15% transduction efficiency that separates the majority of non-responders from responders. Without wishing to be bound to a particular theory, these data indicate that pre-harvest CAR transduction rate is a marker of response to CAR-expressing cell (e.g., T cell, NK cell) therapy in CLL.
  • FIG. 22 is a bar graph depicting the relationship between number of CD27+PD1− CART cells infused and response to therapy.
  • FIG. 23 is a scatter plot depicting the relationship between number of CD27+PD1-CART cells infused and response to therapy.
  • DEFINITIONS
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains.
  • The term “a” and “an” refers to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.
  • The term “about” when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of ±20% or in some instances ±10%, or in some instances ±5%, or in some instances ±1%, or in some instances ±0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.
  • “Acquire” or “acquiring” as the terms are used herein, refer to obtaining possession of a physical entity (e.g., a sample, a polypeptide, a nucleic acid, or a sequence), or a value, e.g., a numerical value, by “directly acquiring” or “indirectly acquiring” the physical entity or value. “Directly acquiring” means performing a process (e.g., performing a synthetic or analytical method) to obtain the physical entity or value. “Indirectly acquiring” refers to receiving the physical entity or value from another party or source (e.g., a third party laboratory that directly acquired the physical entity or value). Directly acquiring a physical entity includes performing a process that includes a physical change in a physical substance, e.g., a starting material. Exemplary changes include making a physical entity from two or more starting materials, shearing or fragmenting a substance, separating or purifying a substance, combining two or more separate entities into a mixture, performing a chemical reaction that includes breaking or forming a covalent or non-covalent bond. Directly acquiring a value includes performing a process that includes a physical change in a sample or another substance, e.g., performing an analytical process which includes a physical change in a substance, e.g., a sample, analyte, or reagent (sometimes referred to herein as “physical analysis”), performing an analytical method, e.g., a method which includes one or more of the following: separating or purifying a substance, e.g., an analyte, or a fragment or other derivative thereof, from another substance; combining an analyte, or fragment or other derivative thereof, with another substance, e.g., a buffer, solvent, or reactant; or changing the structure of an analyte, or a fragment or other derivative thereof, e.g., by breaking or forming a covalent or non-covalent bond, between a first and a second atom of the analyte; or by changing the structure of a reagent, or a fragment or other derivative thereof, e.g., by breaking or forming a covalent or non-covalent bond, between a first and a second atom of the reagent.
  • The term “antibody,” as used herein, refers to a protein, or polypeptide sequence derived from an immunoglobulin molecule which specifically binds with an antigen. Antibodies can be polyclonal or monoclonal, multiple or single chain, or intact immunoglobulins, and may be derived from natural sources or from recombinant sources. Antibodies can be tetramers of immunoglobulin molecules.
  • The term “altered level of expression” of a biomarker as described herein (e.g., a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene signature) refers to an increase (or decrease) in the expression level of a marker in a test sample, such as a sample derived from a patient suffering from cancer (e.g., a hematological cancer such as ALL and CLL) that is greater or less than the standard error of the assay employed to assess expression. In embodiments, the alteration can be at least twice, at least twice three, at least twice four, at least twice five, or at least twice ten or more times greater than or less than the expression level of the biomarkers in a control sample (e.g., a sample from a healthy subject not having the associated disease), or the average expression level in several control samples. An “altered level of expression” can be determined at the protein or nucleic acid (e.g., mRNA) level.
  • The term “antibody fragment” refers to at least one portion of an antibody, that retains the ability to specifically interact with (e.g., by binding, steric hindrance, stabilizing/destabilizing, spatial distribution) an epitope of an antigen. Examples of antibody fragments include, but are not limited to, Fab, Fab, F(ab)2, Fv fragments, scFv antibody fragments, disulfide-linked Fvs (sdFv), a Fd fragment consisting of the VH and CH1 domains, linear antibodies, single domain antibodies such as sdAb (either VL or VH), camelid VHH domains, multi-specific antibodies formed from antibody fragments such as a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region, and an isolated CDR or other epitope binding fragments of an antibody. An antigen binding fragment can also be incorporated into single domain antibodies, maxibodies, minibodies, nanobodies, intrabodies, diabodies, triabodies, tetrabodies, v-NAR and bis-scFv (see, e.g., Hollinger and Hudson, Nature Biotechnology 23:1126-1136, 2005). Antigen binding fragments can also be grafted into scaffolds based on polypeptides such as a fibronectin type III (Fn3)(see U.S. Pat. No. 6,703,199, which describes fibronectin polypeptide minibodies). The term “scFv” refers to a fusion protein comprising at least one antibody fragment comprising a variable region of a light chain and at least one antibody fragment comprising a variable region of a heavy chain, wherein the light and heavy chain variable regions are contiguously linked via a short flexible polypeptide linker, and capable of being expressed as a single chain polypeptide, and wherein the scFv retains the specificity of the intact antibody from which it is derived. Unless specified, as used herein an scFv may have the VL and VH variable regions in either order, e.g., with respect to the N-terminal and C-terminal ends of the polypeptide, the scFv may comprise VL-linker-VH or may comprise VH-linker-VL.
  • The term “antibody heavy chain,” refers to the larger of the two types of polypeptide chains present in antibody molecules in their naturally occurring conformations, and which normally determines the class to which the antibody belongs.
  • The term “antibody light chain,” refers to the smaller of the two types of polypeptide chains present in antibody molecules in their naturally occurring conformations. Kappa (κ) and lambda (λ) light chains refer to the two major antibody light chain isotypes.
  • The term “complementarity determining region” or “CDR,” as used herein, refers to the sequences of amino acids within antibody variable regions which confer antigen specificity and binding affinity. For example, in general, there are three CDRs in each heavy chain variable region (e.g., HCDR1, HCDR2, and HCDR3) and three CDRs in each light chain variable region (LCDR1, LCDR2, and LCDR3). The precise amino acid sequence boundaries of a given CDR can be determined using any of a number of well-known schemes, including those described by Kabat et al. (1991), “Sequences of Proteins of Immunological Interest,” 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (“Kabat” numbering scheme), Al-Lazikani et al., (1997) JMB 273,927-948 (“Chothia” numbering scheme), or a combination thereof Under the Kabat numbering scheme, in some embodiments, the CDR amino acid residues in the heavy chain variable domain (VH) are numbered 31-35 (HCDR1), 50-65 (HCDR2), and 95-102 (HCDR3); and the CDR amino acid residues in the light chain variable domain (VL) are numbered 24-34 (LCDR1), 50-56 (LCDR2), and 89-97 (LCDR3). Under the Chothia numbering scheme, in some embodiments, the CDR amino acids in the VH are numbered 26-32 (HCDR1), 52-56 (HCDR2), and 95-102 (HCDR3); and the CDR amino acid residues in the VL are numbered 26-32 (LCDR1), 50-52 (LCDR2), and 91-96 (LCDR3). In a combined Kabat and Chothia numbering scheme, in some embodiments, the CDRs correspond to the amino acid residues that are part of a Kabat CDR, a Chothia CDR, or both. For instance, in some embodiments, the CDRs correspond to amino acid residues 26-35 (HCDR1), 50-65 (HCDR2), and 95-102 (HCDR3) in a VH, e.g., a mammalian VH, e.g., a human VH; and amino acid residues 24-34 (LCDR1), 50-56 (LCDR2), and 89-97 (LCDR3) in a VL, e.g., a mammalian VL, e.g., a human VL.
  • The term “anti-cancer effect” refers to a biological effect which can be manifested by various means, including but not limited to, e.g., a decrease in tumor volume, a decrease in the number of cancer cells, a decrease in the number of metastases, an increase in life expectancy, decrease in cancer cell proliferation, decrease in cancer cell survival, or amelioration of various physiological symptoms associated with the cancerous condition. An “anti-cancer effect” can also be manifested by the ability of the peptides, polynucleotides, cells and antibodies in prevention of the occurrence of cancer in the first place. The term “anti-tumor effect” refers to a biological effect which can be manifested by various means, including but not limited to, e.g., a decrease in tumor volume, a decrease in the number of tumor cells, a decrease in tumor cell proliferation, or a decrease in tumor cell survival.
  • The term “allogeneic” refers to any material derived from a different animal of the same species as the individual to whom the material is introduced. Two or more individuals are said to be allogeneic to one another when the genes at one or more loci are not identical. In some aspects, allogeneic material from individuals of the same species may be sufficiently unlike genetically to interact antigenically.
  • The term “apheresis” as used herein refers to an extracorporeal process by which the blood of a donor or patient is removed from the donor or patient and passed through an apparatus that separates out selected particular constituent(s) and returns the remainder to the circulation of the donor or patient, e.g., by retransfusion. Thus, in the context of “an apheresis sample” refers to a sample obtained using apheresis.
  • The term “autologous” refers to any material derived from the same individual to whom it is later to be re-introduced into the individual.
  • A “biomarker” or “marker” is a gene, mRNA, or protein that undergoes alterations in expression that are associated with progression of cancer (e.g., a hematological cancer such as ALL and CLL) or responsiveness to treatment. The alteration can be in amount and/or activity in a biological sample (e.g., a blood, plasma, or a serum sample) obtained from a subject having cancer, as compared to its amount and/or activity, in a sample obtained from a baseline or prior value for the subject, the subject at a different time interval, an average or median value for a cancer patient population, a healthy control, or a healthy subject population (e.g., a control); such alterations in expression and/or activity are associated with of the responsiveness of a subject having a cancer disease state (e.g., a hematological cancer such as ALL and CLL) to a CAR-expressing cell (e.g., a CAR-expressing immune effector cell (e.g., a CAR-expressing T cell, NK cell) therapy, e.g., a CD19 CAR-expressing cell therapy. For example, a marker of the invention which is predictive of responsiveness to therapeutics can have an altered expression level, protein level, or protein activity, in a biological sample obtained from a subject having, or suspected of having, cancer as compared to a biological sample obtained from a control subject.
  • The term “cancer” refers to a disease characterized by the uncontrolled growth of aberrant cells. Cancer cells can spread locally or through the bloodstream and lymphatic system to other parts of the body. Examples of various cancers are described herein and include but are not limited to, breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, renal cancer, liver cancer, brain cancer, lymphoma, leukemia, lung cancer and the like. Cancers include, but are not limited to, B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, and Waldenstrom macroglobulinemia. In an embodiment, the cancer is associated with CD19 expression. The terms “tumor” and “cancer” are used interchangeably herein, e.g., both terms encompass solid and liquid tumors. As used herein, the term “cancer” or “tumor” includes premalignant, as well as malignant cancers and tumors.
  • The terms “cancer associated antigen” or “tumor antigen” interchangeably refers to a molecule (typically protein, carbohydrate or lipid) that is preferentially expressed on the surface of a cancer cell, either entirely or as a fragment (e.g., MHC/peptide), in comparison to a normal cell, and which is useful for the preferential targeting of a pharmacological agent to the cancer cell. In some embodiments, a tumor antigen is a marker expressed by both normal cells and cancer cells, e.g., a lineage marker, e.g., CD19 on B cells. In some embodiments, a cancer-associated antigen is a cell surface molecule that is overexpressed in a cancer cell in comparison to a normal cell, for instance, 1-fold over expression, 2-fold overexpression, 3-fold overexpression or more in comparison to a normal cell. In some embodiments, a cancer-associated antigen is a cell surface molecule that is inappropriately synthesized in the cancer cell, for instance, a molecule that contains deletions, additions or mutations in comparison to the molecule expressed on a normal cell. In some embodiments, a tumor antigen will be expressed exclusively on the cell surface of a cancer cell, entirely or as a fragment (e.g., MHC/peptide), and not synthesized or expressed on the surface of a normal cell. In some embodiments, the CARs of the present invention includes CARs comprising an antigen binding domain (e.g., antibody or antibody fragment) that binds to a MHC presented peptide. Normally, peptides derived from endogenous proteins fill the pockets of Major histocompatibility complex (MHC) class I molecules, and are recognized by T cell receptors (TCRs) on CD8+T lymphocytes. The MHC class I complexes are constitutively expressed by all nucleated cells. In cancer, virus-specific and/or tumor-specific peptide/MHC complexes represent a unique class of cell surface targets for immunotherapy. TCR-like antibodies targeting peptides derived from viral or tumor antigens in the context of human leukocyte antigen (HLA)-A1 or HLA-A2 have been described (see, e.g., Sastry et al., J Virol. 2011 85(5):1935-1942; Sergeeva et al., Blood, 2011 117(16):4262-4272; Verma et al., J Immunol 2010 184(4):2156-2165; Willemsen et al., Gene Ther 2001 8(21):1601-1608; Dao et al., Sci Transl Med 2013 5(176):176ra33; Tassev et al., Cancer Gene Ther 2012 19(2):84-100). For example, TCR-like antibody can be identified from screening a library, such as a human scFv phage displayed library.
  • As used herein, the term “CD19” refers to the Cluster of Differentiation 19 protein, which is an antigenic determinant detectable on leukemia precursor cells. The human and murine amino acid and nucleic acid sequences can be found in a public database, such as GenBank, UniProt and Swiss-Prot. For example, the amino acid sequence of human CD19 can be found as UniProt/Swiss-Prot Accession No. P15391 and the nucleotide sequence encoding of the human CD19 can be found at Accession No. NM_001178098. As used herein, “CD19” includes proteins comprising mutations, e.g., point mutations, fragments, insertions, deletions and splice variants of full length wild-type CD19. CD19 is expressed on most B lineage cancers, including, e.g., acute lymphoblastic leukemia, chronic lymphocyte leukemia and non-Hodgkin lymphoma. Other cells which express CD19 are provided below in the definition of “disease associated with expression of CD19.” It is also an early marker of B cell progenitors. See, e.g., Nicholson et al., MOL. IMMUN. 34 (16-17): 1157-1165 (1997). In one aspect the antigen-binding portion of the CAR-expressing cell (e.g., T cell, NK cell) recognizes and binds an antigen within the extracellular domain of the CD19 protein. In one aspect, the CD19 protein is expressed on a cancer cell. In one embodiment, the CD19 has a wild-type sequence, e.g., a wild-type human sequence. In another embodiment, the CD19 has a mutant sequence, e.g., a mutant human sequence.
  • The term “Chimeric Antigen Receptor” or alternatively a “CAR” refers to a set of polypeptides, typically two in the simplest embodiments, which when in an immune effector cell, provides the cell with specificity for a target cell, typically a cancer cell, and with intracellular signal generation. In some embodiments, a CAR comprises at least an extracellular antigen binding domain, a transmembrane domain and a cytoplasmic signaling domain (also referred to herein as “an intracellular signaling domain”) comprising a functional signaling domain derived from a stimulatory molecule and/or costimulatory molecule as defined below. In some embodiments, the set of polypeptides are in the same polypeptide chain (e.g., comprise a chimeric fusion protein). In some embodiments, the set of polypeptides are not contiguous with each other, e.g., are in different polypeptide chains. In some aspects, the set of polypeptides include a dimerization switch that, upon the presence of a dimerization molecule, can couple the polypeptides to one another, e.g., can couple an antigen binding domain to an intracellular signaling domain. In one aspect, the stimulatory molecule of the CAR is the zeta chain associated with the T cell receptor complex. In one aspect, the cytoplasmic signaling domain comprises a primary signaling domain (e.g., a primary signaling domain of CD3-zeta). In one aspect, the cytoplasmic signaling domain further comprises one or more functional signaling domains derived from at least one costimulatory molecule as defined below. In one aspect, the costimulatory molecule is chosen from 4-1BB (i.e., CD137), CD27, ICOS, and/or CD28. In one aspect, the CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a stimulatory molecule. In one aspect, the CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a co-stimulatory molecule and a functional signaling domain derived from a stimulatory molecule. In one aspect, the CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising two functional signaling domains derived from one or more co-stimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule. In one aspect, the CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising at least two functional signaling domains derived from one or more co-stimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule. In one aspect the CAR comprises an optional leader sequence at the amino-terminus (N-ter) of the CAR fusion protein. In one aspect, the CAR further comprises a leader sequence at the N-terminus of the extracellular antigen recognition domain, wherein the leader sequence is optionally cleaved from the antigen recognition domain (e.g., a scFv) during cellular processing and localization of the CAR to the cellular membrane. In an embodiment, the CAR is CTL019.
  • The portion of the CAR composition comprising an antibody or antibody fragment thereof may exist in a variety of forms where the antigen binding domain is expressed as part of a contiguous polypeptide chain including, for example, a single domain antibody fragment (sdAb), a single chain antibody (scFv) and a humanized antibody (Harlow et al., 1999, In: USING ANTIBODIES: A LABORATORY MANUAL, COLD SPRING HARBOR LABORATORY PRESS, NY; Harlow et al., 1989, In: ANTIBODIES: A LABORATORY MANUAL, Cold Spring Harbor, N.Y.; Houston et al., 1988, PROC. NATL. ACAD. SCI. USA 85:5879-5883; Bird et al., 1988, Science 242:423-426). In one aspect, the antigen binding domain of a CAR composition of the invention comprises an antibody fragment. In a further aspect, the CAR comprises an antibody fragment that comprises a scFv.
  • As used herein, the term “binding domain” or “antibody molecule” refers to a protein, e.g., an immunoglobulin chain or fragment thereof, comprising at least one immunoglobulin variable domain sequence. The term “binding domain” or “antibody molecule” encompasses antibodies and antibody fragments. In an embodiment, an antibody molecule is a multispecific antibody molecule, e.g., it comprises a plurality of immunoglobulin variable domain sequences, wherein a first immunoglobulin variable domain sequence of the plurality has binding specificity for a first epitope and a second immunoglobulin variable domain sequence of the plurality has binding specificity for a second epitope. In an embodiment, a multispecific antibody molecule is a bispecific antibody molecule. A bispecific antibody has specificity for no more than two antigens. A bispecific antibody molecule is characterized by a first immunoglobulin variable domain sequence which has binding specificity for a first epitope and a second immunoglobulin variable domain sequence that has binding specificity for a second epitope.
  • The portion of the CAR of the invention comprising an antibody or antibody fragment thereof may exist in a variety of forms where the antigen binding domain is expressed as part of a contiguous polypeptide chain including, for example, a single domain antibody fragment (sdAb), a single chain antibody (scFv), a humanized antibody, or bispecific antibody (Harlow et al., 1999, In: Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY; Harlow et al., 1989, In: Antibodies: A Laboratory Manual, Cold Spring Harbor, N.Y.; Houston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883; Bird et al., 1988, Science 242:423-426). In one aspect, the antigen binding domain of a CAR composition of the invention comprises an antibody fragment. In a further aspect, the CAR comprises an antibody fragment that comprises a scFv.
  • The phrase “disease associated with expression of CD19” includes, but is not limited to, a disease associated with expression of CD19 (e.g., wild type or mutant CD19) or condition associated with cells which express, or at any time expressed, CD19 including, e.g., proliferative diseases such as a cancer or malignancy or a precancerous condition such as a myelodysplasia, a myelodysplastic syndrome or a preleukemia; or a noncancer related indication associated with cells which express CD19. For the avoidance of doubt, a disease associated with expression of CD19 may include a condition associated with cells which do not presently express CD19, e.g., because CD19 expression has been downregulated, e.g., due to treatment with a molecule targeting CD19, e.g., a CD19 CAR, but which at one time expressed CD19. In one aspect, a cancer associated with expression of CD19 is a hematological cancer. In one aspect, the hematological cancer is a leukemia or a lymphoma. In one aspect, a cancer associated with expression of CD19 includes cancers and malignancies including, but not limited to, e.g., one or more acute leukemias including but not limited to, e.g., acute myeloid leukemia (AML), B-cell acute lymphocytic leukemia (“B-ALL”), T-cell acute lymphocytic leukemia (“T-ALL”), acute lymphocytic leukemia (ALL); one or more chronic leukemias including but not limited to, e.g., chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL). Additional cancers or hematologic conditions associated with expression of CD19 comprise, but are not limited to, e.g., B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, myeloproliferative neoplasm; a histiocytic disorder (e.g., a mast cell disorder or a blastic plasmacytoid dendritic cell neoplasm); a mast cell disorder, e.g., systemic mastocytosis or mast cell leukemia; B-cell prolymphocytic leukemia, plasma cell myeloma, and “preleukemia” which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells, and the like. Further diseases associated with expression of CD19 expression include, but not limited to, e.g., atypical and/or non-classical cancers, malignancies, precancerous conditions or proliferative diseases associated with expression of CD19. Non-cancer related indications associated with expression of CD19 include, but are not limited to, e.g., autoimmune disease, (e.g., lupus), inflammatory disorders (allergy and asthma) and transplantation. In some embodiments, the tumor antigen-expressing cells express, or at any time expressed, mRNA encoding the tumor antigen. In an embodiment, the tumor antigen-expressing cells produce the tumor antigen protein (e.g., wild-type or mutant), and the tumor antigen protein may be present at normal levels or reduced levels. In an embodiment, the tumor antigen-expressing cells produced detectable levels of a tumor antigen protein at one point, and subsequently produced substantially no detectable tumor antigen protein. In other embodiments, the disease is a CD19-negative cancer, e.g., a CD19-negative relapsed cancer. In some embodiments, the tumor antigen (e.g., CD19)-expressing cell expresses, or at any time expressed, mRNA encoding the tumor antigen. In an embodiment, the tumor antigen (e.g., CD19)-expressing cell produces the tumor antigen protein (e.g., wild-type or mutant), and the tumor antigen protein may be present at normal levels or reduced levels. In an embodiment, the tumor antigen (e.g., CD19)-expressing cell produced detectable levels of a tumor antigen protein at one point, and subsequently produced substantially no detectable tumor antigen protein.
  • The term “costimulatory molecule” refers to the cognate binding partner on a T cell that specifically binds with a costimulatory ligand, thereby mediating a costimulatory response by the T cell, such as, but not limited to, proliferation. Costimulatory molecules are cell surface molecules other than antigen receptors or their ligands that are required for an efficient immune response. Costimulatory molecules include, but are not limited to MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signalling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM-1, ICOS (CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, NKG2D, NKG2C, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, CD19a, and a ligand that specifically binds with CD83.
  • A costimulatory intracellular signaling domain refers to an intracellular portion of a costimulatory molecule. The intracellular signaling domain can comprise the entire intracellular portion, or the entire native intracellular signaling domain, of the molecule from which it is derived, or a functional fragment thereof.
  • As used herein, a “value of responder or relapser status” includes a measure (e.g., level) predictive of responsiveness or relapse of a subject to a treatment (e.g., a treatment that comprises, or consists of, a CAR-expressing cell therapy as described herein). In some embodiments, the measure is qualitative or quantitative. In some embodiments, the value of responder or relapser status is complete responder, partial responder, non-responder, relapser or non-relapser. In some embodiments, the value of responder or relapser status is a probability of being a complete responder, a partial responder, a non-responder, a relapser or a non-relapser. In some embodiments, the value of responder or relapser status can be determined based on the measure of any of (i)-(viii) as described herein.
  • With respect to responsiveness, a subject responds to treatment if a parameter of a cancer (e.g., a hematological cancer, e.g., cancer cell growth, proliferation and/or survival) in the subject is retarded or reduced by a detectable amount, e.g., about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more as determined by any appropriate measure, e.g., by mass, cell count or volume. In one example, a subject responds to treatment if the subject experiences a life expectancy extended by about 5%, 10%, 20%, 30%, 40%, 50% or more beyond the life expectancy predicted if no treatment is administered. In another example, a subject responds to treatment, if the subject has an increased disease-free survival, overall survival or increased time to progression.
  • Several methods can be used to determine if a patient responds to a treatment including, for example, criteria provided by NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). For example, in the context of B-ALL, a complete response or complete responder, may involve one or more of: <5% BM blast, >1000 neutrophil/ANC (/μL). >100,000 platelets (/μL) with no circulating blasts or extramedullary disease (No lymphadenopathy, splenomegaly, skin/gum infiltration/testicular mass/CNS involvement), Trilineage hematopoiesis, and no recurrence for 4 weeks. A partial responder may involve one or more of ≥50% reduction in BM blast, >1000 neutrophil/ANC (/μL). >100,000 platelets (/μL). A non-responder can show disease progression, e.g., >25% in BM blasts.
  • A “complete responder” as used herein refers to a subject having a disease, e.g., a cancer, who exhibits a complete response, e.g., a complete remission, to a treatment. A complete response may be identified, e.g., using the NCCN Guidelines®, or Cheson et al, J Clin Oncol 17:1244 (1999) and Cheson et al., “Revised Response Criteria for Malignant Lymphoma”, J Clin Oncol 25:579-586 (2007) (both of which are incorporated by reference herein in their entireties), as described herein.
  • A “partial responder” as used herein refers to a subject having a disease, e.g., a cancer, who exhibits a partial response, e.g., a partial remission, to a treatment. A partial response may be identified, e.g., using the NCCN Guidelines®, or Cheson criteria as described herein.
  • A “non-responder” as used herein refers to a subject having a disease, e.g., a cancer, who does not exhibit a response to a treatment, e.g., the patient has stable disease or progressive disease. A non-responder may be identified, e.g., using the NCCN Guidelines®, or Cheson criteria as described herein.
  • The term “relapse” as used herein refers to reappearance of a disease (e.g., cancer) after an initial period of responsiveness, e.g., after prior treatment with a therapy, e.g., cancer therapy (e.g., complete response or partial response). The initial period of responsiveness may involve the level of cancer cells falling below a certain threshold, e.g., below 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%. The reappearance may involve the level of cancer cells rising above a certain threshold, e.g., above 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%. For example, e.g., in the context of B-ALL, the reappearance may involve, e.g., a reappearance of blasts in the blood, bone marrow (>5%), or any extramedullary site, after a complete response. A complete response, in this context, may involve <5% BM blast. More generally, in an embodiment, a response (e.g., complete response or partial response) can involve the absence of detectable MRD (minimal residual disease). In an embodiment, the initial period of responsiveness lasts at least 1, 2, 3, 4, 5, or 6 days; at least 1, 2, 3, or 4 weeks; at least 1, 2, 3, 4, 6, 8, 10, or 12 months; or at least 1, 2, 3, 4, or 5 years.
  • In some embodiments, a therapy that includes a CD19 inhibitor, e.g., a CD19 CAR therapy, may relapse or be refractory to treatment. The relapse or resistance can be caused by CD19 loss (e.g., an antigen loss mutation) or other CD19 alteration that reduces the level of CD19 (e.g., caused by clonal selection of CD19-negative clones). A cancer that harbors such CD19 loss or alteration is referred to herein as a “CD19-negative cancer” or a “CD19-negative relapsed cancer”). It shall be understood that a CD19-negative cancer need not have 100% loss of CD19, but a sufficient reduction to reduce the effectiveness of a CD19 therapy such that the cancer relapses or becomes refractory. In some embodiments, a CD19-negative cancer results from a CD19 CAR therapy. In some embodiments, a CD19-negative multiple myeloma can be treated with a CD19 CAR-expressing therapy, e.g., as described in PCT/US2015/024671, filed Apr. 7, 2015 (e.g., paragraphs 9 and 90, and Example 6 therein), which is incorporated by reference in its entirety. In some embodiments, a CD19-negative cancer can be treated with a CAR-expressing therapy, e.g., a CD123 CAR-expressing therapy, e.g., as described in PCT/US2015/045898 filed Aug. 19, 2015 (e.g., p. 26, p. 30, and Example 7 therein) which is incorporated by reference in its entirety.
  • The term “endogenous” refers to any material from or produced inside an organism, cell, tissue or system.
  • The term “effective amount” or “therapeutically effective amount” are used interchangeably herein, and refer to an amount of a compound, formulation, material, or composition, as described herein effective to achieve a particular biological result.
  • The term “exogenous” refers to any material introduced from or produced outside an organism, cell, tissue or system.
  • The term “expression” refers to the transcription and/or translation of a particular nucleotide sequence driven by a promoter.
  • The term “flexible polypeptide linker” or “linker” as used in the context of a scFv refers to a peptide linker that consists of amino acids such as glycine and/or serine residues used alone or in combination, to link variable heavy and variable light chain regions together. In one embodiment, the flexible polypeptide linker is a Gly/Ser linker and comprises the amino acid sequence (Gly-Gly-Gly-Ser)n, where n is a positive integer equal to or greater than 1. For example, n=1, n=2, n=3, n=4, n=5, n=6, n=7, n=8, n=9 and n=10 (SEQ ID NO:28). In one embodiment, the flexible polypeptide linkers include, but are not limited to, (Gly4Ser)4 (SEQ ID NO:29) or (Gly4 Ser)3 (SEQ ID NO:30). In another embodiment, the linkers include multiple repeats of (Gly2Ser), (GlySer) or (Gly3Ser) (SEQ ID NO:31). Also included within the scope of the invention are linkers described in WO2012/138475, incorporated herein by reference.
  • The terms “homology” or “identity,” as used interchangeably herein, refer to sequence similarity between two polynucleotide sequences or between two polypeptide sequences, with identity being a more strict comparison. The phrases “percent identity or homology” and “% identity or homology” refer to the percentage of sequence similarity found in a comparison of two or more polynucleotide sequences or two or more polypeptide sequences. “Sequence similarity” refers to the percent similarity in base pair sequence (as determined by any suitable method) between two or more polynucleotide sequences. Two or more sequences can be anywhere from 0-100% similar, or any integer value there between. Identity or similarity can be determined by comparing a position in each sequence that can be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same nucleotide base or amino acid, then the molecules are identical at that position. A degree of similarity or identity between polynucleotide sequences is a function of the number of identical or matching nucleotides at positions shared by the polynucleotide sequences. A degree of identity of polypeptide sequences is a function of the number of identical amino acids at positions shared by the polypeptide sequences. A degree of homology or similarity of polypeptide sequences is a function of the number of amino acids at positions shared by the polypeptide sequences. The term “substantial homology,” as used herein, refers to homology of at least 50%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% or more.
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab, F(ab)2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies and antibody fragments thereof are human immunoglobulins (recipient antibody or antibody fragment) in which residues from a complementary-determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, a humanized antibody/antibody fragment can comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications can further refine and optimize antibody or antibody fragment performance. In general, the humanized antibody or antibody fragment thereof will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or a significant portion of the FR regions are those of a human immunoglobulin sequence. The humanized antibody or antibody fragment can also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see Jones et al., NATURE, 321: 522-525, 1986; Reichmann et al., NATURE, 332: 323-329, 1988; Presta, CURR. OP. STRUCT. BIOL., 2: 593-596, 1992.
  • “Immune effector cell,” as that term is used herein, refers to a cell that is involved in an immune response, e.g., in the promotion of an immune effector response. Examples of immune effector cells include T cells, e.g., alpha/beta T cells and gamma/delta T cells, B cells, natural killer (NK) cells, natural killer T (NK-T) cells, mast cells, and myeloid-derived phagocytes.
  • “Immune effector function or immune effector response,” as that term is used herein, refers to function or response, e.g., of an immune effector cell, that enhances or promotes an immune attack of a target cell. E.g., an immune effector function or response refers a property of a T or NK cell that promotes killing or the inhibition of growth or proliferation, of a target cell. In the case of a T cell, primary stimulation and co-stimulation are examples of immune effector function or response.
  • The term “effector function” refers to a specialized function of a cell. Effector function of a T cell, for example, may be cytolytic activity or helper activity including the secretion of cytokines.
  • The term “4-1BB” refers to a member of the TNFR superfamily with an amino acid sequence provided as GenBank Acc. No. AAA62478.2, or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like; and a “4-1BB costimulatory domain” is defined as amino acid residues 214-255 of GenBank Acc No. AAA62478.2, or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like. In one aspect, the “4-1BB costimulatory domain” is the sequence provided as SEQ ID NO:14 or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like.
  • An “intracellular signaling domain,” as the term is used herein, refers to an intracellular portion of a molecule. The intracellular signaling domain can generate a signal that promotes an immune effector function of the CAR containing cell, e.g., a CAR-expressing cell, e.g., a T cell or an NK cell. Examples of immune effector function, e.g., in a CAR-expressing cell include, cytolytic activity and helper activity, including the secretion of cytokines. In embodiments, the intracellular signal domain transduces the effector function signal and directs the cell to perform a specialized function. While the entire intracellular signaling domain can be employed, in many cases it is not necessary to use the entire chain. To the extent that a truncated portion of the intracellular signaling domain is used, such truncated portion may be used in place of the intact chain as long as it transduces the effector function signal. The term intracellular signaling domain is thus meant to include any truncated portion of the intracellular signaling domain sufficient to transduce the effector function signal.
  • In an embodiment, the intracellular signaling domain can comprise a primary intracellular signaling domain. Exemplary primary intracellular signaling domains include those derived from the molecules responsible for primary stimulation, or antigen dependent simulation. In an embodiment, the intracellular signaling domain can comprise a costimulatory intracellular domain. Exemplary costimulatory intracellular signaling domains include those derived from molecules responsible for costimulatory signals, or antigen independent stimulation. For example, in the case of a CAR-expressing cell (e.g., a T cell, an NK cell), a primary intracellular signaling domain can comprise a cytoplasmic sequence of a T cell receptor, and a costimulatory intracellular signaling domain can comprise cytoplasmic sequence from co-receptor or costimulatory molecule.
  • A primary intracellular signaling domain can comprise a signaling motif which is known as an immunoreceptor tyrosine-based activation motif or ITAM. Examples of ITAM containing primary cytoplasmic signaling sequences include, but are not limited to, those derived from CD3 zeta, FcR gamma, FcR beta, CD3 gamma, CD3 delta, CD3 epsilon, CD5, CD22, CD79a, CD79b, CD278 (“ICOS”), FccRI, CD66d, CD32, DAP10, and DAP12.
  • As used herein, “in vitro transcribed RNA” refers to RNA, preferably mRNA, that has been synthesized in vitro. Generally, the in vitro transcribed RNA is generated from an in vitro transcription vector. The in vitro transcription vector comprises a template that is used to generate the in vitro transcribed RNA.
  • The term “isolated” means altered or removed from the natural state. For example, a nucleic acid or a peptide naturally present in a living animal is not “isolated,” but the same nucleic acid or peptide partially or completely separated from the coexisting materials of its natural state is “isolated.” An isolated nucleic acid or protein can exist in substantially purified form, or can exist in a non-native environment such as, for example, a host cell.
  • The term “lentivirus” refers to a genus of the Retroviridae family. Lentiviruses are unique among the retroviruses in being able to infect non-dividing cells; they can deliver a significant amount of genetic information into the DNA of the host cell, so they are one of the most efficient methods of a gene delivery vector. HIV, SIV, and FIV are all examples of lentiviruses.
  • The term “lentiviral vector” refers to a vector derived from at least a portion of a lentivirus genome, including especially a self-inactivating lentiviral vector as provided in Milone et al., MOL. THER. 17(8): 1453-1464 (2009). Other examples of lentivirus vectors that may be used in the clinic, include but are not limited to, e.g., the LENTIVECTOR® gene delivery technology from Oxford BioMedica, the LENTIMAX™ vector system from Lentigen and the like. Nonclinical types of lentiviral vectors are also available and would be known to one skilled in the art.
  • The term ‘low, immune enhancing, dose” when used in conjunction with an mTOR inhibitor, e.g., an allosteric mTOR inhibitor, e.g., RAD001 or rapamycin, or a catalytic mTOR inhibitor, refers to a dose of mTOR inhibitor that partially, but not fully, inhibits mTOR activity, e.g., as measured by the inhibition of P70 S6 kinase activity. Methods for evaluating mTOR activity, e.g., by inhibition of P70 S6 kinase, are discussed herein. The dose is insufficient to result in complete immune suppression but is sufficient to enhance the immune response. In an embodiment, the low, immune enhancing, dose of mTOR inhibitor results in a decrease in the number of PD-1 positive immune effector cells, e.g., T cells or NK cells and/or an increase in the number of PD-1 negative immune effector cells, e.g., T cells or NK cells, or an increase in the ratio of PD-1 negative immune effector cells, e.g., T cells or NK cells/PD-1 positive immune effector cells, e.g., T cells or NK cells.
  • In general, the term “naïve T cell” refers to immune cells that comprise antigen-inexperienced cells, e.g., immune cells that are precursors of memory cells. In some embodiments, naïve T cells may be differentiated, but have not yet encountered their cognate antigen, and therefore are activated T cells or memory T cells. In some embodiments, naïve T cells may be characterized by expression of CD62L, CD27, CCR7, CD45RA, CD28, and CD127, and the absence of CD95, or CD45RO isoform. In certain embodiments, a naïve T cells is a type of younger T cell as described herein.
  • The term “less exhausted” or “less exhausted phenotype” refers to immune effector cells that have reduced (e.g., lack) expression of immune cell exhaustion markers, e.g. PD1, TIM3, and LAG3. In some embodiments, a less exhausted cell may be a younger T cell as described herein.
  • The term “nucleic acid” or “polynucleotide” refers to deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), or a combination of a DNA or RNA thereof, and polymers thereof in either single- or double-stranded form. The term “nucleic acid” includes a gene, CDNA or an mRNA. In one embodiment, the nucleic acid molecule is synthetic (e.g., chemically synthesized) or recombinant. Unless specifically limited, the term encompasses nucleic acids containing analogues or derivatives of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., NUCLEIC ACID RES. 19:5081 (1991); Ohtsuka et al., J. BIOL. CHEM. 260:2605-2608 (1985); and Rossolini et al., MOL. CELL. Probes 8:91-98 (1994)). In the context of the present invention, the following abbreviations for the commonly occurring nucleic acid bases are used. “A” refers to adenosine, “C” refers to cytosine, “G” refers to guanosine, “T” refers to thymidine, and “U” refers to uridine.
  • A “nucleic acid” “marker” or “biomarker” is a nucleic acid (e.g., DNA, mRNA, CDNA) encoded by or corresponding to a marker as described herein. For example, such marker nucleic acid molecules include DNA (e.g., genomic DNA and CDNA) comprising the entire or a partial sequence of any of the nucleic acid sequences set forth, or the complement or hybridizing fragment of such a sequence. The marker nucleic acid molecules also include RNA comprising the entire or a partial sequence of any of the nucleic acid sequences set forth herein, or the complement of such a sequence, wherein all thymidine residues are replaced with uridine residues. A “marker protein” is a protein encoded by or corresponding to a marker of the invention. A marker protein comprises the entire or a partial sequence of a protein encoded by any of the sequences set forth herein, or a fragment thereof. The terms “protein” and “polypeptide” are used interchangeably herein.
  • An “overexpression” or “significantly higher level of expression” of the gene products refers to an expression level or copy number in a test sample that is greater than the standard error of the assay employed to assess the level of expression. In embodiments, the overexpression can be at least two, at least three, at least four, at least five, or at least ten or more times the expression level of the gene in a control sample or the average expression level of gene products in several control samples.
  • The terms “peptide,” “polypeptide,” and “protein” are used interchangeably, and refer to a compound comprised of amino acid residues covalently linked by peptide bonds. A protein or peptide must contain at least two amino acids, and no limitation is placed on the maximum number of amino acids that can comprise a protein's or peptide's sequence. Polypeptides include any peptide or protein comprising two or more amino acids joined to each other by peptide bonds. As used herein, the term refers to both short chains, which also commonly are referred to in the art as peptides, oligopeptides and oligomers, for example, and to longer chains, which generally are referred to in the art as proteins, of which there are many types. “Polypeptides” include, for example, biologically active fragments, substantially homologous polypeptides, oligopeptides, homodimers, heterodimers, variants of polypeptides, modified polypeptides, derivatives, analogs, fusion proteins, among others. A polypeptide includes a natural peptide, a recombinant peptide, or a combination thereof.
  • As used herein, a “poly(A)” is a series of adenosines attached by polyadenylation to the mRNA. In some embodiments of a construct for transient expression, the polyA is between 50 and 5000 (SEQ ID NO: 34) (e.g., 2000; SEQ ID NO: 32), e.g., 64 (SEQ ID NO: 37), e.g., greater than 100 (e.g., 150, SEQ ID NO: 33), e.g., greater than 400 (SEQ ID NO: 38). poly(A) sequences can be modified chemically or enzymatically to modulate mRNA functionality such as localization, stability or efficiency of translation.
  • The term “probe” refers to any molecule which is capable of selectively binding to a specifically intended target molecule, for example a marker of the invention. Probes can be either synthesized by one skilled in the art, or derived from appropriate biological preparations. For purposes of detection of the target molecule, probes can be specifically designed to be labeled, as described herein. Examples of molecules that can be utilized as probes include, but are not limited to, RNA, DNA, proteins, antibodies, and organic monomers.
  • The term “promoter” refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a polynucleotide sequence.
  • The term “promoter/regulatory sequence” refers to a nucleic acid sequence which is required for expression of a gene product operably linked to the promoter/regulatory sequence. In some instances, this sequence may be the core promoter sequence and in other instances, this sequence may also include an enhancer sequence and other regulatory elements which are required for expression of the gene product. The promoter/regulatory sequence may, for example, be one which expresses the gene product in a tissue specific manner.
  • The term “prophylaxis” as used herein means the prevention of or protective treatment for a disease or disease state.
  • Ranges: throughout this disclosure, various aspects of the invention can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6. As another example, a range such as 95-99% identity, includes something with 95%, 96%, 97%, 98% or 99% identity, and includes subranges such as 96-99%, 96-98%, 96-97%, 97-99%, 97-98% and 98-99% identity. This applies regardless of the breadth of the range.
  • The term “recombinant antibody” refers to an antibody which is generated using recombinant DNA technology, such as, for example, an antibody expressed by a bacteriophage or yeast expression system. The term should also be construed to mean an antibody which has been generated by the synthesis of a DNA molecule encoding the antibody and which DNA molecule expresses an antibody protein, or an amino acid sequence specifying the antibody, wherein the DNA or amino acid sequence has been obtained using recombinant DNA or amino acid sequence technology which is available and well known in the art.
  • “Refractory” as used herein refers to a disease, e.g., cancer, that does not respond to a treatment. In embodiments, a refractory cancer can be resistant to a treatment before or at the beginning of the treatment. In other embodiments, the refractory cancer can become resistant during a treatment. A refractory cancer is also called a resistant cancer.
  • In embodiments, a reference or control level or activity is the level and/or activity in a subject, e.g., a sample obtained from one or more of: a baseline or prior value for the subject (e.g., prior to treatment with a CAR-expressing cell); the subject at a different time interval; an average or median value for a cancer patient population; a healthy control; or a healthy subject population (e.g., a control).
  • “Sample,” “tissue sample,” “patient sample,” “patient cell or tissue sample” or “specimen” each refers to a biological sample obtained from a tissue or bodily fluid of a subject or patient. The source of the tissue sample can be solid tissue as from a fresh, frozen and/or preserved organ, tissue sample, biopsy, or aspirate; blood or any blood constituents (e.g., serum, plasma); bodily fluids such as urine, cerebral spinal fluid, whole blood, plasma and serum. The sample can include a non-cellular fraction (e.g., urine, plasma, serum, or other non-cellular body fluid). In one embodiment, the sample is a urine sample. In other embodiments, the body fluid from which the sample is obtained from an individual comprises blood (e.g., whole blood). In an embodiment, the sample is a whole blood sample obtained from the subject. In certain embodiments, the blood can be further processed to obtain plasma or serum. In an embodiment, the sample is an apheresis sample obtained from the blood of the subject. In an embodiment, the sample is a manufactured product sample, e.g., genetically engineered T cells obtained from the blood of the subject, e.g., a manufactured CAR-expressing cell (e.g., T cell, NK cell) product, e.g., a manufactured CD19 CAR-expressing cell product. In another embodiment, the sample contains a tissue, cells (e.g., peripheral blood mononuclear cells (PBMC)). For example, the sample can be a fine needle biopsy sample, an archival sample (e.g., an archived sample with a known diagnosis and/or treatment history), a histological section (e.g., a frozen or formalin-fixed section, e.g., after long term storage), among others. The term sample includes any material obtained and/or derived from a biological sample, including a polypeptide, and nucleic acid (e.g., genomic DNA, CDNA, RNA) purified or processed from the sample. Purification and/or processing of the sample can involve one or more of extraction, concentration, antibody isolation, sorting, concentration, fixation, addition of reagents and the like. The sample can contain compounds that are not naturally intermixed with the tissue in nature such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics or the like.
  • The term “product” or “manufactured product” as used herein, refers to a manufactured composition comprising a genetically engineered cell (e.g., an immune effector cell), e.g., a population of cells in which a plurality of cells are engineered to express a CAR, e.g., a CAR described herein. A manufactured product can be any genetically engineered immune effector cell (e.g., T cell, NK cell), e.g., genetically engineered immune effector cells obtained from the blood of the subject, e.g., a manufactured CAR-expressing cell product, e.g., a manufactured CD19 CAR-expressing cell product. In an embodiment, a cell (e.g., an immune effector cell) engineered to express a CAR may be obtained from an activated cryopreserved expanded cell population (e.g., an expanded immune effector cell population).
  • The term “signaling domain” refers to the functional portion of a protein which acts by transmitting information within the cell to regulate cellular activity via defined signaling pathways by generating second messengers or functioning as effectors by responding to such messengers.
  • The amount of a biomarker, e.g., expression of gene products (e.g., one or more the biomarkers described herein), in a subject is “significantly” higher or lower than the normal amount of a marker, if the amount of the marker is greater or less, respectively, than the normal level by an amount greater than the standard error of the assay employed to assess amount, or at least two, three, four, five, ten or more times that amount. Alternatively, the amount of the marker in the subject can be considered “significantly” higher or lower than the normal amount if the amount is at least about 1.5, two, at least about three, at least about four, or at least about five times, higher or lower, respectively, than the normal amount of the marker.
  • The term “specifically binds,” refers to an antibody, or a ligand, which recognizes and binds with a cognate binding partner protein present in a sample, but which antibody or ligand does not substantially recognize or bind other molecules in the sample.
  • The term “stimulation,” refers to a primary response induced by binding of a stimulatory molecule (e.g., a TCR/CD3 complex) with its cognate ligand thereby mediating a signal transduction event, such as, but not limited to, signal transduction via the TCR/CD3 complex. Stimulation can mediate altered expression of certain molecules, such as down regulation of TGF-β, and/or reorganization of cytoskeletal structures, and the like.
  • The term “stimulatory molecule,” refers to a molecule expressed by a T cell that provides the primary cytoplasmic signaling sequence(s) that regulate primary activation of the TCR complex in a stimulatory way for at least some aspect of the T cell signaling pathway. In one aspect, the primary signal is initiated by, for instance, binding of a TCR/CD3 complex with an MHC molecule loaded with peptide, and which leads to mediation of a T cell response, including, but not limited to, proliferation, activation, differentiation, and the like. A primary cytoplasmic signaling sequence (also referred to as a “primary signaling domain”) that acts in a stimulatory manner may contain a signaling motif which is known as immunoreceptor tyrosine-based activation motif or ITAM. Examples of an ITAM containing cytoplasmic signaling sequence that is of particular use in the invention includes, but is not limited to, those derived from CD3 zeta, common FcR gamma (FCER1G), Fc gamma RIIa, FcR beta (Fc Epsilon Rib), CD3 gamma, CD3 delta, CD3 epsilon, CD79a, CD79b, DAP10, and DAP12. In a specific CAR of the invention, the intracellular signaling domain in any one or more CARS of the invention comprises an intracellular signaling sequence, e.g., a primary signaling sequence of CD3-zeta. In a specific CAR of the invention, the primary signaling sequence of CD3-zeta is the sequence provided as SEQ ID NO:18 (mutant CD3 zeta), or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like. In a specific CAR of the invention, the primary signaling sequence of CD3-zeta is the sequence as provided in SEQ ID NO:20 (wild-type human CD3 zeta), or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like.
  • The term “subject” is intended to include living organisms in which an immune response can be elicited (e.g., mammals, human). In an embodiment, a subject is a mammal. In an embodiment, a subject is a human. In an embodiment, a subject is a patient.
  • The term “therapeutic” as used herein means a treatment. A therapeutic effect is obtained by reduction, suppression, remission, or eradication of a disease state.
  • The term “transfected” or “transformed” or “transduced” refers to a process by which exogenous nucleic acid is transferred or introduced into the host cell. A “transfected” or “transformed” or “transduced” cell is one which has been transfected, transformed or transduced with exogenous nucleic acid. The cell includes the primary subject cell and its progeny.
  • As used herein, “transient” refers to expression of a non-integrated transgene for a period of hours, days or weeks, wherein the period of time of expression is less than the period of time for expression of the gene if integrated into the genome or contained within a stable plasmid replicon in the host cell.
  • The term “transmembrane domain,” refers to a polypeptide that spans the plasma membrane. In an embodiment, it links an extracellular sequence, e.g., a switch domain, an extracellular recognition element, e.g., an antigen binding domain, an inhibitory counter ligand binding domain, or costimulatory ECD domain, to an intracellular sequence, e.g., to a switch domain or an intracellular signaling domain. A transmembrane domain can include one or more additional amino acids adjacent to the transmembrane region, e.g., one or more amino acid associated with the extracellular region of the protein from which the transmembrane was derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the extracellular region) and/or one or more additional amino acids associated with the intracellular region of the protein from which the transmembrane protein is derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the intracellular region). Examples of transmembrane domains are disclosed herein.
  • The terms “treat”, “treatment” and “treating” refer to the reduction or amelioration of the progression, severity and/or duration of a proliferative disorder, or the amelioration of one or more symptoms (e.g., one or more discernible symptoms) of a proliferative disorder resulting from the administration of one or more therapies (e.g., one or more therapeutic agents such as a CAR of the invention). In specific embodiments, the terms “treat”, “treatment” and “treating” refer to the amelioration of at least one measurable physical parameter of a proliferative disorder, such as growth of a tumor, not necessarily discernible by the patient. In other embodiments the terms “treat”, “treatment” and “treating”-refer to the inhibition of the progression of a proliferative disorder, either physically by, e.g., stabilization of a discernible symptom, physiologically by, e.g., stabilization of a physical parameter, or both. In other embodiments the terms “treat”, “treatment” and “treating” refer to the reduction or stabilization of tumor size or cancerous cell count.
  • An “underexpression” or “significantly lower level of expression” of products (e.g., the markers set forth herein) refers to an expression level in a test sample that is greater than the standard error of the assay employed to assess expression, for example, at least 1.5, twice, at least three, at least four, at least five, or at least ten or more times less than the expression level of the gene in a control sample, or the average expression level of gene products in several control samples.
  • The term “xenogeneic” refers to a graft derived from an animal of a different species.
  • As used herein, the term “young T cell” or “younger T cell”, refers to an immune effector cell that comprises a less differentiated phenotype, e.g., a younger cell, e.g., a young T cell. In some embodiments, a younger T cell may be a naïve T cell (TN). In some embodiments, a young T cell may be characterized by expression of CD62L, and the absence of CD25, CD44, or CD45RO isoform. In some embodiments, a younger T cell may be a memory stem cell (TSCM). In some embodiments, a younger T cells may be a central memory T cell (TCM). Phenotypic markers associated with TN, TSCM and TCM are disclosed in, e.g., Maus, M. et al. (2014) Annu. Rev. Immunol. 32:189-225 (see for example, FIG. 3), incorporated by reference herein. Exemplary phenotypes of TN include one or more (or all) of the following: CD45RA+, CD45RO−, CD62Lhigh, CCR7high, CD95−, CD122−, CD27high, CD28+, CD57−, KLRG-1−, or long telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid TN markers). Exemplary phenotypes of TSCM include one or more (or all) of the following: CD45RA+, CD45RO−, CD62Lhigh, CCR7high, CD95+, CD122+, CD27high, CD28high, CD57−, KLRG-1−, or long telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid TSCM markers). Exemplary phenotypes of TCM include one or more (or all) of the following: CD45RA−, CD45ROhigh, CD62Lhigh, CCR7+, CD95+, CD122high, CD27+, CD28high, CD57−, KLRG-1−/+, or long/intermediate telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid TCM markers).
  • As used herein, the term “older T cell” refers to an immune effector cell that comprises a more exhausted phenotype. In some embodiments, an older T cell may be an effector memory T cell (TEM). In other embodiments, an older T cell may be an effector T cell (TEFF). In other embodiments, an older T cell has an exhausted phenotype. Phenotypic markers associated with TEM, TEFF and exhausted T cells are disclosed in, e.g., Maus, M. et al. (2014) Annu. Rev. Immunol. 32:189-225 (see for example, FIG. 3), incorporated by reference herein. Exemplary phenotypes of TEM include one or more (or all) of the following: CD45RA−/+, CD45ROhigh, CD62L−, CCR7−, CD95−, CD122high, CD27−/+, CD28−/+, CD57low, KLRG-1+, or intermediate telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid TEM markers). Exemplary phenotypes of TEFF include one or more (or all) of the following: CD45RA−/+, CD45RO+, CD62L−, CCR7−, CD95high, CD122−/+, CD27−, CD28−, CD57+, KLRG-1high, or short/intermediate telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid TEFF markers). Exemplary phenotypes of an exhausted T cell phenotype include one or more (or all) of the following: CD45RA−/+, CD45RO+, CD62L−, CCR7-, CD95high, CD122low, CD27−, CD28−, CD57high, KLRG-1high, or short telomere length (or any combination of two, three, four, five, six, seven, eight, nine, or all of the aforesaid markers).
  • The term “zeta” or alternatively “zeta chain”, “CD3-zeta” or “TCR-zeta” is defined as the protein provided as GenBank Acc. No. BAG36664.1, or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like, and a “zeta stimulatory domain” or alternatively a “CD3-zeta stimulatory domain” or a “TCR-zeta stimulatory domain” is defined as the amino acid residues from the cytoplasmic domain of the zeta chain that are sufficient to functionally transmit an initial signal necessary for T cell activation. In one aspect the cytoplasmic domain of zeta comprises residues 52 through 164 of GenBank Acc. No. BAG36664.1 or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like, that are functional orthologs thereof. In one aspect, the “zeta stimulatory domain” or a “CD3-zeta stimulatory domain” is the sequence provided as SEQ ID NO:18. In one aspect, the “zeta stimulatory domain” or a “CD3-zeta stimulatory domain” is the sequence provided as SEQ ID NO:20.
  • Various aspects of the invention are described in further detail below. Additional definitions are set out throughout the specification.
  • DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS
  • Biomarkers predicting response to a therapy in subjects having cancer (e.g., a hematological cancer such as chronic lymphocytic leukemia (CLL) and acute lymphoblastic leukemia (ALL)) are provided herein.
  • In one aspect, biomarkers predicting response to a cell expressing a CAR, e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) (e.g., a CD19 CAR-expressing cell described herein such as, e.g., CTL019) in subjects having CLL and ALL are provided herein.
  • Methods are provided for the diagnosis and monitoring of treatment of cancer (e.g., a hematological cancer such as ALL and CLL) based on detection of certain biomarkers in samples from patients who have, or are suspected of having, cancer. Further, expression of one or more such biomarkers can be used to distinguish subjects that respond favorably to a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., “complete responders” or “CR”) from subjects that don't respond to a CAR-expressing cell therapy (e.g., “non-responders” or “NR”) and from subjects that have a partial response to a CAR-expressing cell therapy (e.g., “partial responders” or “PR”).
  • Use of Biomarkers to Evaluate Disease Progression and Predict Subject Response to CAR-Expressing Cell Therapy
  • In an embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more genes in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1 and/or a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene signature can be used with methods of the present disclosure to acquire a disease progression value. The disease progression value can be used for, e.g., in evaluating the effectiveness of therapies in treating cancer (e.g., a hematological cancer such as ALL and CLL). In an embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more genes in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and/or KLRG1 and/or a CD19 CAR-expressing cell gene signature are used to classify a subject as a complete responder, partial responder, or non-responder to CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019). In an embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more genes in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and/or KLRG1, and/or a CD19 CAR-expressing cell gene signature are used to predict a subject's responsiveness to a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019).
  • Subjects
  • For any of the methods and kits disclosed herein, the subject treated, or the subject from which the value is obtained, is a subject having, or at risk of having, cancer at any stage of treatment. Exemplary cancers include, but are not limited to, B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, and Waldenstrom macroglobulinemia. In an embodiment, the cancer is a hematological cancer. In an embodiment, the cancer is ALL. In another embodiment, the cancer is CLL. In an embodiment, the cancer is associated with CD19 expression.
  • In an embodiment, the subject has received a pretreatment of an additional therapy, e.g., a subject that has been identified as a partial responder or non-responder and subsequently has been pretreated with an additional therapy. In an embodiment, the subject receives pretreatment with an mTOR inhibitor. In an embodiment, the mTOR inhibitor is administered at a dose or dosing schedule described herein. In one embodiment, a low, immune enhancing dose of an mTOR inhibitor is given to the subject prior to treatment with a CAR-expressing cell (e.g., a T cell, an NK cell). In an embodiment, administration of a low, immune enhancing, dose of an mTOR inhibitor, e.g., an allosteric inhibitor, e.g., RAD001, or a catalytic inhibitor, is initiated prior to administration of a CAR expressing cell described herein, e.g., T cells. In an embodiment, the CAR cells are administered after a sufficient time, or sufficient dosing, of an mTOR inhibitor, such that the level of PD1 negative immune effector cells, e.g., T cells, or the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells, has been, at least transiently, increased. In an embodiment, the cell, e.g., T cell, to be engineered to express a CAR, is harvested after a sufficient time, or after sufficient dosing of the low, immune enhancing, dose of an mTOR inhibitor, such that the level of PD1 negative immune effector cells, e.g., T cells, or the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells, in the subject or harvested from the subject has been, at least transiently, increased.
  • In one embodiment, the subject has received a pretreatment with a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein. Examples of inhibitory molecules, e.g., checkpoint molecules include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR (e.g., TGFRbeta). In embodiments, an inhibitory nucleic acid, e.g., an inhibitory nucleic acid, e.g., a dsRNA, e.g., an siRNA or shRNA; or e.g., an inhibitory protein or system, e.g., a clustered regularly interspaced short palindromic repeats (CRISPR), a transcription-activator like effector nuclease (TALEN), or a zinc finger endonuclease (ZFN), e.g., as described herein, can be used to inhibit expression of a molecule that modulates or regulates, e.g., inhibits, T-cell function in the CAR-expressing cell. In an embodiment the agent is an shRNA, e.g., an shRNA described herein. In one embodiment, the inhibitor of checkpoint molecule can be, e.g., an antibody or antibody fragment that binds to a checkpoint molecule. For example, the agent can be an antibody or antibody fragment that binds to PD1, PD-L1, PD-L2 (e.g., as described herein) or CTLA4 (e.g., ipilimumab (also referred to as MDX-010 and MDX-101, and marketed as Yervoy®; Bristol-Myers Squibb; Tremelimumab (IgG2 monoclonal antibody available from Pfizer, formerly known as ticilimumab, CP-675,206)). In an embodiment, the agent is an antibody or antibody fragment that binds to TIM3, e.g., as described herein. In an embodiment, the agent is an antibody or antibody fragment that binds to LAG3, e.g., as described herein. In an embodiment, the agent is an antibody or antibody fragment that binds to CEACAM, e.g., as described herein.
  • In an embodiment, the subject receives an additional therapy in combination with CAR-expressing cell (e.g., a T cell, an NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019). In an embodiment, the subject receives an mTOR inhibitor, e.g., an mTOR inhibitor described herein, in combination with CAR-expressing cell therapy. In one embodiment, the mTOR inhibitor is administered at a dose and/or dosing schedule described herein. In one embodiment, the subject receives a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein, in combination with CAR-expressing cell therapy. In one embodiment, the checkpoint inhibitor is administered at a dose and/or dosing schedule described herein. In an embodiment, the subject receives a kinase inhibitor, e.g., a kinase inhibitor described herein. In one embodiment, the kinase inhibitor is administered at a dose and/or dosing schedule described herein.
  • In an embodiment, the subject has been identified as a non-responder and the subject receives a therapy other than a CAR-expressing cell therapy, e.g., a standard of care therapy for the particular cancer type. In one embodiment, the subject receives one or more of an anti-CD20 antibody, or functional fragment thereof (e.g., ofatumumab, rituximab, obinutuzumab), an anti-CD52 antibody or functional fragment thereof (e.g., alemtuzumab), an alkylating agent (e.g., a nitrogen mustard alkylating agent such as, e.g., bendamustine HCl, chlorambucel, cyclophosphamide), a kinase inhibitor (e.g., a kinase inhibitor described herein such as, e.g., a BTK inhibitor described herein or a phosphonositide-3 kinase inhibitor described herein). In one embodiment, the subject receives a stem cell transplant.
  • Biomarkers Assessment Analysis of CTL019 Biomarkers
  • Analysis of levels of expression and/or activity of gene products correlated with a subject's response to CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019) and cancer disease progression (e.g., a hematological cancer such as CLL and ALL) has led to the identification of novel CD19 CAR-expressing cell gene signatures. For example, the present invention provides methods for evaluation of expression level of one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, twenty-five, thirty, thirty-five, forty, forty-five, fifty, one hundred, or more genes from Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 that comprise a CD19 CAR-expressing cell gene signature.
  • Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and/or KLRG1 lists genes (e.g., protein biomarkers) that are differentially expressed by complete responders compared to partial responders and non-responders.
  • In some embodiments, methods of the present disclosure can be used to determine the responsiveness of a subject to treatment with a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy described herein such as, e.g., CTL019), wherein a statistically significant difference in the amount, e.g., expression, and/or activity of a marker disclosed herein relative to a reference, e.g., a median value for a cancer patient population (e.g., a hematological cancer such as CLL and ALL), a median value for a population of healthy, cancer-free subjects, a median value for a population of non-responders or partial responders, in a subjects sample, then the more likely the disease is to respond to CAR-expressing cell therapy.
  • In an embodiment, the disclosure provides a method of, or assay for, identifying a subject having cancer (e.g., a hematological cancer such as CLL and ALL) as having an increased or decreased likelihood to respond to a treatment that comprises a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019), the method comprising:
      • (1) acquiring a sample from the subject;
      • (2) determining a level of one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or more) markers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 in the sample; and
      • (3) comparing the determined level of one or more markers to a reference level; wherein a difference, e.g., statistically significant difference in the determined level to the reference level is predictive of the subjects responsiveness to the CAR-expressing cell therapy; and
      • (4) identifying the subject as a complete responder, partial responder or non-responder to the CAR-expressing cell therapy.
  • In one embodiment, the sample is a blood, plasma or a serum sample. In one embodiment, the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject. In an embodiment, the sample is a manufactured product sample, e.g., genetically engineered T cells obtained from the blood of the subject, e.g., a manufactured CAR-expressing cell product, e.g., a manufactured CD19 CAR-expressing cell product.
  • In an embodiment, the disclosure provides a method of, or assay for, identifying a subject having a cancer including, but not limited to, B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, and Waldenstrom macroglobulinemia. In an embodiment, the cancer is a hematological cancer. In an embodiment, the cancer is ALL. In another embodiment, the cancer is CLL. In an embodiment, the cancer is associated with CD19 expression.
  • In an embodiment, a CAR-expressing cell therapy comprises a CAR-expressing cell therapy described herein, e.g., CTL019.
  • In an embodiment, a CAR-expressing cell therapy consists of a CAR-expressing cell therapy described herein, e.g., CTL019.
  • In an embodiment, the disclosure provides a method of, or assay for, identifying a subject having cancer (e.g., a hematological cancer such as CLL and ALL) as having an increased or decreased likelihood to respond to a treatment that comprises a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019), the method comprising:
      • (1) acquiring a sample from the subject;
      • (2) determining a gene signature of the sample; and
      • (3) comparing the determined gene signature to a reference gene signature;
        wherein a difference, e.g., statistically significant difference in expression level of one or more of the markers in the determined gene signature, e.g., as compared to a predetermined value, is predictive of the subjects responsiveness to the CAR-expressing cell therapy.
  • In an embodiment, the disclosure provides a method of, or assay for, determining the responsiveness of a subject having cancer (e.g., a hematological cancer such as CLL and ALL) to a treatment comprising a cell expressing a CAR (e.g., a cell expressing a CD19 CAR, e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein such as, e.g., CTL019), the method comprising:
  • determining a level of one or more markers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 in a sample obtained prior to treatment;
  • wherein a statistically significant difference in expression level of one or more markers in the sample relative to a predetermined value is indicative of increased responsiveness to the CAR-expressing cell.
  • The methods provided herein are particularly useful for identifying subjects that are likely to respond to CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019) prior to initiation of such treatment (e.g., pre-therapy) or early in the therapeutic regimen. In some embodiments, expression or activity of biomarkers is measured in a subject at least 2 weeks, at least 1 month, at least 3 months, at least 6 months, or at least 1 year prior to initiation of therapy. In some embodiments, expression or activity of biomarkers is measured less than 6 months prior to the initiation of therapy. Thus, in some embodiments, expression or activity of biomarkers is measured within 6 months, 5 months, 4 months, 3 months, 2 months, 1 month, 2 weeks, 1 week, 6 days, 5 days, 4 days, 3 days, 2 days, or 1 day prior to the initiation of therapy. In other embodiments, the expression or activity of biomarkers is determined after initiation of therapy (e.g., 1 month, 2 months, 3 months, 3.5 months, 4 months, 4.5 months, 5 months, 5.5 months, 6 months).
  • In an embodiment, the invention provides a method of evaluating a subject having cancer (e.g., a hematological cancer such as CLL and ALL) comprising:
  • acquiring a value of responder status for the subject that comprises a measure of one or more of the following:
  • one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more) biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature, and a CD19 CAR-expressing cell gene set signature, thereby evaluating the subject.
  • In an embodiment, the disclosure provides a method of evaluating a subject having cancer (e.g., a hematological cancer such as CLL and ALL) comprising acquiring a value of responder status for the subject that comprises a measure of one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature, thereby evaluating the subject.
  • In an embodiment, the disclosure provides a method of evaluating or monitoring the effectiveness of a CAR-expressing cell therapy in a subject having cancer comprising:
  • acquiring a value of responder status for the subject that comprises a measure of one or more of the following:
  • a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature, and a CD19 CAR-expressing cell gene set signature,
  • thereby evaluating or monitoring the effectiveness of the CAR therapy in the subject.
  • In an embodiment, the disclosure provides a method of evaluating or monitoring the effectiveness of a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019) in a subject having cancer (e.g., a hematological cancer such as CLL and ALL) comprising: acquiring a value of responder status for the subject that comprises a measure of one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more) of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene set signature, thereby evaluating or monitoring the effectiveness of the CAR-expressing cell (e.g., T cell, NK cell) therapy in the subject.
  • In an embodiment, the value of responder status comprises a measure of a combination of a gene signature and a biomarker.
  • In an embodiment, the value of the responder status comprises a measure of a CD19 CAR-expressing cell gene set signature and a combination of one or more of: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, and a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature.
  • In an embodiment, the value of the responder status comprises a measure of a CD19 CAR-expressing cell gene set signature and a combination of one or more of: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1.
  • In an embodiment, the value of the responder status comprises a measure of one or more biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B and a combination of one or more of: a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature, and a CD19 CAR-expressing cell gene set signature.
  • In an embodiment, the value of the responder status comprises a measure of one or more biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B and a combination of one or more of: CD57, CD27, CD122, CD62L, and KLRG1.
  • In an embodiment, the CD19 CAR-expressing cell gene signature comprises a value for expression of at least 5, 6, 7, 8, 9 or 10 genes comprising a CD19 CAR-expressing cell gene signature.
  • In an embodiment, the value for expression of the gene comprises a value for a transcriptional parameter, e.g., the level of an mRNA encoded by the gene.
  • In an embodiment, the value for expression of the protein comprises a value for a translational parameter, e.g., the level of a protein.
  • In an embodiment, provided methods further comprise obtaining a sample from the subject, wherein the sample comprises a cellular or tissue fraction. In an embodiment, the cellular fraction comprises blood.
  • In an embodiment, the measure of biomarker and/or gene signature is acquired before, at the same time, or during course of a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019).
  • In an embodiment, the measure of biomarker and/or gene signature is acquired less than 6 months, 5 months, 4 months, 3 months, 2 months, 1 month, 2 weeks, 1 week, 6 days, 5 days, 4 days, 3 days, 2 days prior to the initiation of a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019).
  • The methods described herein can also be used to monitor a positive response of a subject to CAR-expressing cell (e.g., T cell, NK cell) treatment (e.g., a CD19 CAR-expressing cell treatment described herein such as, e.g., CTL019). Such methods are useful for early detection of tolerance to therapy or to predict whether disease in a subject will progress. In such embodiments, the expression or activity of biomarkers is determined e.g., at least every week, at least every 2 weeks, at least every month, at least every 2 months, at least every 3 months, at least every 4 months, at least every 5 months, at least every 6 months, at least every 7 months, at least every 8 months, at least every 9 months, at least every 10 months, at least every 11 months, at least every year, at least every 18 months, at least every 2 years, at least every 3 years, at least every 5 years or more. It is also contemplated that expression or activity of the biomarkers is at irregular intervals e.g., biomarkers can be detected in a subject at 3 months of treatment, at 6 months of treatment, and at 7 months of treatment. Thus, in some embodiments, the expression or activity of the biomarkers is determined when deemed necessary by the skilled physician monitoring treatment of the subject.
  • The methods described herein can be used in treating any subject having cancer (e.g., a hematological cancer such as CLL and ALL). In one aspect, the invention pertains to methods of treating cancer (e.g., a hematological cancer such as CLL and ALL) in a subject.
  • In an embodiment, the disclosure provides methods for treating cancer including, but not limited to, B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, and Waldenstrom macroglobulinemia. In an embodiment, the invention provides methods for treating ALL. In another embodiment, the invention provides methods for treating CLL. In an embodiment, the invention provides methods for treating cancer that is associated with CD19 expression.
  • In an embodiment, provided methods comprise administering to the subject a cell expressing a CAR, e.g. a CAR T cell, e.g. a CD19 CAR T cell, e.g., a CTL019 product, if the subject is identified as having a difference, e.g., statistically significant difference in expression level of one or more markers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 relative to a reference level, such that the cancer (e.g., a hematological cancer such as ALL and CLL) is treated in the subject.
  • As discussed above, an example of a cancer that is treatable by disclosed methods is a cancer associated with expression of CD19. In one aspect, the cancer associated with expression of CD19 is a hematological cancer. In one aspect, the hematological cancer is a leukemia or a lymphoma. In one aspect, a cancer associated with expression of CD19 includes cancers and malignancies including, but not limited to, e.g., one or more acute leukemias including but not limited to, e.g., B-cell acute lymphoid leukemia (“B-ALL”), T-cell acute lymphoid leukemia (“T-ALL”), acute lymphoid leukemia (ALL); one or more chronic leukemias including but not limited to, e.g., chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL). Additional cancers or hematologic conditions associated with expression of CD19 include, but are not limited to, e.g., B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, and “preleukemia” which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells, and the like. Further, a disease associated with CD19 expression include, but not limited to, e.g., atypical and/or non-classical cancers, malignancies, precancerous conditions or proliferative diseases associated with expression of CD19.
  • In an embodiment, the disclosure provides a method for treating a subject having cancer (e.g., a hematological cancer such as ALL and CLL) comprising:
  • determining if the subject has an increased likelihood to respond to a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019) by comparing the level of one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more) markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 in a sample from the subject relative to a reference level, wherein a statistically significant difference in expression level of one or more maker genes relative to the reference level is indicative of an increased likelihood of response; and
  • administering to the subject a therapeutically effective dose of a CAR-expressing cell therapy.
  • In an embodiment, the disclosure provides a method for treating a subject having cancer (e.g., a hematological cancer such as ALL and CLL), comprising:
  • obtaining a sample from the subject;
  • determining a level of one or more markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 in the sample;
  • comparing the determined level of one or more markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 to a reference level; and
  • administering a therapeutically effective dose of a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019), if the subject is identified as having a statistically significant difference in the determined level of one or more markers in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 to a reference level, in the sample.
  • In an embodiment, the disclosure provides a method of treating cancer (e.g., a hematological cancer such as ALL and CLL) in a subject, comprising:
  • acquiring a value of responder status for the subject that comprises a measure of one or more of the following:
  • a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature, and a CD19 CAR-expressing cell gene set signature, and responsive to a determination of responder status, performing one, two, three, four or more of:
  • identifying the subject as a complete responder, partial responder or non-responder;
  • administering a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019);
  • selecting or altering a dosing of a CAR-expressing cell therapy;
  • selecting or altering the schedule or time course of a CAR-expressing cell therapy;
  • administering, e.g., to a non-responder or a partial responder, an additional agent in combination with a CAR-expressing cell therapy, e.g., a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein;
  • administering to a non-responder or partial responder a therapy that increases the number of naïve T cells in the subject prior to treatment with a CAR-expressing cell therapy;
  • modifying a manufacturing process of a CAR-expressing cell therapy, e.g., enrich for naïve T cells prior to introducing a nucleic acid encoding a CAR, e.g., for a subject identified as a non-responder or a partial responder; or selecting an alternative therapy, e.g., for a non-responder or partial responder; or
  • selecting an alternative therapy, e.g., an alternative therapy described herein, e.g., a standard of care therapy for the cancer; thereby treating cancer in a subject.
  • In an embodiment, the disclosure provides a method of treating cancer (e.g., a hematological cancer such as ALL and CLL) in a subject, comprising: acquiring a value of responder status for the subject that comprises a measure of one or more of the following: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and a CD19 CAR-expressing cell gene set signature, and responsive to a determination of responder status, performing one, two, three four or more of: identifying the subject as a complete responder, partial responder or non-responder; administering a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019); selecting or altering a dosing of a CAR-expressing cell therapy; selecting or altering the schedule or time course of a CAR-expressing cell therapy; administering, e.g., to a non-responder or a partial responder, an additional agent in combination with a CAR-expressing cell therapy, e.g., a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein; administering to a non-responder or partial responder a therapy that increases the number of naïve T cells in the subject prior to treatment with a CAR-expressing cell therapy; modifying a manufacturing process of a CAR-expressing cell therapy, e.g., enrich for naïve T cells prior to introducing a nucleic acid encoding a CAR into the T cells, e.g., for a subject identified as a non-responder or a partial responder; selecting an alternative therapy, e.g., a standard of care for the cancer, e.g., for a non-responder or partial responder; or selecting an alternative CAR-expressing cell therapy; thereby treating cancer in a subject.
  • In some embodiments, the amount of the biomarker determined in a sample from a subject is quantified as an absolute measurement (e.g., ng/mL). Absolute measurements can easily be compared to a reference value or cut-off value. For example, a cut-off value can be determined that represents a disease progressing status; any absolute values falling either above (i.e., for biomarkers that increase expression with progression of a cancer, e.g., a hematological cancer such as ALL and CLL) or falling below (i.e., for biomarkers with decreased expression with progression of a cancer, e.g., a hematological cancer such as ALL and CLL) the cut-off value are likely to be disease progressing.
  • Alternatively, the relative amount of a biomarker is determined. In one embodiment, the relative amount is determined by comparing the expression and/or activity of one or more biomarkers in a subject with cancer to the expression of the biomarkers in a reference parameter. In some embodiments, a reference parameter is obtained from one or more of: a baseline or prior value for the subject, the subject at a different time interval, an average or median value for a cancer subject (e.g., patient) population, a healthy control, or a healthy subject population.
  • The present disclosure also pertains to the field of predictive medicine in which diagnostic assays, pharmacogenomics, and monitoring clinical trials are used for predictive purposes to thereby treat an individual prophylactically. Accordingly, one aspect of the present disclosure relates to assays for determining the amount, structure, and/or activity of polypeptides or nucleic acids corresponding to one or more markers described herein, in order to determine whether an individual having cancer (e.g., a hematological cancer such as CLL and ALL) or at risk of developing cancer (e.g., a hematological cancer such as CLL and ALL) will be more likely to respond to CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019).
  • Accordingly, in one aspect, the disclosure provides a method for determining whether a subject with cancer (e.g., a hematological cancer such as CLL and ALL) is likely to respond to a cell expressing a CAR, e.g., a CD19 CAR-expressing cell described herein, such as CTL019. In another aspect, the disclosure is drawn to a method for predicting a time course of disease. In still another aspect, the method is drawn to a method for predicting a probability of a significant event in the time course of the disease (e.g., reoccurrence or remission). In certain embodiments, the method comprises detecting a combination of biomarkers associated with responsiveness to treatment as described herein and determining whether the subject is likely to respond to treatment.
  • In an aspect, the disclosure provides a method for providing a prognosis for success rate of a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019) in a subject having cancer (e.g., a hematological cancer such as ALL and CLL), said method comprising steps of:
  • providing a biological sample from the subject;
  • determining the levels of expression of one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more) genes listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 to obtain a gene expression pattern for the sample; and based on the gene expression pattern obtained, providing a prognosis to the subject.
  • In an embodiment, the step of determining the levels of expression of the set of genes further comprises detecting the expression of mRNA expressed from said genes. In an embodiment, provided methods further comprise a step wherein determining the expression of mRNA comprises exposing said mRNA to a nucleic acid probe complementary to said mRNA.
  • In an embodiment, the step of determining the levels of expression of the set of genes further comprises detecting the expression of a polypeptide encoded by said genes.
  • In an embodiment, provided methods comprise selecting a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy described herein such as, e.g., CTL019) for the subject, based on the prognosis provided.
  • In some embodiments, the methods involve evaluation of a biological sample, e.g., a sample from a subject, e.g., a patient who has been diagnosed with or is suspected of having cancer (e.g., a hematological cancer such as CLL or ALL, e.g., presents with symptoms of CLL or ALL) to detect changes in expression and/or activity of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or more genes in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 and a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene signature.
  • The results of the screening method and the interpretation thereof are predictive of the patient disease progression (e.g., progression of a cancer, e.g., a hematological cancer such as ALL or CLL). According to the present invention, alterations in expression or activity of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or more genes in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 and a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene signature is indicative of cancer progression (e.g., a hematological cancer such as ALL and CLL) relative to an average or median value for a cancer patient population or to an average median for a population of healthy, cancer free subjects.
  • In yet another embodiment, the one or more alterations, e.g., alterations in biomarker expression are assessed at pre-determined intervals, e.g., a first point in time and at least at a subsequent point in time. In one embodiment, a time course is measured by determining the time between significant events in the course of a subject disease, wherein the measurement is predictive of whether a subject has a long time course. In another embodiment, the significant event is the progression from diagnosis to death. In another embodiment, the significant event is the progression from diagnosis to worsening disease.
  • Methods for Detection of Gene Expression
  • Biomarker expression level can also be assayed. Expression of a marker described herein can be assessed by any of a wide variety of known methods for detecting expression of a transcribed molecule or protein. Non-limiting examples of such methods include immunological methods for detection of secreted, cell-surface, cytoplasmic, or nuclear proteins, protein purification methods, protein function or activity assays, nucleic acid hybridization methods, nucleic acid reverse transcription methods, and nucleic acid amplification methods.
  • In certain embodiments, activity of a particular gene is characterized by a measure of gene transcript (e.g., mRNA), by a measure of the quantity of translated protein, or by a measure of gene product activity. Marker expression can be monitored in a variety of ways, including by detecting mRNA levels, protein levels, or protein activity, any of which can be measured using standard techniques. Detection can involve quantification of the level of gene expression (e.g., genomic DNA, CDNA, mRNA, protein, or enzyme activity), or, alternatively, can be a qualitative assessment of the level of gene expression, in particular in comparison with a control level. The type of level being detected will be clear from the context.
  • Methods of detecting and/or quantifying the gene transcript (mRNA or CDNA made therefrom) using nucleic acid hybridization techniques are known to those of skill in the art (see e.g., Sambrook et al. supra). For example, one method for evaluating the presence, absence, or quantity of CDNA involves a Southern transfer as described above. Briefly, the mRNA is isolated (e.g., using an acid guanidinium-phenol-chloroform extraction method, Sambrook et al. supra.) and reverse transcribed to produce CDNA. The CDNA is then optionally digested and run on a gel in buffer and transferred to membranes. Hybridization is then carried out using the nucleic acid probes specific for the target CDNA.
  • A general principle of such diagnostic and prognostic assays involves preparing a sample or reaction mixture that can contain a marker, and a probe, under appropriate conditions and for a time sufficient to allow the marker and probe to interact and bind, thus forming a complex that can be removed and/or detected in the reaction mixture. These assays can be conducted in a variety of ways.
  • For example, one method to conduct such an assay would involve anchoring the marker or probe onto a solid phase support, also referred to as a substrate, and detecting target marker/probe complexes anchored on the solid phase at the end of the reaction. In one embodiment of such a method, a sample from a subject, which is to be assayed for presence and/or concentration of marker, can be anchored onto a carrier or solid phase support. In another embodiment, the reverse situation is possible, in which the probe can be anchored to a solid phase and a sample from a subject can be allowed to react as an unanchored component of the assay.
  • In order to conduct assays with the above-mentioned approaches, the non-immobilized component is added to the solid phase upon which the second component is anchored. After the reaction is complete, uncomplexed components can be removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized upon the solid phase. The detection of marker/probe complexes anchored to the solid phase can be accomplished in a number of methods outlined herein.
  • In another embodiment, the probe, when it is the unanchored assay component, can be labeled for the purpose of detection and readout of the assay, either directly or indirectly, with detectable labels discussed herein and which are well-known to one skilled in the art.
  • It is also possible to directly detect marker/probe complex formation without further manipulation or labeling of either component (marker or probe), for example by utilizing the technique of fluorescence energy transfer (see, for example, Lakowicz et al., U.S. Pat. No. 5,631,169; Stavrianopoulos, et al., U.S. Pat. No. 4,868,103). A fluorophore label on the first, ‘donor’ molecule is selected such that, upon excitation with incident light of appropriate wavelength, its emitted fluorescent energy will be absorbed by a fluorescent label on a second ‘acceptor’ molecule, which in turn is able to fluoresce due to the absorbed energy. Alternately, the ‘donor’ protein molecule can simply utilize the natural fluorescent energy of tryptophan residues. Labels are chosen that emit different wavelengths of light, such that the ‘acceptor’ molecule label can be differentiated from that of the ‘donor’. Since the efficiency of energy transfer between the labels is related to the distance separating the molecules, spatial relationships between the molecules can be assessed. In a situation in which binding occurs between the molecules, the fluorescent emission of the ‘acceptor’ molecule label in the assay should be maximal. An FET binding event can be conveniently measured through standard fluorometric detection means well known in the art (e.g., using a fluorimeter).
  • In another embodiment, determination of the ability of a probe to recognize a marker can be accomplished without labeling either assay component (probe or marker) by utilizing a technology such as real-time Biomolecular Interaction Analysis (BIA) (see, e.g., Sjolander, S. and Urbaniczky, C., 1991, ANAL. CHEM. 63:2338-2345 and Szabo et al., 1995, CURR. OPIN. STRUCT. BIOL. 5:699-705). As used herein, “BIA” or “surface plasmon resonance” is a technology for studying biospecific interactions in real time, without labeling any of the interactants (e.g., BIAcore). Changes in the mass at the binding surface (indicative of a binding event) result in alterations of the refractive index of light near the surface (the optical phenomenon of surface plasmon resonance (SPR)), resulting in a detectable signal which can be used as an indication of real-time reactions between biological molecules.
  • Alternatively, in another embodiment, analogous diagnostic and prognostic assays can be conducted with marker and probe as solutes in a liquid phase. In such an assay, the complexed marker and probe are separated from uncomplexed components by any of a number of standard techniques, including but not limited to: differential centrifugation, chromatography, electrophoresis and immunoprecipitation. In differential centrifugation, marker/probe complexes can be separated from uncomplexed assay components through a series of centrifugal steps, due to the different sedimentation equilibria of complexes based on their different sizes and densities (see, for example, Rivas, G., and Minton, A. P., 1993, Trends Biochem Sci. 18(8):284-7). Standard chromatographic techniques can also be utilized to separate complexed molecules from uncomplexed ones. For example, gel filtration chromatography separates molecules based on size, and through the utilization of an appropriate gel filtration resin in a column format, for example, the relatively larger complex can be separated from the relatively smaller uncomplexed components. Similarly, the relatively different charge properties of the marker/probe complex as compared to the uncomplexed components can be exploited to differentiate the complex from uncomplexed components, for example, through the utilization of ion-exchange chromatography resins. Such resins and chromatographic techniques are well known to one skilled in the art (see, e.g., Heegaard, N. H., 1998, J. MOL. RECOGNIT. Winter 11(1-6):141-8; Hage, D. S., and Tweed, S. A. J CHROMATOGR B BIOMED SCI APPL 1997 Oct. 10; 699(1-2):499-525). Gel electrophoresis can also be employed to separate complexed assay components from unbound components (see, e.g., Ausubel et al., ed., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, 1987-1999). In this technique, protein or nucleic acid complexes are separated based on size or charge, for example. In order to maintain the binding interaction during the electrophoretic process, non-denaturing gel matrix materials and conditions in the absence of reducing agent are typical. Appropriate conditions to the particular assay and components thereof will be well known to one skilled in the art.
  • In a particular embodiment, the level of mRNA corresponding to the marker can be determined both by in situ and by in vitro formats in a biological sample using methods known in the art. The term “biological sample” is intended to include tissues, cells, biological fluids and isolates thereof, isolated from a subject, as well as tissues, cells and fluids present within a subject. Many expression detection methods use isolated RNA. For in vitro methods, any RNA isolation technique that does not select against the isolation of mRNA can be utilized for the purification of RNA from cells (see, e.g., Ausubel et al., ed., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York 1987-1999). Additionally, large numbers of tissue samples can readily be processed using techniques well known to those of skill in the art, such as, for example, the single-step RNA isolation process of Chomczynski (1989, U.S. Pat. No. 4,843,155).
  • The isolated nucleic acid can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, polymerase chain reaction analyses and probe arrays. One diagnostic method for the detection of mRNA levels involves contacting the isolated mRNA with a nucleic acid molecule (probe) that can hybridize to the mRNA encoded by the gene being detected. The nucleic acid probe can be, for example, a full-length CDNA, or a portion thereof, such as an oligonucleotide of at least 7, 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to a mRNA or genomic DNA encoding a marker of the present invention. Other suitable probes for use in the diagnostic assays are described herein. Hybridization of an mRNA with the probe indicates that the marker in question is being expressed.
  • In one format, the mRNA is immobilized on a solid surface and contacted with a probe, for example by running the isolated mRNA on an agarose gel and transferring the mRNA from the gel to a membrane, such as nitrocellulose. In an alternative format, the probe(s) are immobilized on a solid surface and the mRNA is contacted with the probe(s), for example, in an Affymetrix gene chip array. A skilled artisan can readily adapt known mRNA detection methods for use in detecting the level of mRNA encoded by the markers described herein.
  • The probes can be full length or less than the full length of the nucleic acid sequence encoding the protein. Shorter probes are empirically tested for specificity. Exemplary nucleic acid probes are 20 bases or longer in length (See, e.g., Sambrook et al. for methods of selecting nucleic acid probe sequences for use in nucleic acid hybridization). Visualization of the hybridized portions allows the qualitative determination of the presence or absence of CDNA.
  • An alternative method for determining the level of a transcript corresponding to a marker of the present invention in a sample involves the process of nucleic acid amplification, e.g., by rtPCR (the experimental embodiment set forth in Mullis, 1987, U.S. Pat. No. 4,683,202), ligase chain reaction (Barany, 1991, Proc. Natl. Acad. Sci. USA, 88:189-193), self-sustained sequence replication (Guatelli et al., 1990, PROC. NATL. ACAD. SCI. USA 87:1874-1878), transcriptional amplification system (Kwoh et al., 1989, PROC. NATL. ACAD. SCI. USA 86:1173-1177), Q-Beta Replicase (Lizardi et al., 1988, BIO/TECHNOLOGY 6:1197), rolling circle replication (Lizardi et al., U.S. Pat. No. 5,854,033) or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. Fluorogenic rtPCR can also be used in the methods of the invention. In fluorogenic rtPCR, quantitation is based on amount of fluorescence signals, e.g., TaqMan and sybr green. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers. As used herein, amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5′ or 3′ regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between. In general, amplification primers are from about 10 to 30 nucleotides in length and flank a region from about 50 to 200 nucleotides in length. Under appropriate conditions and with appropriate reagents, such primers permit the amplification of a nucleic acid molecule comprising the nucleotide sequence flanked by the primers.
  • For in situ methods, mRNA does not need to be isolated from the cells prior to detection. In such methods, a cell or tissue sample is prepared/processed using known histological methods. The sample is then immobilized on a support, typically a glass slide, and then contacted with a probe that can hybridize to mRNA that encodes the marker.
  • As an alternative to making determinations based on the absolute expression level of the marker, determinations can be based on the normalized expression level of the marker. Expression levels are normalized by correcting the absolute expression level of a marker by comparing its expression to the expression of a gene that is not a marker, e.g., a housekeeping gene that is constitutively expressed. Suitable genes for normalization include housekeeping genes such as the actin gene, or epithelial cell-specific genes. This normalization allows the comparison of the expression level in one sample, e.g., a subject sample, to another sample, e.g., a healthy subject, or between samples from different sources.
  • Alternatively, the expression level can be provided as a relative expression level. To determine a relative expression level of a marker, the level of expression of the marker can be determined for 10 or more samples of normal versus cancer isolates, or even 50 or more samples, prior to the determination of the expression level for the sample in question. The mean expression level of each of the genes assayed in the larger number of samples can be determined and this can be used as a baseline expression level for the marker. The expression level of the marker determined for the test sample (absolute level of expression) then can be divided by the mean expression value obtained for that marker. This provides a relative expression level.
  • In certain embodiments, the samples used in the baseline determination will be from samples derived from a subject having cancer (e.g., a hematological cancer such as ALL and CLL) versus samples from a healthy subject of the same tissue type. The choice of the cell source is dependent on the use of the relative expression level. Using expression found in normal tissues as a mean expression score aids in validating whether the marker assayed is specific to the tissue from which the cell was derived (versus normal cells). In addition, as more data is accumulated, the mean expression value can be revised, providing improved relative expression values based on accumulated data. Expression data from normal cells provides a means for grading the severity of the cancer disease state.
  • In another embodiment, expression of a marker is assessed by preparing genomic DNA or mRNA/CDNA (i.e., a transcribed polynucleotide) from cells in a subject sample, and by hybridizing the genomic DNA or mRNA/CDNA with a reference polynucleotide which is a complement of a polynucleotide comprising the marker, and fragments thereof. CDNA can, optionally, be amplified using any of a variety of polymerase chain reaction methods prior to hybridization with the reference polynucleotide. Expression of one or more markers can likewise be detected using quantitative PCR (QPCR) to assess the level of expression of the marker(s). Alternatively, any of the many known methods of detecting mutations or variants (e.g., single nucleotide polymorphisms, deletions, etc.) of a marker of the invention can be used to detect occurrence of a mutated marker in a subject.
  • In a related embodiment, a mixture of transcribed polynucleotides obtained from the sample is contacted with a substrate having fixed thereto a polynucleotide complementary to or homologous with at least a portion (e.g., at least 7, at least 10, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, at least 100, at least 500, or more nucleotide residues) of a marker described herein. If polynucleotides complementary to, or homologous with, a marker described herein are differentially detectable on the substrate (e.g., detectable using different chromophores or fluorophores, or fixed to different selected positions), then the levels of expression of a plurality of markers can be assessed simultaneously using a single substrate (e.g., a “gene chip” microarray of polynucleotides fixed at selected positions). When a method of assessing marker expression is used which involves hybridization of one nucleic acid with another, the hybridization can be performed under stringent hybridization conditions.
  • In another embodiment, a combination of methods to assess the expression of a marker is utilized.
  • Because the compositions, kits, and methods rely on detection of a difference in expression levels of one or more markers described herein, in certain embodiments the level of expression of the marker is significantly greater than the minimum detection limit of the method used to assess expression in at least one of a biological sample from a subject with cancer (e.g., a hematological cancer such as ALL and CLL) or a reference (e.g., a biological sample from a healthy subject, e.g., a subject without cancer).
  • Nucleic Acid Molecules and Probes
  • One aspect of the disclosure pertains to isolated nucleic acid molecules that correspond to one or markers described herein, including nucleic acids which encode a polypeptide corresponding to one or more markers described herein or a portion of such a polypeptide. The nucleic acid molecules include those nucleic acid molecules which reside in genomic regions identified herein. Isolated nucleic acid molecules also include nucleic acid molecules sufficient for use as hybridization probes to identify nucleic acid molecules that correspond to a marker described herein, including nucleic acid molecules which encode a polypeptide corresponding to a marker described herein, and fragments of such nucleic acid molecules, e.g., those suitable for use as PCR primers for the amplification or mutation of nucleic acid molecules. Nucleic acid molecules can be DNA molecules (e.g., CDNA or genomic DNA) and RNA molecules (e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs. The nucleic acid molecule can be single-stranded or double-stranded; in certain embodiments the nucleic acid molecule is double-stranded DNA.
  • An “isolated” nucleic acid molecule is one which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid molecule. In certain embodiments, an “isolated” nucleic acid molecule is free of sequences (such as protein-encoding sequences) which naturally flank the nucleic acid (i.e., sequences located at the 5 and 3 ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived.
  • The language “substantially free of other cellular material or culture medium” includes preparations of nucleic acid molecule in which the molecule is separated from cellular components of the cells from which it is isolated or recombinantly produced. Thus, nucleic acid molecule that is substantially free of cellular material includes preparations of nucleic acid molecule having less than about 30%, less than about 20%, less than about 10%, or less than about 5% (by dry weight) of other cellular material or culture medium.
  • If so desired, a nucleic acid molecule, e.g., the marker gene products identified herein, can be isolated using standard molecular biology techniques and the sequence information in the database records described herein. Using all or a portion of such nucleic acid sequences, nucleic acid molecules can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook et al., ed., MOLECULAR CLONING: A LABORATORY MANUAL, 2nd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989).
  • A nucleic acid molecule can be amplified using CDNA, mRNA, or genomic DNA as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques. The nucleic acid molecules so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis. Furthermore, oligonucleotides corresponding to all or a portion of a nucleic acid molecule of the invention can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
  • Probes based on the sequence of a nucleic acid molecule of the invention can be used to detect transcripts (e.g., mRNA) or genomic sequences corresponding to one or more markers described herein. The probe comprises a label group attached thereto, e.g., a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Such probes can be used as part of a diagnostic test kit for identifying cells or tissues which mis-express the protein, such as by measuring levels of a nucleic acid molecule encoding the protein in a sample of cells from a subject, e.g., detecting mRNA levels or determining whether a gene encoding the protein has been mutated or deleted.
  • Polypeptide Detection
  • Methods to measure biomarkers described herein, include, but are not limited to: Western blot, immunoblot, enzyme-linked immunosorbant assay (ELISA), radioimmunoassay (RIA), immunoprecipitation, surface plasmon resonance, chemiluminescence, fluorescent polarization, phosphorescence, immunohistochemical analysis, liquid chromatography mass spectrometry (LC-MS), matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry, microcytometry, microarray, microscopy, fluorescence activated cell sorting (FACS), magnetic activated cell sorting (MACS), flow cytometry, laser scanning cytometry, hematology analyzer and assays based on a property of the protein including but not limited to DNA binding, ligand binding, or interaction with other protein partners.
  • The activity or level of a marker protein can also be detected and/or quantified by detecting or quantifying the expressed polypeptide. The polypeptide can be detected and quantified by any of a number of means well known to those of skill in the art. These can include analytic biochemical methods such as electrophoresis, capillary electrophoresis, high performance liquid chromatography (HPLC), thin layer chromatography (TLC), hyperdiffusion chromatography, and the like, or various immunological methods such as fluid or gel precipitin reactions, immunodiffusion (single or double), immunoelectrophoresis, radioimmunoassay (RIA), enzyme-linked immunosorbent assays (ELISAs), immunofluorescent assays, Western blotting, immunohistochemistry and the like. A skilled artisan can readily adapt known protein/antibody detection methods for use in determining the expression level of one or more biomarkers in a serum sample.
  • Another agent for detecting a polypeptide is an antibody capable of binding to a polypeptide corresponding to a marker described herein, e.g., an antibody with a detectable label. Antibodies can be polyclonal or monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab)2) can be used. The term “labeled”, with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently labeled streptavidin.
  • In another embodiment, the antibody is labeled, e.g., a radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody. In another embodiment, an antibody derivative (e.g., an antibody conjugated with a substrate or with the protein or ligand of a protein-ligand pair (e.g., biotin-streptavidin)), or an antibody fragment (e.g., a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically with a protein corresponding to the marker, such as the protein encoded by the open reading frame corresponding to the marker or such a protein which has undergone all or a portion of its normal post-translational modification, is used.
  • Proteins from cells can be isolated using techniques that are well known to those of skill in the art. The protein isolation methods employed can, for example, be such as those described in Harlow and Lane (Harlow and Lane, 1988, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.).
  • In one format, antibodies, or antibody fragments, can be used in methods such as Western blots or immunofluorescence techniques to detect the expressed proteins. In such uses, one can immobilize either the antibody or proteins on a solid support. Suitable solid phase supports or carriers include any support capable of binding an antigen or an antibody. Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.
  • In another embodiment, the polypeptide is detected using an immunoassay. As used herein, an immunoassay is an assay that utilizes an antibody to specifically bind to the analyte. The immunoassay is thus characterized by detection of specific binding of a polypeptide to an anti-antibody as opposed to the use of other physical or chemical properties to isolate, target, and quantify the analyte.
  • The polypeptide is detected and/or quantified using any of a number of well recognized immunological binding assays (see, e.g., U.S. Pat. Nos. 4,366,241; 4,376,110; 4,517,288; and 4,837,168). For a review of the general immunoassays, see also Asai (1993) Methods in Cell Biology Volume 37: Antibodies in Cell Biology, Academic Press, Inc. New York; Stites & Terr (1991) Basic and Clinical Immunology 7th Edition.
  • In another embodiment, the polypeptide is detected and/or quantified using Luminex® assay technology. The Luminex® assay separates tiny color-coded beads into e.g., distinct sets that are each coated with a reagent for a particular bioassay, allowing the capture and detection of specific analytes from a sample in a multiplex manner. The Luminex® assay technology can be compared to a multiplex ELISA assay using bead-based fluorescence cytometry to detect analytes such as biomarkers.
  • The disclosure also encompasses kits for detecting the presence of a polypeptide or nucleic acid corresponding to a marker described herein in a biological sample, e.g., a sample containing tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, and bone marrow. Such kits can be used to determine if a subject is suffering from or is at increased risk of developing cancer (e.g., a hematological cancer such as CLL and ALL). For example, the kit can comprise a labeled compound or agent capable of detecting a polypeptide or an mRNA encoding a polypeptide corresponding to a marker described herein in a biological sample and means for determining the amount of the polypeptide or mRNA in the sample (e.g., an antibody which binds the polypeptide or an oligonucleotide probe which binds to DNA or mRNA encoding the polypeptide). Kits can also include instructions for interpreting the results obtained using the kit.
  • The disclosure thus includes a kit for assessing the disease progression of a subject having cancer (e.g., a hematological cancer such as CLL and ALL).
  • In an embodiment, a kit can be used to assess the disease progression of a cancer including, but not limited to, B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma, marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, and Waldenstrom macroglobulinemia. In an embodiment, the disclosure provides a kit for assessing the disease progression of a subject having a hematological cancer. In an embodiment, the disclosure provides a kit for assessing the disease progression of a subject having ALL. In another embodiment, the disclosure provides a kit for assessing the disease progression of a subject having CLL. In an embodiment, the disclosure provides a kit for assessing the disease progression of a subject having cancer that is associated with CD19 expression.
  • In an embodiment, the disclosure provides a kit for assessing and characterizing responder status (e.g., compete responder, partial responder or non-responder) of a subject having a hematological cancer to a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy as described herein, such as e.g., CTL019). In an embodiment, the disclosure provides a kit for assessing and characterizing responder status (e.g., compete responder, partial responder or non-responder) of a subject having ALL to a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy as described herein, such as e.g., CTL019). In an embodiment, the disclosure provides a kit for assessing and characterizing responder status (e.g., compete responder, partial responder or non-responder) of a subject having CLL to a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy as described herein, such as e.g., CTL019).
  • Suitable reagents for binding with a polypeptide corresponding to a marker described herein include antibodies, antibody derivatives, antibody fragments, and the like. Suitable reagents for binding with a nucleic acid (e.g., a genomic DNA, an mRNA, a spliced mRNA, a CDNA, or the like) include complementary nucleic acids. For example, the nucleic acid reagents can include oligonucleotides (labeled or non-labeled) fixed to a substrate, labeled oligonucleotides not bound with a substrate, pairs of PCR primers, molecular beacon probes, and the like.
  • The kit can optionally comprise additional components useful for performing the methods described herein. By way of example, the kit can comprise fluids (e.g., SSC buffer) suitable for annealing complementary nucleic acids or for binding an antibody with a protein with which it specifically binds, one or more sample compartments, an instructional material which describes performance of a method of the invention, a reference sample for comparison of expression levels of the biomarkers described herein, and the like.
  • A kit of the invention can comprise a reagent useful for determining protein level or protein activity of a marker.
  • In an embodiment, a kit is provided for providing a prognosis for success rate of a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell therapy as described herein, such as e.g., CTL019) in a subject having cancer (e.g., a hematological cancer such as CLL and ALL), said kit comprising:
  • a set of reagents that specifically detects expression levels of one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 50 or more) genes listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 and a CD19 CAR-expressing cell gene set signature; and
  • instructions for using said kit;
  • wherein said instructions for use provide that if one or more of the detected expression levels is greater than a reference level, the subject is more likely to respond positively to a CAR-expressing cell therapy.
  • In an embodiment, the set of reagents detects the expression of mRNA expressed from said set of genes.
  • In an embodiment, the set of reagents comprises nucleic acid probes complementary to mRNA expressed from said set of genes.
  • In an embodiment, the nucleic acid probes complementary to mRNA are CDNA or oligonucleotides.
  • In an embodiment, the nucleic acid probes complementary to mRNA are immobilized on a substrate surface.
  • In an embodiment, the set of reagents detects the expression of polypeptides encoded by said set of genes.
  • Therapeutic Agents, Compositions and Administration
  • The methods described herein can be used to assess a responder status to a cell expressing a CAR. In one embodiment, the cell expresses a CAR molecule comprising an antigen binding domain (e.g., an antibody or antibody fragment that specifically binds to a tumor antigen), a transmembrane domain, and an intracellular signaling domain (e.g., an intracellular signaling domain comprising a costimulatory domain and/or a primary signaling domain). In an embodiment, the antigen binding domain comprises any antibody, or a fragment thereof, e.g., an scFv, known in the art that targets or specifically binds to any of the tumor antigens described herein. For example, the tumor antigen is BCMA (also known as TNFRSF17, Tumor Necrosis Factor Receptor Superfamily, Member 17, or B Cell Maturation Antigen), CD33, CLL-1 (also known as C-type Lectin-Like domain family 1, or CLECL1) or claudin-6 (CLDN6). The antibody, or fragment thereof, can be a murine, humanized, or fully human antibody or fragment thereof, e.g., an scFv.
  • In one embodiment, the CAR comprises an antibody or antibody fragment which includes an anti-CD19 binding domain described herein (e.g., a murine or humanized antibody or antibody fragment that specifically binds to CD19 as described herein), a transmembrane domain described herein, and an intracellular signaling domain described herein (e.g., an intracellular signaling domain comprising a costimulatory domain and/or a primary signaling domain described herein).
  • Antigen Binding Domain
  • In one aspect, the CAR of the invention comprises a target-specific binding element otherwise referred to as an antigen binding domain. The choice of moiety depends upon the type and number of ligands that define the surface of a target cell. For example, the antigen binding domain may be chosen to recognize a ligand that acts as a cell surface marker on target cells associated with a particular disease state. Thus, examples of cell surface markers that may act as ligands for the antigen binding domain in a CAR of the invention include those associated with viral, bacterial and parasitic infections, autoimmune disease and cancer cells.
  • In one aspect, the CAR-mediated T-cell response can be directed to an antigen of interest by way of engineering an antigen binding domain that specifically binds a desired antigen into the CAR.
  • In one aspect, the portion of the CAR comprising the antigen binding domain comprises an antigen binding domain that targets a tumor antigen, e.g., a tumor antigen described herein.
  • The antigen binding domain can be any domain that binds to the antigen including but not limited to a monoclonal antibody, a polyclonal antibody, a recombinant antibody, a human antibody, a humanized antibody, and a functional fragment thereof, including but not limited to a single-domain antibody such as a heavy chain variable domain (VH), a light chain variable domain (VL) and a variable domain (VHH) of camelid derived nanobody, and to an alternative scaffold known in the art to function as antigen binding domain, such as a recombinant fibronectin domain, a T cell receptor (TCR), or a fragment there of, e.g., single chain TCR, and the like. In some instances, it is beneficial for the antigen binding domain to be derived from the same species in which the CAR will ultimately be used in. For example, for use in humans, it may be beneficial for the antigen binding domain of the CAR to comprise human or humanized residues for the antigen binding domain of an antibody or antibody fragment.
  • Any known CD19 CAR, e.g., the CD19 antigen binding domain of any known CD19 CAR, in the art can be used in accordance with the instant invention. For example, LG-740; CD19 CAR described in the U.S. Pat. Nos. 8,399,645; 7,446,190; Xu et al., Leuk Lymphoma. 2013 54(2):255-260(2012); Cruz et al., Blood 122(17):2965-2973 (2013); Brentjens et al., Blood, 118(18):4817-4828 (2011); Kochenderfer et al., Blood 116(20):4099-102 (2010); Kochenderfer et al., Blood 122 (25):4129-39(2013); and 16th Annu Meet Am Soc Gen Cell Ther (ASGCT) (May 15-18, Salt Lake City) 2013, Abst 10.
  • Exemplary target antigens that can be targeted using the CAR-expressing cells, include, but are not limited to, CD19, CD123, EGFRvIII, mesothelin, among others, as described in, for example, WO 2014/130635, WO 2014/130657, and WO 2015/090230, each of which is herein incorporated by reference in its entirety.
  • In one embodiment, the CAR T cell that specifically binds to CD19 has the USAN designation TISAGENLECLEUCEL-T. CTL019 is made by a gene modification of T cells is mediated by stable insertion via transduction with a self-inactivating, replication deficient Lentiviral (LV) vector containing the CTL019 transgene under the control of the EF-1 alpha promoter. CTL019 can be a mixture of transgene positive and negative T cells that are delivered to the subject on the basis of percent transgene positive T cells.
  • In other embodiments, the CAR-expressing cells can specifically bind to human CD19, e.g., can include a CAR molecule, or an antigen binding domain (e.g., a humanized antigen binding domain) according to Table 3 of WO2014/153270, incorporated herein by reference.
  • In other embodiments, the CAR-expressing cells can specifically bind to CD123, e.g., can include a CAR molecule (e.g., any of the CAR1-CAR8), or an antigen binding domain according to Tables 1-2 of WO 2014/130635, incorporated herein by reference.
  • In other embodiments, the CAR-expressing cells can specifically bind to EGFRvIII, e.g., can include a CAR molecule, or an antigen binding domain according to Table 2 or SEQ ID NO:11 of WO 2014/130657, incorporated herein by reference.
  • In other embodiments, the CAR-expressing cells can specifically bind to mesothelin, e.g., can include a CAR molecule, or an antigen binding domain according to Tables 2-3 of WO 2015/090230, incorporated herein by reference.
  • In one embodiment, the antigen binding domain comprises one, two three (e.g., all three) heavy chain CDRs, HC CDR1, HC CDR2 and HC CDR3, from an antibody listed above, and/or one, two, three (e.g., all three) light chain CDRs, LC CDR1, LC CDR2 and LC CDR3, from an antibody listed above. In one embodiment, the antigen binding domain comprises a heavy chain variable region and/or a variable light chain region of an antibody listed or described above.
  • In some embodiments, the tumor antigen is a tumor antigen described in International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety. In some embodiments, the tumor antigen is chosen from one or more of: CD19; CD123; CD22; CD30; CD171; CS-1 (also referred to as CD2 subset 1, CRACC, SLAMF7, CD319, and 19A24); C-type lectin-like molecule-1 (CLL-1 or CLECL1); CD33; epidermal growth factor receptor variant III (EGFRvIII); ganglioside G2 (GD2); ganglioside GD3 (aNeu5 Ac(2-8)aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer); TNF receptor family member B cell maturation (BCMA); Tn antigen ((Tn Ag) or (GalNAcα-Ser/Thr)); prostate-specific membrane antigen (PSMA); Receptor tyrosine kinase-like orphan receptor 1 (ROR1); Fms-Like Tyrosine Kinase 3 (FLT3); Tumor-associated glycoprotein 72 (TAG72); CD38; CD44v6; Carcinoembryonic antigen (CEA); Epithelial cell adhesion molecule (EPCAM); B7H3 (CD276); KIT (CD117); Interleukin-13 receptor subunit alpha-2 (IL-13Ra2 or CD213A2); Mesothelin; Interleukin 11 receptor alpha (IL-11Ra); prostate stem cell antigen (PSCA); Protease Serine 21 (Testisin or PRSS21); vascular endothelial growth factor receptor 2 (VEGFR2); Lewis(Y) antigen; CD24; Platelet-derived growth factor receptor beta (PDGFR-beta); Stage-specific embryonic antigen-4 (SSEA-4); CD20; Folate receptor alpha; Receptor tyrosine-protein kinase ERBB2 (Her2/neu); Mucin 1, cell surface associated (MUC1); epidermal growth factor receptor (EGFR); neural cell adhesion molecule (NCAM); Prostase; prostatic acid phosphatase (PAP); elongation factor 2 mutated (ELF2M); Ephrin B2; fibroblast activation protein alpha (FAP); insulin-like growth factor 1 receptor (IGF-I receptor), carbonic anhydrase IX (CAIX); Proteasome (Prosome, Macropain) Subunit, Beta Type, 9 (LMP2); glycoprotein 100 (gp100); oncogene fusion protein consisting of breakpoint cluster region (BCR) and Abelson murine leukemia viral oncogene homolog 1 (Abl) (bcr-abl); tyrosinase; ephrin type-A receptor 2 (EphA2); Fucosyl GM1; sialyl Lewis adhesion molecule (sLe); ganglioside GM3 (aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer); transglutaminase 5 (TGS5); high molecular weight-melanoma-associated antigen (HMWMAA); o-acetyl-GD2 ganglioside (OAcGD2); Folate receptor beta; tumor endothelial marker 1 (TEM1/CD248); tumor endothelial marker 7-related (TEM7R); claudin 6 (CLDN6); thyroid stimulating hormone receptor (TSHR); G protein-coupled receptor class C group 5, member D (GPRC5D); chromosome X open reading frame 61 (CXORF61); CD97; CD179a; anaplastic lymphoma kinase (ALK); Polysialic acid; placenta-specific 1 (PLAC1); hexasaccharide portion of globoH glycoceramide (GloboH); mammary gland differentiation antigen (NY-BR-1); uroplakin 2 (UPK2); Hepatitis A virus cellular receptor 1 (HAVCR1); adrenoceptor beta 3 (ADRB3); pannexin 3 (PANX3); G protein-coupled receptor 20 (GPR20); lymphocyte antigen 6 complex, locus K 9 (LY6K); Olfactory receptor 51E2 (OR51E2); TCR Gamma Alternate Reading Frame Protein (TARP); Wilms tumor protein (WT1); Cancer/testis antigen 1 (NY-ESO-1); Cancer/testis antigen 2 (LAGE-1a); Melanoma-associated antigen 1 (MAGE-A1); ETS translocation-variant gene 6, located on chromosome 12p (ETV6-AML); sperm protein 17 (SPA17); X Antigen Family, Member 1A (XAGE1); angiopoietin-binding cell surface receptor 2 (Tie 2); melanoma cancer testis antigen-1 (MAD-CT-1); melanoma cancer testis antigen-2 (MAD-CT-2); Fos-related antigen 1; tumor protein p53 (p53); p53 mutant; prostein; surviving; telomerase; prostate carcinoma tumor antigen-1 (PCTA-1 or Galectin 8), melanoma antigen recognized by T cells 1 (MelanA or MART1); Rat sarcoma (Ras) mutant; human Telomerase reverse transcriptase (hTERT); sarcoma translocation breakpoints; melanoma inhibitor of apoptosis (ML-IAP); ERG (transmembrane protease, serine 2 (TMPRSS2) ETS fusion gene); N-Acetyl glucosaminyl-transferase V (NA17); paired box protein Pax-3 (PAX3); Androgen receptor; Cyclin B1; v-myc avian myelocytomatosis viral oncogene neuroblastoma derived homolog (MYCN); Ras Homolog Family Member C (RhoC); Tyrosinase-related protein 2 (TRP-2); Cytochrome P450 1B1 (CYP1B1); CCCTC-Binding Factor (Zinc Finger Protein)-Like (BORIS or Brother of the Regulator of Imprinted Sites), Squamous Cell Carcinoma Antigen Recognized By T Cells 3 (SART3); Paired box protein Pax-5 (PAX5); proacrosin binding protein sp32 (OY-TES1); lymphocyte-specific protein tyrosine kinase (LCK); A kinase anchor protein 4 (AKAP-4); synovial sarcoma, X breakpoint 2 (SSX2); Receptor for Advanced Glycation Endproducts (RAGE-1); renal ubiquitous 1 (RU1); renal ubiquitous 2 (RU2); legumain; human papilloma virus E6 (HPV E6); human papilloma virus E7 (HPV E7); intestinal carboxyl esterase; heat shock protein 70-2 mutated (mut hsp70-2); CD79a; CD79b; CD72; Leukocyte-associated immunoglobulin-like receptor 1 (LAIR1); Fc fragment of IgA receptor (FCAR or CD89); Leukocyte immunoglobulin-like receptor subfamily A member 2 (LILRA2); CD300 molecule-like family member f (CD300LF); C-type lectin domain family 12 member A (CLEC12A); bone marrow stromal cell antigen 2 (BST2); EGF-like module-containing mucin-like hormone receptor-like 2 (EMR2); lymphocyte antigen 75 (LY75); Glypican-3 (GPC3); Fc receptor-like 5 (FCRL5); and immunoglobulin lambda-like polypeptide 1 (IGLL1).
  • CD19 CAR Constructs
  • Murine CD19 CAR constructs are described in PCT publication WO 2012/079000, incorporated herein by reference, and the amino acid sequence of the murine CD19 CAR and scFv constructs are shown in Table 2 below.
  • TABLE 2
    Murine CD19 CAR Constructs
    CTL019 Full- SEQ ID MALPVTALLLPLALLLHAARPdiqmtqttsslsaslgdrvtiscrasqdiskylnw
    aa NO: 81 yqqkpdgtvklliyhtsrlhsgvpsrfsgsgsgtdysltisnleqediatyfcqqg
    ntlpytfgggtkleitggggsggggsggggsevklqesgpglvapsqslsvtctvs
    gvslpdygvswirqpprkglewlgviwgsettyynsalksrltiikdnsksqvflk
    mnslqtddtaiyycakhyyyggsyamdywgqgtsvtvsstttpaprpptpaptias
    qplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitlyckrgr
    kkllyifkqpfmrpvqttqeedgcscrfpeeeeggcelrvkfsrsadapaykqgqn
    qlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeaysei
    gmkgerrrgkghdglyqglstatkdtydalhmqalppr
    CTL019 scFv SEQ ID Diqmtqttsslsaslgdrvtiscrasqdiskylnwyqqkpdgtvklliyhtsrlhs
    domain NO: 52 gvpsrfsgsgsgtdysltisnleqediatyfcqqgntlpytfgggtkleitggggs
    ggggsggggsevklqesgpglvapsgslsvtctvsgvslpdygvswirqpprkgle
    wlgviwgsettyynsalksrltiikdnsksqvflkmnslqtddtaiyycakhyyyg
    gsyamdywgqgtsvtvss
    mCAR1 scFv SEQ ID QVQLLESGAELVRPGSSVKISCKASGYAFSSYWMNWVKQRPGQGLEWIGQIYPGDG
    NO: 84 DTNYNGKFKGQATLTADKSSSTAYMQLSGLTSEDSAVYSCARKTISSVVDFYFDYW
    GQGTTVTGGGSGGGSGGGSGGGSELVLTQSPKFMSTSVGDRVSVTCKASQNVGTNV
    AWYQQKPGQSPKPLIYSATYRNSGVPDRFTGSGSGTDFTLTITNVQSKDLADYFCQ
    YNRYPYTSFFFTKLEIKRRS
    mCAR1 Full- SEQ ID QVQLLESGAELVRPGSSVKISCKASGYAFSSYWMNWVKQRPGQGLEWIGQIYPGDG
    aa NO: 85 DTNYNGKFKGQATLTADKSSSTAYMQLSGLTSEDSAVYSCARKTISSVVDFYFDYW
    GQGTTVTGGGSGGGSGGGSGGGSELVLTQSPKFMSTSVGDRVSVTCKASQNVGTNV
    AWYQQKPGQSPKPLIYSATYRNSGVPDRFTGSGSGTDFTLTITNVQSKDLADYFCQ
    YNRYPYTSFFFTKLEIKRRSKIEVMYPPPYLDNEKSNGTIIHVKGKHLCPSPLFPG
    PSKPFWVLVVVGGVLACYSLLVTVAFTIFWVRSKRSRLLHSDYMNMTPRRPGPTRK
    HYQPYAPPRDFAAYRSRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRG
    RDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTA
    TKDTYDALHMQALPPR
    mCAR2 scFv SEQ ID DIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWYQQKPDGTVKLLIYHTSRLHS
    NO: 86 GVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNTLPYTFGGGTKLEITGSTSG
    SGKPGSGEGSTKGEVKLQESGPGLVAPSQSLSVTCTVSGVSLPDYGVSWIRQPPRK
    GLEWLGVIWGSETTYYNSALKSRLTIIKDNSKSQVFLKMNSLQTDDTAIYYCAKHY
    YYGGSYAMDYWGQGTSVTVSSE
    mCAR2 CAR- SEQ ID DIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWYQQKPDGTVKLLIYHTSRLHS
    aa NO: 87 GVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNTLPYTFGGGTKLEITGSTSG
    SGKPGSGEGSTKGEVKLQESGPGLVAPSQSLSVTCTVSGVSLPDYGVSWIRQPPRK
    GLEWLGVIWGSETTYYNSALKSRLTIIKDNSKSQVFLKMNSLQTDDTAIY
    YCAKHYYYGGSYAMDYWGQGTSVTVSSESKYGPPCPPCPMFWVLVVVGGVLACYSL
    LVTVAFTIFWVKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFEEEEGGCELRVKF
    SRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYN
    ELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPRL
    mCAR2 Full- SEQ ID DIQMTQTT SSLSASLGDR VTISCRASQD ISKYLNWYQQ KPDGTVKLLI
    aa NO: 88 YHTSRLHSGV PSRFSGSGSG TDYSLTISNL EQEDIATYFC QQGNTLPYTF
    GGGTKLEITG STSGSGKPGS GEGSTKGEVK LQESGPGLVA PSQSLSVTCT
    VSGVSLPDYG VSWIRQPPRK GLEWLGVIWG SETTYYNSAL KSRLTIIKDN
    SKSQVFLKMN SLQTDDTAIY YCAKHYYYGG SYAMDYWGQG TSVTVSSESK
    YGPPCPPCPM FWVLVVVGGV LACYSLLVTV
    AFIIFWVKRG RKKLLYIFKQ PFMRPVQTTQ EEDGCSCRFE EEEGGCELRV
    KFSRSADAPA YQQGQNQLYN ELNLGRREEY DVLDKRRGRD PEMGGKPRRK
    NPQEGLYNEL QKDKMAEAYS EIGMKGERRR GKGHDGLYQG LSTATKDTYD
    ALHMQALPPR LEGGGEGRGS LLTCGDVEEN PGPRMLLLVT SLLLCELPHP
    AFLLIPRKVC NGIGIGEFKD SLSINATNIK HFKNCTSISG DLHILPVAFR
    GDSFTHTPPL DPQELDILKT VKEITGFLLI QAWPENRTDL HAFENLEIIR
    GRTKQHGQFS LAVVSLNITS LGLRSLKEIS DGDVIISGNK NLCYANTINW
    KKLFGTSGQK TKIISNRGEN SCKATGQVCH ALCSPEGCWG PEPRDCVSCR
    NVSRGRECVD KCNLLEGEPR EFVENSECIQ CHPECLPQAM NITCTGRGPD
    NCIQCAHYID GPHCVKTCPA GVMGENNTLV WKYADAGHVC HLCHPNCTYG
    CTGPGLEGCP TNGPKIPSIA TGMVGALLLL LVVALGIGLF M
    mCAR3 scFv SEQ ID DIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWYQQKPDGTVKLLIYHTSRLHS
    NO: 89 GVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNTLPYTFGGGTKLEITGSTSG
    SGKPGSGEGSTKGEVKLQESGPGLVAPSQSLSVTCTVSGVSLPDYGVSWIRQPPRK
    GLEWLGVIWGSETTYYNSALKSRLTIIKDNSKSQVFLKMNSLQTDDTAIYYCAKHY
    YYGGSYAMDYWGQGTSVTVSS
    mCAR3 Full- SEQ ID DIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWYQQKPDGTVKLLIYHTSRLHS
    aa NO: 90 GVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNTLPYTFGGGTKLEITGSTSG
    SGKPGSGEGSTKGEVKLQESGPGLVAPSQSLSVTCTVSGVSLPDYGVSWIRQPPRK
    GLEWLGVIWGSETTYYNSALKSRLTIIKDNSKSQVFLKMNSLQTDDTAIYYCAKHY
    YYGGSYAMDYWGQGTSVTVSSAAAIEVMYPPPYLDNEKSNGTIIHVKGKHLCPSPL
    FPGPSKPFWVLVVVGGVLACYSLLVTVAFTIFWVRSKRSRLLHSDYMNMTPRRPGP
    TRKHYQPYAPPRDFAAYRSRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDK
    RRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGL
    STATKDTYDALHMQALPPR
  • CD19 CAR constructs containing humanized anti-CD19 scFv domains are described in PCT publication WO 2014/153270, incorporated herein by reference.
  • In an embodiment, the antigen binding domain comprises an anti-CD19 antibody, or fragment thereof, e.g., an scFv. For example, the antigen binding domain comprises a variable heavy chain and a variable light chain listed in Table 12. The linker sequence joining the variable heavy and variable light chains can be, e.g., any of the linker sequences described herein, or alternatively, can be GSTSGSGKPGSGEGSTKG (SEQ ID NO:45).
  • TABLE 12
    Anti-CD 19 antibody binding domains
    CD19 huscFv1 EIVMTQSPATLSLSPGERATLSCRASQDISKYLNWYQQKPGQAPRLLTYHTSRL
    HSGIPARFSGSGSGTDYTLTISSLQPEDFAVYFCQQGNTLPYTFGQGTKLEIKG
    GGGSGGGGSGGGGSQVQLQESGPGLVKPSETLSLICTVSGVSLPDYGVSWIRQP
    PGKGLEWIGVIWGSETTYYSSSLKSRVTISKDNSKNQVSLKLSSVTAADTAVYY
    CAKHYYYGGSYAMDYWGQGTLVTVSS (SEQ ID NO: 24)
    CD19 huscFv2 Eivmtqspatlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgip
    arfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikggggsggggsg
    gggsqvqlqesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgse
    ttyygsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgqgt
    lvtvss (SEQ ID NO: 25)
    CD19 huscFv3 Qvqlqesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgsettyy
    ssslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgqgtlvtv
    ssggggsggggsggggseivmtqspatlslspgeratlscrasqdiskylnwyqqkpgq
    aprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqg
    tkleik (SEQ ID NO: 26)
    CD19 huscFv4 Qvqlqesqpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgsettyy
    qsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgqgtlvtv
    ssggggsggggsggggseivmtqspatlslspgeratlscrasqdiskylnwyqqkpgq
    aprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqg
    tkleik (SEQ ID NO: 27)
    CD19 huscFv5 Eivmtqspatlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgip
    arfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikggggsggggsg
    gggsggggsqvqlgesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigv
    iwgsettyyssslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdy
    wgqgtlvtvss (SEQ ID NO: 39)
    CD19 huscFv6 Eivmtgspatlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgip
    arfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikggggsggggsg
    gggsggggsqvglgesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigv
    iwgsettyygsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdy
    wgqgtivtvss (SEQ ID NO: 43)
    CD19 huscFv7 Qvglgesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgsettyy
    ssslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgqgtivtv
    ssggggsggggsggggsggggseivmtgspat1s1spgeratlscrasqdiskylnwyq
    qkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqgntlpy
    tfgqgtkleik (SEQ ID NO: 46)
    CD19 huscFv8 Qvglgesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgsettyy
    gsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgqgtivtv
    ssggggsggggsggggsggggseivmtgspat1s1spgeratlscrasqdiskylnwyq
    qkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqgntlpy
    tfgqgtkleik (SEQ ID NO: 47)
    CD19 huscFv9 Eivmtqspatlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgip
    arfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikggggsggggsg
    gggsggggsqvqlgesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigv
    iwgsettyynsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdy
    wgqgtivtvss (SEQ ID NO: 48)
    CD19 Hu Qvglqesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgsettyy
    scFv10 nsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgqgtlvtv
    ssggggsggggsggggsggggseivmtqspatlslspgeratlscrasqdiskylnwyq
    qkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqgntlpy
    tfgqgtkleik (SEQ ID NO: 49)
    CD19 Hu Eivmtqspatlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgip
    scFv11 arfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikggggsggggsg
    gggsqvqlgesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgse
    ttyynsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgqgt
    lvtvss (SEQ ID NO: 50)
    CD19 Hu Qvqlqesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgsettyy
    scFv12 nsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgqgtlvtv
    ssggggsggggsggggseivmtgspat1s1spgeratlscrasqdiskylnwyqqkpgq
    aprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqg
    tkleik (SEQ ID NO: 51)
    CD19 muCTL019 Diqmtqttsslsaslgdrvtiscrasqdiskylnwyqqkpdgtvklliyhtsrlhsgvp
    srfsgsgsgtdysltisnleqediatyfcqqgntlpytfgggtkleitggggsggggsg
    gggsevklqesgpglvapsqslsvtctvsgvslpdygvswirqpprkglewlgviwgse
    ttyynsalksrltiikdnsksqvflkmnslqtddtaiyycakhyyyggsyamdywgqgt
    svtvss (SEQ ID NO: 52)
    CD19 SSJ25- QVQLLESGAELVRPGSSVKISCKASGYAFSSYWMNWVKQRPGQGLEWIGQIYPGDGDTN
    Cl VH YNGKFKGQATLTADKSSSTAYMQLSGLTSEDSAVYSCARKTISSVVDFYFDYWGQGTTV
    sequence T (SEQ ID NO: 53)
    CD19 SSJ25- ELVLTQSPKFMSTSVGDRVSVTCKASQNVGTNVAWYQQKPGQSPKPLIYSATYRNSGVP
    Cl VL DRFTGSGSGTDFTLTITNVQSKDLADYFYFCQYNRYPYTSGGGTKLEIKRRS (SEQ
    sequence ID NO: 54)
  • Any known CD19 CAR, e.g., the CD19 antigen binding domain of any known CD19 CAR, in the art can be used in accordance with the present disclosure. For example, LG-740; CD19 CAR described in the U.S. Pat. Nos. 8,399,645; 7,446,190; Xu et al., LEUK LYMPHOMA. 2013 54(2):255-260(2012); Cruz et al., BLOOD 122(17):2965-2973 (2013); Brentjens et al., BLOOD, 118(18):4817-4828 (2011); Kochenderfer et al., BLOOD 116(20):4099-102 (2010); Kochenderfer et al., BLOOD 122 (25):4129-39(2013); and 16th Annu Meet Am Soc Gen Cell Ther (ASGCT) (May 15-18, Salt Lake City) 2013, Abst 10.
  • In one embodiment, the antigen binding domain comprises one, two three (e.g., all three) heavy chain CDRs, HC CDR1, HC CDR2 and HC CDR3, from an antibody listed above, and/or one, two, three (e.g., all three) light chain CDRs, LC CDR1, LC CDR2 and LC CDR3, from an antibody listed above. In one embodiment, the antigen binding domain comprises a heavy chain variable region and/or a variable light chain region of an antibody listed or described above.
  • In an embodiment, an antigen binding domain against CD22 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Haso et al., BLOOD, 121(7): 1165-1174 (2013); Wayne et al., CLIN CANCER RES 16(6): 1894-1903 (2010); Kato et al., LEUK RES 37(1):83-88 (2013); Creative BioMart (creativebiomart.net): MOM-18047-S(P).
  • In an embodiment, an antigen binding domain against CD20 is an antigen binding portion, e.g., CDRs, of the anitbody Rituximab, Ofatumumab, Ocrelizumab, Veltuzumab, or GA101.
  • In an embodiment, an antigen binding domain against ROR1 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Hudecek et al., CLIN CANCER RES 19(12):3153-3164 (2013); WO 2011159847; and US20130101607.
  • Bispecific CARS
  • In an embodiment a multispecific antibody molecule is a bispecific antibody molecule. A bispecific antibody has specificity for no more than two antigens. A bispecific antibody molecule is characterized by a first immunoglobulin variable domain sequence which has binding specificity for a first epitope and a second immunoglobulin variable domain sequence that has binding specificity for a second epitope. In an embodiment the first and second epitopes are on the same antigen, e.g., the same protein (or subunit of a multimeric protein). In an embodiment the first and second epitopes overlap. In an embodiment the first and second epitopes do not overlap. In an embodiment the first and second epitopes are on different antigens, e.g., different proteins (or different subunits of a multimeric protein). In an embodiment a bispecific antibody molecule comprises a heavy chain variable domain sequence and a light chain variable domain sequence which have binding specificity for a first epitope and a heavy chain variable domain sequence and a light chain variable domain sequence which have binding specificity for a second epitope. In an embodiment a bispecific antibody molecule comprises a half antibody having binding specificity for a first epitope and a half antibody having binding specificity for a second epitope. In an embodiment a bispecific antibody molecule comprises a half antibody, or fragment thereof, having binding specificity for a first epitope and a half antibody, or fragment thereof, having binding specificity for a second epitope. In an embodiment a bispecific antibody molecule comprises a scFv, or fragment thereof, have binding specificity for a first epitope and a scFv, or fragment thereof, have binding specificity for a second epitope.
  • In certain embodiments, the antibody molecule is a multi-specific (e.g., a bispecific or a trispecific) antibody molecule. Protocols for generating bispecific or heterodimeric antibody molecules, and various configurations for bispecific antibody molecules, are described in, e.g., paragraphs 455-458 of WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.
  • In one aspect, the bispecific antibody molecule is characterized by a first immunoglobulin variable domain sequence, e.g., a scFv, which has binding specificity for CD19, e.g., comprises a scFv as described herein, or comprises the light chain CDRs and/or heavy chain CDRs from a scFv described herein, and a second immunoglobulin variable domain sequence that has binding specificity for a second epitope on a different antigen.
  • Chimeric TCR
  • In one aspect, the antibodies and antibody fragments of the present invention (e.g., CD19 antibodies and fragments) can be grafted to one or more constant domain of a T cell receptor (“TCR”) chain, for example, a TCR alpha or TCR beta chain, to create a chimeric TCR. Without being bound by theory, it is believed that chimeric TCRs will signal through the TCR complex upon antigen binding. For example, an scFv as disclosed herein, can be grafted to the constant domain, e.g., at least a portion of the extracellular constant domain, the transmembrane domain and the cytoplasmic domain, of a TCR chain, for example, the TCR alpha chain and/or the TCR beta chain. As another example, an antibody fragment, for example a VL domain as described herein, can be grafted to the constant domain of a TCR alpha chain, and an antibody fragment, for example a VH domain as described herein, can be grafted to the constant domain of a TCR beta chain (or alternatively, a VL domain may be grafted to the constant domain of the TCR beta chain and a VH domain may be grafted to a TCR alpha chain). As another example, the CDRs of an antibody or antibody fragment may be grafted into a TCR alpha and/or beta chain to create a chimeric TCR. For example, the LCDRs disclosed herein may be grafted into the variable domain of a TCR alpha chain and the HCDRs disclosed herein may be grafted to the variable domain of a TCR beta chain, or vice versa. Such chimeric TCRs may be produced, e.g., by methods known in the art (For example, Willemsen R A et al, Gene Therapy 2000; 7: 1369-1377; Zhang T et al, Cancer Gene Ther 2004; 11: 487-496; Aggen et al, Gene Ther. 2012 April; 19(4):365-74).
  • Non Antibody Scaffolds
  • In embodiments, the antigen binding domain comprises a non-antibody scaffold, e.g., a fibronectin, ankyrin, domain antibody, lipocalin, small modular immuno-pharmaceutical, maxybody, Protein A, or affilin. The non-antibody scaffold has the ability to bind to target antigen on a cell. In embodiments, the antigen binding domain is a polypeptide or fragment thereof of a naturally occurring protein expressed on a cell. In some embodiments, the antigen binding domain comprises a non-antibody scaffold. A wide variety of non-antibody scaffolds can be employed so long as the resulting polypeptide includes at least one binding region which specifically binds to the target antigen on a target cell.
  • Non-antibody scaffolds include: fibronectin (Novartis, MA), ankyrin (Molecular Partners AG, Zurich, Switzerland), domain antibodies (Domantis, Ltd., Cambridge, Mass., and Ablynx nv, Zwijnaarde, Belgium), lipocalin (Pieris Proteolab AG, Freising, Germany), small modular immuno-pharmaceuticals (Trubion Pharmaceuticals Inc., Seattle, Wash.), maxybodies (Avidia, Inc., Mountain View, Calif.), Protein A (Affibody AG, Sweden), and affilin (gamma-crystallin or ubiquitin) (Scil Proteins GmbH, Halle, Germany).
  • In an embodiment the antigen binding domain comprises the extracellular domain, or a counter-ligand binding fragment thereof, of molecule that binds a counterligand on the surface of a target cell.
  • Transmembrane Domain
  • In embodiments, a CAR described herein comprises a transmembrane domain that is fused to an extracellular sequence, e.g., an extracellular recognition element, which can comprise an antigen binding domain. In an embodiment, the transmembrane domain is one that naturally is associated with one of the domains in the CAR. In an embodiment, the transmembrane domain is one that is not naturally associated with one of the domains in the CAR.
  • A transmembrane domain can include one or more additional amino acids adjacent to the transmembrane region, e.g., one or more amino acid associated with the extracellular region of the protein from which the transmembrane was derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the extracellular region) and/or one or more additional amino acids associated with the intracellular region of the protein from which the transmembrane protein is derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the intracellular region).
  • In embodiments, the transmembrane domain is one which minimizes interactions with other elements, e.g., other transmembrane domains. In some instances, the transmembrane domain minimizes binding of such domains to the transmembrane domains of the same or different surface membrane proteins, e.g., to minimize interactions with other members of the receptor complex. Suitable examples can be derived by selection or modification of amino acid substitution of a known transmembrane domain. In an embodiment, the transmembrane domain is capable of promoting homodimerization with another CAR on the cell surface.
  • The transmembrane domain may comprise a naturally occurring, or a non-naturally occurring synthetic sequence. Where naturally occurring, the transmembrane domain may be derived from any membrane-bound or transmembrane protein.
  • Transmembrane regions suitable for use in molecules described herein may be derived from any one or more of e.g., the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154. In some embodiments, a transmembrane domain may include at least the transmembrane region(s) of, e.g., KIRDS2, OX40, CD2, CD27, LFA-1 (CD11a, CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD160, CD19, IL2R beta, IL2R gamma, IL7R a, ITGA1, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, PAG/Cbp, NKG2D, NKG2C, or CD19. In an embodiment the transmembrane domain is derived from CD8. In an embodiment the transmembrane domain is derived from CD28. In one aspect, the transmembrane domain is a transmembrane domain from the sequence provided as SEQ ID NO: 12 or SEQ ID NO: 42.
  • In an embodiment, a sequence, e.g., a hinge or spacer sequence, can be disposed between a transmembrane domain and another sequence or domain to which it is fused. In embodiments, a variety of human hinges (aka “spacers”) can be employed as well, e.g., including but not limited to the human Ig (immunoglobulin) hinge. Optionally, a short oligo- or polypeptide linker, between 2 and 10 amino acids in length may form the linkage between the transmembrane domain and another domain, e.g., an intracellular signaling domain or costimulatory domain, of a CAR. A glycine-serine doublet provides a particularly suitable linker. In one aspect, the hinge or spacer is the amino acid sequence provided as SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 8. In one aspect, the hinge or spacer comprises a KIR2DS2 hinge.
  • In an embodiment, the transmembrane domain may be a non-naturally occurring sequence, in which case can comprise predominantly hydrophobic residues such as leucine and valine. In an embodiment, a triplet of phenylalanine, tryptophan and valine will be found at each end of a transmembrane domain.
  • Optionally, a short oligo- or polypeptide linker, between 2 and 10 amino acids in length may form the linkage between the transmembrane domain and the cytoplasmic region of the CAR. A glycine-serine doublet provides a particularly suitable linker. For example, in one aspect, the linker comprises the amino acid sequence of GGGGSGGGGS (SEQ ID NO:10). In some embodiments, the linker is encoded by a nucleotide sequence of GGTGGCGGAGGTTCTGGAGGTGGAGGTTCC (SEQ ID NO:11).
  • Cytoplasmic Domain
  • The cytoplasmic domain or region of the CAR includes an intracellular signaling domain. An intracellular signaling domain is generally responsible for activation of at least one of the normal effector functions of the immune cell in which the CAR has been introduced.
  • Examples of intracellular signaling domains for use in the CAR of the invention include the cytoplasmic sequences of the T cell receptor (TCR) and co-receptors that act in concert to initiate signal transduction following antigen receptor engagement, as well as any derivative or variant of these sequences and any recombinant sequence that has the same functional capability.
  • It is known that signals generated through the TCR alone are insufficient for full activation of the T cell and that a secondary and/or costimulatory signal is also required. Thus, T cell activation can be said to be mediated by two distinct classes of cytoplasmic signaling sequences: those that initiate antigen-dependent primary activation through the TCR (primary intracellular signaling domains) and those that act in an antigen-independent manner to provide a secondary or costimulatory signal (secondary cytoplasmic domain, e.g., a costimulatory domain).
  • Primary Signaling Domain
  • A primary signaling domain regulates primary activation of the TCR complex either in a stimulatory way, or in an inhibitory way. Primary intracellular signaling domains that act in a stimulatory manner may contain signaling motifs which are known as immunoreceptor tyrosine-based activation motifs or ITAMs.
  • Examples of ITAM containing primary intracellular signaling domains that are of particular use in the invention include those of TCR zeta, FcR gamma, FcR beta, CD3 gamma, CD3 delta, CD3 epsilon, CD5, CD22, CD79a, CD79b, CD278 (also known as “ICOS”), FccRI, DAP10, DAP12, and CD66d. In one embodiment, a CAR of the invention comprises an intracellular signaling domain, e.g., a primary signaling domain of CD3-zeta, e.g., a CD3-zeta sequence described herein.
  • In one embodiment, a primary signaling domain comprises a modified ITAM domain, e.g., a mutated ITAM domain which has altered (e.g., increased or decreased) activity as compared to the native ITAM domain. In one embodiment, a primary signaling domain comprises a modified ITAM-containing primary intracellular signaling domain, e.g., an optimized and/or truncated ITAM-containing primary intracellular signaling domain. In an embodiment, a primary signaling domain comprises one, two, three, four or more ITAM motifs. Further examples of molecules containing a primary intracellular signaling domain that are of particular use in the invention include those of DAP10, DAP12, and CD32.
  • A primary intracellular signaling domain comprises a functional fragment, or analog, of a primary stimulatory molecule (e.g., CD3 zeta-GenBank Acc. No. BAG36664.1). The primary intracellular signaling domain can comprise the entire intracellular region or a fragment of the intracellular region which is sufficient for generation of an intracellular signal when an antigen binding domain to which it is fused binds cognate antigen. In embodiments the primary intracellular signaling domain has at least 70, 75, 80, 85, 90, 95, 98, or 99% sequence identity with the entire intracellular region, or a fragment of the intracellular region which is sufficient for generation of an intracellular signal, of a naturally occurring primary stimulatory molecule, e.g., a human (GenBank Acc No. BAG36664.1), or other mammalian, e.g., a nonhuman species, e.g., rodent, monkey, ape or murine intracellular primary stimulatory molecule. In embodiments the primary intracellular signaling domain has at least 70, 75, 80, 85, 90, 95, 98, or 99% sequence identity with SEQ ID NO: 18 or SEQ ID NO: 20.
  • In embodiments, the primary intracellular signaling domain, has at least 70, 75, 80, 85, 90, 95, 96, 97, 98, or 99% identity with, or differs by no more than 30, 25, 20, 15, 10, 5, 4, 3, 2, or 1 amino acid residues from the corresponding residues of the entire intracellular region, or a fragment of the intracellular region which is sufficient for generation of an intracellular signal, of a naturally occurring human primary stimulatory molecule, e.g., a naturally occurring human primary stimulatory molecule disclosed herein.
  • Costimulatory Signaling Domain
  • The intracellular signalling domain of the CAR can comprise the CD3-zeta signalling domain by itself or it can be combined with any other desired intracellular signalling domain(s) useful in the context of a CAR of the invention. For example, the intracellular signalling domain of the CAR can comprise a CD3 zeta chain portion and a costimulatory signaling domain. The costimulatory signaling domain refers to a portion of the CAR comprising the intracellular domain of a costimulatory molecule. In one embodiment, the intracellular domain is designed to comprise the signaling domain of CD3-zeta and the signaling domain of CD28. In one aspect, the intracellular domain is designed to comprise the signaling domain of CD3-zeta and the signaling domain of ICOS.
  • A costimulatory molecule is a cell surface molecule other than an antigen receptor or its ligands that is required for an efficient response of lymphocytes to an antigen. Examples of such molecules include MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM-1, ICOS (CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, NKG2D, NKG2C, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, CD19a, and a ligand that specifically binds with CD83, and the like. For example, CD27 costimulation has been demonstrated to enhance expansion, effector function, and survival of human CAR-expressing cell (e.g., T cell, NK cell) cells in vitro and augments human T cell persistence and antitumor activity in vivo (Song et al. BLOOD. 2012; 119(3):696-706). Further examples of such costimulatory molecules include CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKG2D and NKG2C.
  • The intracellular signaling sequences within the cytoplasmic portion of the CAR of the invention may be linked to each other in a random or specified order. Optionally, a short oligo- or polypeptide linker, for example, between 2 and 10 amino acids (e.g., 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) in length may form the linkage between intracellular signaling sequence. In one embodiment, a glycine-serine doublet can be used as a suitable linker. In one embodiment, a single amino acid, e.g., an alanine, a glycine, can be used as a suitable linker.
  • In one aspect, the intracellular signaling domain is designed to comprise two or more, e.g., 2, 3, 4, 5, or more, costimulatory signaling domains. In an embodiment, the two or more, e.g., 2, 3, 4, 5, or more, costimulatory signaling domains, are separated by a linker molecule, e.g., a linker molecule described herein. In one embodiment, the intracellular signaling domain comprises two costimulatory signaling domains. In some embodiments, the linker molecule is a glycine residue. In some embodiments, the linker is an alanine residue.
  • A costimulatory domain comprises a functional fragment, or analog, of a costimulatory molecule (e.g., ICOS, CD28, or 4-1BB). It can comprise the entire intracellular region or a fragment of the intracellular region which is sufficient for generation of an intracellular signal, e.g., when an antigen binding domain to which it is fused binds cognate antigen. In embodiments the costimulatory domain has at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity with the entire intracellular region, or a fragment of the intracellular region which is sufficient for generation of an intracellular signal, of a naturally occurring costimulatory molecule as described herein, e.g., a human, or other mammalian, e.g., a nonhuman species, e.g., rodent, monkey, ape or murine intracellular costimulatory molecule. In embodiments the costimulatory domain has at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity with SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 40, or SEQ ID NO: 44.
  • In embodiments the costimulatory signaling domain, has at least 70, 75, 80, 85, 90, 95, 96, 97, 98, or 99% identity with, or differs by no more than 30, 25, 20, 15, 10, 5, 4, 3, 2, or 1 amino acid residues from the corresponding residues of the entire intracellular region, or a fragment of the intracellular region which is sufficient for generation of an intracellular signal, of, a naturally occurring human costimulatory molecule, e.g., a naturally occurring human costimulatory molecule disclosed herein.
  • Any of the CARs described herein can include one or more of the components listed in Table 11.
  • TABLE 11
    Sequences of various components of CAR
    (aa—amino acids, na—nucleic acids
    that encodes the corresponding protein)
    SEQ
    ID
    NO description Sequence
    1 EF-1 CGTGAGGCTCCGGTGCCCGTCAGTGGGCAGAGCGCACATCGCCCACAGTCC
    promoter CCGAGAAGTTGGGGGGAGGGGTCGGCAATTGAACCGGTGCCTAGAGAAGG
    TGGCGCGGGGTAAACTGGGAAAGTGATGTCGTGTACTGGCTCCGCCTTTTTC
    CCGAGGGTGGGGGAGAACCGTATATAAGTGCAGTAGTCGCCGTGAACGTTC
    TTTTTCGCAACGGGTTTGCCGCCAGAACACAGGTAAGTGCCGTGTGTGGTTC
    CCGCGGGCCTGGCCTCTTTACGGGTTATGGCCCTTGCGTGCCTTGAATTACTT
    CCACCTGGCTGCAGTACGTGATTCTTGATCCCGAGCTTCGGGTTGGAAGTGG
    GTGGGAGAGTTCGAGGCCTTGCGCTTAAGGAGCCCCTTCGCCTCGTGCTTGA
    GTTGAGGCCTGGCCTGGGCGCTGGGGCCGCCGCGTGCGAATCTGGTGGCAC
    CTTCGCGCCTGTCTCGCTGCTTTCGATAAGTCTCTAGCCATTTAAAATTTTTGA
    TGACCTGCTGCGACGCTTTTTTTCTGGCAAGATAGTCTTGTAAATGCGGGCC
    AAGATCTGCACACTGGTATTTCGGTTTTTGGGGCCGCGGGCGGCGACGGGG
    CCCGTGCGTCCCAGCGCACATGTTCGGCGAGGCGGGGCCTGCGAGCGCGGC
    CACCGAGAATCGGACGGGGGTAGTCTCAAGCTGGCCGGCCTGCTCTGGTGC
    CTGGCCTCGCGCCGCCGTGTATCGCCCCGCCCTGGGCGGCAAGGCTGGCCC
    GGTCGGCACCAGTTGCGTGAGCGGAAAGATGGCCGCTTCCCGGCCCTGCTG
    CAGGGAGCTCAAAATGGAGGACGCGGCGCTCGGGAGAGCGGGCGGGTGA
    GTCACCCACACAAAGGAAAAGGGCCTTTCCGTCCTCAGCCGTCGCTTCATGT
    GACTCCACGGAGTACCGGGCGCCGTCCAGGCACCTCGATTAGTTCTCGAGCT
    TTTGGAGTACGTCGTCTTTAGGTTGGGGGGAGGGGTTTTATGCGATGGAGT
    TTCCCCACACTGAGTGGGTGGAGACTGAAGTTAGGCCAGCTTGGCACTTGAT
    GTAATTCTCCTTGGAATTTGCCCTTTTTGAGTTTGGATCTTGGTTCATTCTCAA
    GCCTCAGACAGTGGTTCAAAGTTTTTTTCTTCCATTTCAGGTGTCGTGA
    2 Leader (aa) MALPVTALLLPLALLLHAARP
    3 Leader (na) ATGGCCCTGCCTGTGACAGCCCTGCTGCTGCCTCTGGCTCTGCTGCTGCATGC
    CGCTAGACCC
    4 CD 8 hinge TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD
    (aa)
    5 CD8 hinge ACCACGACGCCAGCGCCGCGACCACCAACACCGGCGCCCACCATCGCGTCG
    (na) CAGCCCCTGTCCCTGCGCCCAGAGGCGTGCCGGCCAGCGGCGGGGGGCGCA
    GTGCACACGAGGGGGCTGGACTTCGCCTGTGAT
    6 Ig4 hinge ESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEV
    (aa) QFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSN
    KGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVE
    WESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALH
    NHYTQKSLSLSLGKM
    7 Ig4 hinge GAGAGCAAGTACGGCCCTCCCTGCCCCCCTTGCCCTGCCCCCGAGTTCCTGG
    (na) GCGGACCCAGCGTGTTCCTGTTCCCCCCCAAGCCCAAGGACACCCTGATGAT
    CAGCCGGACCCCCGAGGTGACCTGTGTGGTGGTGGACGTGTCCCAGGAGGA
    CCCCGAGGTCCAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCACAACGC
    CAAGACCAAGCCCCGGGAGGAGCAGTTCAATAGCACCTACCGGGTGGTGTC
    CGTGCTGACCGTGCTGCACCAGGACTGGCTGAACGGCAAGGAATACAAGTG
    TAAGGTGTCCAACAAGGGCCTGCCCAGCAGCATCGAGAAAACCATCAGCAA
    GGCCAAGGGCCAGCCTCGGGAGCCCCAGGTGTACACCCTGCCCCCTAGCCA
    AGAGGAGATGACCAAGAACCAGGTGTCCCTGACCTGCCTGGTGAAGGGCTT
    CTACCCCAGCGACATCGCCGTGGAGTGGGAGAGCAACGGCCAGCCCGAGAA
    CAACTACAAGACCACCCCCCCTGTGCTGGACAGCGACGGCAGCTTCTTCCTG
    TACAGCCGGCTGACCGTGGACAAGAGCCGGTGGCAGGAGGGCAACGTCTTT
    AGCTGCTCCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGAGCC
    TGAGCCTGTCCCTGGGCAAGATG
    8 IgD hinge RWPESPKAQASSVPTAQPQAEGSLAKATTAPATTRNTGRGGEEKKKEKEKEEQ
    (aa) EERETKTPECPSHTQPLGVYLLTPAVQDLWLRDKATFTCFVVGSDLKDAHLTWE
    VAGKVPTGGVEEGLLERHSNGSQSQHSRLTLPRSLWNAGTSVTCTLNHPSLPPQ
    RLMALREPAAQAPVKLSLNLLASSDPPEAASWLLCEVSGFSPPNILLMWLEDQR
    EVNTSGFAPARPPPQPGSTTFWAWSVLRVPAPPSPQPATYTCVVSHEDSRTLL
    NASRSLEVSYVTDH
    9 IgD hinge AGGTGGCCCGAAAGTCCCAAGGCCCAGGCATCTAGTGTTCCTACTGCACAGC
    (na) CCCAGGCAGAAGGCAGCCTAGCCAAAGCTACTACTGCACCTGCCACTACGCG
    CAATACTGGCCGTGGCGGGGAGGAGAAGAAAAAGGAGAAAGAGAAAGAA
    GAACAGGAAGAGAGGGAGACCAAGACCCCTGAATGTCCATCCCATACCCAG
    CCGCTGGGCGTCTATCTCTTGACTCCCGCAGTACAGGACTTGTGGCTTAGAG
    ATAAGGCCACCTTTACATGTTTCGTCGTGGGCTCTGACCTGAAGGATGCCCA
    TTTGACTTGGGAGGTTGCCGGAAAGGTACCCACAGGGGGGGTTGAGGAAG
    GGTTGCTGGAGCGCCATTCCAATGGCTCTCAGAGCCAGCACTCAAGACTCAC
    CCTTCCGAGATCCCTGTGGAACGCCGGGACCTCTGTCACATGTACTCTAAATC
    ATCCTAGCCTGCCCCCACAGCGTCTGATGGCCCTTAGAGAGCCAGCCGCCCA
    GGCACCAGTTAAGCTTAGCCTGAATCTGCTCGCCAGTAGTGATCCCCCAGAG
    GCCGCCAGCTGGCTCTTATGCGAAGTGTCCGGCTTTAGCCCGCCCAACATCT
    TGCTCATGTGGCTGGAGGACCAGCGAGAAGTGAACACCAGCGGCTTCGCTC
    CAGCCCGGCCCCCACCCCAGCCGGGTTCTACCACATTCTGGGCCTGGAGTGT
    CTTAAGGGTCCCAGCACCACCTAGCCCCCAGCCAGCCACATACACCTGTGTT
    GTGTCCCATGAAGATAGCAGGACCCTGCTAAATGCTTCTAGGAGTCTGGAG
    GTTTCCTACGTGACTGACCATT
    10 GS GGGGSGGGGS
    hinge/linker
    (aa)
    11 GS GGTGGCGGAGGTTCTGGAGGTGGAGGTTCC
    hinge/linker
    (na)
    12 CD8TM (aa) IYIWAPLAGTCGVLLLSLVITLYC
    13 CD8 TM (na) ATCTACATCTGGGCGCCCTTGGCCGGGACTTGTGGGGTCCTTCTCCTGTCACT
    GGTTATCACCCTTTACTGC
    14 4-1BB KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL
    intracellular
    domain (aa)
    15 4-1BB AAACGGGGCAGAAAGAAACTCCTGTATATATTCAAACAACCATTTATGAGAC
    intracellular CAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCTGCCGATTTCCAGAAG
    domain (na) AAGAAGAAGGAGGATGTGAACTG
    16 CD27 (aa) QRRKYRSNKGESPVEPAEPCRYSCPREEEGSTIPIQEDYRKPEPACSP
    17 CD 27 (na) AGGAGTAAGAGGAGCAGGCTCCTGCACAGTGACTACATGAACATGACTCCC
    CGCCGCCCCGGGCCCACCCGCAAGCATTACCAGCCCTATGCCCCACCACGCG
    ACTTCGCAGCCTATCGCTCC
    18 CD3-zeta RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRK
    (aa) NPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDAL
    HMQALPPR
    19 CD3-zeta AGAGTGAAGTTCAGCAGGAGCGCAGACGCCCCCGCGTACAAGCAGGGCCA
    (na) GAACCAGCTCTATAACGAGCTCAATCTAGGACGAAGAGAGGAGTACGATGT
    TTTGGACAAGAGACGTGGCCGGGACCCTGAGATGGGGGGAAAGCCGAGAA
    GGAAGAACCCTCAGGAAGGCCTGTACAATGAACTGCAGAAAGATAAGATGG
    CGGAGGCCTACAGTGAGATTGGGATGAAAGGCGAGCGCCGGAGGGGCAAG
    GGGCACGATGGCCTTTACCAGGGTCTCAGTACAGCCACCAAGGACACCTAC
    GACGCCCTTCACATGCAGGCCCTGCCCCCTCGC
    20 CD3-zeta RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRK
    (aa) NPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDAL
    HMQALPPR
    21 CD3-zeta AGAGTGAAGTTCAGCAGGAGCGCAGACGCCCCCGCGTACCAGCAGGGCCA
    (na) GAACCAGCTCTATAACGAGCTCAATCTAGGACGAAGAGAGGAGTACGATGT
    TTTGGACAAGAGACGTGGCCGGGACCCTGAGATGGGGGGAAAGCCGAGAA
    GGAAGAACCCTCAGGAAGGCCTGTACAATGAACTGCAGAAAGATAAGATGG
    CGGAGGCCTACAGTGAGATTGGGATGAAAGGCGAGCGCCGGAGGGGCAAG
    GGGCACGATGGCCTTTACCAGGGTCTCAGTACAGCCACCAAGGACACCTAC
    GACGCCCTTCACATGCAGGCCCTGCCCCCTCGC
    22 linker GGGGS
    23 linker GGTGGCGGAGGTTCTGGAGGTGGAGGTTCC
    28 linker (Gly-Gly-Gly-Ser)n, where n = 1-10
    29 linker (Gly4 Ser)4
    30 linker (Gly4 Ser)3
    31 linker (Gly3Ser)
    32 polyA A2000
    33 polyA A150
    34 polyA A5000
    35 polyT T100
    36 polyT T5000
    37 polyA A64
    38 polyA A400
  • Combination of CARs
  • In one aspect, the CAR-expressing cell described herein can further comprise a second CAR, e.g., a second CAR that includes a different antigen binding domain, e.g., to the same target or a different target (e.g., a target other than a cancer associated antigen described herein or a different cancer associated antigen described herein, e.g., CD19, CD33, CLL-1, CD34, FLT3, or folate receptor beta). In one embodiment, the second CAR includes an antigen binding domain to a target expressed the same cancer cell type as the cancer associated antigen. In one embodiment, the CAR-expressing cell comprises a first CAR that targets a first antigen and includes an intracellular signaling domain having a costimulatory signaling domain but not a primary signaling domain, and a second CAR that targets a second, different, antigen and includes an intracellular signaling domain having a primary signaling domain but not a costimulatory signaling domain. While not wishing to be bound by theory, placement of a costimulatory signaling domain, e.g., 4-1BB, CD28, ICOS, CD27 or OX-40, onto the first CAR, and the primary signaling domain, e.g., CD3 zeta, on the second CAR can limit the CAR activity to cells where both targets are expressed. In one embodiment, the CAR expressing cell comprises a first cancer associated antigen CAR that includes an antigen binding domain that binds a target antigen described herein, a transmembrane domain and a costimulatory domain and a second CAR that targets a different target antigen (e.g., an antigen expressed on that same cancer cell type as the first target antigen) and includes an antigen binding domain, a transmembrane domain and a primary signaling domain. In another embodiment, the CAR expressing cell comprises a first CAR that includes an antigen binding domain that binds a target antigen described herein, a transmembrane domain and a primary signaling domain and a second CAR that targets an antigen other than the first target antigen (e.g., an antigen expressed on the same cancer cell type as the first target antigen) and includes an antigen binding domain to the antigen, a transmembrane domain and a costimulatory signaling domain.
  • In one embodiment, the CAR-expressing cell comprises a CAR described herein (e.g., a CD19 CAR) and an inhibitory CAR. In one embodiment, the inhibitory CAR comprises an antigen binding domain that binds an antigen found on normal cells but not cancer cells, e.g., normal cells that also express CLL. In one embodiment, the inhibitory CAR comprises the antigen binding domain, a transmembrane domain and an intracellular domain of an inhibitory molecule. For example, the intracellular domain of the inhibitory CAR can be an intracellular domain of PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR (e.g., TGFRbeta).
  • In one embodiment, when the CAR-expressing cell comprises two or more different CARs, the antigen binding domains of the different CARs can be such that the antigen binding domains do not interact with one another. For example, a cell expressing a first and second CAR can have an antigen binding domain of the first CAR, e.g., as a fragment, e.g., an scFv, that does not form an association with the antigen binding domain of the second CAR, e.g., the antigen binding domain of the second CAR is a VHH.
  • In some embodiments, when present on the surface of a cell, binding of the antigen binding domain of the first CAR to its cognate antigen is not substantially reduced by the presence of the second CAR. In some embodiments, binding of the antigen binding domain of the first CAR to its cognate antigen in the presence of the second CAR is 85%, 90%, 95%, 96%, 97%, 98% or 99% of binding of the antigen binding domain of the first CAR to its cognate antigen in the absence of the second CAR.
  • In some embodiments, when present on the surface of a cell, the antigen binding domains of the first CAR said second CAR, associate with one another less than if both were scFv antigen binding domains. In some embodiments, the antigen binding domains of the first CAR and the second CAR, associate with one another 85%, 90%, 95%, 96%, 97%, 98% or 99% less than if both were scFv antigen binding domains.
  • CAR-Expressing Cells
  • The CARs described herein are expressed on cells, e.g., immune effector cells, e.g., T cells. For example, a nucleic acid construct of a CAR described herein is transduced to a T cell. In embodiments, the cells expressing the CARs described herein are an in vitro transcribed RNA CAR T cell.
  • Sources of Cells, e.g., T Cells
  • Prior to expansion and genetic modification or other modification, a source of cells, e.g., immune effector cells, e.g., T cells or NK cells, can be obtained from a subject. Examples of subjects include humans, monkeys, chimpanzees, dogs, cats, mice, rats, and transgenic species thereof. T cells can be obtained from a number of sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors. In some embodiments, the cells obtained as described in this section are subjected to an assay described herein, e.g., one or more biomarkers are assayed.
  • In certain aspects of the present disclosure, immune effector cells, e.g., T cells or NK cells, can be obtained from a unit of blood collected from a subject using any number of techniques known to the skilled artisan, such as Ficoll™ separation. In one aspect, cells from the circulating blood of an individual are obtained by apheresis. The apheresis product typically contains lymphocytes, including T cells, monocytes, granulocytes, B cells, other nucleated white blood cells, red blood cells, and platelets. In one aspect, the cells collected by apheresis may be washed to remove the plasma fraction and, optionally, to place the cells in an appropriate buffer or media for subsequent processing steps. In one embodiment, the cells are washed with phosphate buffered saline (PBS). In an alternative embodiment, the wash solution lacks calcium and may lack magnesium or may lack many if not all divalent cations.
  • Initial activation steps in the absence of calcium can lead to magnified activation. As those of ordinary skill in the art would readily appreciate a washing step may be accomplished by methods known to those in the art, such as by using a semi-automated “flow-through” centrifuge (for example, the Cobe 2991 cell processor, the Baxter CytoMate, or the Haemonetics Cell Saver 5) according to the manufacturer's instructions. After washing, the cells may be resuspended in a variety of biocompatible buffers, such as, for example, Ca-free, Mg-free PBS, PlasmaLyte A, or other saline solution with or without buffer. Alternatively, the undesirable components of the apheresis sample may be removed and the cells directly resuspended in culture media.
  • It is recognized that the methods of the application can utilize culture media conditions comprising 5% or less, for example 2%, human AB serum, and employ known culture media conditions and compositions, for example those described in Smith et al., “Ex vivo expansion of human T cells for adoptive immunotherapy using the novel Xeno-free CTS Immune Cell Serum Replacement” Clinical & Translational Immunology (2015) 4, e31; doi:10.1038/cti.2014.31.
  • In one aspect, T cells are isolated from peripheral blood lymphocytes by lysing the red blood cells and depleting the monocytes, for example, by centrifugation through a PERCOLL™ gradient or by counterflow centrifugal elutriation.
  • The methods described herein can include, e.g., selection of a specific subpopulation of immune effector cells, e.g., T cells, that are a T regulatory cell-depleted population, CD25+ depleted cells, using, e.g., a negative selection technique, e.g., described herein. In embodiments, the population of T regulatory depleted cells contains less than 30%, 25%, 20%, 15%, 10%, 5%, 4%, 3%, 2%, 1% of CD25+ cells.
  • In one embodiment, T regulatory cells, e.g., CD25+ T cells, are removed from the population using an anti-CD25 antibody, or fragment thereof, or a CD25-binding ligand, e.g., IL-2. In one embodiment, the anti-CD25 antibody, or fragment thereof, or CD25-binding ligand is conjugated to a substrate, e.g., a bead, or is otherwise coated on a substrate, e.g., a bead. In one embodiment, the anti-CD25 antibody, or fragment thereof, is conjugated to a substrate as described herein.
  • In one embodiment, the T regulatory cells, e.g., CD25+ T cells, are removed from the population using CD25 depletion reagent from Miltenyi™. In one embodiment, the ratio of cells to CD25 depletion reagent is 1e7 cells to 20 uL, or 1e7 cells to15 uL, or 1e7 cells to 10 uL, or 1e7 cells to 5 uL, or 1e7 cells to 2.5 uL, or 1e7 cells to 1.25 uL. In one embodiment, e.g., for T regulatory cells, e.g., CD25+ depletion, greater than 500 million cells/ml is used. In a further aspect, a concentration of cells of 600, 700, 800, or 900 million cells/ml is used.
  • In one embodiment, the population of immune effector cells to be depleted includes about 6×109 CD25+ T cells. In other aspects, the population of immune effector cells to be depleted include about 1×109 to 1×1010 CD25+ T cell, and any integer value in between. In one embodiment, the resulting population T regulatory depleted cells has 2×109 T regulatory cells, e.g., CD25+ cells, or less (e.g., 1×109, 5×108, 1×108, 5×107, 1×107, or less CD25+ cells).
  • In one embodiment, the T regulatory cells, e.g., CD25+ cells, are removed from the population using the CliniMAC system with a depletion tubing set, such as, e.g., tubing 162-01. In one embodiment, the CliniMAC system is run on a depletion setting such as, e.g., DEPLETION2.1.
  • Without wishing to be bound by a particular theory, decreasing the level of negative regulators of immune cells (e.g., decreasing the number of unwanted immune cells, e.g., TREG cells), in a subject prior to apheresis or during manufacturing of a CAR-expressing cell product can reduce the risk of subject relapse. In an embodiment, a patient is pre-treated with one or more therapies that reduce TREG cells prior to collection of cells for CAR-expressing cell (e.g., T cell, NK cell) product manufacturing, thereby reducing the risk of patient relapse to CAR-expressing cell (e.g., T cell, NK cell) treatment (e.g., CTL019 treatment). Methods of depleting TREG cells are known in the art. Methods of decreasing TREG cells include, but are not limited to, cyclophosphamide, anti-GITR antibody, CD25-depletion, and combinations thereof.
  • In some embodiments, the manufacturing methods comprise reducing the number of (e.g., depleting) TREG cells prior to manufacturing of the CAR-expressing cell. For example, manufacturing methods comprise contacting the sample, e.g., the apheresis sample, with an anti-GITR antibody and/or an anti-CD25 antibody (or fragment thereof, or a CD25-binding ligand), e.g., to deplete TREG cells prior to manufacturing of the CAR-expressing cell (e.g., T cell, NK cell) product.
  • In an embodiment, a patient is pre-treated with cyclophosphamide prior to collection of cells for CAR-expressing cell (e.g., T cell, NK cell) product manufacturing, thereby reducing the risk of patient relapse to CAR-expressing cell treatment (e.g., CTL019 treatment). In an embodiment, a patient is pre-treated with an anti-GITR antibody prior to collection of cells for CAR-expressing cell (e.g., T cell, NK cell) product manufacturing, thereby reducing the risk of patient relapse to CAR-expressing cell treatment (e.g., CTL019 treatment).
  • In an embodiment, the CAR-expressing cell (e.g., T cell, NK cell) manufacturing process is modified to deplete TREG cells prior to manufacturing of the CAR-expressing cell (e.g., T cell, NK cell) product (e.g., a CTL019 product). In an embodiment, CD25-depletion is used to deplete TREG cells prior to manufacturing of the CAR-expressing cell (e.g., T cell, NK cell) product (e.g., a CTL019 product).
  • The methods described herein can include more than one selection step, e.g., more than one depletion step. Enrichment of a T cell population by negative selection can be accomplished, e.g., with a combination of antibodies directed to surface markers unique to the negatively selected cells. One method is cell sorting and/or selection via negative magnetic immunoadherence or flow cytometry that uses a cocktail of monoclonal antibodies directed to cell surface markers present on the cells negatively selected. For example, to enrich for CD4+ cells by negative selection, a monoclonal antibody cocktail can include antibodies to CD14, CD20, CD11b, CD16, HLA-DR, and CD8.
  • The methods described herein can further include removing cells from the population which express a tumor antigen, e.g., a tumor antigen that does not comprise CD25, e.g., CD19, CD30, CD38, CD123, CD20, CD14 or CD11b, to thereby provide a population of T regulatory depleted, e.g., CD25+ depleted, and tumor antigen depleted cells that are suitable for expression of a CAR, e.g., a CAR described herein. In one embodiment, tumor antigen expressing cells are removed simultaneously with the T regulatory, e.g., CD25+ cells. For example, an anti-CD25 antibody, or fragment thereof, and an anti-tumor antigen antibody, or fragment thereof, can be attached to the same substrate, e.g., bead, which can be used to remove the cells or an anti-CD25 antibody, or fragment thereof, or the anti-tumor antigen antibody, or fragment thereof, can be attached to separate beads, a mixture of which can be used to remove the cells. In other embodiments, the removal of T regulatory cells, e.g., CD25+ cells, and the removal of the tumor antigen expressing cells is sequential, and can occur, e.g., in either order.
  • Also provided are methods that include removing cells from the population which express a check point inhibitor, e.g., a check point inhibitor described herein, e.g., one or more of PD1+ cells, LAG3+ cells, and TIM3+ cells, to thereby provide a population of T regulatory depleted, e.g., CD25+ depleted cells, and check point inhibitor depleted cells, e.g., PD1+, LAG3+ and/or TIM3+ depleted cells. Exemplary check point inhibitors include B7-H1, B&-1, CD160, P1H, 2B4, PD1, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, TIGIT, CTLA-4, BTLA and LAIR1. In one embodiment, check point inhibitor expressing cells are removed simultaneously with the T regulatory, e.g., CD25+ cells. For example, an anti-CD25 antibody, or fragment thereof, and an anti-check point inhibitor antibody, or fragment thereof, can be attached to the same bead which can be used to remove the cells, or an anti-CD25 antibody, or fragment thereof, and the anti-check point inhibitor antibody, or fragment there, can be attached to separate beads, a mixture of which can be used to remove the cells. In other embodiments, the removal of T regulatory cells, e.g., CD25+ cells, and the removal of the check point inhibitor expressing cells is sequential, and can occur, e.g., in either order.
  • Methods described herein can include a positive selection step. For example, T cells can isolated by incubation with anti-CD3/anti-CD28 (e.g., 3×28)-conjugated beads, such as DYNABEADS® M-450 CD3/CD28 T, for a time period sufficient for positive selection of the desired T cells. In one embodiment, the time period is about 30 minutes. In a further embodiment, the time period ranges from 30 minutes to 36 hours or longer and all integer values there between. In a further embodiment, the time period is at least 1, 2, 3, 4, 5, or 6 hours. In yet another embodiment, the time period is 10 to 24 hours, e.g., 24 hours. Longer incubation times may be used to isolate T cells in any situation where there are few T cells as compared to other cell types, such in isolating tumor infiltrating lymphocytes (TIL) from tumor tissue or from immunocompromised individuals. Further, use of longer incubation times can increase the efficiency of capture of CD8+ T cells. Thus, by simply shortening or lengthening the time T cells are allowed to bind to the CD3/CD28 beads and/or by increasing or decreasing the ratio of beads to T cells (as described further herein), subpopulations of T cells can be preferentially selected for or against at culture initiation or at other time points during the process. Additionally, by increasing or decreasing the ratio of anti-CD3 and/or anti-CD28 antibodies on the beads or other surface, subpopulations of T cells can be preferentially selected for or against at culture initiation or at other desired time points.
  • In one embodiment, a T cell population can be selected that expresses one or more of IFN-γ, TNFα, IL-17A, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-21, CCL20,GM-CSF, IL-10, IL-13, granzyme B, and perforin, or other appropriate molecules, e.g., other cytokines. Methods for screening for cell expression can be determined, e.g., by the methods described in PCT Publication No.: WO 2013/126712. In an embodiment, the T cell population expresses cytokine CCL20, IL-17a, IL-6, and combinations thereof.
  • For isolation of a desired population of cells by positive or negative selection, the concentration of cells and surface (e.g., particles such as beads) can be varied. In certain aspects, it may be desirable to significantly decrease the volume in which beads and cells are mixed together (e.g., increase the concentration of cells), to ensure maximum contact of cells and beads. For example, in one aspect, a concentration of 10 billion cells/ml, 9 billion/ml, 8 billion/ml, 7 billion/ml, 6 billion/ml, or 5 billion/ml is used. In one aspect, a concentration of 1 billion cells/ml is used. In yet one aspect, a concentration of cells from 75, 80, 85, 90, 95, or 100 million cells/ml is used. In further aspects, concentrations of 125 or 150 million cells/ml can be used.
  • Using high concentrations can result in increased cell yield, cell activation, and cell expansion. Further, use of high cell concentrations allows more efficient capture of cells that may weakly express target antigens of interest, such as CD28-negative T cells, or from samples where there are many tumor cells present (e.g., leukemic blood, tumor tissue, etc.). Such populations of cells may have therapeutic value and would be desirable to obtain. For example, using high concentration of cells allows more efficient selection of CD8+ T cells that normally have weaker CD28 expression.
  • In a related aspect, it may be desirable to use lower concentrations of cells. By significantly diluting the mixture of T cells and surface (e.g., particles such as beads), interactions between the particles and cells is minimized. This selects for cells that express high amounts of desired antigens to be bound to the particles. For example, CD4+ T cells express higher levels of CD28 and are more efficiently captured than CD8+ T cells in dilute concentrations. In one aspect, the concentration of cells used is 5×106/ml. In other aspects, the concentration used can be from about 1×105/ml to 1×106/ml, and any integer value in between.
  • In other aspects, the cells may be incubated on a rotator for varying lengths of time at varying speeds at either 2-10° C. or at room temperature.
  • T cells for stimulation can also be frozen after a washing step. Wishing not to be bound by theory, the freeze and subsequent thaw step provides a more uniform product by removing granulocytes and to some extent monocytes in the cell population. After the washing step that removes plasma and platelets, the cells may be suspended in a freezing solution. While many freezing solutions and parameters are known in the art and will be useful in this context, one method involves using PBS containing 20% DMSO and 8% human serum albumin, or culture media containing 10 % Dextran 40 and 5% Dextrose, 20% Human Serum Albumin and 7.5% DMSO, or 31.25% Plasmalyte-A, 31.25% Dextrose 5%, 0.45% NaCl, 10 % Dextran 40 and 5% Dextrose, 20% Human Serum Albumin, and 7.5% DMSO or other suitable cell freezing media containing for example, Hespan and PlasmaLyte A, the cells then are frozen to −80° C. at a rate of 1° per minute and stored in the vapor phase of a liquid nitrogen storage tank. Other methods of controlled freezing may be used as well as uncontrolled freezing immediately at −20° C. or in liquid nitrogen.
  • In certain aspects, cryopreserved cells are thawed and washed as described herein and allowed to rest for one hour at room temperature prior to activation using the methods of the present invention.
  • Also contemplated in the context of the invention is the collection of blood samples or apheresis product from a subject at a time period prior to when the expanded cells as described herein might be needed. As such, the source of the cells to be expanded can be collected at any time point necessary, and desired cells, such as T cells, isolated and frozen for later use in immune effector cell therapy for any number of diseases or conditions that would benefit from immune effector cell therapy, such as those described herein. In one aspect a blood sample or an apheresis is taken from a generally healthy subject. In certain aspects, a blood sample or an apheresis is taken from a generally healthy subject who is at risk of developing a disease, but who has not yet developed a disease, and the cells of interest are isolated and frozen for later use. In certain aspects, the T cells may be expanded, frozen, and used at a later time. In certain aspects, samples are collected from a patient shortly after diagnosis of a particular disease as described herein but prior to any treatments. In a further aspect, the cells are isolated from a blood sample or an apheresis from a subject prior to any number of relevant treatment modalities, including but not limited to treatment with agents such as natalizumab, efalizumab, antiviral agents, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies, cytoxan, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, and irradiation.
  • In a further aspect of the present invention, T cells are obtained from a patient directly following treatment that leaves the subject with functional T cells. In this regard, it has been observed that following certain cancer treatments, in particular treatments with drugs that damage the immune system, shortly after treatment during the period when patients would normally be recovering from the treatment, the quality of T cells obtained may be optimal or improved for their ability to expand ex vivo. Likewise, following ex vivo manipulation using the methods described herein, these cells may be in a preferred state for enhanced engraftment and in vivo expansion. Thus, it is contemplated within the context of the present invention to collect blood cells, including T cells, dendritic cells, or other cells of the hematopoietic lineage, during this recovery phase. Further, in certain aspects, mobilization (for example, mobilization with GM-CSF) and conditioning regimens can be used to create a condition in a subject wherein repopulation, recirculation, regeneration, and/or expansion of particular cell types is favored, especially during a defined window of time following therapy. Illustrative cell types include T cells, B cells, dendritic cells, and other cells of the immune system.
  • In one embodiment, the immune effector cells expressing a CAR molecule, e.g., a CAR molecule described herein, are obtained from a subject that has received a low, immune enhancing dose of an mTOR inhibitor. In an embodiment, the population of immune effector cells, e.g., T cells, to be engineered to express a CAR, are harvested after a sufficient time, or after sufficient dosing of the low, immune enhancing, dose of an mTOR inhibitor, such that the level of PD1 negative immune effector cells, e.g., T cells, or the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells, in the subject or harvested from the subject has been, at least transiently, increased.
  • In other embodiments, population of immune effector cells, e.g., T cells, which have, or will be engineered to express a CAR, can be treated ex vivo by contact with an amount of an mTOR inhibitor that increases the number of PD1 negative immune effector cells, e.g., T cells or increases the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells.
  • In one embodiment, a T cell population is diacylglycerol kinase (DGK)-deficient. DGK-deficient cells include cells that do not express DGK RNA or protein, or have reduced or inhibited DGK activity. DGK-deficient cells can be generated by genetic approaches, e.g., administering RNA-interfering agents, e.g., siRNA, shRNA, miRNA, to reduce or prevent DGK expression. Alternatively, DGK-deficient cells can be generated by treatment with DGK inhibitors described herein.
  • In one embodiment, a T cell population is Ikaros-deficient. Ikaros-deficient cells include cells that do not express Ikaros RNA or protein, or have reduced or inhibited Ikaros activity, Ikaros-deficient cells can be generated by genetic approaches, e.g., administering RNA-interfering agents, e.g., siRNA, shRNA, miRNA, to reduce or prevent Ikaros expression. Alternatively, Ikaros-deficient cells can be generated by treatment with Ikaros inhibitors, e.g., lenalidomide.
  • In embodiments, a T cell population is DGK-deficient and Ikaros-deficient, e.g., does not express DGK and Ikaros, or has reduced or inhibited DGK and Ikaros activity. Such DGK and Ikaros-deficient cells can be generated by any of the methods described herein.
  • Allogeneic CAR
  • In embodiments described herein, the immune effector cell can be an allogeneic immune effector cell, e.g., T cell. For example, the cell can be an allogeneic T cell, e.g., an allogeneic T cell lacking expression of a functional T cell receptor (TCR) and/or human leukocyte antigen (HLA), e.g., HLA class I and/or HLA class II.
  • A T cell lacking a functional TCR can be, e.g., engineered such that it does not express any functional TCR on its surface, engineered such that it does not express one or more subunits that comprise a functional TCR (e.g., engineered such that it does not express (or exhibits reduced expression) of TCR alpha, TCR beta, TCR gamma, TCR delta, TCR epsilon, and/or TCR zeta) or engineered such that it produces very little functional TCR on its surface. Alternatively, the T cell can express a substantially impaired TCR, e.g., by expression of mutated or truncated forms of one or more of the subunits of the TCR. The term “substantially impaired TCR” means that this TCR will not elicit an adverse immune reaction in a host.
  • A T cell described herein can be, e.g., engineered such that it does not express a functional HLA on its surface. For example, a T cell described herein, can be engineered such that cell surface expression HLA, e.g., HLA class 1 and/or HLA class II, is downregulated. In some embodiments, downregulation of HLA may be accomplished by reducing or eliminating expression of beta-2 microglobulin (B2M).
  • In some embodiments, the T cell can lack a functional TCR and a functional HLA, e.g., HLA class I and/or HLA class II.
  • Modified T cells that lack expression of a functional TCR and/or HLA can be obtained by any suitable means, including a knock out or knock down of one or more subunit of TCR or HLA. For example, the T cell can include a knock down of TCR and/or HLA using siRNA, shRNA, clustered regularly interspaced short palindromic repeats (CRISPR) transcription-activator like effector nuclease (TALEN), or zinc finger endonuclease (ZFN).
  • In some embodiments, the allogeneic cell can be a cell which does not expresses or expresses at low levels an inhibitory molecule, e.g. by any method described herein. For example, the cell can be a cell that does not express or expresses at low levels an inhibitory molecule, e.g., that can decrease the ability of a CAR-expressing cell to mount an immune effector response. Examples of inhibitory molecules include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR (e.g., TGFRbeta). Inhibition of an inhibitory molecule, e.g., by inhibition at the DNA, RNA or protein level, can optimize a CAR-expressing cell performance. In embodiments, an inhibitory nucleic acid, e.g., an inhibitory nucleic acid, e.g., a dsRNA, e.g., an siRNA or shRNA, a clustered regularly interspaced short palindromic repeats (CRISPR), a transcription-activator like effector nuclease (TALEN), or a zinc finger endonuclease (ZFN), e.g., as described herein, can be used.
  • siRNA and shRNA to Inhibit TCR or HLA
  • In some embodiments, TCR expression and/or HLA expression can be inhibited using siRNA or shRNA that targets a nucleic acid encoding a TCR and/or HLA, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, WIC class I, WIC class II, GAL9, adenosine, and TGFR beta), in a cell, e.g., T cell.
  • Expression systems for siRNA and shRNAs, and exemplary shRNAs, are described, e.g., in paragraphs 649 and 650 of International Application WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.
  • CRISPR to inhibit TCR or HLA
  • “CRISPR” or “CRISPR to TCR and/or HLA” or “CRISPR to inhibit TCR and/or HLA” as used herein refers to a set of clustered regularly interspaced short palindromic repeats, or a system comprising such a set of repeats. “Cas”, as used herein, refers to a CRISPR-associated protein. A “CRISPR/Cas” system refers to a system derived from CRISPR and Cas which can be used to silence or mutate a TCR and/or HLA gene, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MEW class II, GAL9, adenosine, and TGFR beta), in a cell, e.g., T cell.
  • The CRISPR/Cas system, and uses thereof, are described, e.g., in paragraphs 651-658 of International Application WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.
  • TALEN to Inhibit TCR and/or HLA
  • “TALEN” or “TALEN to HLA and/or TCR” or “TALEN to inhibit HLA and/or TCR” refers to a transcription activator-like effector nuclease, an artificial nuclease which can be used to edit the HLA and/or TCR gene, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, WIC class II, GAL9, adenosine, and TGFR beta), in a cell, e.g., T cell.
  • TALENs, and uses thereof, are described, e.g., in paragraphs 659-665 of International Application WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.
  • Zinc Finger Nuclease to Inhibit HLA and/or TCR
  • “ZFN” or “Zinc Finger Nuclease” or “ZFN to HLA and/or TCR” or “ZFN to inhibit HLA and/or TCR” refer to a zinc finger nuclease, an artificial nuclease which can be used to edit the HLA and/or TCR gene, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, WIC class I, MHC class II, GAL9, adenosine, and TGFR beta), in a cell, e.g., T cell.
  • ZFNs, and uses thereof, are described, e.g., in paragraphs 666-671 of International Application WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.
  • Telomerase Expression
  • While not wishing to be bound by any particular theory, in some embodiments, a therapeutic T cell has short term persistence in a patient, due to shortened telomeres in the T cell; accordingly, transfection with a telomerase gene can lengthen the telomeres of the T cell and improve persistence of the T cell in the patient. See Carl June, “Adoptive T cell therapy for cancer in the clinic”, Journal of Clinical Investigation, 117:1466-1476 (2007). Thus, in an embodiment, an immune effector cell, e.g., a T cell, ectopically expresses a telomerase subunit, e.g., the catalytic subunit of telomerase, e.g., TERT, e.g., hTERT. In some aspects, this disclosure provides a method of producing a CAR-expressing cell, comprising contacting a cell with a nucleic acid encoding a telomerase subunit, e.g., the catalytic subunit of telomerase, e.g., TERT, e.g., hTERT. The cell may be contacted with the nucleic acid before, simultaneous with, or after being contacted with a construct encoding a CAR.
  • In one aspect, the disclosure features a method of making a population of immune effector cells (e.g., T cells, NK cells). In an embodiment, the method comprises: providing a population of immune effector cells (e.g., T cells or NK cells), contacting the population of immune effector cells with a nucleic acid encoding a CAR; and contacting the population of immune effector cells with a nucleic acid encoding a telomerase subunit, e.g., hTERT, under conditions that allow for CAR and telomerase expression.
  • In an embodiment, the nucleic acid encoding the telomerase subunit is DNA. In an embodiment, the nucleic acid encoding the telomerase subunit comprises a promoter capable of driving expression of the telomerase subunit.
  • In an embodiment, hTERT has the amino acid sequence of GenBank Protein ID AAC51724.1 (Meyerson et al., “hEST2, the Putative Human Telomerase Catalytic Subunit Gene, Is Up-Regulated in Tumor Cells and during Immortalization” Cell Volume 90, Issue 4, 22 Aug. 1997, Pages 785-795) as set out in SEQ ID NO: 82 herein.
  • In an embodiment, the hTERT has a sequence at least 80%, 85%, 90%, 95%, 96{circumflex over ( )}, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 82. In an embodiment, the hTERT has a sequence of SEQ ID NO: 82. In an embodiment, the hTERT comprises a deletion (e.g., of no more than 5, 10, 15, 20, or 30 amino acids) at the N-terminus, the C-terminus, or both. In an embodiment, the hTERT comprises a transgenic amino acid sequence (e.g., of no more than 5, 10, 15, 20, or 30 amino acids) at the N-terminus, the C-terminus, or both.
  • In an embodiment, the hTERT is encoded by the nucleic acid sequence of GenBank Accession No. AF018167 (Meyerson et al., “hEST2, the Putative Human Telomerase Catalytic Subunit Gene, Is Up-Regulated in Tumor Cells and during Immortalization” Cell Volume 90, Issue 4, 22 Aug. 1997, Pages 785-795) as set out in SEQ ID NO: 83 herein.
  • In an embodiment, the hTERT is encoded by a nucleic acid having a sequence at least 80%, 85%, 90%, 95%, 96, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 83. In an embodiment, the hTERT is encoded by a nucleic acid of SEQ ID NO: 83.
  • Activation and Expansion of Immune Effector Cells (e.g., T Cells)
  • Immune effector cells such as T cells may be activated and expanded generally using methods as described, for example, in U.S. Pat. Nos. 6,352,694; 6,534,055; 6,905,680; 6,692,964; 5,858,358; 6,887,466; 6,905,681; 7,144,575; 7,067,318; 7,172,869; 7,232,566; 7,175,843; 5,883,223; 6,905,874; 6,797,514; 6,867,041; and U.S. Patent Application Publication No. 20060121005. In some embodiments, immune effector cells are subjected to an assay as described herein (e.g., one or more biomarkers are assayed) before, during, or after activation, or before, during, or after expansion.
  • Generally, a population of immune effector cells may be expanded by contact with a surface having attached thereto an agent that stimulates a CD3/TCR complex associated signal and a ligand that stimulates a costimulatory molecule on the surface of the T cells. In particular, T cell populations may be stimulated as described herein, such as by contact with an anti-CD3 antibody, or antigen-binding fragment thereof, or an anti-CD2 antibody immobilized on a surface, or by contact with a protein kinase C activator (e.g., bryostatin) in conjunction with a calcium ionophore. For co-stimulation of an accessory molecule on the surface of the T cells, a ligand that binds the accessory molecule is used. For example, a population of T cells can be contacted with an anti-CD3 antibody and an anti-CD28 antibody, under conditions appropriate for stimulating proliferation of the T cells. To stimulate proliferation of either CD4+ T cells or CD8+ T cells, an anti-CD3 antibody and an anti-CD28 antibody can be used. Examples of an anti-CD28 antibody include 9.3, B-T3, XR-CD28 (Diaclone, Besancon, France) can be used as can other methods commonly known in the art (Berg et al., Transplant Proc. 30(8):3975-3977, 1998; Haanen et al., J. Exp. Med. 190(9):13191328, 1999; Garland et al., J. Immunol Meth. 227(1-2):53-63, 1999).
  • In certain aspects, the primary stimulatory signal and the costimulatory signal for the T cell may be provided by different protocols. For example, the agents providing each signal may be in solution or coupled to a surface. When coupled to a surface, the agents may be coupled to the same surface (i.e., in “cis” formation) or to separate surfaces (i.e., in “trans” formation). Alternatively, one agent may be coupled to a surface and the other agent in solution. In one aspect, the agent providing the costimulatory signal is bound to a cell surface and the agent providing the primary activation signal is in solution or coupled to a surface. In certain aspects, both agents can be in solution. In one aspect, the agents may be in soluble form, and then cross-linked to a surface, such as a cell expressing Fc receptors or an antibody or other binding agent which will bind to the agents. In this regard, see for example, U.S. Patent Application Publication Nos. 20040101519 and 20060034810 for artificial antigen presenting cells (aAPCs) that are contemplated for use in activating and expanding T cells in the present invention.
  • In one aspect, the two agents are immobilized on beads, either on the same bead, i.e., “cis,” or to separate beads, i.e., “trans.” By way of example, the agent providing the primary activation signal is an anti-CD3 antibody or an antigen-binding fragment thereof and the agent providing the costimulatory signal is an anti-CD28 antibody or antigen-binding fragment thereof; and both agents are co-immobilized to the same bead in equivalent molecular amounts. In one aspect, a 1:1 ratio of each antibody bound to the beads for CD4+ T cell expansion and T cell growth is used. In certain aspects of the present invention, a ratio of anti CD3:CD28 antibodies bound to the beads is used such that an increase in T cell expansion is observed as compared to the expansion observed using a ratio of 1:1. In one particular aspect an increase of from about 1 to about 3 fold is observed as compared to the expansion observed using a ratio of 1:1. In one aspect, the ratio of CD3:CD28 antibody bound to the beads ranges from 100:1 to 1:100 and all integer values there between. In one aspect, more anti-CD28 antibody is bound to the particles than anti-CD3 antibody, i.e., the ratio of CD3:CD28 is less than one. In certain aspects, the ratio of anti CD28 antibody to anti CD3 antibody bound to the beads is greater than 2:1. In one particular aspect, a 1:100 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:75 CD3:CD28 ratio of antibody bound to beads is used. In a further aspect, a 1:50 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:30 CD3:CD28 ratio of antibody bound to beads is used. In one preferred aspect, a 1:10 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:3 CD3:CD28 ratio of antibody bound to the beads is used. In yet one aspect, a 3:1 CD3:CD28 ratio of antibody bound to the beads is used.
  • Ratios of particles to cells from 1:500 to 500:1 and any integer values in between may be used to stimulate T cells or other target cells. As those of ordinary skill in the art can readily appreciate, the ratio of particles to cells may depend on particle size relative to the target cell. For example, small sized beads could only bind a few cells, while larger beads could bind many. In certain aspects the ratio of cells to particles ranges from 1:100 to 100:1 and any integer values in-between and in further aspects the ratio comprises 1:9 to 9:1 and any integer values in between, can also be used to stimulate T cells. The ratio of anti-CD3- and anti-CD28-coupled particles to T cells that result in T cell stimulation can vary as noted above, however certain suitable values include 1:100, 1:50, 1:40, 1:30, 1:20, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, and 15:1 with one suitable ratio being at least 1:1 particles per T cell. In one aspect, a ratio of particles to cells of 1:1 or less is used. In one particular aspect, a suitable particle: cell ratio is 1:5. In further aspects, the ratio of particles to cells can be varied depending on the day of stimulation. For example, in one aspect, the ratio of particles to cells is from 1:1 to 10:1 on the first day and additional particles are added to the cells every day or every other day thereafter for up to 10 days, at final ratios of from 1:1 to 1:10 (based on cell counts on the day of addition). In one particular aspect, the ratio of particles to cells is 1:1 on the first day of stimulation and adjusted to 1:5 on the third and fifth days of stimulation. In one aspect, particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:5 on the third and fifth days of stimulation. In one aspect, the ratio of particles to cells is 2:1 on the first day of stimulation and adjusted to 1:10 on the third and fifth days of stimulation. In one aspect, particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:10 on the third and fifth days of stimulation. One of skill in the art will appreciate that a variety of other ratios may be suitable for use in the present invention. In particular, ratios will vary depending on particle size and on cell size and type. In one aspect, the most typical ratios for use are in the neighborhood of 1:1, 2:1 and 3:1 on the first day.
  • In further aspects, the cells, such as T cells, are combined with agent-coated beads, the beads and the cells are subsequently separated, and then the cells are cultured. In an alternative aspect, prior to culture, the agent-coated beads and cells are not separated but are cultured together. In a further aspect, the beads and cells are first concentrated by application of a force, such as a magnetic force, resulting in increased ligation of cell surface markers, thereby inducing cell stimulation.
  • By way of example, cell surface proteins may be ligated by allowing paramagnetic beads to which anti-CD3 and anti-CD28 are attached (3×28 beads) to contact the T cells. In one aspect the cells (for example, 104 to 109 T cells) and beads (for example, DYNABEADS® M-450 CD3/CD28 T paramagnetic beads at a ratio of 1:1) are combined in a buffer, for example PBS (without divalent cations such as, calcium and magnesium). Again, those of ordinary skill in the art can readily appreciate any cell concentration may be used. For example, the target cell may be very rare in the sample and comprise only 0.01% of the sample or the entire sample (i.e., 100%) may comprise the target cell of interest. Accordingly, any cell number is within the context of the present invention. In certain aspects, it may be desirable to significantly decrease the volume in which particles and cells are mixed together (i.e., increase the concentration of cells), to ensure maximum contact of cells and particles. For example, in one aspect, a concentration of about 10 billion cells/ml, 9 billion/ml, 8 billion/ml, 7 billion/ml, 6 billion/ml, 5 billion/ml, or 2 billion cells/ml is used. In one aspect, greater than 100 million cells/ml is used. In a further aspect, a concentration of cells of 10, 15, 20, 25, 30, 35, 40, 45, or 50 million cells/ml is used. In yet one aspect, a concentration of cells from 75, 80, 85, 90, 95, or 100 million cells/ml is used. In further aspects, concentrations of 125 or 150 million cells/ml can be used. Using high concentrations can result in increased cell yield, cell activation, and cell expansion. Further, use of high cell concentrations allows more efficient capture of cells that may weakly express target antigens of interest, such as CD28-negative T cells. Such populations of cells may have therapeutic value and would be desirable to obtain in certain aspects. For example, using high concentration of cells allows more efficient selection of CD8+ T cells that normally have weaker CD28 expression.
  • In one embodiment, cells transduced with a nucleic acid encoding a CAR, e.g., a CAR described herein, are expanded, e.g., by a method described herein. In one embodiment, the cells are expanded in culture for a period of several hours (e.g., about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 18, 21 hours) to about 14 days (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days). In one embodiment, the cells are expanded for a period of 4 to 9 days. In one embodiment, the cells are expanded for a period of 8 days or less, e.g., 7, 6 or 5 days. In one embodiment, the cells, e.g., a CD19 CAR cell described herein, are expanded in culture for 5 days, and the resulting cells are more potent than the same cells expanded in culture for 9 days under the same culture conditions. Potency can be defined, e.g., by various T cell functions, e.g. proliferation, target cell killing, cytokine production, activation, migration, or combinations thereof. In one embodiment, the cells, e.g., a CD19 CAR cell described herein, expanded for 5 days show at least a one, two, three or four fold increase in cells doublings upon antigen stimulation as compared to the same cells expanded in culture for 9 days under the same culture conditions. In one embodiment, the cells, e.g., the cells expressing a CD19 CAR described herein, are expanded in culture for 5 days, and the resulting cells exhibit higher proinflammatory cytokine production, e.g., IFN-γ and/or GM-CSF levels, as compared to the same cells expanded in culture for 9 days under the same culture conditions. In one embodiment, the cells, e.g., a CD19 CAR cell described herein, expanded for 5 days show at least a one, two, three, four, five, tenfold or more increase in pg/ml of proinflammatory cytokine production, e.g., IFN-γ and/or GM-CSF levels, as compared to the same cells expanded in culture for 9 days under the same culture conditions.
  • Several cycles of stimulation may also be desired such that culture time of T cells can be 60 days or more. Conditions appropriate for T cell culture include an appropriate media (e.g., Minimal Essential Media or RPMI Media 1640 or, X-vivo 15, (Lonza)) that may contain factors necessary for proliferation and viability, including serum (e.g., fetal bovine or human serum), interleukin-2 (IL-2), insulin, IFN-γ, IL-4, IL-7, GM-CSF, IL-10, IL-12, IL-15, TGFβ, and TNF-α or any other additives for the growth of cells known to the skilled artisan. Other additives for the growth of cells include, but are not limited to, surfactant, plasmanate, and reducing agents such as N-acetyl-cysteine and 2-mercaptoethanol. Media can include RPMI 1640, AIM-V, DMEM, MEM, α-MEM, F-12, X-Vivo 15, and X-Vivo 20, Optimizer, with added amino acids, sodium pyruvate, and vitamins, either serum-free or supplemented with an appropriate amount of serum (or plasma) or a defined set of hormones, and/or an amount of cytokine(s) sufficient for the growth and expansion of T cells. Antibiotics, e.g., penicillin and streptomycin, are included only in experimental cultures, not in cultures of cells that are to be infused into a subject. The target cells are maintained under conditions necessary to support growth, for example, an appropriate temperature (e.g., 37° C.) and atmosphere (e.g., air plus 5% CO2).
  • In one embodiment, the cells are expanded in an appropriate media (e.g., media described herein) that includes one or more interleukin that result in at least a 200-fold (e.g., 200-fold, 250-fold, 300-fold, 350-fold) increase in cells over a 14 day expansion period, e.g., as measured by a method described herein such as flow cytometry. In one embodiment, the cells are expanded in the presence IL-15 and/or IL-7 (e.g., IL-15 and IL-7).
  • In embodiments, methods described herein, e.g., CAR-expressing cell manufacturing methods, comprise removing T regulatory cells, e.g., CD25+ T cells, from a cell population, e.g., using an anti-CD25 antibody, or fragment thereof, or a CD25-binding ligand, IL-2. Methods of removing T regulatory cells, e.g., CD25+ T cells, from a cell population are described herein. In embodiments, the methods, e.g., manufacturing methods, further comprise contacting a cell population (e.g., a cell population in which T regulatory cells, such as CD25+ T cells, have been depleted; or a cell population that has previously contacted an anti-CD25 antibody, fragment thereof, or CD25-binding ligand) with IL-15 and/or IL-7. For example, the cell population (e.g., that has previously contacted an anti-CD25 antibody, fragment thereof, or CD25-binding ligand) is expanded in the presence of IL-15 and/or IL-7.
  • In some embodiments a CAR-expressing cell described herein is contacted with a composition comprising a interleukin-15 (IL-15) polypeptide, a interleukin-15 receptor alpha (IL-15Ra) polypeptide, or a combination of both a IL-15 polypeptide and a IL-15Ra polypeptide e.g., hetIL-15, during the manufacturing of the CAR-expressing cell, e.g., ex vivo. In embodiments, a CAR-expressing cell described herein is contacted with a composition comprising a IL-15 polypeptide during the manufacturing of the CAR-expressing cell, e.g., ex vivo. In embodiments, a CAR-expressing cell described herein is contacted with a composition comprising a combination of both a IL-15 polypeptide and a IL-15 Ra polypeptide during the manufacturing of the CAR-expressing cell, e.g., ex vivo. In embodiments, a CAR-expressing cell described herein is contacted with a composition comprising hetlL-15 during the manufacturing of the CAR-expressing cell, e.g., ex vivo.
  • In one embodiment the CAR-expressing cell described herein is contacted with a composition comprising hetIL-15 during ex vivo expansion. In an embodiment, the CAR-expressing cell described herein is contacted with a composition comprising an IL-15 polypeptide during ex vivo expansion. In an embodiment, the CAR-expressing cell described herein is contacted with a composition comprising both an IL-15 polypeptide and an IL-15Ra polypeptide during ex vivo expansion. In one embodiment the contacting results in the survival and proliferation of a lymphocyte subpopulation, e.g., CD8+ T cells.
  • In one embodiment, the cells are cultured (e.g., expanded, simulated, and/or transduced) in media comprising serum. The serum may be, e.g., human AB serum (hAB). In some embodiments, the hAB serum is present at about 2%, about 5%, about 2-3%, about 3-4%, about 4-5%, or about 2-5%. As shown in Example 15 herein, 2% and 5% serum are each suitable levels that allow for many fold expansion of T cells. Furthermore, as shown in Smith et al., “Ex vivo expansion of human T cells for adoptive immunotherapy using the novel Xeno-free CTS Immune Cell Serum Replacement” Clinical & Translational Immunology (2015) 4, e31; doi:10.1038/cti.2014.31, medium containing 2% human AB serum is suitable for ex vivo expansion of T cells.
  • T cells that have been exposed to varied stimulation times may exhibit different characteristics. For example, typical blood or apheresed peripheral blood mononuclear cell products have a helper T cell population (TH, CD4+) that is greater than the cytotoxic or suppressor T cell population (TC, CD8+). Ex vivo expansion of T cells by stimulating CD3 and CD28 receptors produces a population of T cells that prior to about days 8-9 consists predominately of TH cells, while after about days 8-9, the population of T cells comprises an increasingly greater population of TC cells. Accordingly, depending on the purpose of treatment, infusing a subject with a T cell population comprising predominately of TH cells may be advantageous. Similarly, if an antigen-specific subset of TC cells has been isolated it may be beneficial to expand this subset to a greater degree.
  • Further, in addition to CD4 and CD8 markers, other phenotypic markers vary significantly, but in large part, reproducibly during the course of the cell expansion process. Thus, such reproducibility enables the ability to tailor an activated T cell product for specific purposes.
  • In some embodiments, cells transduced with a nucleic acid encoding a CAR, e.g., a CAR described herein, can be selected for administration based upon, e.g., protein expression levels of one or more of CCL20, GM-CSF, IFNγ, IL-10, IL-13, IL-17a, IL-2, IL-21, IL-4, IL-5, IL-6, IL-9, TNFα and/or combinations thereof. In some embodiments, cells transduced with a nucleic acid encoding a CAR, e.g., a CAR described herein, can be selected for administration based upon, e.g., protein expression levels of CCL20, IL-17a, IL-6 and combinations thereof.
  • Once a CAR described herein is constructed, various assays can be used to evaluate the activity of the molecule, such as but not limited to, the ability to expand T cells following antigen stimulation, sustain T cell expansion in the absence of re-stimulation, and anti-cancer activities in appropriate in vitro and animal models. Assays to evaluate the effects of a CAR are described in further detail below.
  • Western blot analysis of CAR expression in primary T cells can be used to detect the presence of monomers and dimers, e.g., as described in paragraph 695 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • In vitro expansion of CAR+ T cells following antigen stimulation can be measured by flow cytometry. For example, a mixture of CD4+ and CD8+ T cells are stimulated with αCD3/αCD28 aAPCs followed by transduction with lentiviral vectors expressing GFP under the control of the promoters to be analyzed. Exemplary promoters include the CMV IE gene, EF-1α, ubiquitin C, or phosphoglycerokinase (PGK) promoters. GFP fluorescence is evaluated on day 6 of culture in the CD4+ and/or CD8+ T cell subsets by flow cytometry. See, e.g., Milone et al., MOLECULAR THERAPY 17(8): 1453-1464 (2009). Alternatively, a mixture of CD4+ and CD8+ T cells are stimulated with αCD3/αCD28 coated magnetic beads on day 0, and transduced with CAR on day 1 using a bicistronic lentiviral vector expressing CAR along with eGFP using a 2A ribosomal skipping sequence. Cultures are re-stimulated with either a cancer associate antigen as described herein +K562 cells (K562-a cancer associate antigen as described herein), wild-type K562 cells (K562 wild type) or K562 cells expressing hCD32 and 4-1BBL in the presence of antiCD3 and anti-CD28 antibody (K562-BBL-3/28) following washing. Exogenous IL-2 is added to the cultures every other day at 100 IU/ml. GFP+ T cells are enumerated by flow cytometry using bead-based counting. See, e.g., Milone et al., MOLECULAR THERAPY 17(8): 1453-1464 (2009).
  • Sustained CAR+ T cell expansion in the absence of re-stimulation can also be measured. See, e.g., Milone et al., MOLECULAR THERAPY 17(8): 1453-1464 (2009). Briefly, mean T cell volume (fl) is measured on day 8 of culture using a Coulter Multisizer III particle counter, a Nexcelom Cellometer Vision or Millipore Scepter, following stimulation with αCD3/αCD28 coated magnetic beads on day 0, and transduction with the indicated CAR on day 1.
  • Animal models can also be used to measure a CAR-expressing cell (e.g., T cell, NK cell) activity, e.g., as described in paragraph 698 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • Dose dependent CAR treatment response can be evaluated, e.g., as described in paragraph 699 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • Assessment of cell proliferation and cytokine production has been previously described, e.g., as described in paragraph 700 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • In another embodiment, potency of a cell (e.g., T cell, NK cell) population (e.g. a CAR-expressing cell) product, e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) cell product, e.g., CTL019 cells) is assessed using a Luminex® panel of cytokines to determine cytokine expression levels. Cell (e.g., T cell, NK cell) populations (e.g, a manufactured CAR-expressing cell) cell product, e.g., a CD19 CAR-expressing cell product, e.g., CTL019 cells) are activated in vitro by CD19-expressing K562 (K562-19) cells, which mimic CD19-expressing B cells in CLL. Following cell (e.g., T cell, NK cell) activation, cytokine expression profiles are measured in the co-cultured cell media and potency of activated cells (e.g., a CAR-expressing cell product, e.g., a CD19 CAR-expressing cell product, e.g., CTL019 cells) is correlated with expression of different cytokines including, but not limited to CCL-20/MIP-3a, GM-CSF, IFNγ, IL-10, IL-13, IL-17a, IL-2, IL-21, IL-4, IL-5, IL-6, IL-9, TNFα and/or combinations thereof.
  • In an embodiment, cytokine expression levels are informative with regards to the potency of a cell (e.g., T cell, NK cell) population (e.g., to kill tumor cells). In an embodiment, cytokine expression levels described herein are used to improve a cell (e.g., T cell, NK cell) population (e.g., a CAR-expressing cell product, e.g., a CD 19 CAR-expressing cell product, e.g., CTL019 cells) prior to infusion in patients. In an embodiment, cytokine expression levels described herein provide an endpoint during optimization of the manufacturing process.
  • Cytotoxicity can be assessed by a standard 51Cr-release assay, e.g., as described in paragraph 701 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • Imaging technologies can be used to evaluate specific trafficking and proliferation of CARs in tumor-bearing animal models, e.g., as described in paragraph 702 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • Other assays, including those described in the Example section herein as well as those that are known in the art can also be used to evaluate the CARs described herein.
  • Alternatively, or in combination to the methods disclosed herein, methods and compositions for one or more of: detection and/or quantification of CAR-expressing cells (e.g., in vitro or in vivo (e.g., clinical monitoring)); immune cell expansion and/or activation; and/or CAR-specific selection, that involve the use of a CAR ligand, are disclosed. In one exemplary embodiment, the CAR ligand is an antibody that binds to the CAR molecule, e.g., binds to the extracellular antigen binding domain of CAR (e.g., an antibody that binds to the antigen binding domain, e.g., an anti-idiotypic antibody; or an antibody that binds to a constant region of the extracellular binding domain). In other embodiments, the CAR ligand is a CAR antigen molecule (e.g., a CAR antigen molecule as described herein).
  • In one aspect, a method for detecting and/or quantifying CAR-expressing cells is disclosed. For example, the CAR ligand can be used to detect and/or quantify CAR-expressing cells in vitro or in vivo (e.g., clinical monitoring of CAR-expressing cells in a patient, or dosing a patient). The method includes:
  • providing the CAR ligand (optionally, a labelled CAR ligand, e.g., a CAR ligand that includes a tag, a bead, a radioactive or fluorescent label);
  • acquiring the CAR-expressing cell (e.g., acquiring a sample containing CAR-expressing cells, such as a manufacturing sample or a clinical sample);
  • contacting the CAR-expressing cell with the CAR ligand under conditions where binding occurs, thereby detecting the level (e.g., amount) of the CAR-expressing cells present. Binding of the CAR-expressing cell with the CAR ligand can be detected using standard techniques such as FACS, ELISA and the like.
  • In another aspect, a method of expanding and/or activating cells (e.g., immune effector cells) is disclosed. The method includes:
  • providing a CAR-expressing cell (e.g., a first CAR-expressing cell or a transiently expressing CAR cell);
  • contacting said CAR-expressing cell with a CAR ligand, e.g., a CAR ligand as described herein), under conditions where immune cell expansion and/or proliferation occurs, thereby producing the activated and/or expanded cell population.
  • In certain embodiments, the CAR ligand is present on (e.g., is immobilized or attached to a substrate, e.g., a non-naturally occurring substrate). In some embodiments, the substrate is a non-cellular substrate. The non-cellular substrate can be a solid support chosen from, e.g., a plate (e.g., a microtiter plate), a membrane (e.g., a nitrocellulose membrane), a matrix, a chip or a bead. In embodiments, the CAR ligand is present in the substrate (e.g., on the substrate surface). The CAR ligand can be immobilized, attached, or associated covalently or non-covalently (e.g., cross-linked) to the substrate. In one embodiment, the CAR ligand is attached (e.g., covalently attached) to a bead. In the aforesaid embodiments, the immune cell population can be expanded in vitro or ex vivo. The method can further include culturing the population of immune cells in the presence of the ligand of the CAR molecule, e.g., using any of the methods described herein.
  • In other embodiments, the method of expanding and/or activating the cells further comprises addition of a second stimulatory molecule, e.g., CD28. For example, the CAR ligand and the second stimulatory molecule can be immobilized to a substrate, e.g., one or more beads, thereby providing increased cell expansion and/or activation.
  • In yet another aspect, a method for selecting or enriching for a CAR expressing cell is provided. The method includes contacting the CAR expressing cell with a CAR ligand as described herein; and selecting the cell on the basis of binding of the CAR ligand.
  • In yet other embodiments, a method for depleting, reducing and/or killing a CAR expressing cell is provided. The method includes contacting the CAR expressing cell with a CAR ligand as described herein; and targeting the cell on the basis of binding of the CAR ligand, thereby reducing the number, and/or killing, the CAR-expressing cell. In one embodiment, the CAR ligand is coupled to a toxic agent (e.g., a toxin or a cell ablative drug). In another embodiment, the anti-idiotypic antibody can cause effector cell activity, e.g., ADCC or ADC activities.
  • Exemplary anti-CAR antibodies that can be used in the methods disclosed herein are described, e.g., in WO 2014/190273 and by Jena et al., “Chimeric Antigen Receptor (CAR)-Specific Monoclonal Antibody to Detect CD19-Specific T cells in Clinical Trials”, PLOS March 2013 8:3 e57838, the contents of which are incorporated by reference.
  • In some aspects and embodiments, the compositions and methods herein are optimized for a specific subset of T cells, e.g., as described in US Serial No. PCT/US2015/043219 filed Jul. 31, 2015, the contents of which are incorporated herein by reference in their entirety. In some embodiments, the optimized subsets of T cells display an enhanced persistence compared to a control T cell, e.g., a T cell of a different type (e.g., CD8+ or CD4+) expressing the same construct.
  • In some embodiments, a CD4+ T cell comprises a CAR described herein, which CAR comprises an intracellular signaling domain suitable for (e.g., optimized for, e.g., leading to enhanced persistence in) a CD4+ T cell, e.g., an ICOS domain. In some embodiments, a CD8+ T cell comprises a CAR described herein, which CAR comprises an intracellular signaling domain suitable for (e.g., optimized for, e.g., leading to enhanced persistence of) a CD8+ T cell, e.g., a 4-1BB domain, a CD28 domain, or another costimulatory domain other than an ICOS domain. In some embodiments, the CAR described herein comprises an antigen binding domain described herein, e.g., a CAR comprising an antigen binding domain.
  • In an aspect, described herein is a method of treating a subject, e.g., a subject having cancer. The method includes administering to said subject, an effective amount of:
      • 1) a CD4+ T cell comprising a CAR (the CARCD4+)
  • comprising:
  • an antigen binding domain, e.g., an antigen binding domain described herein;
  • a transmembrane domain; and
  • an intracellular signaling domain, e.g., a first costimulatory domain, e.g., an ICOS domain; and
      • 2) a CD8+ T cell comprising a CAR (the CARCD8+) comprising:
  • an antigen binding domain, e.g., an antigen binding domain described herein;
  • a transmembrane domain; and
  • an intracellular signaling domain, e.g., a second costimulatory domain, e.g., a 4-1BB domain, a CD28 domain, or another costimulatory domain other than an ICOS domain;
  • wherein the CARCD4+ and the CARCD8+ differ from one another.
  • Optionally, the method further includes administering:
      • 3) a second CD8+ T cell comprising a CAR (the second CARCD8+) comprising:
  • an antigen binding domain, e.g., an antigen binding domain described herein;
  • a transmembrane domain; and
  • an intracellular signaling domain, wherein the second CARCD8+ comprises an intracellular signaling domain, e.g., a costimulatory signaling domain, not present on the CARCD8+, and, optionally, does not comprise an ICOS signaling domain.
  • RNA Transfection
  • Disclosed herein are methods for producing an in vitro transcribed RNA CAR. RNA CAR and methods of using the same are described, e.g., in paragraphs 553-570 of in International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • In one embodiment, the in vitro transcribed RNA CAR can be introduced to a cell as a form of transient transfection. The RNA may have a 3′ UTR, a 5′ UTR, or both. The 5′ UTR may contain a Kozak sequence. The RNA may comprise an IRES. The RNA may comprise a 5′ cap. The RNA may comprise a polyA sequence. RNA can be produced using a DNA template that comprises a promoter, e.g., a T7, T7, or SP6 promoter. RNA can be introduced into target cells using any of a number of different methods, for instance, commercially available methods which include, but are not limited to, electroporation, the Gene Pulser II, Multiporator, cationic liposome mediated transfection using lipofection, polymer encapsulation, peptide mediated transfection, or biolistic particle delivery systems such as “gene guns”.
  • Non-Viral Delivery Methods
  • In some aspects, non-viral methods can be used to deliver a nucleic acid encoding a CAR described herein into a cell or tissue or a subject. Suitable non-viral delivery methods include transposons (e.g., Sleeping Beauty, piggyBac, and pT2-based transposons). Exemplary non-viral delivery methods and methods of using the same are described, e.g., in paragraphs 571-579 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • Methods of Manufacture/Production
  • In one aspect, methods of manufacturing a CAR-expressing cell according to the invention are disclosed herein (e.g., in “Source of Cells” and “Activation and Expansion of Cells”).
  • In an embodiment, a method of manufacturing a CAR-expressing cell is provided. The method comprises:
      • providing a preparation of a CAR-expressing cell (e.g., a plurality of CAR-expressing immune effector cells, such as a T cells, or an NK cells) (e.g., a CD19 CAR-expressing cell as described herein, such as, e.g., CTL019);
      • acquiring a value for the level of (e.g., determining the level of expression of) one or more genes listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 to obtain a gene expression pattern for the sample;
      • (optionally) comparing the obtained gene expression pattern to that of a historical record of gene expression;
  • determining a difference between the obtained and historical gene expression; and
      • recording the determined difference in a quality control record.
  • In an embodiment, provided methods comprise steps of providing a CAR-expressing cell (e.g., T cell, NK cell) preparation (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019);
      • determining the levels of expression of one or more genes listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1 to obtain a gene expression pattern (e.g., a gene signature) for the sample;
      • correlating the gene signature with patient response to a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g. a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019);
      • and optimizing the CAR-expressing cell (e.g., T cell, NK cell) preparation based on the correlation of the gene signature and patient response prior to infusion into patients.
  • In an embodiment, provided methods comprise acquiring a value for the level (e.g., determining the expression level) of a cytokine, e.g., one or more cytokines listed in Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), and Table 17, Table 18, Table 20, secreted by CAR-expressing cells (e.g., T cell, NK cell) in response to antigen recognition. In an embodiment, provided methods comprise determining the expression levels of one or more cytokines CCL20/MIP3a, IL-17a, IL-6 and/or combinations thereof, secreted by CAR-expressing cells (e.g., T cell, NK cell) in response to antigen recognition. In an embodiment, provided methods further comprise integration of cytokines secreted by CAR-expressing cells (e.g., T cells, NK cells), e.g., one or more cytokines listed in Table 14, Table 15 and Table 16, in a potency assay. In an embodiment, provided methods further comprise integration of cytokines CCL20/MIP3a, IL-17a, IL-6 and/or combinations thereof, in a potency assay.
  • In an embodiment, provided methods comprise integration of cytokines secreted by CAR-expressing cells (e.g., T cells, NK cells), e.g., one or more cytokines listed in Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), and Table 17 in a potency assay, and determining whether a CAR-expressing cell (e.g., T cell, NK cell) preparation (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019) may have a clinical effect. In an embodiment, CCL20/MIP3a, IL-17a, IL-6 and/or combinations thereof are used in a potency assay to determine whether a CAR-expressing cell (e.g., T cell, NK cell) preparation (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019) may have a clinical effect. In an embodiment, provided methods further comprise adjusting the CAR-expressing cell (e.g., T cell, NK cell) infusion dose to achieve clinical efficacy.
  • In an embodiment, provided methods comprise a step of providing a blood sample, e.g., a T cell sample, from a subject having cancer.
  • In an embodiment, provided methods further comprise a step of comparing the obtained gene expression pattern difference with that of a reference sample.
  • In an embodiment, a reference sample is a CAR-expressing cell (e.g., T cell, NK cell) preparation (e.g., a CD19 CAR-expressing cell as described herein, such as, e.g., CTL019) from a different batch of cells producing the therapeutic CAR-expressing cell preparation.
  • In an embodiment, a reference sample is a healthy donor sample with a manufactured CAR-expressing cell (e.g., T cell, NK cell) product (e.g., a CD19 CAR-expressing cell as described herein, such as, e.g., CTL019). In an embodiment, a reference sample is a healthy donor sample with a manufactured CD19 CAR-expressing cell product, such as, e.g., CTL019 product.
  • In an embodiment, provided methods further comprise a step of recording the result of the comparing in a quality control record for the therapeutic CAR-expressing cell (e.g., T cell, NK cell) preparation.
  • In an embodiment, the determined difference is compared with a historical record of the reference sample.
  • In an embodiment, the CAR-expressing cell (e.g., T cell, NK cell) preparation is a CD19 CAR-expressing cell (e.g., CTL019) preparation.
  • In an embodiment, the CAR-expressing cell (e.g., T cell, NK cell) preparation comprises a CD19 CAR-expressing cell (e.g., CTL019) preparation.
  • In an embodiment, the CAR-expressing cell (e.g., T cell, NK cell) preparation consists of a CD19 CAR-expressing cell (e.g., CTL019) preparation.
  • In an aspect, a method is provided, comprising:
  • providing a blood sample, e.g., a T cell sample, from a subject having cancer;
  • determining the levels of expression of one or more genes listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, or KLRG1 to obtain a gene expression pattern for the sample;
  • comparing the obtained gene expression pattern to that of a reference value, e.g., a historical record of gene expression;
  • determining a difference between the obtained and the reference value; and
  • recording the determined difference in a quality control record.
  • The method can comprise a step of comparing the obtained gene expression pattern difference with that of a reference sample.
  • In some embodiments, the methods disclosed herein further include administering a T cell depleting agent after treatment with the cell (e.g., an immune effector cell as described herein), thereby reducing (e.g., depleting) the CAR-expressing cells (e.g., the CD19CAR-expressing cells). Such T cell depleting agents can be used to effectively deplete CAR-expressing cells (e.g., CD19CAR-expressing cells) to mitigate toxicity. In some embodiments, the CAR-expressing cells were manufactured according to a method herein, e.g., assayed (e.g., before or after transfection or transduction) according to a method herein.
  • In some embodiments, the T cell depleting agent is administered one, two, three, four, or five weeks after administration of the cell, e.g., the population of immune effector cells, described herein.
  • In one embodiment, the T cell depleting agent is an agent that depletes CAR-expressing cells, e.g., by inducing antibody dependent cell-mediated cytotoxicity (ADCC) and/or complement-induced cell death. For example, CAR-expressing cells described herein may also express an antigen (e.g., a target antigen) that is recognized by molecules capable of inducing cell death, e.g., ADCC or complement-induced cell death. For example, CAR expressing cells described herein may also express a target protein (e.g., a receptor) capable of being targeted by an antibody or antibody fragment. Examples of such target proteins include, but are not limited to, EpCAM, VEGFR, integrins (e.g., integrins αvβ3, α4, αI3/4β3, α4β7, α5β1, αvβ3, αv), members of the TNF receptor superfamily (e.g., TRAIL-R1, TRAIL-R2), PDGF Receptor, interferon receptor, folate receptor, GPNMB, ICAM-1, HLA-DR, CEA, CA-125, MUC1, TAG-72, IL-6 receptor, 5T4, GD2, GD3, CD2, CD3, CD4, CD5, CD11, CD11a/LFA-1, CD15, CD18/ITGB2, CD19, CD20, CD22, CD23/1gE Receptor, CD25, CD28, CD30, CD33, CD38, CD40, CD41, CD44, CD51, CD52, CD62L, CD74, CD80, CD125, CD147/basigin, CD152/CTLA-4, CD154/CD40L, CD195/CCR5, CD319/SLAMF7, and EGFR, and truncated versions thereof (e.g., versions preserving one or more extracellular epitopes but lacking one or more regions within the cytoplasmic domain).
  • In some embodiments, the CAR expressing cell co-expresses the CAR and the target protein, e.g., naturally expresses the target protein or is engineered to express the target protein. For example, the cell, e.g., the population of immune effector cells, can include a nucleic acid (e.g., vector) comprising the CAR nucleic acid (e.g., a CAR nucleic acid as described herein) and a nucleic acid encoding the target protein.
  • In one embodiment, the T cell depleting agent is a CD52 inhibitor, e.g., an anti-CD52 antibody molecule, e.g., alemtuzumab.
  • In other embodiments, the cell, e.g., the population of immune effector cells, expresses a CAR molecule as described herein (e.g., CD19CAR) and the target protein recognized by the T cell depleting agent. In one embodiment, the target protein is CD20. In embodiments where the target protein is CD20, the T cell depleting agent is an anti-CD20 antibody, e.g., rituximab.
  • In further embodiments of any of the aforesaid methods, the methods further include transplanting a cell, e.g., a hematopoietic stem cell, or a bone marrow, into the mammal.
  • In another aspect, the invention features a method of conditioning a mammal prior to cell transplantation. The method includes administering to the mammal an effective amount of the cell comprising a CAR nucleic acid or polypeptide, e.g., a CD19 CAR nucleic acid or polypeptide. In some embodiments, the cell transplantation is a stem cell transplantation, e.g., a hematopoietic stem cell transplantation, or a bone marrow transplantation. In other embodiments, conditioning a subject prior to cell transplantation includes reducing the number of target-expressing cells in a subject, e.g., CD19-expressing normal cells or CD19-expressing cancer cells.
  • Nucleic Acid Constructs Encoding a CAR
  • Nucleic acid molecules encoding one or more CAR constructs can be introduced into an immune effector cell (e.g., a T cell) as described herein. In one aspect, the nucleic acid molecule is provided as a messenger RNA transcript. In one aspect, the nucleic acid molecule is provided as a DNA construct.
  • In some embodiments, a nucleic acid described herein is introduced into a cell that has been assayed by a method described herein, e.g., one or more biomarkers has been assayed. In some embodiments, a cell comprising a nucleic acid described herein is assayed by a method described herein, e.g., one or more biomarkers has been assayed.
  • The nucleic acid molecules described herein can be a DNA molecule, an RNA molecule, or a combination thereof. In one embodiment, the nucleic acid molecule is an mRNA encoding a CAR polypeptide as described herein. In other embodiments, the nucleic acid molecule is a vector that includes any of the aforesaid nucleic acid molecules.
  • Nucleic acid molecules can encode, e.g., a CAR molecule described herein, and can comprise, e.g., a nucleic acid sequence described herein, e.g., in Table 11, Table 12 or Table 13.
  • The nucleic acid sequences coding for the desired molecules can be obtained using recombinant methods known in the art, such as, for example by screening libraries from cells expressing the gene, by deriving the gene from a vector known to include the same, or by isolating directly from cells and tissues containing the same, using standard techniques. Alternatively, the gene of interest can be produced synthetically, rather than cloned.
  • Also described are vectors in which a nucleic acid of the present disclosure is inserted. Vectors derived from retroviruses such as the lentivirus are suitable tools to achieve long-term gene transfer since they allow long-term, stable integration of a transgene and its propagation in daughter cells. Lentiviral vectors have the added advantage over vectors derived from onco-retroviruses such as murine leukemia viruses in that they can transduce non-proliferating cells, such as hepatocytes. They also have the added advantage of low immunogenicity. A retroviral vector may also be, e.g., a gammaretroviral vector. A gammaretroviral vector may include, e.g., a promoter, a packaging signal (w), a primer binding site (PBS), one or more (e.g., two) long terminal repeats (LTR), and a transgene of interest, e.g., a gene encoding a CAR. A gammaretroviral vector may lack viral structural gens such as gag, pol, and env. Exemplary gammaretroviral vectors include Murine Leukemia Virus (MLV), Spleen-Focus Forming Virus (SFFV), and Myeloproliferative Sarcoma Virus (MPSV), and vectors derived therefrom. Other gammaretroviral vectors are described, e.g., in Tobias Maetzig et al., “Gammaretroviral Vectors: Biology, Technology and Application” Viruses. 2011 June; 3(6): 677-713.
  • In another embodiment, the vector comprising the nucleic acid encoding the desired CAR of the invention is an adenoviral vector (A5/35). In another embodiment, the expression of nucleic acids encoding CARs can be accomplished using of transposons such as sleeping beauty, CRISPR, CAS9, and zinc finger nucleases. See below June et al. 2009 NATURE REVIEWS IMMUNOLOGY 9.10: 704-716, is incorporated herein by reference.
  • In brief summary, the expression of natural or synthetic nucleic acids encoding CARs is typically achieved by operably linking a nucleic acid encoding the CAR polypeptide or portions thereof to a promoter, and incorporating the construct into an expression vector. The vectors can be suitable for replication and integration eukaryotes. Typical cloning vectors contain transcription and translation terminators, initiation sequences, and promoters useful for regulation of the expression of the desired nucleic acid sequence.
  • The expression constructs may also be used for nucleic acid immunization and gene therapy, using standard gene delivery protocols. Methods for gene delivery are known in the art. See, e.g., U.S. Pat. Nos. 5,399,346, 5,580,859, 5,589,466, incorporated by reference herein in their entireties. In another embodiment, the invention provides a gene therapy vector.
  • The nucleic acid can be cloned into a number of types of vectors. For example, the nucleic acid can be cloned into a vector including, but not limited to a plasmid, a phagemid, a phage derivative, an animal virus, and a cosmid. Vectors of particular interest include expression vectors, replication vectors, probe generation vectors, and sequencing vectors.
  • Further, the expression vector may be provided to a cell in the form of a viral vector. Viral vector technology is well known in the art and is described, for example, in Sambrook et al., 2012, MOLECULAR CLONING: A LABORATORY MANUAL, volumes 1-4, Cold Spring Harbor Press, NY), and in other virology and molecular biology manuals. Viruses, which are useful as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, and lentiviruses. In general, a suitable vector contains an origin of replication functional in at least one organism, a promoter sequence, convenient restriction endonuclease sites, and one or more selectable markers, (e.g., WO 01/96584; WO 01/29058; and U.S. Pat. No. 6,326,193).
  • A number of viral based systems have been developed for gene transfer into mammalian cells. For example, retroviruses provide a convenient platform for gene delivery systems. A selected gene can be inserted into a vector and packaged in retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to cells of the subject either in vivo or ex vivo. A number of retroviral systems are known in the art. In some embodiments, adenovirus vectors are used. A number of adenovirus vectors are known in the art. In one embodiment, lentivirus vectors are used.
  • Additional promoter elements, e.g., enhancers, regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have been shown to contain functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription. Exemplary promoters include the CMV IE gene, EF-1α, ubiquitin C, or phosphoglycerokinase (PGK) promoters.
  • An example of a promoter that is capable of expressing a CAR transgene in a mammalian T cell is the EF1a promoter. The native EF1a promoter drives expression of the alpha subunit of the elongation factor-1 complex, which is responsible for the enzymatic delivery of aminoacyl tRNAs to the ribosome. The EF1a promoter has been extensively used in mammalian expression plasmids and has been shown to be effective in driving CAR expression from transgenes cloned into a lentiviral vector. See, e.g., Milone et al., MOL. THER. 17(8): 1453-1464 (2009). In one aspect, the EF1a promoter comprises the sequence provided as SEQ ID NO:11.
  • Another example of a promoter is the immediate early cytomegalovirus (CMV) promoter sequence. This promoter sequence is a strong constitutive promoter sequence capable of driving high levels of expression of any polynucleotide sequence operatively linked thereto. However, other constitutive promoter sequences may also be used, including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, an avian leukemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the elongation factor-1a promoter, the hemoglobin promoter, and the creatine kinase promoter. Further, the invention should not be limited to the use of constitutive promoters. Inducible promoters are also contemplated as part of the invention. The use of an inducible promoter provides a molecular switch capable of turning on expression of the polynucleotide sequence which it is operatively linked when such expression is desired, or turning off the expression when expression is not desired. Examples of inducible promoters include, but are not limited to a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline promoter.
  • Another example of a promoter is the phosphoglycerate kinase (PGK) promoter. In embodiments, a truncated PGK promoter (e.g., a PGK promoter with one or more, e.g., 1, 2, 5, 10, 100, 200, 300, or 400, nucleotide deletions when compared to the wild-type PGK promoter sequence) may be desired.
  • A vector may also include, e.g., a signal sequence to facilitate secretion, a polyadenylation signal and transcription terminator (e.g., from Bovine Growth Hormone (BGH) gene), an element allowing episomal replication and replication in prokaryotes (e.g. SV40 origin and ColE1 or others known in the art) and/or elements to allow selection (e.g., ampicillin resistance gene and/or zeocin marker).
  • In order to assess the expression of a CAR polypeptide or portions thereof, the expression vector to be introduced into a cell can also contain either a selectable marker gene or a reporter gene or both to facilitate identification and selection of expressing cells from the population of cells sought to be transfected or infected through viral vectors. In other aspects, the selectable marker may be carried on a separate piece of DNA and used in a co-transfection procedure. Both selectable markers and reporter genes may be flanked with appropriate regulatory sequences to enable expression in the host cells. Useful selectable markers include, for example, antibiotic-resistance genes, such as neo and the like.
  • Reporter genes are used for identifying potentially transfected cells and for evaluating the functionality of regulatory sequences. Reporter genes are described, e.g., in paragraph 599 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • In embodiments, the vector may comprise two or more nucleic acid sequences encoding a CAR, e.g., a CAR described herein, e.g., a CD19 CAR, and a second CAR, e.g., an inhibitory CAR or a CAR that specifically binds to an antigen other than CD19. In such embodiments, the two or more nucleic acid sequences encoding the CAR are encoded by a single nucleic molecule in the same frame and as a single polypeptide chain. In this aspect, the two or more CARs, can, e.g., be separated by one or more peptide cleavage sites. (e.g., an auto-cleavage site or a substrate for an intracellular protease). Examples of peptide cleavage sites include T2A, P2A, E2A, or F2A sites.
  • Methods of introducing and expressing genes into a cell are known in the art. In the context of an expression vector, the vector can be readily introduced into a host cell, e.g., mammalian, bacterial, yeast, or insect cell by any method in the art. For example, the expression vector can be transferred into a host cell by physical, chemical, or biological means, e.g., those described in paragraphs 601-603 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • In the case where a non-viral delivery system is utilized, an exemplary delivery vehicle is a liposome. The use of lipid formulations is contemplated for the introduction of the nucleic acids into a host cell (in vitro, ex vivo or in vivo), and is described, e.g., in paragraphs 604-605 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • Regardless of the method used to introduce exogenous nucleic acids into a host cell or otherwise expose a cell to the inhibitor of the present invention, in order to confirm the presence of the recombinant DNA sequence in the host cell, a variety of assays may be performed. Such assays include, for example, “molecular biological” assays well known to those of skill in the art, such as Southern and Northern blotting, RT-PCR and PCR; “biochemical” assays, such as detecting the presence or absence of a particular peptide, e.g., by immunological means (ELISAs and Western blots) or by assays described herein to identify agents falling within the scope of the invention.
  • Therapeutic Methods
  • In one aspect, the disclosure provides methods for treating a disease associated with expression of a tumor antigen described herein. In some embodiments, immune effector cells are assayed by a method described herein, e.g., one or more biomarkers is assayed, and the cells are administered to a subject as part of a treatment described herein. For example, the immune effector cells can be administered as part of a combination therapy described herein.
  • In one aspect, the present disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CAR. In one embodiment, the cancer to be treated is a B cell malignancy. In one embodiment, the cancer to be treated is ALL (acute lymphoblastic leukemia), CLL (chronic lymphocytic leukemia), DLBCL (diffuse large B-cell lymphoma), MCL (Mantle cell lymphoma, or MM (multiple myeloma).
  • In one aspect, the disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CD19 CAR, wherein the cancer cells express CD19. In one embodiment, the cancer to be treated is a B cell malignancy. In one embodiment, the cancer to be treated is ALL (acute lymphoblastic leukemia), CLL (chronic lymphocytic leukemia), DLBCL (diffuse large B-cell lymphoma), MCL (Mantle cell lymphoma), Hodgkin lymphoma, or MM (multiple myeloma).
  • In one aspect, the present invention provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CD22 CAR, wherein the cancer cells express CD22. In one embodiment, the cancer to be treated is a B cell malignancy. In one embodiment, the cancer to be treated is ALL (acute lymphoblastic leukemia), CLL (chronic lymphocytic leukemia), DLBCL (diffuse large B-cell lymphoma), MCL (Mantle cell lymphoma), Hodgkin lymphoma, or MM (multiple myeloma).
  • In one aspect, the present invention provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CD20 CAR, wherein the cancer cells express CD20. In one embodiment, the cancer to be treated is a B cell malignancy. In one embodiment, the cancer to be treated is ALL (acute lymphoblastic leukemia), CLL (chronic lymphocytic leukemia), DLBCL (diffuse large B-cell lymphoma), MCL (Mantle cell lymphoma), Hodgkin lymphoma, or MM (multiple myeloma).
  • In one aspect, the present invention provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a ROR1 CAR, wherein the cancer cells express ROR1. In one embodiment, the cancer to be treated is a B cell malignancy. In one embodiment, the cancer to be treated is ALL (acute lymphoblastic leukemia), CLL (chronic lymphocytic leukemia), DLBCL (diffuse large B-cell lymphoma), MCL (Mantle cell lymphoma), Hodgkin lymphoma, or MM (multiple myeloma).
  • The disclosure includes a type of cellular therapy where immune effector cells (e.g., T cells, NK cells) are genetically modified (e.g., via transduction of a lentiviral vector) to express a CAR and the CAR-expressing cell is infused to a recipient in need thereof. The infused cell is able to kill tumor cells in the recipient. Unlike antibody therapies, CAR-modified immune effector cells (e.g., T cells, NK cells) are able to replicate in vivo resulting in long-term persistence that can lead to sustained tumor control. CAR-expressing cells (e.g., T cells or NK cells) generated using lentiviral vectors will have stable CAR expression. In various aspects, the immune effector cells (e.g., T cells, NK cells) administered to the patient, or their progeny, persist in the patient for at least four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, twelve months, thirteen months, fourteen month, fifteen months, sixteen months, seventeen months, eighteen months, nineteen months, twenty months, twenty-one months, twenty-two months, twenty-three months, two years, three years, four years, or five years after administration of the T cell to the patient.
  • The invention also includes a type of cellular therapy where immune effector cells (e.g., T cells, NK cells) are modified, e.g., by in vitro transcribed RNA, to transiently express a CAR and the CAR-expressing cell is infused to a recipient in need thereof. CAR-expressing cells (e.g., T cells, NK cells) generated through transduction of CAR RNA (e.g., by transfection or electroporation) transiently express RNA CARs for 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 days after transduction. The infused cell is able to kill tumor cells in the recipient. Thus, in various aspects, the immune effector cells (e.g., T cells, NK cells) administered to the patient, is present for less than one month, e.g., three weeks, two weeks, one week, after administration of the T cell to the patient.
  • In one aspect, the present disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CAR, e.g., a CAR described herein.
  • In one embodiment, the present disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL and CLL) by providing to the subject in need thereof immune effector cells (e.g., T cells, NK cells) that are engineered to express a CAR that specifically targets or binds to a tumor antigen (or cancer associated antigen) described herein, wherein the subject has been identified as a responder or partial responder. In other embodiments, the methods provide treating a cancer (e.g., a hematological cancer such as ALL and CLL) as a partial responder or non-responder by providing to the subject a cancer therapy other than a CAR therapy, e.g., providing the subject a treatment that is the standard of care for that particular type of cancer. In yet another embodiment, the method of treatment includes altering the manufacturing of a CAR-expressing cell to enrich for naïve T cells, e.g., as described herein, for a subject identified as a partial responder or non-responder prior to administering a CAR-expressing cell, e.g., a CAR-expressing cell described herein.
  • In one embodiment, the immune effector cells (e.g., T cells, NK cells) are engineered to express CD19 CAR, for treating a subject having cancer (e.g., a hematological cancer such as ALL and CLL), wherein the cancer cells express CD19. In one embodiment, the cancer to be treated is ALL or CLL. The CD19 CAR molecules to be expressed in an immune effector cell can comprise any anti-CD19 antigen binding domain in the art (e.g., those provided in Table 12) in combination with any of the CAR domains described herein to generate a full CAR construct. For example, the full CAR construct is a CAR listed in Table 13. Table 13 provides the exemplary full CD19 CAR constructs generated using the various CAR domains (e.g., transmembrane and intracellular signaling domains) listed in Table 12, and the anti-CD19 antigen binding domains listed in Table 12. Amino acid sequences are designated (aa) and nucleic acid sequences are designated (nt).
  • TABLE 13
    CD19 CAR Constructs
    Name Sequence
    CAR
     1
    104875 atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgctcggcccga
    CAR 1- aattgtgatgacccagtcacccgccactcttagcctttcacccggtgagcgcgcaaccctgtctt
    Full-nt gcagagcctcccaagacatctcaaaataccttaattggtatcaacagaagcccggacaggctcct
    cgccttctgatctaccacaccagccggctccattctggaatccctgccaggttcagcggtagcgg
    atctgggaccgactacaccctcactatcagctcactgcagccagaggacttcgctgtctatttct
    gtcagcaagggaacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtgga
    ggtggcagcggaggaggtgggtccggcggtggaggaagccaggtccaactccaagaaagcggacc
    gggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagcggagtgtctctccccg
    attacggggtgtcttggatcagacagccaccggggaagggtctggaatggattggagtgatttgg
    ggctctgagactacttactactcttcatccctcaagtcacgcgtcaccatctcaaaggacaactc
    taagaatcaggtgtcactgaaactgtcatctgtgaccgcagccgacaccgccgtgtactattgcg
    ctaagcattactattatggcgggagctacgcaatggattactggggacagggtactctggtcacc
    gtgtccagcaccactaccccagcaccgaggccacccaccccggctcctaccatcgcctcccagcc
    tctgtccctgcgtccggaggcatgtagacccgcagctggtggggccgtgcatacccggggtcttg
    acttcgcctgcgatatctacatttgggcccctctggctggtacttgcggggtcctgctgctttca
    ctcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacatctttaagcaaccctt
    catgaggcctgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg
    aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctacaagcagggg
    cagaaccagctctacaacgaactcaatcttggtcggagagaggagtacgacgtgctggacaagcg
    gagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatccccaagagggcctgtaca
    acgagctccaaaaggataagatggcagaagcctatagcgagattggtatgaaaggggaacgcaga
    agaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaaggacacctatgacgc
    tcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 55)
    104875 MALPVTALLLPLALLLHAARPeivmtqspatlslspgeratlsc rasgdiskyln wyqqkpgqap
    CAR 1- rlliy htsrlhs giparfsgsgsgtdytltisslqpedfavyfc qqgntlpyt fgqgtkleikgg
    Full-aa ggsggggsggggsqvqlqesgpglvkpsetlsltctvsgvslp dygvs wirqppgkglewig viw
    gsettyyssslks rvtiskdnsknqvslklssvtaadtavyycak hyyyggsyamdy wgqgtlvt
    vsstttpaprpptpaptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvllls
    lvitlyckrgrkkllyifkqpfmrpvqttqeedgcscrfpeeeeggcelrykfsrsadapaykqg
    qnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeayseigmkgerr
    rgkghdglyqglstatkdtydalhmgalppr (SEQ ID NO: 56)
    CAR 2
    104876 atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgctcggcccga
    CAR 2- aattgtgatgacccagtcacccgccactcttagcctttcacccggtgagcgcgcaaccctgtctt
    Full-nt gcagagcctcccaagacatctcaaaataccttaattggtatcaacagaagcccggacaggctcct
    cgccttctgatctaccacaccagccggctccattctggaatccctgccaggttcagcggtagcgg
    atctgggaccgactacaccctcactatcagctcactgcagccagaggacttcgctgtctatttct
    gtcagcaagggaacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtgga
    ggtggcagcggaggaggtgggtccggcggtggaggaagccaggtccaactccaagaaagcggacc
    gggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagcggagtgtctctccccg
    attacggggtgtcttggatcagacagccaccggggaagggtctggaatggattggagtgatttgg
    ggctctgagactacttactaccaatcatccctcaagtcacgcgtcaccatctcaaaggacaactc
    taagaatcaggtgtcactgaaactgtcatctgtgaccgcagccgacaccgccgtgtactattgcg
    ctaagcattactattatggcgggagctacgcaatggattactggggacagggtactctggtcacc
    gtgtccagcaccactaccccagcaccgaggccacccaccccggctcctaccatcgcctcccagcc
    tctgtccctgcgtccggaggcatgtagacccgcagctggtggggccgtgcatacccggggtcttg
    acttcgcctgcgatatctacatttgggcccctctggctggtacttgcggggtcctgctgctttca
    ctcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacatctttaagcaaccctt
    catgaggcctgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg
    aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctacaagcagggg
    cagaaccagctctacaacgaactcaatcttggtcggagagaggagtacgacgtgctggacaagcg
    gagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatccccaagagggcctgtaca
    acgagctccaaaaggataagatggcagaagcctatagcgagattggtatgaaaggggaacgcaga
    agaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaaggacacctatgacgc
    tcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 57)
    104876 MALPVTALLLPLALLLHAARPeivmtqspatlslspgeratlsc rasqdiskyln wyqqkpgqap
    CAR 2- rlliy htsrlhs giparfsgsgsgtdytltisslqpedfavyfc qqgntlpyt fgqgtkleikgg
    Full-aa ggsggggsggggsqvglqesgpglvkpsetlsltctvsgvslp dygvs wirqppgkglewig viw
    gsettyyqsslks rvtiskdnsknqvslklssvtaadtavyycak hyyyggsy amdywgqgtlvt
    vsstttpaprpptpaptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvllls
    lvitlyckrgrkkllyifkqpfmrpvgttgeedgcscrfpeeeeggcelrykfsrsadapaykqg
    qnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeayseigmkgerr
    rgkghdglyqglstatkdtydalhmqalppr (SEQ ID NO: 58)
    CAR 3
    104877 atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgctcgcccaca
    CAR 3- agtccagcttcaagaatcagggcctggtctggtgaagccatctgagactctgtccctcacttgca
    Full-nt ccgtgagcggagtgtccctcccagactacggagtgagctggattagacagcctcccggaaaggga
    ctggagtggatcggagtgatttggggtagcgaaaccacttactattcatcttccctgaagtcacg
    ggtcaccatttcaaaggataactcaaagaatcaagtgagcctcaagctctcatcagtcaccgccg
    ctgacaccgccgtgtattactgtgccaagcattactactatggagggtcctacgccatggactac
    tggggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcgggagcgg
    tggaggtggctccgaaatcgtgatgacccagagccctgcaaccctgtccctttctcccggggaac
    gggctaccctttcttgtcgggcatcacaagatatctcaaaatacctcaattggtatcaacagaag
    ccgggacaggcccctaggcttcttatctaccacacctctcgcctgcatagcgggattcccgcacg
    ctttagcgggtctggaagcgggaccgactacactctgaccatctcatctctccagcccgaggact
    tcgccgtctacttctgccagcagggtaacaccctgccgtacaccttcggccagggcaccaagctt
    gagatcaaaaccactactcccgctccaaggccacccacccctgccccgaccatcgcctctcagcc
    gctttccctgcgtccggaggcatgtagacccgcagctggtggggccgtgcatacccggggtcttg
    acttcgcctgcgatatctacatttgggcccctctggctggtacttgcggggtcctgctgctttca
    ctcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacatctttaagcaaccctt
    catgaggcctgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg
    aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctacaagcagggg
    cagaaccagctctacaacgaactcaatcttggtcggagagaggagtacgacgtgctggacaagcg
    gagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatccccaagagggcctgtaca
    acgagctccaaaaggataagatggcagaagcctatagcgagattggtatgaaaggggaacgcaga
    agaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaaggacacctatgacgc
    tcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 59)
    104877 MALPVTALLLPLALLLHAARPqvqlqesgpglvkpsetlsltctvsgvslp dygvs wirqppgkg
    CAR 3- lewig viwgsettyyssslks rvtiskdnsknqvslklssvtaadtavyycak hyyyggsyamdy
    Full-aa wgggtivtvssggggsggggsggggseivmtgspatls1spgeratlsc rasqdiskyln wyggk
    pgqaprlliy htsrlhs giparfsgsgsgtdytltisslqpedfavyfc qqgntlpyt fgqgtkl
    eiktttpaprpptpaptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvllls
    lvitlyckrgrkkllyifkgpfmrpvqttqeedgcscrfpeeeeggcelrykfsrsadapaykqg
    qnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeayseigmkgerr
    rgkghdglyqglstatkdtydalhmqalppr (SEQ ID NO: 60)
    CAR 4
    104878 atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgctcgcccaca
    CAR 4- agtccagcttcaagaatcagggcctggtctggtgaagccatctgagactctgtccctcacttgca
    Full-nt ccgtgagcggagtgtccctcccagactacggagtgagctggattagacagcctcccggaaaggga
    ctggagtggatcggagtgatttggggtagcgaaaccacttactatcaatcttccctgaagtcacg
    ggtcaccatttcaaaggataactcaaagaatcaagtgagcctcaagctctcatcagtcaccgccg
    ctgacaccgccgtgtattactgtgccaagcattactactatggagggtcctacgccatggactac
    tggggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcgggagcgg
    tggaggtggctccgaaatcgtgatgacccagagccctgcaaccctgtccctttctcccggggaac
    gggctaccctttcttgtcgggcatcacaagatatctcaaaatacctcaattggtatcaacagaag
    ccgggacaggcccctaggcttcttatctaccacacctctcgcctgcatagcgggattcccgcacg
    ctttagcgggtctggaagcgggaccgactacactctgaccatctcatctctccagcccgaggact
    tcgccgtctacttctgccagcagggtaacaccctgccgtacaccttcggccagggcaccaagctt
    gagatcaaaaccactactcccgctccaaggccacccacccctgccccgaccatcgcctctcagcc
    gctttccctgcgtccggaggcatgtagacccgcagctggtggggccgtgcatacccggggtcttg
    acttcgcctgcgatatctacatttgggcccctctggctggtacttgcggggtcctgctgctttca
    ctcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacatctttaagcaaccctt
    catgaggcctgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg
    aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctacaagcagggg
    cagaaccagctctacaacgaactcaatcttggtcggagagaggagtacgacgtgctggacaagcg
    gagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatccccaagagggcctgtaca
    acgagctccaaaaggataagatggcagaagcctatagcgagattggtatgaaaggggaacgcaga
    agaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaaggacacctatgacgc
    tcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 61)
    104878 MALPVTALLLPLALLLHAARPqvqlqesgpglvkpsetlsltctvsgvslp dygys wirqppgkg
    CAR 4- lewig viwgsettyyqsslks rvtiskdnsknqvslklssvtaadtavyycak hyyyggsyamdy
    Full-aa wgqgtlvtvssggggsggggsggggseivmtgspatlslspgeratlsc rasqdiskyln wyqqk
    pgqaprlliy htsrlhs giparfsgsgsgtdytltisslqpedfavyfc qqgntlpyt fgqgtkl
    eiktttpaprpptpaptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvllls
    lvitlyckrgrkkllyifkqpfmrpvqttqeedgcscrfpeeeeggcelrvkfsrsadapaykqg
    qnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeayseigmkgerr
    rgkghdglygglstatkdtydalhmqalppr (SEQ ID NO: 62)
    CAR 5
    CAR5 scFv eivmtqspatlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgiparfsgs
    domain gsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikggggsggggsggggsggggsqvq
    lqesgpglvkpsetlsltctvsgvslpdygyswirqppgkglewigviwgsettyyssslksrvt
    iskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgqgtlvtvss (SEQ ID NO:
    63)
    104879 atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgctcggcccga
    CAR 5- aattgtgatgacccagtcacccgccactcttagcctttcacccggtgagcgcgcaaccctgtctt
    Full-nt gcagagcctcccaagacatctcaaaataccttaattggtatcaacagaagcccggacaggctcct
    cgccttctgatctaccacaccagccggctccattctggaatccctgccaggttcagcggtagcgg
    atctgggaccgactacaccctcactatcagctcactgcagccagaggacttcgctgtctatttct
    gtcagcaagggaacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtgga
    ggtggcagcggaggaggtgggtccggcggtggaggaagcggcggaggcgggagccaggtccaact
    ccaagaaagcggaccgggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagcg
    gagtgtctctccccgattacggggtgtcttggatcagacagccaccggggaagggtctggaatgg
    attggagtgatttggggctctgagactacttactactcttcatccctcaagtcacgcgtcaccat
    ctcaaaggacaactctaagaatcaggtgtcactgaaactgtcatctgtgaccgcagccgacaccg
    ccgtgtactattgcgctaagcattactattatggcgggagctacgcaatggattactggggacag
    ggtactctggtcaccgtgtccagcaccactaccccagcaccgaggccacccaccccggctcctac
    catcgcctcccagcctctgtccctgcgtccggaggcatgtagacccgcagctggtggggccgtgc
    atacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggtacttgcggg
    gtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacat
    ctttaagcaacccttcatgaggcctgtgcagactactcaagaggaggacggctgttcatgccggt
    tcccagaggaggaggaaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctcca
    gcctacaagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagtacga
    cgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatcccc
    aagagggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagattggtatg
    aaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaa
    ggacacctatgacgctcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 64)
    104879 MALPVTALLLPLALLLHAARPeivmtqspatlslspgerat1sc rasgdiskyln wyqqkpgqap
    CAR 5- rlliy htsrlhs giparfsgsgsgtdytltisslqpedfavyfc qqgntlpyt fgqgtkleikgg
    Full-aa ggsggggsggggsggggsqvqlqesgpglvkpsetlsltctvsgvslp dygvs wirqppgkglew
    ig viwgsettyyssslks rvtiskdnsknqvslklssvtaadtavyycak hyyyggsyamdy wgq
    gtivtvsstttpaprpptpaptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcg
    vlllslvitlyckrgrkkllyifkgpfmrpvqttqeedgcscrfpeeeeggcelrykfsrsadap
    aykqgqnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeayseigm
    kgerrrgkghdglyqglstatkdtydalhmgalppr (SEQ ID NO: 65)
    CAR 6
    104880 atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgctcggcccga
    CAR6- aattgtgatgacccagtcacccgccactcttagcctttcacccggtgagcgcgcaaccctgtctt
    Full-nt gcagagcctcccaagacatctcaaaataccttaattggtatcaacagaagcccggacaggctcct
    cgccttctgatctaccacaccagccggctccattctggaatccctgccaggttcagcggtagcgg
    atctgggaccgactacaccctcactatcagctcactgcagccagaggacttcgctgtctatttct
    gtcagcaagggaacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtgga
    ggtggcagcggaggaggtgggtccggcggtggaggaagcggaggcggagggagccaggtccaact
    ccaagaaagcggaccgggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagcg
    gagtgtctctccccgattacggggtgtcttggatcagacagccaccggggaagggtctggaatgg
    attggagtgatttggggctctgagactacttactaccaatcatccctcaagtcacgcgtcaccat
    ctcaaaggacaactctaagaatcaggtgtcactgaaactgtcatctgtgaccgcagccgacaccg
    ccgtgtactattgcgctaagcattactattatggcgggagctacgcaatggattactggggacag
    ggtactctggtcaccgtgtccagcaccactaccccagcaccgaggccacccaccccggctcctac
    catcgcctcccagcctctgtccctgcgtccggaggcatgtagacccgcagctggtggggccgtgc
    atacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggtacttgcggg
    gtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacat
    ctttaagcaacccttcatgaggcctgtgcagactactcaagaggaggacggctgttcatgccggt
    tcccagaggaggaggaaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctcca
    gcctacaagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagtacga
    cgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatcccc
    aagagggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagattggtatg
    aaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaa
    ggacacctatgacgctcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 66)
    104880 MALPVTALLLPLALLLHAARPeivmtqspatlslspgeratlsc rasqdiskyln wyqqkpgqap
    CAR6- rlliy htsrlhs giparfsgsgsgtdytltisslqpedfavyfc qqgntlpyt fgqgtkleikgg
    Full-aa ggsggggsggggsggggsqvqlqesgpglvkpsetlsltctvsgvslp dygvs wirqppgkglew
    ig viwgsettyyqsslks rvtiskdnsknqvslklssvtaadtavyycak hyyyggsyamdy wgq
    gtlvtvsstttpaprpptpaptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcg
    vlllslvitlyckrgrkkllyifkqpfmrpvgttgeedgcscrfpeeeeggcelrykfsrsadap
    aykqgqnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelgkdkmaeayseigm
    kgerrrgkghdglyqglstatkdtydalhmqalppr (SEQ ID NO: 67)
    CAR 7
    104881 atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgctcgcccaca
    CAR 7 agtccagcttcaagaatcagggcctggtctggtgaagccatctgagactctgtccctcacttgca
    Full-nt ccgtgagcggagtgtccctcccagactacggagtgagctggattagacagcctcccggaaaggga
    ctggagtggatcggagtgatttggggtagcgaaaccacttactattcatcttccctgaagtcacg
    ggtcaccatttcaaaggataactcaaagaatcaagtgagcctcaagctctcatcagtcaccgccg
    ctgacaccgccgtgtattactgtgccaagcattactactatggagggtcctacgccatggactac
    tggggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcgggagcgg
    tggaggtggctccggaggtggcggaagcgaaatcgtgatgacccagagccctgcaaccctgtccc
    tttctcccggggaacgggctaccctttcttgtcgggcatcacaagatatctcaaaatacctcaat
    tggtatcaacagaagccgggacaggcccctaggcttcttatctaccacacctctcgcctgcatag
    cgggattcccgcacgctttagcgggtctggaagcgggaccgactacactctgaccatctcatctc
    tccagcccgaggacttcgccgtctacttctgccagcagggtaacaccctgccgtacaccttcggc
    cagggcaccaagcttgagatcaaaaccactactcccgctccaaggccacccacccctgccccgac
    catcgcctctcagccgctttccctgcgtccggaggcatgtagacccgcagctggtggggccgtgc
    atacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggtacttgcggg
    gtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacat
    ctttaagcaacccttcatgaggcctgtgcagactactcaagaggaggacggctgttcatgccggt
    tcccagaggaggaggaaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctcca
    gcctacaagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagtacga
    cgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatcccc
    aagagggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagattggtatg
    aaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaa
    ggacacctatgacgctcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 68)
    104881 MALPVTALLLPLALLLHAARPqvqlqesgpglvkpsetlsltctvsgvslp dygvs wirqppgkg
    CAR 7 lewig viwgsettyyssslks rvtiskdnsknqvslklssvtaadtavyycak hyyyggsyamdy
    Full-aa wgqgtlvtvssggggsggggsggggsggggseivmtqspatlslspgeratlsc rasqdiskyln
    wyqqkpgqaprlliy htsrlhs giparfsgsgsgtdytltisslqpedfavyfc qqgntlpyt fg
    ggtkleiktttpaprpptpaptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcg
    vlllslvitlyckrgrkkllyifkqpfmrpvqttqeedgcscrfpeeeeggcelrykfsrsadap
    aykqgqnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeayseigm
    kgerrrgkghdglyqglstatkdtydalhmgalppr (SEQ ID NO: 69)
    CAR 8
    104882 atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgctcgcccaca
    CAR 8- agtccagcttcaagaatcagggcctggtctggtgaagccatctgagactctgtccctcacttgca
    Full-nt ccgtgagcggagtgtccctcccagactacggagtgagctggattagacagcctcccggaaaggga
    ctggagtggatcggagtgatttggggtagcgaaaccacttactatcaatcttccctgaagtcacg
    ggtcaccatttcaaaggataactcaaagaatcaagtgagcctcaagctctcatcagtcaccgccg
    ctgacaccgccgtgtattactgtgccaagcattactactatggagggtcctacgccatggactac
    tggggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcgggagcgg
    tggaggtggctccggaggcggtgggtcagaaatcgtgatgacccagagccctgcaaccctgtccc
    tttctcccggggaacgggctaccctttcttgtcgggcatcacaagatatctcaaaatacctcaat
    tggtatcaacagaagccgggacaggcccctaggcttcttatctaccacacctctcgcctgcatag
    cgggattcccgcacgctttagcgggtctggaagcgggaccgactacactctgaccatctcatctc
    tccagcccgaggacttcgccgtctacttctgccagcagggtaacaccctgccgtacaccttcggc
    cagggcaccaagcttgagatcaaaaccactactcccgctccaaggccacccacccctgccccgac
    catcgcctctcagccgctttccctgcgtccggaggcatgtagacccgcagctggtggggccgtgc
    atacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggtacttgcggg
    gtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacat
    ctttaagcaacccttcatgaggcctgtgcagactactcaagaggaggacggctgttcatgccggt
    tcccagaggaggaggaaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctcca
    gcctacaagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagtacga
    cgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatcccc
    aagagggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagattggtatg
    aaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaa
    ggacacctatgacgctcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 70)
    104882 MALPVTALLLPLALLLHAARPqvqlqesgpglvkpsetlsltctvsgvslp dygvs wirqppgkg
    CAR 8- lewig viwgsettyyqsslks rvtiskdnskngvslklssvtaadtavyycak hyyyggsyamdy
    Full-aa wgqgtlvtvssggggsggggsggggsggggseivmtqspatlslspgeratlsc rasqdiskyln
    wyqqkpgqaprlliy htsrlhs giparfsgsgsgtdytltisslqpedfavyfc qqgntlpyt fg
    qgtkleiktttpaprpptpaptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcg
    vlllslvitlyckrgrkkllyifkqpfmrpvqttqeedgcscrfpeeeeggcelrvkfsrsadap
    aykqgqnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeayseigm
    kgerrrgkghdglyqglstatkdtydalhmqalppr (SEQ ID NO: 71)
    CAR 9
    105974 atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgctcggcccga
    CAR 9 aattgtgatgacccagtcacccgccactcttagcctttcacccggtgagcgcgcaaccctgtctt
    Full-nt gcagagcctcccaagacatctcaaaataccttaattggtatcaacagaagcccggacaggctcct
    cgccttctgatctaccacaccagccggctccattctggaatccctgccaggttcagcggtagcgg
    atctgggaccgactacaccctcactatcagctcactgcagccagaggacttcgctgtctatttct
    gtcagcaagggaacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtgga
    ggtggcagcggaggaggtgggtccggcggtggaggaagcggaggcggtgggagccaggtccaact
    ccaagaaagcggaccgggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagcg
    gagtgtctctccccgattacggggtgtcttggatcagacagccaccggggaagggtctggaatgg
    attggagtgatttggggctctgagactacttactacaactcatccctcaagtcacgcgtcaccat
    ctcaaaggacaactctaagaatcaggtgtcactgaaactgtcatctgtgaccgcagccgacaccg
    ccgtgtactattgcgctaagcattactattatggcgggagctacgcaatggattactggggacag
    ggtactctggtcaccgtgtccagcaccactaccccagcaccgaggccacccaccccggctcctac
    catcgcctcccagcctctgtccctgcgtccggaggcatgtagacccgcagctggtggggccgtgc
    atacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggtacttgcggg
    gtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacat
    ctttaagcaacccttcatgaggcctgtgcagactactcaagaggaggacggctgttcatgccggt
    tcccagaggaggaggaaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctcca
    gcctacaagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagtacga
    cgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatcccc
    aagagggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagattggtatg
    aaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaa
    ggacacctatgacgctcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 72)
    105974 MALPVTALLLPLALLLHAARPeivmtqspatlslspgeratlsc rasqdiskyln wyqqkpgqap
    CAR 9- rlliy htsrlhs giparfsgsgsgtdytltisslqpedfavyfc qqgntlpyt fgqgtkleikgg
    Full-aa ggsggggsggggsggggsqlqlqesgpglvkpsetlsltctvsgvslp dygvs wirqppgkglew
    ig viwgsettyynsslks rvtiskdnsknqvslklssvtaadtavyycak hyyyggsyamdy wgq
    gtlvtvsstttpaprpptpaptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcg
    vlllslvitlyckrgrkkllyifkqpfmrpvqttgeedgcscrfpeeeeggcelrykfsrsadap
    aykqgqnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeayseigm
    kgerrrgkghdglyqglstatkdtydalhmqalppr (SEQ ID NO: 73)
    CAR10
    105975 atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgctcggcccga
    CAR 10 aattgtgatgacccagtcacccgccactcttagcctttcacccggtgagcgcgcaaccctgtctt
    Full-nt gcagagcctcccaagacatctcaaaataccttaattggtatcaacagaagcccggacaggctcct
    cgccttctgatctaccacaccagccggctccattctggaatccctgccaggttcagcggtagcgg
    atctgggaccgactacaccctcactatcagctcactgcagccagaggacttcgctgtctatttct
    gtcagcaagggaacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtgga
    ggtggcagcggaggaggtgggtccggcggtggaggaagcggaggcggtgggagccaggtccaact
    ccaagaaagcggaccgggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagcg
    gagtgtctctccccgattacggggtgtcttggatcagacagccaccggggaagggtctggaatgg
    attggagtgatttggggctctgagactacttactacaactcatccctcaagtcacgcgtcaccat
    ctcaaaggacaactctaagaatcaggtgtcactgaaactgtcatctgtgaccgcagccgacaccg
    ccgtgtactattgcgctaagcattactattatggcgggagctacgcaatggattactggggacag
    ggtactctggtcaccgtgtccagcaccactaccccagcaccgaggccacccaccccggctcctac
    catcgcctcccagcctctgtccctgcgtccggaggcatgtagacccgcagctggtggggccgtgc
    atacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggtacttgcggg
    gtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacat
    ctttaagcaacccttcatgaggcctgtgcagactactcaagaggaggacggctgttcatgccggt
    tcccagaggaggaggaaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctcca
    gcctacaagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagtacga
    cgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatcccc
    aagagggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagattggtatg
    aaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaa
    ggacacctatgacgctcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 74)
    105975 MALPVTALLLPLALLLHAARPEIVMTQSPATLSLSPGERATLSC RASQDISKYLN WYQQKPGQAP
    CAR 10 RLLIY HTSRLHS GIPARFSGSGSGTDYTLTISSLQPEDFAVYFC QQGNTLPYT FGQGTKLEIKGG
    Full-aa GGSGGGGSGGGGSGGGGSQVQLQESGPGLVKPSETLSLTCTVSGVSLP DYGVS WIRQPPGKGLEW
    IG VIWGSETTYYNSSLKS RVTISKDNSKNQVSLKLSSVTAADTAVYYCAK HYYYGGSYAMDY WGQ
    GTLVTVSSTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCG
    VLLLSLVITLYCKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAP
    AYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGM
    KGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NO: 75)
    CAR11
    105976 atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgctcgcccaca
    CAR 11 agtccagcttcaagaatcagggcctggtctggtgaagccatctgagactctgtccctcacttgca
    Full-nt ccgtgagcggagtgtccctcccagactacggagtgagctggattagacagcctcccggaaaggga
    ctggagtggatcggagtgatttggggtagcgaaaccacttactataactcttccctgaagtcacg
    ggtcaccatttcaaaggataactcaaagaatcaagtgagcctcaagctctcatcagtcaccgccg
    ctgacaccgccgtgtattactgtgccaagcattactactatggagggtcctacgccatggactac
    tggggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcgggagcgg
    tggaggtggctccggaggtggcggaagcgaaatcgtgatgacccagagccctgcaaccctgtccc
    tttctcccggggaacgggctaccctttcttgtcgggcatcacaagatatctcaaaatacctcaat
    tggtatcaacagaagccgggacaggcccctaggcttcttatctaccacacctctcgcctgcatag
    cgggattcccgcacgctttagcgggtctggaagcgggaccgactacactctgaccatctcatctc
    tccagcccgaggacttcgccgtctacttctgccagcagggtaacaccctgccgtacaccttcggc
    cagggcaccaagcttgagatcaaaaccactactcccgctccaaggccacccacccctgccccgac
    catcgcctctcagccgctttccctgcgtccggaggcatgtagacccgcagctggtggggccgtgc
    atacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggtacttgcggg
    gtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacat
    ctttaagcaacccttcatgaggcctgtgcagactactcaagaggaggacggctgttcatgccggt
    tcccagaggaggaggaaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctcca
    gcctacaagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagtacga
    cgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatcccc
    aagagggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagattggtatg
    aaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaa
    ggacacctatgacgctcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 76)
    105976 MALPVTALLLPLALLLHAARPQVQLQESGPGLVKPSETLSLTCTVSGVSLP DYGVS WIRQPPGKG
    CAR
     11 LEWIG VIWGSETTYYNSSLKS RVTISKDNSKNQVSLKLSSVTAADTAVYYCAK HYYYGGSYAMDY
    Full-aa WGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVMTQSPATLSLSPGERATLSC RASQDISKYLN
    WYQQKPGQAPRLLIY HTSRLHS GIPARFSGSGSGTDYTLTISSLQPEDFAVYFC QQGNTLPYT FG
    QGTKLEIKTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCG
    VLLLSLVITLYCKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAP
    AYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGM
    KGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NO: 77)
    CAR12
    105977 atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgctcggcccga
    CAR 12- aattgtgatgacccagtcacccgccactcttagcctttcacccggtgagcgcgcaaccctgtctt
    Full-nt gcagagcctcccaagacatctcaaaataccttaattggtatcaacagaagcccggacaggctcct
    cgccttctgatctaccacaccagccggctccattctggaatccctgccaggttcagcggtagcgg
    atctgggaccgactacaccctcactatcagctcactgcagccagaggacttcgctgtctatttct
    gtcagcaagggaacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtgga
    ggtggcagcggaggaggtgggtccggcggtggaggaagccaggtccaactccaagaaagcggacc
    gggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagcggagtgtctctccccg
    attacggggtgtcttggatcagacagccaccggggaagggtctggaatggattggagtgatttgg
    ggctctgagactacttactacaactcatccctcaagtcacgcgtcaccatctcaaaggacaactc
    taagaatcaggtgtcactgaaactgtcatctgtgaccgcagccgacaccgccgtgtactattgcg
    ctaagcattactattatggcgggagctacgcaatggattactggggacagggtactctggtcacc
    gtgtccagcaccactaccccagcaccgaggccacccaccccggctcctaccatcgcctcccagcc
    tctgtccctgcgtccggaggcatgtagacccgcagctggtggggccgtgcatacccggggtcttg
    acttcgcctgcgatatctacatttgggcccctctggctggtacttgcggggtcctgctgctttca
    ctcgtgatcactctttactgtaagcgcggtcggaagaagctgctgtacatctttaagcaaccctt
    catgaggcctgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg
    aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctacaagcagggg
    cagaaccagctctacaacgaactcaatcttggtcggagagaggagtacgacgtgctggacaagcg
    gagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatccccaagagggcctgtaca
    acgagctccaaaaggataagatggcagaagcctatagcgagattggtatgaaaggggaacgcaga
    agaggcaaaggccacgacggactgtaccagggactcagcaccgccaccaaggacacctatgacgc
    tcttcacatgcaggccctgccgcctcgg (SEQ ID NO: 78)
    105977 MALPVTALLLPLALLLHAARPEIVMTQSPATLSLSPGERATLSC RASQDISKYLN WYQQKPGQAP
    CAR 12- RLLIY HTSRLHS GIPARFSGSGSGTDYTLTISSLQPEDFAVYFC QQGNTLPYT FGQGTKLEIKGG
    Full-aa GGSGGGGSGGGGSQVQLQESGPGLVKPSETLSLTCTVSGVSLP DYGVS WIRQPPGKGLEWIG VIW
    GSETTYYNSSLKS RVTISKDNSKNQVSLKLSSVTAADTAVYYCAK HYYYGGSYAMDY WGQGTLVT
    VSSTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLS
    LVITLYCKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYKQG
    QNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERR
    RGKGHDGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NO: 79)
    CTL019
    CTL019 atggccttaccagtgaccgccttgctcctgccgctggccttgctgctccacgccgccaggccgga
    Full-nt catccagatgacacagactacatcctccctgtctgcctctctgggagacagagtcaccatcagtt
    gcagggcaagtcaggacattagtaaatatttaaattggtatcagcagaaaccagatggaactgtt
    aaactcctgatctaccatacatcaagattacactcaggagtcccatcaaggttcagtggcagtgg
    gtctggaacagattattctctcaccattagcaacctggagcaagaagatattgccacttactttt
    gccaacagggtaatacgcttccgtacacgttcggaggggggaccaagctggagatcacaggtggc
    ggtggctcgggcggtggtgggtcgggtggcggcggatctgaggtgaaactgcaggagtcaggacc
    tggcctggtggcgccctcacagagcctgtccgtcacatgcactgtctcaggggtctcattacccg
    actatggtgtaagctggattcgccagcctccacgaaagggtctggagtggctgggagtaatatgg
    ggtagtgaaaccacatactataattcagctctcaaatccagactgaccatcatcaaggacaactc
    caagagccaagttttcttaaaaatgaacagtctgcaaactgatgacacagccatttactactgtg
    ccaaacattattactacggtggtagctatgctatggactactggggccaaggaacctcagtcacc
    gtctcctcaaccacgacgccagcgccgcgaccaccaacaccggcgcccaccatcgcgtcgcagcc
    cctgtccctgcgcccagaggcgtgccggccagcggcggggggcgcagtgcacacgagggggctgg
    acttcgcctgtgatatctacatctgggcgcccttggccgggacttgtggggtccttctcctgtca
    ctggttatcaccctttactgcaaacggggcagaaagaaactcctgtatatattcaaacaaccatt
    tatgagaccagtacaaactactcaagaggaagatggctgtagctgccgatttccagaagaagaag
    aaggaggatgtgaactgagagtgaagttcagcaggagcgcagacgcccccgcgtacaagcagggc
    cagaaccagctctataacgagctcaatctaggacgaagagaggagtacgatgttttggacaagag
    acgtggccgggaccctgagatggggggaaagccgagaaggaagaaccctcaggaaggcctgtaca
    atgaactgcagaaagataagatggcggaggcctacagtgagattgggatgaaaggcgagcgccgg
    aggggcaaggggcacgatggcctttaccagggtctcagtacagccaccaaggacacctacgacgc
    ccttcacatgcaggccctgccccctcgc (SEQ ID NO: 80)
    CTL019 MALPVTALLLPLALLLHAARPdiqmtqttsslsaslgdrvtiscrasqdiskylnwyqqkpdgtv
    Full-aa klliyhtsrlhsgvpsrfsgsgsgtdysltisnleqediatyfcqqgntlpytfgggtkleitgg
    ggsggggsggggsevklqesgpglvapsqslsvtctvsgvslpdygvswirqpprkglewlgviw
    gsettyynsalksrltiikdnsksqvflkmnslqtddtaiyycakhyyyggsyamdywgqgtsvt
    vsstttpaprpptpaptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvllls
    lvitlyckrgrkkllyifkgpfmrpvqttqeedgcscrfpeeeeggcelrvkfsrsadapaykqg
    qnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeayseigmkgerr
    rgkghdglyqglstatkdtydalhmgalppr (SEQ ID NO: 81)

    CD19 Associated Diseases and/or Disorders
  • In one aspect, the disclosure provides methods for treating cancer, e.g., a cancer associated with CD19 expression, with a CAR-expressing cell (e.g., T cell, NK cell) therapy. Exemplary cancers include, but are not limited to e.g., one or more acute leukemias including but not limited to, e.g., B-cell acute lymphocytic leukemia (“B-ALL”), T-cell acute lymphocytic leukemia (“T-ALL”), acute lymphocytic leukemia (ALL); one or more chronic leukemias including but not limited to, e.g., chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL). Additional cancers or hematologic conditions associated with expression of CD19 include, but are not limited to, e.g., B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, and “preleukemia” which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells, and the like. Further, a disease associated with CD19 expression include, but not limited to, e.g., atypical and/or non-classical cancers, malignancies, precancerous conditions or proliferative diseases associated with expression of CD19.
  • In one embodiment, the disclosure provides methods for treating CLL.
  • In another embodiment, the disclosure provides methods for treating ALL.
  • In another embodiment, the disclosure provides methods for treating B-cell ALL.
  • In one aspect, the disclosure provides methods of treating a responder (e.g., a complete responder and partial responder) having cancer (e.g., a hematological cancer such as ALL and CLL) with a CAR-expressing cell (e.g., T cell, NK cell) (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019). In an embodiment, the disclosure provides methods of treating a responder (e.g., a complete responder and partial responder) with a CAR-expressing cell (e.g., T cell, NK cell) in combination with another therapeutic agent, e.g., another therapeutic agent described herein (e.g., another CAR, e.g., another CAR described herein, an inhibitory CAR, e.g., an inhibitory CAR described herein, a kinase inhibitor (e.g., a kinase inhibitor described herein, e.g., an mTOR inhibitor, a BTK inhibitor), a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein, a standard of care therapy, etc.). The combination can be, e.g., with any agent described herein. In an embodiment, after a CAR-expressing cell (e.g., T cell, NK cell) treatment, e.g., an initial CAR-expressing cell (e.g., T cell, NK cell) treatment, a partial responder is tested by any one of the methods described herein, such as, e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene set signature, and if status has not changed and/or is down-graded to, e.g., a non-responder, then the subject is administered an alternative therapy, e.g., a standard of care for the particular cancer.
  • In one aspect, the disclosure provides methods of treating a non-responder having cancer (e.g., a hematological cancer such as ALL and CLL) with a CAR-expressing cell (e.g., T cell, NK cell) (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019). In an embodiment, the disclosure provides methods of treating a non-responder with a CAR-expressing cell (e.g., T cell, NK cell) in combination with another therapeutic agent, e.g., another therapeutic agent described herein (e.g., another CAR, e.g., another CAR described herein, an inhibitory CAR, e.g., an inhibitory CAR described herein, a kinase inhibitor (e.g., a kinase inhibitor described herein, e.g., an mTOR inhibitor, a BTK inhibitor), a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein, a standard of care therapy, etc.). The combination can be, e.g., with any agent described herein. In an embodiment, after a CAR-expressing cell (e.g., T cell, NK cell) treatment, e.g., an initial CAR-expressing cell (e.g., T cell, NK cell) treatment, a non-responder is tested by any one of the methods described herein, such as, e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene set signature, and if status has changed and/or is up-graded to, e.g., a partial-responder, e.g., a complete responder, then the subject is administered an alternative therapy described herein.
  • In an embodiment, the disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL or CLL) comprising steps of: (1) identifying a partial responder subject and/or non-responder subject, (2) administering to the partial responder subject and/or non-responder subject an mTOR inhibitor described herein, such as, e.g., RAD001 and rapamycin, e.g., at a dose and/or dosing schedule described herein; and (3) administering a CAR (e.g., a CD19 CAR described herein, such as, e.g., CTL019), e.g., subsequent to the administration of the mTOR inhibitor, thus treating the cancer. In an embodiment, the method further includes administering the mTOR inhibitor and/or the CAR in combination with one or more checkpoint inhibitors described here, such as, e.g., a PD1inhibitor.
  • In an embodiment, the disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL or CLL) comprising steps of: (1) identifying a partial responder subject (e.g., patient) and/or non-responder subject (e.g., patient), (2) enriching the T cell population of the partial responder subject and/or non-responder subject by selecting for a less exhausted and/or more naïve T cell population, (3) introducing (e.g., by transforming, transducing, infecting, electroporating, etc.) a CAR (e.g., a CD19 CAR described herein, such as, e.g., CTL019) into said enriched T cell population thus transforming the subject's T cell population; and (4) administering the CAR-expressing T cell population into the partial responder subject and/or non-responder subject, thus treating the cancer.
  • In an embodiment, the disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL or CLL) comprising steps of: (1) identifying a partial responder subject (e.g., patient) and/or non-responder subject (e.g., patient), (2) reevaluating a partial responder subject and/or non-responder subject (e.g., patient) at a later time period for naïve T cells and/or less exhausted phenotype, and (3), e.g., if the subject has an increase in naïve T cells and/or a less exhausted phenotype, administering a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) as described herein, such as, e.g., CTL019), thus treating the cancer. In an embodiment, a later time period comprises at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 5 hours, at least 6 hours, at least 7 hours, at least 8 hours, at least 9 hours, at least 10 hours, at least 11 hours, at least 12 hours, at least 1 day, at least 2 days, at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least 1 week, at least 2 weeks, at least 3 weeks, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months at least 8 months, at least 9 months, at least 10 months, at least 11 months, at least 1 year, at least 2 years, at least 3 years, at least 4 years, at least 5 years, at least 6 years, at least 7 years, at least 8 years, at least 9 years, at least 10 years, or 11 years or more.
  • In an embodiment, the disclosure provides methods of treating cancer (e.g., a hematological cancer such as ALL or CLL) comprising the steps of (1) identifying partial responders and/or non-responders; and (2) treating with an alternative therapy, e.g., a standard of care for the particular cancer (e.g., the standard of care for ALL or CLL). In an embodiment, a partial responder is treated only with the standard of care (e.g., the standard of care for a hematological cancer such as ALL or CLL) in the absence of treatment with a CAR. In an embodiment, a non-responder is treated only with the standard of care (e.g., the standard of care for a hematological cancer such as ALL or CLL) in the absence of treatment with a CAR.
  • In an embodiment, standard of care for CLL includes, but is not limited to exemplary therapies described herein, e.g., described in Table A, and combinations thereof.
  • TABLE A
    Exemplary therapies for CLL
    w/o del
    (11q) or del del
    del(17p) (17p) (11q)
    First line ≥70 yrs with comorbidities
    Obinutuzumab + chlorambucil X X X
    Rituxan + chlorambucil X X
    Rituxan X
    Chlorambucil X
    Fludarabine ± Rituxan X X
    Cladribine X
    Bendamustine ± Rituxan X X
    PCR (pentostatin, cyclophosphamide, Rituxan) X X
    First Line <70 years without significant comorbidities
    FCR (Fludarabine, cyclophosphamide, Rituxan) X X X
    FR (Fludarabine, Rituxan) X X
    PCR X X
    Bendamustine ± Rituxan X X
    Obinutuzumab + chlorambucil X X X
    Second line- Relapsed/Refractory ≥70 years
    Imbruvica X X X
    Reduced-dose FCR X X
    Reduced-dose PCRR X X
    Bendamustine ± Rituxan X X
    Ofatumumab X X X
    Alemutuzumab + Rituxan X X X
    High dose methylprednisone (HDMP) + rituximab X X X
    Lenalidomide + Rituxan X X X
    Dose dense rituximab X X
    Second line- Relapsed/Refractory < years without significant comorbiditites
    Imbruvica X X X
    FCR (Fludarabine, cyclophosphaide, Rituxan) X X
    PCR X X
    Bendamustine ± Rituxan X X
    Fludarabine + alemtuzumab X X
    R-CHOP (Rituxan, cyclophosphamide, X X X
    dosorubicin, vincristine, prednisone)
    Ofatumumab X X X
    OFAR (oxaliplatin, Fludara, cytarabine, Rituxan) X X X
    HDMP + rituximab X X X
    Lenalidomide + Rituxan X X X
  • In an embodiment, standard of care for CLL includes (1) radiation therapy, (2) chemotherapy, (3) surgery (e.g., removal of the spleen), (4) targeted therapy, (5) stem cell transplantation, and combinations thereof. In an embodiment, the standard of care comprises external radiation therapy. In an embodiment, the standard of care comprises internal radiation therapy (e.g., a radioactive substance sealed in needles, wires or catheters, for example, that are placed directly into or near the cancer).
  • In an embodiment, standard of care for ALL includes, but is not limited to exemplary therapies described herein, e.g., described in Table B, and combinations thereof.
  • TABLE B
    Exemplary therapies for ALL
    First Line
    RCHOP (Rituxan, cyclophosphamide, doxorubicin, vincristine,
    prednisone)
    Dose dense RCHOP 14 (category 3)
    Dose adjusted EPOCH (etoposide, prednisone, vincristine,
    cyclophosphamide, doxorubicin) + Rituxan
    First Line Therapy for subjects with Poor left
    ventricular function or very frail
    RCEPP (rituximab, cyclophosphamide, etoposide, prednisone,
    procarbazine)
    RCEOP (rituximab, cyclophosphamide, etoposide, vincristine,
    prednisone)
    RCNOP (rituximab, cyclophosphamide, mitoxantrone, vincristine,
    prednisone)
    RCEOP (rituximab, cyclophosphamide, etoposide, vincristine, prednisone)
    Dose adjusted EPOCH (etoposide, prednisone, vincristine,
    cyclophosphamide, doxorubicin) + Rituxan
    Second line- proceed to high dose therapy with autologous
    stem cell rescue
    DHAP (dexamethasone, cisplatin, cytarabine) ± Rituxan
    ESHAP (etoposide, methylprednisolone, cytarabine, cisplatin) ± Rituxan
    GDP (gemcitabine, dexamethasone, cisplatin) ± Rituxan
    GemOx(gemcitabine, oxaliplatin) ± Rituxan
    ICE (ifosfamide, carboplatin, etoposide) + Rituxan
    MINE (mesna, ifosfamide, mitoxantrone, etoposide) ± Rituxan
    Second-line therapy (non-candidates for high-dose therapy)
    CEPP (cyclophosphamide, etoposide, prednisone, procarbazine) ± Rituxan
    CEOP (cyclophosphamide, etoposide, vincristine, prednisone) ± Rituxan
    DA-EPOCH ± Rituxan
    Revlimid ± Rituxan
    Rituxan
    GemOx ± Rituxan
    GDP ± Rituxan
    Bendamustine + Rituxan
  • In an embodiment, standard of care for ALL includes (1) chemotherapy, (2) radiation therapy, (3) stem cell transplantation, (4) biological therapy, (5) targeted therapy, and combinations thereof.
  • In an embodiment, the standard of care includes, but is not limited to, fludarabine with cyclophosphamide (FC); fludarabine with rituximab (FR); fludarabine, cyclophosphamide, and rituximab (FCR); cyclophosphamide, doxorubicin, vincristine and prednisone (CHOP); and combinations thereof. General chemotherapeutic agents considered for use include, but are not limited to anastrozole (Arimidex®), bicalutamide (Casodex®), bleomycin sulfate (Blenoxane®), busulfan (Myleran®), busulfan injection (Busulfex®), capecitabine (Xeloda®), N4-pentoxycarbonyl-5-deoxy-5-fluorocytidine, carboplatin (Paraplatin®), carmustine (BiCNU®), chlorambucil (Leukeran®), cisplatin (Platinol®), cladribine (Leustatin®), cyclophosphamide (Cytoxan® or Neosar®), cytarabine, cytosine arabinoside (Cytosar-U®), cytarabine liposome injection (DepoCyt®), dacarbazine (DTIC-Dome®), dactinomycin (Actinomycin D, Cosmegan), daunorubicin hydrochloride (Cerubidine®), daunorubicin citrate liposome injection (DaunoXome®), dexamethasone, docetaxel (Taxotere®), doxorubicin hydrochloride (Adriamycin®, Rubex®), etoposide (Vepesid®), fludarabine phosphate (Fludara®), 5-fluorouracil (Adrucil®, Efudex®), flutamide (Eulexin®), tezacitibine, Gemcitabine (difluorodeoxycitidine), hydroxyurea (Hydrea®), Idarubicin (Idamycin®), ifosfamide (IFEX®), irinotecan (Camptosar®), L-asparaginase (ELSPAR®), leucovorin calcium, melphalan (Alkeran®), 6-mercaptopurine (Purinethol®), methotrexate (Folex®), mitoxantrone (Novantrone®), mylotarg, paclitaxel (Taxol®), phoenix (Yttrium90/MX-DTPA), pentostatin, polifeprosan 20 with carmustine implant (Gliadel®), tamoxifen citrate (Nolvadex®), teniposide (Vumon®), 6-thioguanine, thiotepa, tirapazamine (Tirazone®), topotecan hydrochloride for injection (Hycamptin®), vinblastine (Velban®), vincristine (Oncovin®), vinorelbine (Navelbine®), and combinations thereof.
  • In an embodiment, chemotherapy comprises an antimetabolite, including, but not limited to, folic acid antagonists (also referred to herein as antifolates), pyrimidine analogs, purine analogs and adenosine deaminase inhibitors): methotrexate (Rheumatrex®, Trexall®), 5-fluorouracil (Adrucil®, Efudex®, Fluoroplex®), floxuridine (FUDF®), cytarabine (Cytosar-U®, Tarabine PFS), 6-mercaptopurine (Puri-Nethol®)), 6-thioguanine (Thioguanine Tabloid®), fludarabine phosphate (Fludara®), pentostatin (Nipent®), pemetrexed (Alimta®), raltitrexed (Tomudex®), cladribine (Leustatin®), clofarabine (Clofarex®, Clolar®), cytarabine liposomal (also known as Liposomal Ara-C, DepoCyt™); decitabine (Dacogen®); hydroxyurea (Hydrea®, Droxia™ and Mylocel™); mercaptopurine (Puri-Nethol®), pralatrexate (Folotyn™) capecitabine (Xeloda®), nelarabine (Arranon®), azacitidine (Vidaza®) and gemcitabine (Gemzar®). Suitable antimetabolites include, e.g., 5-fluorouracil (Adrucil®, Efudex®, Fluoroplex®), floxuridine (FUDF®), capecitabine (Xeloda®), pemetrexed (Alimta®), raltitrexed (Tomudex®) and gemcitabine (Gemzar®), and combinations thereof. In an embodiment, the purine analogue is fludarabine.
  • In an embodiment, chemotherapy comprises an alkylating agent including, but not limited to nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes, uracil mustard (Aminouracil Mustard®, Chlorethaminacil®, Demethyldopan®, Desmethyldopan®, Haemanthamine®, Nordopan®, Uracil nitrogen Mustard®, Uracillost®, Uracilmostaza®, Uramustin®, Uramustine®), chlormethine (Mustargen®), cyclophosphamide (Cytoxan®, Neosar®, Clafen®, Endoxan®, Procytox®, Revimmune™), ifosfamide (Mitoxana®), melphalan (Alkeran®), Chlorambucil (Leukeran®), pipobroman (Amedel®, Vercyte®), triethylenemelamine (Hemel®, Hexalen®, Hexastat®), triethylenethiophosphoramine, Temozolomide (Temodar®), thiotepa (Thioplex®), busulfan (Busilvex®, Myleran®), carmustine (BiCNU®), lomustine (CeeNU®), streptozocin (Zanosar®), and Dacarbazine (DTIC-Dome®) and combinations thereof. Additional exemplary alkylating agents include, without limitation, Oxaliplatin (Eloxatin®); Temozolomide (Temodar® and Temodal®); Dactinomycin (also known as actinomycin-D, Cosmegen®); Melphalan (also known as L-PAM, L-sarcolysin, and phenylalanine mustard, Alkeran®); Altretamine (also known as hexamethylmelamine (HMM), Hexalen®); Carmustine (BiCNU®); Bendamustine (Treanda®); Busulfan (Busulfex® and Myleran®); Carboplatin (Paraplatin®); Lomustine (also known as CCNU, CeeNU®); Cisplatin (also known as CDDP, Platinol® and Platinol®-AQ); Chlorambucil (Leukeran®); Cyclophosphamide (Cytoxan® and Neosar®); Dacarbazine (also known as DTIC, DIC and imidazole carboxamide, DTIC-Dome®); Altretamine (also known as hexamethylmelamine (HMM), Hexalen®); Ifosfamide (Ifex®); Prednumustine; Procarbazine (Matulane®); Mechlorethamine (also known as nitrogen mustard, mustine and mechloroethamine hydrochloride, Mustargen®); Streptozocin (Zanosar®); Thiotepa (also known as thiophosphoamide, TESPA and TSPA, Thioplex®); Cyclophosphamide (Endoxan®, Cytoxan®, Neosar®, Procytox®, Revimmune®); Bendamustine HCl (Treanda®) and combinations thereof. In an embodiment, the alkylating agent is bendamustine. In an embodiment, the alkylating agent is cyclophosphamide.
  • In an embodiment, the chemotherapeutic agent is a kinase inhibitor, e.g., a tyrosine kinase inhibitor including, but not limited to, erlotinib hydrochloride (Tarceva®); linifanib (N-[4-(3-amino-1H-indazol-4-yl)phenyl]-N-2-fluoro-5-methylphenyl)urea, also known as ABT 869, available from Genentech); sunitinib malate (Sutent®); bosutinib (4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-methoxy-7-[3-(4-methylpiperazin-1-yl)propoxy]quinoline-3-carbonitrile, also known as SKI-606, and described in U.S. Pat. No. 6,780,996); dasatinib (Sprycel®); pazopanib (Votrient®); sorafenib (Nexavar®); zactima (ZD6474); and imatinib or imatinib mesylate (Gilvec® and Gleevec®). In one embodiment, the kinase inhibitor is a BTK inhibitor selected from ibrutinib (PCI-32765); GDC-0834; RN-486; CGI-560; CGI-1764; HM-71224; CC-292; ONO-4059; CNX-774; and LFM-A13. In one embodiment, the kinase inhibitor is a CDK4 inhibitor selected from aloisine A; flavopiridol or HMR-1275, 2-(2-chlorophenyl)-5,7-dihydroxy-8-[(3S,4R)-3-hydroxy-1-methyl-4-piperidinyl]-4-chromenone; crizotinib (PF-02341066; 2-(2-Chlorophenyl)-5,7-dihydroxy-8-[(2R,3S)-2-(hydroxymethyl)-1-methyl-3-pyrrolidinyl]-4H-1-benzopyran-4-one, hydrochloride (P276-00); 1-methyl-5-[[2-[5-(trifluoromethyl)-1H-imidazol-2-yl]-4-pyridinyl]oxy]-N-[4-(trifluoromethyl)phenyl]-1H-benzimidazol-2-amine (RAF265); indisulam (E7070); roscovitine (CYC202); palbociclib (PD0332991); dinaciclib (SCH727965); N-[5-[[(5-tert-butyloxazol-2-yl)methyl]thio]thiazol-2-yl]piperidine-4-carboxamide (BMS 387032); 4-[[9-chloro-7-(2,6-difluorophenyl)-5H-1.5 pyrimido[5,4-d][2]benzazepin-2-yl]amino]-benzoic acid (MLN8054); 5-[3-(4,6-difluoro-1H-benzimidazol-2-yl)-1H-indazol-5-yl]-N-ethyl-4-methyl-3-pyridinemethanamine (AG-024322); 4-(2,6-dichlorobenzoylamino)-1H-pyrazole-3-carboxylic acid N-(piperidin-4-yl)amide (AT7519); 4-[2-methyl-1-(1-methylethyl)-1H-imidazol-5-yl]-N-[4-(methylsulfonyl)phenyl]-2-pyrimidinamine (AZD5438); and XL281 (BMS908662). In one embodiment, the kinase inhibitor is an MNK inhibitor selected from CGP052088; 4-amino-3-(p-fluorophenylamino)-pyrazolo [3,4-d] pyrimidine (CGP57380); cercosporamide; ETC-1780445-2; and 4-amino-5-(4-fluoroanilino)-pyrazolo [3,4-d] pyrimidine.
  • In an embodiment, targeted therapy includes, but is not limited to an anti-CD20 antibody or functional fragment thereof, such as, e.g., rituximab (Riuxan® and MabThera®); tositumomab (Bexxar®); and ofatumumab (Arzerra®), and combinations thereof. In one embodiment, the targeted therapy includes, but is not limited to, an anti-CD52 antibody or functional fragment thereof such as, e.g., alemtuzumab (Campath®).
  • In an embodiment, biologic therapy comprises immunotherapy. Exemplary anthracyclines include, without limitation, doxorubicin (Adriamycin® and Rubex®); bleomycin (Lenoxane®); daunorubicin (dauorubicin hydrochloride, daunomycin, and rubidomycin hydrochloride, Cerubidine®); daunorubicin liposomal (daunorubicin citrate liposome, DaunoXome®); mitoxantrone (DHAD, Novantrone®); epirubicin (Ellence™); idarubicin (Idamycin®, Idamycin PFS®); mitomycin C (Mutamycin®); geldanamycin; herbimycin; ravidomycin; desacetylravidomycin and combinations thereof.
  • In an embodiment, stem cell transplantation comprises an autogeneic stem cell transplant. In an embodiment, stem cell transplantation comprises an allogenic stem cell transplant. In an embodiment, stem cell transplantation comprises allogeneic bone marrow transplantation. In an embodiment, stem cell transplantation comprises a hematopoietic stem cell transplantation (HSCT). In an embodiment, hematopoietic stem cells are derived from various tissues including, but not limited to bone marrow, peripheral blood, umbilical cord blood, and combinations thereof.
  • In an embodiment, the provided methods comprise determining if the subject is identified as having a statistically significant difference in expression level of one or more markers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15 and Table 16, or a PD-1 biomarker, LAG-3 biomarker, TIM-3 biomarker, CD57 biomarker CD27 biomarker, CD122 biomarker, CD62L biomarker and a KLRG1 biomarker, relative to a reference level, and administering to the subject a therapeutically effective dose of a CAR-expressing cell, e.g., a T cell or NK cell. In an embodiment, a CAR-expressing cell (e.g., T cell, NK cell) is a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein such as, e.g., CTL019.
  • In one aspect, the disclosure provides methods for treating a disease associated with CD19 expression. In one aspect, the invention provides methods for treating a disease wherein part of the tumor is negative for CD19 and part of the tumor is positive for CD19. For example, provided methods are useful for treating subjects that have undergone treatment for a disease associated with elevated expression of CD19, wherein the subject that has undergone treatment for elevated levels of CD19 exhibits a disease associated with elevated levels of CD19.
  • In one aspect, provided methods comprise a vector comprising CD19 CAR operably linked to promoter for expression in mammalian cells (e.g., T cells or NK cells). In one aspect, provided methods comprise a recombinant cell (e.g., T cell or NK cell) expressing a CD19 CAR for use in treating CD19-expressing tumors, wherein the recombinant T cell expressing the CD19 CAR is termed a CD19 CAR-expressing cell. In one aspect, a CD19 CAR-expressing cell (e.g., T cell, NK cell) administered according to provided methods is capable of contacting a tumor cell with at least one CD19 CAR expressed on its surface such that the CAR-expressing cell targets the tumor cell and growth of the tumor is inhibited.
  • In one aspect, the disclosure features to a method of inhibiting growth of a CD19-expressing tumor cell, comprising contacting the tumor cell with a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein such that the CAR-expressing cell is activated in response to the antigen and targets the cancer cell, wherein the growth of the tumor is inhibited.
  • In one aspect, the disclosure includes a type of cellular therapy where T cells are genetically modified to express a CAR and the CAR-expressing cell (e.g., T cell, NK cell) is infused to a recipient in need thereof. The infused cell is able to kill tumor cells in the recipient. Unlike antibody therapies, CAR-modified cells (e.g., T cells or NK cells) are able to replicate in vivo resulting in long-term persistence that can lead to sustained tumor control. In various aspects, the cells administered to the patient, or their progeny, persist in the patient for at least four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, twelve months, thirteen months, fourteen month, fifteen months, sixteen months, seventeen months, eighteen months, nineteen months, twenty months, twenty-one months, twenty-two months, twenty-three months, two years, three years, four years, or five years after administration of the cell to the patient.
  • The disclosure also includes a type of cellular therapy where cells (e.g., T cells, NK cells) are modified, e.g., by in vitro transcribed RNA, to transiently express a chimeric antigen receptor (CAR) and the CAR-expressing cell (e.g., T cell, NK cell) is infused to a recipient in need thereof. The infused cell is able to kill tumor cells in the recipient. Thus, in various aspects, the cells administered to the patient, are present for less than one month, e.g., three weeks, two weeks, one week, after administration of the cell (e.g., T cell, NK cell) to the patient.
  • Without wishing to be bound by any particular theory, the anti-tumor immunity response elicited by the CAR-modified cells (e.g., T cells, NK cells) may be an active or a passive immune response, or alternatively may be due to a direct vs indirect immune response. In one aspect, the CAR transduced T cells exhibit specific proinflammatory cytokine secretion and potent cytolytic activity in response to human cancer cells expressing the CD19, resist soluble CD19 inhibition, mediate bystander killing and mediate regression of an established human tumor. For example, antigen-less tumor cells within a heterogeneous field of CD19-expressing tumor may be susceptible to indirect destruction by CD19-redirected T cells that has previously reacted against adjacent antigen-positive cancer cells.
  • In one aspect, the fully-human CAR-modified cells (e.g., T cells, NK cells) described herein may be a type of vaccine for ex vivo immunization and/or in vivo therapy in a mammal. In one aspect, the mammal is a human.
  • With respect to ex vivo immunization, at least one of the following occurs in vitro prior to administering the cell into a subject: i) expansion of the cells, ii) introducing a nucleic acid encoding a CAR to the cells or iii) cryopreservation of the cells.
  • Ex vivo procedures are well known in the art and are discussed more fully below. Briefly, cells are isolated from a subject (e.g., a human) and genetically modified (i.e., transduced or transfected in vitro) with a vector expressing a CAR disclosed herein. The CAR-modified cell can be administered to a mammalian recipient to provide a therapeutic benefit. The mammalian recipient may be a human and the CAR-modified cell can be autologous with respect to the recipient. Alternatively, the cells can be allogeneic, syngeneic or xenogeneic with respect to the recipient.
  • Hematologic Cancer
  • Hematological cancer conditions are types of cancer such as leukemia and malignant lymphoproliferative conditions that affect blood, bone marrow and the lymphatic system.
  • Leukemia can be classified as acute leukemia and chronic leukemia. Acute leukemia can be further classified as acute myelogenous leukemia (AML) and acute lymphoid leukemia (ALL). Chronic leukemia includes chronic myelogenous leukemia (CML) and chronic lymphoid leukemia (CLL). Other related conditions include myelodysplastic syndromes (MDS, formerly known as “preleukemia”) which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells and risk of transformation to AML.
  • Lymphoma is a group of blood cell tumors that develop from lymphocytes. Exemplary lymphomas include non-Hodgkin lymphoma and Hodgkin lymphoma.
  • The present disclosure provides for compositions and methods for treating cancer. In one aspect, the cancer is a hematologic cancer including but is not limited to a leukemia or a lymphoma. In one aspect, the CAR-expressing cells (e.g., T cells, NK cells) of the invention may be used to treat cancers and malignancies such as, but not limited to, e.g., acute leukemias including but not limited to, e.g., B-cell acute lymphoid leukemia (“B-ALL”), T-cell acute lymphoid leukemia (“T-ALL”), acute lymphoid leukemia (ALL); one or more chronic leukemias including but not limited to, e.g., chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL); additional hematologic cancers or hematologic conditions including, but not limited to, e.g., B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitts lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, and “preleukemia” which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells, and the like. Further a disease associated with CD19 expression includes, but not limited to, e.g., atypical and/or non-classical cancers, malignancies, precancerous conditions or proliferative diseases expressing CD19.
  • The present disclosure also provides methods for inhibiting the proliferation or reducing a CD19-expressing cell population, the methods comprising contacting a population of cells comprising a CD19-expressing cell with a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell. In a specific aspect, the disclosure provides methods for inhibiting the proliferation or reducing the population of cancer cells expressing CD19, the methods comprising contacting the CD19-expressing cancer cell population with a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell. In one aspect, the present disclosure provides methods for inhibiting the proliferation or reducing the population of cancer cells expressing CD19, the methods comprising contacting the CD19-expressing cancer cell population with a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell. In certain aspects, the anti-CD19 CAR-expressing cell (e.g., T cell, NK cell) reduces the quantity, number, amount or percentage of cells and/or cancer cells by at least 25%, at least 30%, at least 40%, at least 50%, at least 65%, at least 75%, at least 85%, at least 95%, or at least 99% in a subject with or animal model for myeloid leukemia or another cancer associated with CD19-expressing cells relative to a negative control. In one aspect, the subject is a human.
  • The present disclosure also provides methods for preventing, treating and/or managing a disease associated with CD19-expressing cells (e.g., a hematologic cancer or atypical cancer expressing CD19), the methods comprising administering to a subject in need a CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell. In one aspect, the subject is a human. Non-limiting examples of disorders associated with CD19-expressing cells include autoimmune disorders (such as lupus), inflammatory disorders (such as allergies and asthma) and cancers (such as hematological cancers or atypical cancers expressing CD19).
  • The present disclosure also provides methods for preventing, treating and/or managing a disease associated with CD19-expressing cells, the methods comprising administering to a subject in need a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell. In one aspect, the subject is a human.
  • The present disclosure provides methods for preventing relapse of cancer associated with CD19-expressing cells (e.g., a hematological cancer such as ALL and CLL), the methods comprising administering to a subject in need thereof a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell. In one aspect, the methods comprise administering to the subject in need thereof an effective amount of a CD19 CAR-expressing cell (e.g., T cell, NK cell) described herein that binds to the CD19-expressing cell in combination with an effective amount of another therapy.
  • Combination Therapy
  • It will be appreciated that any cancer therapy as described above and herein, can be administered in combination with one or more additional therapies to treat and/or reduce the symptoms of cancer described herein. The pharmaceutical compositions can be administered concurrently with, prior to, or subsequent to, one or more other additional therapies or therapeutic agents. In an embodiment, a CAR-expressing cell described herein may be used in combination with other known agents and therapies. Administered “in combination”, as used herein, means that two (or more) different treatments are delivered to the subject during the course of the subject affliction with the disorder, e.g., the two or more treatments are delivered after the subjects has been diagnosed with the disorder and before the disorder has been cured or eliminated or treatment has ceased for other reasons. In some embodiments, the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap in terms of administration. This is sometimes referred to herein as “simultaneous” or “concurrent delivery”. In other embodiments, the delivery of one treatment ends before the delivery of the other treatment begins. In some embodiments of either case, the treatment is more effective because of combined administration. For example, the second treatment is more effective, e.g., an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment or the analogous situation is seen with the first treatment. In some embodiments, delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or greater than additive. The delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered.
  • A CAR-expressing cell described herein and the at least one additional therapeutic agent can be administered simultaneously, in the same or in separate compositions, or sequentially. For sequential administration, the CAR-expressing cell described herein can be administered first, and the additional agent can be administered second, or the order of administration can be reversed.
  • The CAR therapy and/or other therapeutic agents, procedures or modalities can be administered during periods of active disorder, or during a period of remission or less active disease. The CAR therapy can be administered before the other treatment, concurrently with the treatment, post-treatment, or during remission of the disorder.
  • When administered in combination, the CAR therapy and the additional agent (e.g., second or third agent), or all, can be administered in an amount or dose that is higher, lower or the same than the amount or dosage of each agent used individually, e.g., as a monotherapy. In certain embodiments, the administered amount or dosage of the CAR therapy, the additional agent (e.g., second or third agent), or all, is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50%) than the amount or dosage of each agent used individually, e.g., as a monotherapy. In other embodiments, the amount or dosage of the CAR therapy, the additional agent (e.g., second or third agent), or all, that results in a desired effect (e.g., treatment of cancer) is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50% lower) than the amount or dosage of each agent used individually, e.g., as a monotherapy, required to achieve the same therapeutic effect.
  • In further aspects, a CAR-expressing cell described herein may be used in a treatment regimen in combination with surgery, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies or other antibody therapies, cytoxin, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, cytokines, and irradiation peptide vaccine, such as that described in Izumoto et al. 2008 J NEUROSURG 108:963-971.
  • In one embodiment, a CAR-expressing cell described herein can be used in combination with a chemotherapeutic agent. Exemplary chemotherapeutic agents include those described in paragraphs 873-874 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.
  • Exemplary alkylating agents include, without limitation, those described in paragraph 875 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.
  • Exemplary mTOR inhibitors include, without limitation, RAD001, temsirolimus; ridaforolimus (formally known as deferolimus, (1R,2R,4S)-4-[(2R)-2 [(1R,9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28Z,30S,32S,35R)-1,18-dihydroxy-19,30-dimethoxy-15,17,21,23, 29,35-hexamethyl-2,3,10,14,20-pentaoxo-11,36-dioxa-4-azatricyclo[30.3.1.04,9] hexatriaconta-16,24,26,28-tetraen-12-yl]propyl]-2-methoxycyclohexyl dimethylphosphinate, also known as AP23573 and MK8669, and described in PCT Publication No. WO 03/064383); everolimus (Afinitor® or RAD001); rapamycin (AY22989, Sirolimus®); simapimod (CAS 164301-51-3); emsirolimus, (5-{2,4-Bis[(3S)-3-methylmorpholin-4-yl]pyrido[2,3-d]pyrimidin-7-yl}-2-methoxyphenyl)methanol (AZD8055); 2-Amino-8-[trans-4-(2-hydroxyethoxy)cyclohexyl]-6-(6-methoxy-3-pyridinyl)-4-methyl-pyrido[2,3-d]pyrimidin-7(8H)-one (PF04691502, CAS 1013101-36-4); and N2-[1,4-dioxo-4-[[4-(4-oxo-8-phenyl-4H-1-benzopyran-2-yl)morpholinium-4-yl]methoxy]butyl]-arginylglycyl-L-α-aspartylL-serine-(SEQ ID NO: 91), inner salt (SF1126, CAS 936487-67-1), XL765 and combinations thereof.
  • Exemplary immunomodulators include, without limitation, those described in paragraph 882 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.
  • Exemplary anthracyclines include, without limitation, those described in paragraph 883 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.
  • Exemplary vinca alkaloids include, without limitation, those described in paragraph 884 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.
  • Exemplary proteosome inhibitors include, without limitation, those described in paragraph 884 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety, and combinations thereof.
  • In some embodiments, a CAR-expressing cell described herein is administered in combination with an oncolytic virus. In embodiments, oncolytic viruses are capable of selectively replicating in and triggering the death of or slowing the growth of a cancer cell. In some cases, oncolytic viruses have no effect or a minimal effect on non-cancer cells. An oncolytic virus includes but is not limited to an oncolytic adenovirus, oncolytic Herpes Simplex Viruses, oncolytic retrovirus, oncolytic parvovirus, oncolytic vaccinia virus, oncolytic Sinbis virus, oncolytic influenza virus, or oncolytic RNA virus (e.g., oncolytic reovirus, oncolytic Newcastle Disease Virus (NDV), oncolytic measles virus, or oncolytic vesicular stomatitis virus (VSV)).
  • In an embodiment, cells expressing a CAR described herein are administered to a subject in combination with a molecule that decreases the Treg cell population. Methods that decrease the number of (e.g., deplete) Treg cells are known in the art and include, e.g., CD25 depletion, cyclophosphamide administration, modulating GITR function. Without wishing to be bound by theory, it is believed that reducing the number of Treg cells in a subject prior to apheresis or prior to administration of a CAR-expressing cell described herein reduces the number of unwanted immune cells (e.g., Tregs) in the tumor microenvironment and reduces the subject's risk of relapse.
  • In one embodiment, a CAR expressing cell described herein are administered to a subject in combination with a molecule targeting GITR and/or modulating GITR functions, such as a GITR agonist and/or a GITR antibody that depletes regulatory T cells (Tregs). In embodiments, cells expressing a CAR described herein are administered to a subject in combination with cyclophosphamide. In one embodiment, the GITR binding molecules and/or molecules modulating GITR functions (e.g., GITR agonist and/or Treg depleting GITR antibodies) are administered prior to administration of the CAR-expressing cell. For example, in one embodiment, the GITR agonist can be administered prior to apheresis of the cells. In embodiments, cyclophosphamide is administered to the subject prior to administration (e.g., infusion or re-infusion) of the CAR-expressing cell or prior to apheresis of the cells. In embodiments, cyclophosphamide and an anti-GITR antibody are administered to the subject prior to administration (e.g., infusion or re-infusion) of the CAR-expressing cell or prior to apheresis of the cells. In one embodiment, the subject has cancer (e.g., a solid cancer or a hematological cancer such as ALL or CLL). In an embodiment, the subject has CLL. In embodiments, the subject has ALL. In embodiments, the subject has a solid cancer, e.g., a solid cancer described herein. Exemplary GITR agonists include, without limitation, GITR fusion proteins and anti-GITR antibodies (e.g., bivalent anti-GITR antibodies) such as, e.g., a GITR fusion protein described in U.S. Pat. No. 6,111,090, European Patent No.: 090505B1, U.S. Pat. No. 8,586,023, PCT Publication Nos.: WO 2010/003118 and 2011/090754, or an anti-GITR antibody described, e.g., in U.S. Pat. No. 7,025,962, European Patent No.: 1947183B1, U.S. Pat. Nos. 7,812,135, 8,388,967, 8,591,886, European Patent No.: EP 1866339, PCT Publication No.: WO 2011/028683, PCT Publication No.:WO 2013/039954, PCT Publication No.: WO2005/007190, PCT Publication No.: WO 2007/133822, PCT Publication No.: WO2005/055808, PCT Publication No.: WO 99/40196, PCT Publication No.: WO 2001/03720, PCT Publication No.: WO99/20758, PCT Publication No.: WO2006/083289, PCT Publication No.: WO 2005/115451, U.S. Pat. No. 7,618,632, and PCT Publication No.: WO 2011/051726.
  • In an embodiment, a CAR expressing cell described herein, such as, e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell), e.g., CTL019, is administered to a subject, e.g., a subject identified as a partial responder or non-responder, in combination with a GITR agonist, e.g., a GITR agonist described herein. In an embodiment, the GITR agonist is administered prior to the CAR-expressing cell. For example, in an embodiment, the GITR agonist can be administered prior to apheresis of the cells. In an embodiment, the subject has cancer (e.g., a hematological cancer such as ALL and CLL). In an embodiment, the subject has ALL. In an embodiment, the subject has CLL.
  • In an embodiment, a CAR expressing cell described herein, such as, e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell), e.g., CTL019 is administered to a subject, e.g., a subject identified as a partial responder or non-responder, in combination with an mTOR inhibitor, e.g., an mTOR inhibitor described herein, e.g., a target of the rapamycin signaling pathway such as RAD001. In an embodiment, the mTOR inhibitor is administered prior to the CAR-expressing cell. For example, in an embodiment, the mTOR inhibitor can be administered prior to apheresis of the cells. In an embodiment, the subject has cancer (e.g., a hematological cancer such as ALL and CLL). In an embodiment, the subject has ALL. In an embodiment, the subject has CLL.
  • Kinase Inhibitor
  • In one embodiment, a CAR-expressing cell described herein may be used in a treatment regimen in combination with a kinase inhibitor, e.g., a CDK4 inhibitor, a BTK inhibitor, an MNK inhibitor, an mTOR inhibitor, an ITK inhibitor, etc. In one embodiment, the subject is a complete responder, and the subject is administered a treatment regimen that includes administration of a CAR-expressing cell described herein in combination with a kinase inhibitor, e.g., a kinase inhibitor described herein, e.g., at a dose or dosing schedule described herein. In one embodiment, the subject is a partial responder or a non-responder, and the subject is administered a treatment regimen that includes administration of a CAR-expressing cell described herein in combination with a kinase inhibitor, e.g., a kinase inhibitor described herein, e.g., at a dose or dosing schedule described herein.
  • In an embodiment, the kinase inhibitor is a CDK4 inhibitor, e.g., a CDK4 inhibitor described herein, e.g., a CDK4/6 inhibitor, such as, e.g., 6-Acetyl-8-cyclopentyl-5-methyl-2-(5-piperazin-1-yl-pyridin-2-ylamino)-8H-pyrido[2,3-d]pyrimidin-7-one, hydrochloride (also referred to as palbociclib or PD0332991). In one embodiment, the kinase inhibitor is a BTK inhibitor, e.g., a BTK inhibitor described herein, such as, e.g., ibrutinib. In one embodiment, the kinase inhibitor is an mTOR inhibitor, e.g., an mTOR inhibitor described herein, such as, e.g., rapamycin, a rapamycin analog, OSI-027. The mTOR inhibitor can be, e.g., an mTORC1 inhibitor and/or an mTORC2 inhibitor, e.g., an mTORC1 inhibitor and/or mTORC2 inhibitor described herein. In one embodiment, the kinase inhibitor is a MNK inhibitor, e.g., a MNK inhibitor described herein, such as, e.g., 4-amino-5-(4-fluoroanilino)-pyrazolo [3,4-d] pyrimidine. The MNK inhibitor can be, e.g., a MNKla, MNK1b, MNK2a and/or MNK2b inhibitor.
  • In one embodiment, the kinase inhibitor is a CDK4 inhibitor selected from aloisine A; flavopiridol or HMR-1275, 2-(2-chlorophenyl)-5,7-dihydroxy-8-[(3S,4R)-3-hydroxy-1-methyl-4-piperidinyl]-4-chromenone; crizotinib (PF-02341066; 2-(2-Chlorophenyl)-5,7-dihydroxy-8-[(2R,3S)-2-(hydroxymethyl)-1-methyl-3-pyrrolidinyl]-4H-1-benzopyran-4-one, hydrochloride (P276-00); 1-methyl-5-[[2-[5-(trifluoromethyl)-1H-imidazol-2-yl]-4-pyridinyl]oxy]-N-[4-(trifluoromethyl)phenyl]-1H-benzimidazol-2-amine (RAF265); indisulam (E7070); roscovitine (CYC202); palbociclib (PD0332991); dinaciclib (SCH727965); N-[5-[[(5-tert-butyloxazol-2-yl)methyl]thio]thiazol-2-yl]piperidine-4-carboxamide (BMS 387032); 4-[[9-chloro-7-(2,6-difluorophenyl)-5H-pyrimido[5,4-d][2]benzazepin-2-yl]amino]-benzoic acid (MLN8054); 5-[3-(4,6-difluoro-1H-benzimidazol-2-yl)-1H-indazol-5-yl]-N-ethyl-4-methyl-3-pyridinemethanamine (AG-024322); 4-(2,6-dichlorobenzoylamino)-1H-pyrazole-3-carboxylic acid N-(piperidin-4-yl)amide (AT7519); 4-[2-methyl-1-(1-methylethyl)-1H-imidazol-5-yl]-N-[4-(methylsulfonyl)phenyl]-2-pyrimidinamine (AZD5438); and XL281 (BMS908662).
  • In one embodiment, the kinase inhibitor is a CDK4 inhibitor, e.g., palbociclib (PD0332991), and the palbociclib is administered at a dose of about 50 mg, 60 mg, 70 mg, 75 mg, 80 mg, 90 mg, 100 mg, 105 mg, 110 mg, 115 mg, 120 mg, 125 mg, 130 mg, 135 mg (e.g., 75 mg, 100 mg or 125 mg) daily for a period of time, e.g., daily for 14-21 days of a 28 day cycle, or daily for 7-12 days of a 21 day cycle. In one embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more cycles of palbociclib are administered.
  • In one embodiment, the kinase inhibitor is a BTK inhibitor selected from ibrutinib (PCI-32765); GDC-0834; RN-486; CGI-560; CGI-1764; HM-71224; CC-292; ONO-4059; CNX-774; and LFM-A13.
  • In one embodiment, the kinase inhibitor is a BTK inhibitor, e.g., ibrutinib (PCI-32765), and the ibrutinib is administered at a dose of about 250 mg, 300 mg, 350 mg, 400 mg, 420 mg, 440 mg, 460 mg, 480 mg, 500 mg, 520 mg, 540 mg, 560 mg, 580 mg, 600 mg (e.g., 250 mg, 420 mg or 560 mg) daily for a period of time, e.g., daily for 21 day cycle, or daily for 28 day cycle. In one embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more cycles of ibrutinib are administered.
  • In one embodiment, the kinase inhibitor is an mTOR inhibitor selected from temsirolimus; ridaforolimus (1R,2R,4S)-4-[(2R)-2 [(1R,9S,12S,15R,16E,18R,19R,21R, 23S,24E,26E,28Z,30S,32S,35R)-1,18-dihydroxy-19,30-dimethoxy-15,17,21,23, 29,35-hexamethyl-2,3,10,14,20-pentaoxo-11,36-dioxa-4-azatricyclo[30.3.1.04,9] hexatriaconta-16,24,26,28-tetraen-12-yl]propyl]-2-methoxycyclohexyl dimethylphosphinate, also known as AP23573 and MK8669; everolimus (RAD001); rapamycin (AY22989); simapimod; (5-{2,4-bis[(3S)-3-methylmorpholin-4-yl]pyrido[2,3-d]pyrimidin-7-yl}-2-methoxyphenyl)methanol (AZD8055); 2-mmino-8-[trans-4-(2-hydroxyethoxy)cyclohexyl]-6-(6-methoxy-3-pyridinyl)-4-methyl-pyrido[2,3-d]pyrimidin-7(8H)-one (PF04691502); and N2-[1,4-dioxo-4-[[4-(4-oxo-8-phenyl-4H-1-benzopyran-2-yl)morpholinium-4-yl]methoxy]butyl]L-arginylglycyl-L-α-aspartylL-serine-, inner salt (SF1126); and XL765.
  • In one embodiment, the kinase inhibitor is an mTOR inhibitor, e.g., rapamycin, and the rapamycin is administered at a dose of about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg (e.g., 6 mg) daily for a period of time, e.g., daily for 21 day cycle, or daily for 28 day cycle. In one embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more cycles of rapamycin are administered. In one embodiment, the kinase inhibitor is an mTOR inhibitor, e.g., everolimus and the everolimus is administered at a dose of about 2 mg, 2.5 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg (e.g., 10 mg) daily for a period of time, e.g., daily for 28 day cycle. In one embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more cycles of everolimus are administered.
  • In one embodiment, the kinase inhibitor is an MNK inhibitor selected from CGP052088; 4-amino-3-(p-fluorophenylamino)-pyrazolo [3,4-d] pyrimidine (CGP57380); cercosporamide; ETC-1780445-2; and 4-amino-5-(4-fluoroanilino)-pyrazolo [3,4-d] pyrimidine.
  • In some embodiments of the methods, uses, and compositions herein, the BTK inhibitor is a BTK inhibitor described in International Application WO/2015/079417, which is herein incorporated by reference in its entirety. For instance, in some embodiments, the BTK inhibitor is a compound of formula (I) or a pharmaceutically acceptable salt thereof;
  • Figure US20210172020A1-20210610-C00001
  • wherein,
  • R1 is hydrogen, C1-C6 alkyl optionally substituted by hydroxy;
  • R2 is hydrogen or halogen;
  • R3 is hydrogen or halogen;
  • R4 is hydrogen;
  • R5 is hydrogen or halogen;
  • or R4 and R5 are attached to each other and stand for a bond, —CH2-, —CH2-CH2-, —CH═CH—, —CH═CH—CH2-; —CH2-CH═CH—; or —CH2-CH2-CH2-;
  • R6 and R7 stand independently from each other for H, C1-C6 alkyl optionally substituted by hydroxyl, C3-C6 cycloalkyl optionally substituted by halogen or hydroxy, or halogen;
  • R8, R9, R, R′, R10 and R11 independently from each other stand for H, or C1-C6 alkyl optionally substituted by C1-C6 alkoxy; or any two of R8, R9, R, R′, R10 and R11 together with the carbon atom to which they are bound may form a 3-6 membered saturated carbocyclic ring;
  • R12 is hydrogen or C1-C6 alkyl optionally substituted by halogen or C1-C6 alkoxy;
  • or R12 and any one of R8, R9, R, R′, R10 or R11 together with the atoms to which they are bound may form a 4, 5, 6 or 7 membered azacyclic ring, which ring may optionally be substituted by halogen, cyano, hydroxyl, C1-C6 alkyl or C1-C6 alkoxy;
  • n is 0 or 1; and
  • R13 is C2-C6 alkenyl optionally substituted by C1-C6 alkyl, C1-C6 alkoxy or N,N-di-C1-C6 alkyl amino; C2-C6 alkynyl optionally substituted by C1-C6 alkyl or C1-C6 alkoxy; or C2-C6 alkylenyl oxide optionally substituted by C1-C6 alkyl.
  • Low, Immune Enhancing, Dose of an mTOR Inhibitor
  • Methods described herein can use a low, immune enhancing, dose of an mTOR inhibitor e.g., an allosteric mTOR inhibitor, including rapalogs such as RAD001. Administration of a low, immune enhancing, dose of an mTOR inhibitor (e.g., a dose that is insufficient to completely suppress the immune system, but sufficient to improve immune function) can optimize the performance of immune effector cells, e.g., T cells or CAR-expressing cells, in the subject. Methods for measuring mTOR inhibition, dosages, treatment regimens, and suitable pharmaceutical compositions are described in U.S. Patent Application No. 2015/0140036, filed Nov. 13, 2014, hereby incorporated by reference.
  • In an embodiment, administration of a low, immune enhancing, dose of an mTOR inhibitor can result in one or more of the following:
      • i) a decrease in the number of PD-1 positive immune effector cells;
      • ii) an increase in the number of PD-1 negative immune effector cells;
      • iii) an increase in the ratio of PD-1 negative immune effector cells/PD-1 positive immune effector cells;
      • iv) an increase in the number of naive T cells;
      • v) an increase in the expression of one or more of the following markers: CD62Lhigh, CD127high, CD27+, and BCL2, e.g., on memory T cells, e.g., memory T cell precursors;
      • vi) a decrease in the expression of KLRG1, e.g., on memory T cells, e.g., memory T cell precursors; or
      • vii) an increase in the number of memory T cell precursors, e.g., cells with any one or combination of the following characteristics: increased CD62Lhigh, increased CD127high, increased CD27+, decreased KLRG1, and increased BCL2;
        and wherein any of the foregoing, e.g., i), ii), iii), iv), v), vi), or vii), occurs e.g., at least transiently, e.g., as compared to a non-treated subject.
  • In another embodiment, administration of a low, immune enhancing, dose of an mTOR inhibitor results in increased or prolonged proliferation of CAR-expressing cells, e.g., in culture or in a subject, e.g., as compared to non-treated CAR-expressing cells or a non-treated subject. In embodiments, increased proliferation is associated with in an increase in the number of CAR-expressing cells. In another embodiment, administration of a low, immune enhancing, dose of an mTOR inhibitor results in increased killing of cancer cells by CAR-expressing cells, e.g., in culture or in a subject, e.g., as compared to non-treated CAR-expressing cells or a non-treated subject. In embodiments, increased killing of cancer cells is associated with in a decrease in tumor volume.
  • In one embodiment, the cells expressing a CAR molecule, e.g., a CAR molecule described herein, are administered in combination with a low, immune enhancing dose of an mTOR inhibitor, e.g., an allosteric mTOR inhibitor, e.g., RAD001, or a catalytic mTOR inhibitor. For example, administration of the low, immune enhancing, dose of the mTOR inhibitor can be initiated prior to administration of a CAR-expressing cell described herein; completed prior to administration of a CAR-expressing cell described herein; initiated at the same time as administration of a CAR-expressing cell described herein; overlapping with administration of a CAR-expressing cell described herein; or continuing after administration of a CAR-expressing cell described herein.
  • Alternatively or in addition, administration of a low, immune enhancing, dose of an mTOR inhibitor can optimize immune effector cells to be engineered to express a CAR molecule described herein. In such embodiments, administration of a low, immune enhancing, dose of an mTOR inhibitor, e.g., an allosteric inhibitor, e.g., RAD001, or a catalytic inhibitor, is initiated or completed prior to harvest of immune effector cells, e.g., T cells or NK cells, to be engineered to express a CAR molecule described herein, from a subject.
  • In another embodiment, immune effector cells, e.g., T cells or NK cells, to be engineered to express a CAR molecule described herein, e.g., after harvest from a subject, or CAR-expressing immune effector cells, e.g., T cells or NK cells, e.g., prior to administration to a subject, can be cultured in the presence of a low, immune enhancing, dose of an mTOR inhibitor.
  • In an embodiment, administering to the subject a low, immune enhancing, dose of an mTOR inhibitor comprises administering, e.g., once per week, e.g., in an immediate release dosage form, 0.1 to 20, 0.5 to 10, 2.5 to 7.5, 3 to 6, or about 5, mgs of RAD001, or a bioequivalent dose thereof. In an embodiment, administering to the subject a low, immune enhancing, dose of an mTOR inhibitor comprises administering, e.g., once per week, e.g., in a sustained release dosage form, 0.3 to 60, 1.5 to 30, 7.5 to 22.5, 9 to 18, or about 15 mgs of RAD001, or a bioequivalent dose thereof.
  • In an embodiment, a dose of an mTOR inhibitor is associated with, or provides, mTOR inhibition of at least 5 but no more than 90%, at least 10 but no more than 90%, at least 15, but no more than 90%, at least 20 but no more than 90%, at least 30 but no more than 90%, at least 40 but no more than 90%, at least 50 but no more than 90%, at least 60 but no more than 90%, at least 70 but no more than 90%, at least 5 but no more than 80%, at least 10 but no more than 80%, at least 15, but no more than 80%, at least 20 but no more than 80%, at least 30 but no more than 80%, at least 40 but no more than 80%, at least 50 but no more than 80%, at least 60 but no more than 80%, at least 5 but no more than 70%, at least 10 but no more than 70%, at least 15, but no more than 70%, at least 20 but no more than 70%, at least 30 but no more than 70%, at least 40 but no more than 70%, at least 50 but no more than 70%, at least 5 but no more than 60%, at least 10 but no more than 60%, at least 15, but no more than 60%, at least 20 but no more than 60%, at least 30 but no more than 60%, at least 40 but no more than 60%, at least 5 but no more than 50%, at least 10 but no more than 50%, at least 15, but no more than 50%, at least 20 but no more than 50%, at least 30 but no more than 50%, at least 40 but no more than 50%, at least 5 but no more than 40%, at least 10 but no more than 40%, at least 15, but no more than 40%, at least 20 but no more than 40%, at least 30 but no more than 40%, at least 35 but no more than 40%, at least 5 but no more than 30%, at least 10 but no more than 30%, at least 15, but no more than 30%, at least 20 but no more than 30%, or at least 25 but no more than 30%.
  • The extent of mTOR inhibition can be conveyed as, or corresponds to, the extent of P70 S6 kinase inhibition, e.g., the extent of mTOR inhibition can be determined by the level of decrease in P70 S6 kinase activity, e.g., by the decrease in phosphorylation of a P70 S6 kinase substrate. The level of mTOR inhibition can be evaluated by various methods, such as measuring P70 S6 kinase activity by the Boulay assay, as described in U.S. Patent Application No. 2015/01240036, hereby incorporated by reference, or as described in U.S. Pat. No. 7,727,950, hereby incorporated by reference; measuring the level of phosphorylated S6 by western blot; or evaluating a change in the ratio of PD1 negative immune effector cells to PD1 positive immune effector cells.
  • As used herein, the term “mTOR inhibitor” refers to a compound or ligand, or a pharmaceutically acceptable salt thereof, which inhibits the mTOR kinase in a cell. In an embodiment, an mTOR inhibitor is an allosteric inhibitor. Allosteric mTOR inhibitors include the neutral tricyclic compound rapamycin (sirolimus), rapamycin-related compounds, that is compounds having structural and functional similarity to rapamycin including, e.g., rapamycin derivatives, rapamycin analogs (also referred to as rapalogs) and other macrolide compounds that inhibit mTOR activity. In an embodiment, an mTOR inhibitor is a catalytic inhibitor.
  • Rapamycin is a known macrolide antibiotic produced by Streptomyces hygroscopicus having the structure shown in Formula A.
  • Figure US20210172020A1-20210610-C00002
  • See, e.g., McAlpine, J. B., et al., J. Antibiotics (1991) 44: 688; Schreiber, S. L., et al., J. Am. Chem. Soc. (1991) 113: 7433; U.S. Pat. No. 3,929,992. There are various numbering schemes proposed for rapamycin. To avoid confusion, when specific rapamycin analogs are named herein, the names are given with reference to rapamycin using the numbering scheme of formula A.
  • Rapamycin analogs useful in the invention are, for example, O-substituted analogs in which the hydroxyl group on the cyclohexyl ring of rapamycin is replaced by OR1 in which R1 is hydroxyalkyl, hydroxyalkoxyalkyl, acylaminoalkyl, or aminoalkyl; e.g. RAD001, also known as, everolimus as described in U.S. Pat. No. 5,665,772 and WO94/09010 the contents of which are incorporated by reference. Other suitable rapamycin analogs include those substituted at the 26- or 28-position. The rapamycin analog may be an epimer of an analog mentioned above, particularly an epimer of an analog substituted in position 40, 28 or 26, and may optionally be further hydrogenated, e.g. as described in U.S. Pat. No. 6,015,815, WO95/14023 and WO99/15530 the contents of which are incorporated by reference, e.g. ABT578 also known as zotarolimus or a rapamycin analog described in U.S. Pat. No. 7,091,213, WO98/02441 and WO01/14387 the contents of which are incorporated by reference, e.g. AP23573 also known as ridaforolimus.
  • Examples of rapamycin analogs suitable for use in the present invention from U.S. Pat. No. 5,665,772 include, but are not limited to, 40-O-benzyl-rapamycin, 40-O-(4′-hydroxymethyl)benzyl-rapamycin, 40-O-[4′-(1,2-dihydroxyethyl)]benzyl-rapamycin, 40-O-allyl-rapamycin, 40-O-[3′-(2,2-dimethyl-1,3-dioxolan-4(S)-yl)-prop-2′-en-1′-yl]-rapamycin, (2′E,4'S)-40-O-(4′,5′-dihydroxypent-2′-en-1′-yl)-rapamycin, 40-O-(2-hydroxy)ethoxycarbonylmethyl-rapamycin, 40-O(2-hydroxy)ethyl-rapamycin, 40-O-(3-hydroxy)propyl-rapamycin, 40-O(6-hydroxy)hexyl-rapamycin, 40-O-[2-(2-hydroxy)ethoxy]ethyl-rapamycin, 40-O-[(3S)-2,2-dimethyldioxolan-3-yl]methyl-rapamycin, 40-O-[(2S)-2,3-dihydroxyprop-1-yl]-rapamycin, 40-O-(2-acetoxy)ethyl-rapamycin, 40-O-(2-nicotinoyloxy)ethyl-rapamycin, 40-O[2-(N-morpholino)acetoxy]ethyl-rapamycin, 40-O-(2-N-imidazolylacetoxy)ethyl-rapamycin, 40-O-[2-(N-methyl-N′-piperazinyl)acetoxy]ethyl-rapamycin, 39-O-desmethyl-39,40-O,O-ethylene-rapamycin, (26R)-26-dihydro-40-O-(2-hydroxy)ethyl-rapamycin, 40-O(2-aminoethyl)-rapamycin, 40-O(2-acetaminoethyl)-rapamycin, 40-O(2-nicotinamidoethyl)-rapamycin, 40-O-(2-N-methyl-imidazo-2′-ylcarbethoxamido)ethyl)-rapamycin, 40-O-(2-ethoxycarbonylaminoethyl)-rapamycin, 40-O-(2-tolylsulfonamidoethyl)-rapamycin and 40-O-[2-4′,5′-dicarboethoxy-1′,2′,3′-triazol-1′-yl)-ethyl]-rapamycin.
  • Other rapamycin analogs useful in the present invention are analogs where the hydroxyl group on the cyclohexyl ring of rapamycin and/or the hydroxy group at the 28 position is replaced with an hydroxyester group are known, for example, rapamycin analogs found in U.S. RE44,768, e.g. temsirolimus.
  • Other rapamycin analogs useful in the preset invention include those wherein the methoxy group at the 16 position is replaced with another substituent, preferably (optionally hydroxy-substituted) alkynyloxy, benzyl, orthomethoxybenzyl or chlorobenzyl and/or wherein the mexthoxy group at the 39 position is deleted together with the 39 carbon so that the cyclohexyl ring of rapamycin becomes a cyclopentyl ring lacking the 39 position methyoxy group; e.g. as described in WO95/16691 and WO96/41807 the contents of which are incorporated by reference. The analogs can be further modified such that the hydroxy at the 40-position of rapamycin is alkylated and/or the 32-carbonyl is reduced.
  • Rapamycin analogs from WO95/16691 include, but are not limited to, 16-demthoxy-16-(pent-2-ynyl)oxy-rapamycin, 16-demthoxy-16-(but-2-ynyl)oxy-rapamycin, 16-demthoxy-16-(propargyl)oxy-rapamycin, 16-demethoxy-16-(4-hydroxy-but-2-ynyl)oxy-rapamycin, 16-demthoxy-16-benzyloxy-40-O-(2-hydroxyethyl)-rapamycin, 16-demthoxy-16-benzyloxy-rapamycin, 16-demethoxy-16-ortho-methoxybenzyl-rapamycin, 16-demethoxy-40-O-(2-methoxyethyl)-16-pent-2-ynyl)oxy-rapamycin, 39-demethoxy-40-desoxy-39-formyl-42-nor-rapamycin, 39-demethoxy-40-desoxy-39-hydroxymethyl-42-nor-rapamycin, 39-demethoxy-40-desoxy-39-carboxy-42-nor-rapamycin, 39-demethoxy-40-desoxy-39-(4-methyl-piperazin-1-yl)carbonyl-42-nor-rapamycin, 39-demethoxy-40-desoxy-39-(morpholin-4-yl)carbonyl-42-nor-rapamycin, 39-demethoxy-40-desoxy-39-[N-methyl, N-(2-pyridin-2-yl-ethyl)]carbamoyl-42-nor-rapamycin and 39-demethoxy-40-desoxy-39-(p-toluenesulfonylhydrazonomethyl)-42-nor-rapamycin.
  • Rapamycin analogs from WO96/41807 include, but are not limited to, 32-deoxo-rapamycin, 16-O-pent-2-ynyl-32-deoxo-rapamycin, 16-O-pent-2-ynyl-32-deoxo-40-O-(2-hydroxy-ethyl)-rapamycin, 16-O-pent-2-ynyl-32-(S)-dihydro-40-O-(2-hydroxyethyl)-rapamycin, 32(S)-dihydro-40-O-(2-methoxy)ethyl-rapamycin and 32(S)-dihydro-40-O-(2-hydroxyethyl)-rapamycin.
  • Another suitable rapamycin analog is umirolimus as described in US2005/0101624 the contents of which are incorporated by reference.
  • RAD001, otherwise known as everolimus (Afinitor®), has the chemical name (1R,9S,12S,15R,16E,18R,19R,21R,23 S,24E,26E,28E,30S,32S,35R)-1,18-dihydroxy-12-{(1R)-2-[(1S,3R,4R)-4-(2-hydroxyethoxy)-3-methoxycyclohexyl]-1-methylethyl}-19,30-dimethoxy-15,17,21,23,29,35-hexamethyl-11,36-dioxa-4-aza-tricyclo[30.3.1.04,9]hexatriaconta-16,24,26,28-tetraene-2,3,10,14,20-pentaone
  • Further examples of allosteric mTOR inhibitors include sirolimus (rapamycin, AY-22989), 40-[3-hydroxy-2-(hydroxymethyl)-2-methylpropanoate]-rapamycin (also called temsirolimus or CCI-779) and ridaforolimus (AP-23573/MK-8669). Other examples of allosteric mTor inhibitors include zotarolimus (ABT578) and umirolimus.
  • Alternatively or additionally, catalytic, ATP-competitive mTOR inhibitors have been found to target the mTOR kinase domain directly and target both mTORC1 and mTORC2. These are also more effective inhibitors of mTORC1 than such allosteric mTOR inhibitors as rapamycin, because they modulate rapamycin-resistant mTORC1 outputs such as 4EBP1-T37/46 phosphorylation and cap-dependent translation.
  • Catalytic inhibitors include: BEZ235 or 2-methyl-2-[4-(3-methyl-2-oxo-8-quinolin-3-yl-2,3-dihydro-imidazo[4,5-c]quinolin-1-yl)-phenyl]-propionitrile, or the monotosylate salt form. the synthesis of BEZ235 is described in WO2006/122806; CCG168 (otherwise known as AZD-8055, Chresta, C. M., et al., Cancer Res, 2010, 70(1), 288-298) which has the chemical name {5-[2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3 d]pyrimidin-7-yl]-2-methoxy-phenyl}-methanol; 3-[2,4-bis[(3 S)-3-methylmorpholin-4-yl]pyrido[2,3-d]pyrimidin-7-yl]-N-methylbenzamide (WO09104019); 3-(2-aminobenzo[d]oxazol-5-yl)-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-4-amine (WO10051043 and WO2013023184); A N-(3-(N-(3-((3,5-dimethoxyphenyl)amino)quinoxaline-2-yl)sulfamoyl)phenyl)-3-methoxy-4-methylbenzamide (WO07044729 and WO12006552); PKI-587 (Venkatesan, A. M., J. Med. Chem., 2010, 53, 2636-2645) which has the chemical name 1-[4-[4-(dimethylamino)piperidine-1-carbonyl]phenyl]-3-[4-(4,6-dimorpholino-1,3,5-triazin-2-yl)phenyl]urea; GSK-2126458 (ACS Med. Chem. Lett., 2010, 1, 39-43) which has the chemical name 2,4-difluoro-N-{2-methoxy-5-[4-(4-pyridazinyl)-6-quinolinyl]-3-pyridinyl}benzenesulfonamide; 5-(9-isopropyl-8-methyl-2-morpholino-9H-purin-6-yl)pyrimidin-2-amine (WO10114484); (E)-N-(8-(6-amino-5-(trifluoromethyl)pyridin-3-yl)-1-(6-(2-cyanopropan-2-yl)pyridin-3-yl)-3-methyl-1H-imidazo[4,5-c]quinolin-2(3H)-ylidene)cyanamide (WO12007926).
  • Further examples of catalytic mTOR inhibitors include 8-(6-methoxy-pyridin-3-yl)-3-methyl-1-(4-piperazin-1-yl-3-trifluoromethyl-phenyl)-1,3-dihydro-imidazo[4,5-c]quinolin-2-one (WO2006/122806) and Ku-0063794 (Garcia-Martinez J M, et al., Biochem J., 2009, 421(1), 29-42. Ku-0063794 is a specific inhibitor of the mammalian target of rapamycin (mTOR).) WYE-354 is another example of a catalytic mTor inhibitor (Yu K, et al. (2009). Biochemical, Cellular, and In vivo Activity of Novel ATP-Competitive and Selective Inhibitors of the Mammalian Target of Rapamycin. Cancer Res. 69(15): 6232-6240).
  • mTOR inhibitors useful according to the present invention also include prodrugs, derivatives, pharmaceutically acceptable salts, or analogs thereof of any of the foregoing.
  • mTOR inhibitors, such as RAD001, may be formulated for delivery based on well-established methods in the art based on the particular dosages described herein. In particular, U.S. Pat. No. 6,004,973 (incorporated herein by reference) provides examples of formulations useable with the mTOR inhibitors described herein.
  • Inhibitory Molecule Inhibitors/Checkpoint Inhibitors
  • In one embodiment, the subject can be administered an agent which enhances the activity of a CAR-expressing cell. For example, in one embodiment, the agent can be an agent which inhibits a checkpoint molecule. Checkpoint molecules, e.g., Programmed Death 1 (PD1), can, in some embodiments, decrease the ability of a CAR-expressing cell to mount an immune effector response. Examples of inhibitory molecules, e.g., checkpoint molecules include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR (e.g., TGFRbeta). In embodiments, the CAR-expressing cell described herein comprises a switch costimulatory receptor, e.g., as described in WO 2013/019615, which is incorporated herein by reference in its entirety.
  • The methods described herein can include administration of a CAR-expressing cell in combination with a checkpoint inhibitor. In one embodiment, the subject is a complete responder. In another embodiment, the subject is a partial responder or non-responder, and, e.g., in some embodiments, the checkpoint inhibitor is administered prior to the CAR-expressing cell, e.g., two weeks, 12 days, 10 days, 8 days, one week, 6 days, 5 days, 4 days, 3 days, 2 days or 1 day before administration of the CAR-expressing cell. In some embodiments, the checkpoint inhibitor is administered concurrently with the CAR-expressing cell.
  • Inhibition of a checkpoint molecule, e.g., by inhibition at the DNA, RNA or protein level, can optimize a CAR-expressing cell performance. In embodiments, an inhibitory nucleic acid, e.g., an inhibitory nucleic acid, e.g., a dsRNA, e.g., a siRNA or shRNA, a clustered regularly interspaced short palindromic repeats (CRISPR), a transcription-activator like effector nuclease (TALEN), or a zinc finger endonuclease (ZFN), e.g., as described herein, can be used to inhibit expression of a checkpoint molecule in the CAR-expressing cell. In an embodiment, the inhibitor is a shRNA. In an embodiment, the checkpoint molecule is inhibited within a CAR-expressing cell. In these embodiments, a dsRNA molecule that inhibits expression of the checkpoint molecule is linked to the nucleic acid that encodes a component, e.g., all of the components, of the CAR.
  • In one embodiment, the inhibitor of an inhibitory signal can be, e.g., an antibody or antibody fragment that binds to a checkpoint molecule. For example, the agent can be an antibody or antibody fragment that binds to PD1, PD-L1, PD-L2 or CTLA4 (e.g., ipilimumab (also referred to as MDX-010 and MDX-101, and marketed as Yervoy®; Bristol-Myers Squibb; Tremelimumab (IgG2 monoclonal antibody available from Pfizer, formerly known as ticilimumab, CP-675,206). In an embodiment, the agent is an antibody or antibody fragment that binds to TIM3. In an embodiment, the agent is an antibody or antibody fragment that binds to LAG3. In an embodiment, the agent is an antibody or antibody fragment that binds to CEACAM.
  • PD1 is an inhibitory member of the CD28 family of receptors that also includes CD28, CTLA-4, ICOS, and BTLA. PD1 is expressed on activated B cells, T cells and myeloid cells (Agata et al. 1996 INT. IMMUNOL 8:765-75). Two ligands for PD1, PD-L1 and PD-L2 have been shown to downregulate T cell activation upon binding to PD1 (Freeman et a. 2000 J Exp Med 192:1027-34; Latchman et al. 2001 NAT IMMUNOL 2:261-8; Carter et al. 2002 EUR J IMMUNOL 32:634-43). PD-L1 is abundant in human cancers (Dong et al. 2003 J MOL MED 81:281-7; Blank et al. 2005 CANCER IMMUNOL. IMMUNOTHER. 54:307-314; Konishi et al. 2004 CLIN CANCER RES 10:5094). Immune suppression can be reversed by inhibiting the local interaction of PD1 with PD-L1.
  • Antibodies, antibody fragments, and other inhibitors of PD1, PD-L1 and PD-L2 are available in the art and may be used combination with a CAR described herein, e.g., a CD19 CAR described herein. For example, nivolumab (also referred to as BMS-936558 or MDX1106; Bristol-Myers Squibb) is a fully human IgG4 monoclonal antibody which specifically blocks PD1. Nivolumab (clone 5C4) and other human monoclonal antibodies that specifically bind to PD1 are disclosed in U.S. Pat. No. 8,008,449 and WO2006/121168. Pidilizumab (CT-011; Cure Tech) is a humanized IgGlk monoclonal antibody that binds to PD1Pidilizumab and other humanized anti-PD1 monoclonal antibodies are disclosed in WO2009/101611. Lambrolizumab (also referred to as MK03475; Merck) is a humanized IgG4 monoclonal antibody that binds to PD1. Lambrolizumab and other humanized anti-PD1 antibodies are disclosed in U.S. Pat. No. 8,354,509 and WO2009/114335. MDPL3280A (Genentech/Roche) is a human Fc optimized IgG1 monoclonal antibody that binds to PD-L1. MDPL3280A and other human monoclonal antibodies to PD-L1 are disclosed in U.S. Pat. No. 7,943,743 and U.S Publication No.: 20120039906. Other anti-PD-L1 binding agents include YW243.55.S70 (heavy and light chain variable regions are shown in SEQ ID NOs 20 and 21 in WO2010/077634) and MDX-1 105 (also referred to as BMS-936559, and, e.g., anti-PD-L1 binding agents disclosed in WO2007/005874). AMP-224 (B7-DCIg; Amplimmune; e.g., disclosed in WO2010/027827 and WO2011/066342), is a PD-L2 Fc fusion soluble receptor that blocks the interaction between PD1 and B7-H1. Other anti-PD1 antibodies include AMP 514 (Amplimmune), among others, e.g., anti-PD1 antibodies disclosed in U.S. Pat. No. 8,609,089, US 2010028330, and/or US 20120114649.
  • In one embodiment, the anti-PD-1 antibody or fragment thereof is an anti-PD-1 antibody molecule as described in US 2015/0210769, entitled “Antibody Molecules to PD-1 and Uses Thereof,” incorporated by reference in its entirety. In one embodiment, the anti-PD-1 antibody molecule includes at least one, two, three, four, five or six CDRs (or collectively all of the CDRs) from a heavy and light chain variable region from an antibody chosen from any of BAP049-hum01, BAP049-hum02, BAP049-hum03, BAP049-hum04, BAP049-hum05, BAP049-hum06, BAP049-hum07, BAP049-hum08, BAP049-hum09, BAP049-hum10, BAP049-hum11, BAP049-hum12, BAP049-hum13, BAP049-hum14, BAP049-hum15, BAP049-hum16, BAP049-Clone-A, BAP049-Clone-B, BAP049-Clone-C, BAP049-Clone-D, or BAP049-Clone-E; or as described in Table 1 of US 2015/0210769, or encoded by the nucleotide sequence in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or closely related CDRs, e.g., CDRs which are identical or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions).
  • In yet another embodiment, the anti-PD-1 antibody molecule comprises at least one, two, three or four variable regions from an antibody described herein, e.g., an antibody chosen from any of BAP049-hum01, BAP049-hum02, BAP049-hum03, BAP049-hum04, BAP049-hum05, BAP049-hum06, BAP049-hum07, BAP049-hum08, BAP049-hum09, BAP049-hum10, BAP049-hum11, BAP049-hum12, BAP049-hum13, BAP049-hum14, BAP049-hum15, BAP049-hum16, BAP049-Clone-A, BAP049-Clone-B, BAP049-Clone-C, BAP049-Clone-D, or BAP049-Clone-E; or as described in Table 1 of US 2015/0210769, or encoded by the nucleotide sequence in Table 1; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.
  • TIM3 (T cell immunoglobulin-3) also negatively regulates T cell function, particularly in IFN-g-secreting CD4+T helper 1 and CD8+T cytotoxic 1 cells, and plays a critical role in T cell exhaustion. Inhibition of the interaction between TIM3 and its ligands, e.g., galectin-9 (Gal9), phosphotidylserine (PS), and HMGB1, can increase immune response. Antibodies, antibody fragments, and other inhibitors of TIM3 and its ligands are available in the art and may be used combination with a CD19 CAR described herein. For example, antibodies, antibody fragments, small molecules, or peptide inhibitors that target TIM3 binds to the IgV domain of TIM3 to inhibit interaction with its ligands. Antibodies and peptides that inhibit TIM3 are disclosed in WO2013/006490 and US20100247521. Other anti-TIM3 antibodies include humanized versions of RMT3-23 (disclosed in Ngiow et al., 2011, Cancer Res, 71:3540-3551), and clone 8B.2C12 (disclosed in Monney et al., 2002, Nature, 415:536-541). Bi-specific antibodies that inhibit TIM3 and PD-1 are disclosed in US20130156774.
  • In one embodiment, the anti-TIM3 antibody or fragment thereof is an anti-TIM3 antibody molecule as described in US 2015/0218274, entitled “Antibody Molecules to TIM3 and Uses Thereof,” incorporated by reference in its entirety. In one embodiment, the anti-TIM3 antibody molecule includes at least one, two, three, four, five or six CDRs (or collectively all of the CDRs) from a heavy and light chain variable region from an antibody chosen from any of ABTIM3, ABTIM3-hum01, ABTIM3-hum02, ABTIM3-hum03, ABTIM3-hum04, ABTIM3-hum05, ABTIM3-hum06, ABTIM3-hum07, ABTIM3-hum08, ABTIM3-hum09, ABTIM3-hum10, ABTIM3-hum11, ABTIM3-hum12, ABTIM3-hum13, ABTIM3-hum14, ABTIM3-hum15, ABTIM3-hum16, ABTIM3-hum17, ABTIM3-hum18, ABTIM3-hum19, ABTIM3-hum20, ABTIM3-hum21, ABTIM3-hum22, ABTIM3-hum23; or as described in Tables 1-4 of US 2015/0218274; or encoded by the nucleotide sequence in Tables 1-4; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences, or closely related CDRs, e.g., CDRs which are identical or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions).
  • In yet another embodiment, the anti-TIM3 antibody molecule comprises at least one, two, three or four variable regions from an antibody described herein, e.g., an antibody chosen from any of ABTIM3, ABTIM3-hum01, ABTIM3-hum02, ABTIM3-hum03, ABTIM3-hum04, ABTIM3-hum05, ABTIM3-hum06, ABTIM3-hum07, ABTIM3-hum08, ABTIM3-hum09, ABTIM3-hum10, ABTIM3-hum11, ABTIM3-hum12, ABTIM3-hum13, ABTIM3-hum14, ABTIM3-hum15, ABTIM3-hum16, ABTIM3-hum17, ABTIM3-hum18, ABTIM3-hum19, ABTIM3-hum20, ABTIM3-hum21, ABTIM3-hum22, ABTIM3-hum23; or as described in Tables 1-4 of US 2015/0218274; or encoded by the nucleotide sequence in Tables 1-4; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.
  • In other embodiments, the agent which enhances the activity of a CAR-expressing cell is a CEACAM inhibitor (e.g., CEACAM-1, CEACAM-3, and/or CEACAM-5 inhibitor). In one embodiment, the inhibitor of CEACAM is an anti-CEACAM antibody molecule. Exemplary anti-CEACAM-1 antibodies are described in WO 2010/125571, WO 2013/082366 WO 2014/059251 and WO 2014/022332, e.g., a monoclonal antibody 34B1, 26H7, and 5F4; or a recombinant form thereof, as described in, e.g., US 2004/0047858, U.S. Pat. No. 7,132,255 and WO 99/052552. In other embodiments, the anti-CEACAM antibody binds to CEACAM-5 as described in, e.g., Zheng et al. PLoS One. 2010 Sep. 2; 5(9). pii: e12529 (DOI:10:1371/journal.pone.0021146), or crossreacts with CEACAM-1 and CEACAM-5 as described in, e.g., WO 2013/054331 and US 2014/0271618.
  • Without wishing to be bound by theory, carcinoembryonic antigen cell adhesion molecules (CEACAM), such as CEACAM-1 and CEACAM-5, are believed to mediate, at least in part, inhibition of an anti-tumor immune response (see e.g., Markel et al. J Immunol. 2002 Mar. 15; 168(6):2803-10; Markel et al. J Immunol. 2006 Nov. 1; 177(9):6062-71; Markel et al. Immunology. 2009 February; 126(2):186-200; Markel et al. Cancer Immunol Immunother. 2010 February; 59(2):215-30; Ortenberg et al. Mol Cancer Ther. 2012 June; 11(6):1300-10; Stern et al. J Immunol. 2005 Jun. 1; 174(11):6692-701; Zheng et al. PLoS One. 2010 Sep. 2; 5(9). pii: e12529). For example, CEACAM-1 has been described as a heterophilic ligand for TIM-3 and as playing a role in TIM-3-mediated T cell tolerance and exhaustion (see e.g., WO 2014/022332; Huang, et al. (2014) Nature doi:10.1038/nature13848). In embodiments, co-blockade of CEACAM-1 and TIM-3 has been shown to enhance an anti-tumor immune response in xenograft colorectal cancer models (see e.g., WO 2014/022332; Huang, et al. (2014), supra). In other embodiments, co-blockade of CEACAM-1 and PD-1 reduce T cell tolerance as described, e.g., in WO 2014/059251. Thus, CEACAM inhibitors can be used with the other immunomodulators described herein (e.g., anti-PD-1 and/or anti-TIM-3 inhibitors) to enhance an immune response against a cancer, e.g., a melanoma, a lung cancer (e.g., NSCLC), a bladder cancer, a colon cancer an ovarian cancer, and other cancers as described herein.
  • LAG3 (lymphocyte activation gene-3 or CD223) is a cell surface molecule expressed on activated T cells and B cells that has been shown to play a role in CD8+ T cell exhaustion. Antibodies, antibody fragments, and other inhibitors of LAG3 and its ligands are available in the art and may be used combination with a CD19 CAR described herein. For example, BMS-986016 (Bristol-Myers Squib) is a monoclonal antibody that targets LAG3. IMP701 (Immutep) is an antagonist LAG3 antibody and IMP731 (Immutep and GlaxoSmithKline) is a depleting LAG3 antibody. Other LAG3 inhibitors include IMP321 (Immutep), which is a recombinant fusion protein of a soluble portion of LAG3 and Ig that binds to MHC class II molecules and activates antigen presenting cells (APC). Other antibodies are disclosed, in WO2010/019570.
  • In some embodiments, the agent which enhances the activity of a CAR-expressing cell can be, e.g., a fusion protein comprising a first domain and a second domain, wherein the first domain is a checkpoint molecule, or fragment thereof, and the second domain is a polypeptide that is associated with a positive signal, e.g., a polypeptide comprising an intracellular signaling domain as described herein (also referred to herein as an inhibitory CAR or iCAR). In some embodiments, the polypeptide that is associated with a positive signal can include a costimulatory domain of CD28, CD27, ICOS, e.g., an intracellular signaling domain of CD28, CD27 and/or ICOS, and/or a primary signaling domain, e.g., of CD3 zeta, e.g., described herein. In one embodiment, the fusion protein is expressed by the same cell that expressed the CAR. In another embodiment, the fusion protein is expressed by a cell, e.g., a T cell that does not express a CAR, e.g., a CD19 CAR.
  • In one embodiment, the extracellular domain (ECD) of a checkpoint molecule, e.g., a checkpoint molecule described herein such as, e.g., Programmed Death 1 (PD1), can be fused to a transmembrane domain and intracellular signaling domain described herein, e.g., an intracellular signaling domain comprising a costimulatory signaling domain such as, e.g., 41BB OX40, Cd28, CD27, and/or a primary signaling domain, e.g., of CD3 zeta. In one embodiment, the inhibitory CAR, e.g., e.g., PD1 CAR, can be used in combination with another CAR, e.g., CD19CAR (e.g., a CD19RCAR). In one embodiment, the PD1 RCAR (or PD1 CAR) improves the persistence of the T cell. Examples of inhibitory molecules include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR (e.g., TGFRbeta). In one embodiment, the inhibitory molecule CAR comprises a first polypeptide, e.g., of an inhibitory molecule such as PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MEW class I, MEW class II, GAL9, adenosine, and TGFR (e.g., TGFRbeta), or a fragment of any of these (e.g., at least a portion of an extracellular domain of any of these), and a second polypeptide which is an intracellular signaling domain described herein (e.g., comprising a costimulatory domain (e.g., 41BB, CD27 or CD28, e.g., as described herein) and/or a primary signaling domain (e.g., a CD3 zeta signaling domain described herein).
  • In one embodiment, the inhibitory molecule CAR comprises the extracellular domain (ECD) of PD1fused to a transmembrane domain and intracellular signaling domains such as 41BB and CD3 zeta (also referred to herein as a PD1 CAR). In one embodiment, the PD1 CAR improves the persistence of the cell CAR-expressing cell. In one embodiment, the PD1 CAR comprises the extracellular domain of PD1 indicated in SEQ ID NO: 44. In one embodiment, the PD1 CAR comprises, the amino acid sequence of SEQ ID NO:40.
  • In one embodiment, the PD1 CAR comprises the amino acid sequence provided as SEQ ID NO: 41.
  • In one embodiment, the PD1 CAR, e.g., the PD1 CAR described herein, is encoded by a nucleic acid sequence provided as SEQ ID NO: 42, or at least the comprises the nucleic acid sequence encoding the extracellular domain of PD1 (provided as SEQ ID NO: 101).
  • In embodiments, the inhibitory extracellular domain, has at least 70, 75, 80, 85, 90, 95, 96, 97, 98, or 99% identity with, or differs by no more than 30, 25, 20, 15, 10, 5, 4, 3, 2, or 1 amino acid residues from the corresponding residues of a naturally occurring human inhibitory molecule, e.g., a naturally occurring human primary stimulatory molecule disclosed herein.
  • In an embodiment, a nucleic acid molecule that encodes a dsRNA molecule that inhibits expression of the molecule that modulates or regulates, e.g., inhibits, T-cell function is operably linked to a promoter, e.g., a H1- or a U6-derived promoter such that the dsRNA molecule that inhibits expression of the molecule that modulates or regulates, e.g., inhibits, T-cell function is expressed, e.g., is expressed within a CAR-expressing cell. See e.g., Tiscornia G., “Development of Lentiviral Vectors Expressing siRNA,” Chapter 3, in Gene Transfer: Delivery and Expression of DNA and RNA (eds. Friedmann and Rossi). Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA, 2007; Brummelkamp T R, et al. (2002) Science 296: 550-553; Miyagishi M, et al. (2002) Nat. Biotechnol. 19: 497-500. In an embodiment the nucleic acid molecule that encodes a dsRNA molecule that inhibits expression of the molecule that modulates or regulates, e.g., inhibits, T-cell function is present on the same vector, e.g., a lentiviral vector, that comprises a nucleic acid molecule that encodes a component, e.g., all of the components, of the CAR. In such an embodiment, the nucleic acid molecule that encodes a dsRNA molecule that inhibits expression of the molecule that modulates or regulates, e.g., inhibits, T-cell function is located on the vector, e.g., the lentiviral vector, 5′- or 3′- to the nucleic acid that encodes a component, e.g., all of the components, of the CAR. The nucleic acid molecule that encodes a dsRNA molecule that inhibits expression of the molecule that modulates or regulates, e.g., inhibits, T-cell function can be transcribed in the same or different direction as the nucleic acid that encodes a component, e.g., all of the components, of the CAR.
  • In an embodiment the nucleic acid molecule that encodes a dsRNA molecule that inhibits expression of the molecule that modulates or regulates, e.g., inhibits, T-cell function is present on a vector other than the vector that comprises a nucleic acid molecule that encodes a component, e.g., all of the components, of the CAR. In an embodiment, the nucleic acid molecule that encodes a dsRNA molecule that inhibits expression of the molecule that modulates or regulates, e.g., inhibits, T-cell function it transiently expressed within a CAR-expressing cell. In an embodiment, the nucleic acid molecule that encodes a dsRNA molecule that inhibits expression of the molecule that modulates or regulates, e.g., inhibits, T-cell function is stably integrated into the genome of a CAR-expressing cell. In an embodiment, the molecule that modulates or regulates, e.g., inhibits, T-cell function is PD-1.
  • In embodiments, the agent that enhances the activity of a CAR-expressing cell, e.g., inhibitor of an inhibitory molecule, is administered in combination with an allogeneic CAR, e.g., an allogeneic CAR described herein (e.g., described in the Allogeneic CAR section herein).
  • Natural Killer Cell Receptor (NKR) CARs
  • In an embodiment, the CAR molecule described herein comprises one or more components of a natural killer cell receptor (NKR), thereby forming an NKR-CAR. The NKR component can be a transmembrane domain, a hinge domain, or a cytoplasmic domain from any of the following natural killer cell receptors: killer cell immunoglobulin-like receptor (KIR), e.g., KIR2DL1, KIR2DL2/L3, KIR2DL4, KIR2DL5A, KIR2DL5B, KIR2DS1, KIR2DS2, KIR2DS3, KIR2DS4, DIR2DS5, KIR3DL1/S1, KIR3DL2, KIR3DL3, KIR2DP1, and KIR3DP1; natural cytotoxicity receptor (NCR), e.g., NKp30, NKp44, NKp46; signaling lymphocyte activation molecule (SLAM) family of immune cell receptors, e.g., CD48, CD229, 2B4, CD84, NTB-A, CRACC, BLAME, and CD2F-10; Fc receptor (FcR), e.g., CD16, and CD64; and Ly49 receptors, e.g., LY49A, LY49C. The NKR-CAR molecules described herein may interact with an adaptor molecule or intracellular signaling domain, e.g., DAP12. Exemplary configurations and sequences of CAR molecules comprising NKR components are described in International Publication No. WO2014/145252, the contents of which are hereby incorporated by reference.
  • Split CAR
  • In some embodiments, the CAR-expressing cell uses a split CAR. The split CAR approach is described in more detail in publications WO2014/055442 and WO2014/055657, incorporated herein by reference. Briefly, a split CAR system comprises a cell expressing a first CAR having a first antigen binding domain and a costimulatory domain (e.g., 41BB), and the cell also expresses a second CAR having a second antigen binding domain and an intracellular signaling domain (e.g., CD3 zeta). When the cell encounters the first antigen, the costimulatory domain is activated, and the cell proliferates. When the cell encounters the second antigen, the intracellular signaling domain is activated and cell-killing activity begins. Thus, the CAR-expressing cell is only fully activated in the presence of both antigens.
  • Strategies for Regulating Chimeric Antigen Receptors
  • There are many ways CAR activities can be regulated. In some embodiments, a regulatable CAR (RCAR) where the CAR activity can be controlled is desirable to optimize the safety and efficacy of a CAR therapy. There are many ways CAR activities can be regulated. For example, inducible apoptosis using, e.g., a caspase fused to a dimerization domain (see, e.g., Di Stasa et al., N Engl. J. Med. 2011 Nov. 3; 365(18):1673-1683), can be used as a safety switch in the CAR therapy of the instant invention. In one embodiment, the cells (e.g., T cells or NK cells) expressing a CAR of the present invention further comprise an inducible apoptosis switch, wherein a human caspase (e.g., caspase 9) or a modified version is fused to a modification of the human FKB protein that allows conditional dimerization. In the presence of a small molecule, such as a rapalog (e.g., AP 1903, AP20187), the inducible caspase (e.g., caspase 9) is activated and leads to the rapid apoptosis and death of the cells (e.g., T cells or NK cells) expressing a CAR of the present invention. Examples of a caspase-based inducible apoptosis switch (or one or more aspects of such a switch) have been described in, e.g., US2004040047; US20110286980; US20140255360; WO1997031899; WO2014151960; WO2014164348; WO2014197638; WO2014197638; all of which are incorporated by reference herein.
  • In another example, CAR-expressing cells can also express an inducible Caspase-9 (iCaspase-9) molecule that, upon administration of a dimerizer drug (e.g., rimiducid (also called AP1903 (Bellicum Pharmaceuticals) or AP20187 (Ariad)) leads to activation of the Caspase-9 and apoptosis of the cells. The iCaspase-9 molecule contains a chemical inducer of dimerization (CID) binding domain that mediates dimerization in the presence of a CID. This results in inducible and selective depletion of CAR-expressing cells. In some cases, the iCaspase-9 molecule is encoded by a nucleic acid molecule separate from the CAR-encoding vector(s). In some cases, the iCaspase-9 molecule is encoded by the same nucleic acid molecule as the CAR-encoding vector. The iCaspase-9 can provide a safety switch to avoid any toxicity of CAR-expressing cells. See, e.g., Song et al. Cancer Gene Ther. 2008; 15(10):667-75; Clinical Trial Id. No. NCT02107963; and Di Stasi et al. N. Engl. J. Med. 2011; 365:1673-83.
  • Alternative strategies for regulating the CAR therapy of the instant invention include utilizing small molecules or antibodies that deactivate or turn off CAR activity, e.g., by deleting CAR-expressing cells, e.g., by inducing antibody dependent cell-mediated cytotoxicity (ADCC).
  • For example, CAR-expressing cells described herein may also express an antigen that is recognized by molecules capable of inducing cell death, e.g., ADCC or complement-induced cell death. For example, CAR expressing cells described herein may also express a receptor capable of being targeted by an antibody or antibody fragment. Examples of such receptors include EpCAM, VEGFR, integrins (e.g., integrins αvβ3, α4, αI¾β3, α4β7, α5β1, αvβ3, αv), members of the TNF receptor superfamily (e.g., TRAIL-R1, TRAIL-R2), PDGF Receptor, interferon receptor, folate receptor, GPNMB, ICAM-1, HLA-DR, CEA, CA-125, MUC1, TAG-72, IL-6 receptor, 5T4, GD2, GD3, CD2, CD3, CD4, CD5, CD1 1, CD1 1 a/LFA-1, CD15, CD18/ITGB2, CD19, CD20, CD22, CD23/1gE Receptor, CD25, CD28, CD30, CD33, CD38, CD40, CD41, CD44, CD51, CD52, CD62L, CD74, CD80, CD125, CD147/basigin, CD152/CTLA-4, CD154/CD40L, CD195/CCR5, CD319/SLAMF7, and EGFR, and truncated versions thereof (e.g., versions preserving one or more extracellular epitopes but lacking one or more regions within the cytoplasmic domain).
  • For example, a CAR-expressing cell described herein may also express a truncated epidermal growth factor receptor (EGFR) which lacks signaling capacity but retains the epitope that is recognized by molecules capable of inducing ADCC, e.g., cetuximab (ERBITUX®), such that administration of cetuximab induces ADCC and subsequent depletion of the CAR-expressing cells (see, e.g., WO2011/056894, and Jonnalagadda et al., Gene Ther. 2013; 20(8)853-860). Another strategy includes expressing a highly compact marker/suicide gene that combines target epitopes from both CD32 and CD20 antigens in the CAR-expressing cells described herein, which binds rituximab, resulting in selective depletion of the CAR-expressing cells, e.g., by ADCC (see, e.g., Philip et al., Blood. 2014; 124(8)1277-1287). Other methods for depleting CAR-expressing cells described herein include administration of CAMPATH, a monoclonal anti-CD52 antibody that selectively binds and targets mature lymphocytes, e.g., CAR-expressing cells, for destruction, e.g., by inducing ADCC. In other embodiments, the CAR-expressing cell can be selectively targeted using a CAR ligand, e.g., an anti-idiotypic antibody. In some embodiments, the anti-idiotypic antibody can cause effector cell activity, e.g., ADCC or ADC activities, thereby reducing the number of CAR-expressing cells. In other embodiments, the CAR ligand, e.g., the anti-idiotypic antibody, can be coupled to an agent that induces cell killing, e.g., a toxin, thereby reducing the number of CAR-expressing cells. Alternatively, the CAR molecules themselves can be configured such that the activity can be regulated, e.g., turned on and off, as described below.
  • In other embodiments, a CAR-expressing cell described herein may also express a target protein recognized by the T cell depleting agent. In one embodiment, the target protein is CD20 and the T cell depleting agent is an anti-CD20 antibody, e.g., rituximab. In such embodiment, the T cell depleting agent is administered once it is desirable to reduce or eliminate the CAR-expressing cell, e.g., to mitigate the CAR induced toxicity. In other embodiments, the T cell depleting agent is an anti-CD52 antibody, e.g., alemtuzumab, as described in the Examples herein.
  • In other embodiments, an RCAR comprises a set of polypeptides, typically two in the simplest embodiments, in which the components of a standard CAR described herein, e.g., an antigen binding domain and an intracellular signalling domain, are partitioned on separate polypeptides or members. In some embodiments, the set of polypeptides include a dimerization switch that, upon the presence of a dimerization molecule, can couple the polypeptides to one another, e.g., can couple an antigen binding domain to an intracellular signalling domain. In one embodiment, a CAR of the present invention utilizes a dimerization switch as those described in, e.g., WO2014127261, which is incorporated by reference herein. Additional description and exemplary configurations of such regulatable CARs are provided herein and in, e.g., paragraphs 527-551 of International Publication No. WO 2015/090229 filed Mar. 13, 2015, which is incorporated by reference in its entirety. In some embodiments, an RCAR involves a switch domain, e.g., a FKBP switch domain, as set out SEQ ID NO: 92, or comprise a fragment of FKBP having the ability to bind with FRB, e.g., as set out in SEQ ID NO: 93. In some embodiments, the RCAR involves a switch domain comprising a FRB sequence, e.g., as set out in SEQ ID NO: 94, or a mutant FRB sequence, e.g., as set out in any of SEQ ID Nos. 95-100.
  • Co-expression of CAR with a Chemokine Receptor
  • In embodiments, the CAR-expressing cell described herein further comprises a chemokine receptor molecule. Transgenic expression of chemokine receptors CCR2b or CXCR2 in T cells enhances trafficking to CCL2- or CXCL1-secreting solid tumors including melanoma and neuroblastoma (Craddock et al., J Immunother. 2010 October; 33(8):780-8 and Kershaw et al., Hum Gene Ther. 2002 Nov. 1; 13(16):1971-80). Thus, without wishing to be bound by theory, it is believed that chemokine receptors expressed in CAR-expressing cells that recognize chemokines secreted by tumors, e.g., solid tumors, can improve homing of the CAR-expressing cell to the tumor, facilitate the infiltration of the CAR-expressing cell to the tumor, and enhances antitumor efficacy of the CAR-expressing cell. The chemokine receptor molecule can comprise a naturally occurring or recombinant chemokine receptor or a chemokine-binding fragment thereof. A chemokine receptor molecule suitable for expression in a CAR-expressing cell described herein include a CXC chemokine receptor (e.g., CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, or CXCR7), a CC chemokine receptor (e.g., CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CCR10, or CCR11), a CX3C chemokine receptor (e.g., CX3CR1), a XC chemokine receptor (e.g., XCR1), or a chemokine-binding fragment thereof. In one embodiment, the chemokine receptor molecule to be expressed with a CAR described herein is selected based on the chemokine(s) secreted by the tumor. In one embodiment, the CAR-expressing cell described herein further comprises, e.g., expresses, a CCR2b receptor or a CXCR2 receptor. In an embodiment, the CAR described herein and the chemokine receptor molecule are on the same vector or are on two different vectors. In embodiments where the CAR described herein and the chemokine receptor molecule are on the same vector, the CAR and the chemokine receptor molecule are each under control of two different promoters or are under the control of the same promoter.
  • Split CAR
  • In some embodiments, the CAR-expressing cell uses a split CAR. The split CAR approach is described in more detail in publications WO2014/055442 and WO2014/055657. Briefly, a split CAR system comprises a cell expressing a first CAR having a first antigen binding domain and a costimulatory domain (e.g., 41BB), and the cell also expresses a second CAR having a second antigen binding domain and an intracellular signaling domain (e.g., CD3 zeta). When the cell encounters the first antigen, the costimulatory domain is activated, and the cell proliferates. When the cell encounters the second antigen, the intracellular signaling domain is activated and cell-killing activity begins. Thus, the CAR-expressing cell is only fully activated in the presence of both antigens.
  • Pharmaceutical Compositions and Treatments
  • Pharmaceutical compositions may comprise a CAR-expressing cell, e.g., a plurality of CAR-expressing cells, as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients. Such compositions may comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives. Compositions can be, e.g., formulated for intravenous administration.
  • Pharmaceutical compositions of the present disclosure may be administered in a manner appropriate to the disease to be treated (or prevented). The quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease, although appropriate dosages may be determined by clinical trials.
  • In one embodiment, the pharmaceutical composition is substantially free of, e.g., there are no detectable levels of a contaminant, e.g., a contaminant described in paragraph 1009 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • When “an immunologically effective amount,” “an anti-tumor effective amount,” “a tumor-inhibiting effective amount,” or “therapeutic amount” is indicated, the precise amount of the compositions of the present invention to be administered can be determined by a physician with consideration of individual differences in age, weight, tumor size, extent of infection or metastasis, and condition of the patient (subject). It can generally be stated that a pharmaceutical composition comprising the immune effector cells (e.g., T cells, NK cells) described herein may be administered at a dosage of 104 to 109 cells/kg body weight, in some instances 105 to 106 cells/kg body weight, including all integer values within those ranges. T cell compositions may also be administered multiple times at these dosages. The cells can be administered by using infusion techniques that are commonly known in immunotherapy (see, e.g., Rosenberg et al., NEW ENG. J. OF MED. 319:1676, 1988).
  • In some embodiments, a dose of CAR cells (e.g., CD19 CAR cells) comprises about 1×106, 1.1×106, 2×106, 3.6×106, 5×106, 1×107, 1.8×107, 2×107, 5×107, 1×108, 2×108, or 5×108 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19 CAR cells) comprises at least about 1×106, 1.1×106, 2×106, 3.6×106, 5×106, 1×107, 1.8×107, 2×107, 5×107, 1×108, 2×108, or 5×108 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19 CAR cells) comprises up to about 1×106, 1.1×106, 2×106, 3.6×106, 5×106, 1×107, 1.8×107, 2×107, 5×107, 1×108, 2×108, or 5×108 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19 CAR cells) comprises about 1.1×106-1.8×107 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19 CAR cells) comprises about 1×107, 2×107, 5×107, 1×108, 2×108, 5×108, 1×109, 2×109, or 5×109 cells. In some embodiments, a dose of CAR cells (e.g., CD19 CAR cells) comprises at least about 1×107, 2×107, 5×107, 1×108, 2×108, 5×108, 1×109, 2×109, or 5×109 cells. In some embodiments, a dose of CAR cells (e.g., CD19 CAR cells) comprises up to about 1×107, 2×107, 5×107, 1×108, 2×108, 5×108, 1×109, 2×109, or 5×109 cells.
  • In certain aspects, it may be desired to administer activated immune effector cells (e.g., T cells, NK cells) to a subject and then subsequently redraw blood (or have an apheresis performed), activate immune effector cells (e.g., T cells, NK cells) therefrom according to the present disclosure, and reinfuse the patient with these activated and expanded immune effector cells (e.g., T cells, NK cells). This process can be carried out multiple times every few weeks. In certain aspects, immune effector cells (e.g., T cells, NK cells) can be activated from blood draws of from 10 cc to 400 cc. In certain aspects, immune effector cells (e.g., T cells, NK cells) are activated from blood draws of 20 cc, 30 cc, 40 cc, 50 cc, 60 cc, 70 cc, 80 cc, 90 cc, or 100 cc.
  • The administration of the subject compositions may be carried out in any convenient manner, including by aerosol inhalation, injection, ingestion, transfusion, implantation or transplantation. The compositions described herein may be administered to a patient trans arterially, subcutaneously, intradermally, intratumorally, intranodally, intramedullary, intramuscularly, by intravenous (i.v.) injection, or intraperitoneally. In one aspect, the T cell compositions described herein are administered to a patient by intradermal or subcutaneous injection. In one aspect, the T cell compositions described herein are administered by i.v. injection. The compositions of immune effector cells (e.g., T cells, NK cells) may be injected directly into a tumor, lymph node, or site of infection.
  • In a particular exemplary aspect, subjects may undergo leukapheresis, wherein leukocytes are collected, enriched, or depleted ex vivo to select and/or isolate the cells of interest, e.g., T cells. These T cell isolates may be expanded by methods known in the art and treated such that one or more CAR constructs described herein may be introduced, thereby creating a CAR T cell of the present disclosure. Subjects in need thereof may subsequently undergo standard treatment with high dose chemotherapy followed by peripheral blood stem cell transplantation. In certain aspects, following or concurrent with the transplant, subjects receive an infusion of the expanded CAR T cells described herein. In an additional aspect, expanded cells are administered before or following surgery.
  • The dosage of the above treatments to be administered to a patient will vary with the precise nature of the condition being treated and the recipient of the treatment. The scaling of dosages for human administration can be performed according to art-accepted practices. The dose for CAMPATH, for example, will generally be in the range 1 to about 100 mg for an adult patient, usually administered daily for a period between 1 and 30 days. A suitable daily dose is 1 to 10 mg per day although in some instances larger doses of up to 40 mg per day may be used (described in U.S. Pat. No. 6,120,766).
  • In one embodiment, the CAR is introduced into immune effector cells (e.g., T cells, NK cells), e.g., using in vitro transcription, and the subject (e.g., human) receives an initial administration of CAR immune effector cells (e.g., T cells, NK cells) of the invention, and one or more subsequent administrations of the CAR immune effector cells (e.g., T cells, NK cells) of the invention, wherein the one or more subsequent administrations are administered less than 15 days, e.g., 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 days after the previous administration. In one embodiment, more than one administration of the CAR immune effector cells (e.g., T cells, NK cells) described herein are administered to the subject (e.g., human) per week, e.g., 2, 3, or 4 administrations of the CAR immune effector cells (e.g., T cells, NK cells) of the invention are administered per week. In one embodiment, the subject (e.g., human subject) receives more than one administration of the CAR immune effector cells (e.g., T cells, NK cells) per week (e.g., 2, 3 or 4 administrations per week) (also referred to herein as a cycle), followed by a week of no CAR immune effector cells (e.g., T cells, NK cells) administrations, and then one or more additional administration of the CAR immune effector cells (e.g., T cells, NK cells) (e.g., more than one administration of the CAR immune effector cells (e.g., T cells, NK cells) per week) is administered to the subject. In another embodiment, the subject (e.g., human subject) receives more than one cycle of CAR immune effector cells (e.g., T cells, NK cells), and the time between each cycle is less than 10, 9, 8, 7, 6, 5, 4, or 3 days. In one embodiment, the CAR immune effector cells (e.g., T cells, NK cells) are administered every other day for 3 administrations per week. In one embodiment, the CAR immune effector cells (e.g., T cells, NK cells) described herein are administered for at least two, three, four, five, six, seven, eight or more weeks.
  • In one aspect, CAR-expressing cells (e.g., T cells, NK cells) as described herein such as, e.g., CD19 CAR-expressing cells, e.g., CTL019 are generated using lentiviral viral vectors, such as lentivirus. CAR-expressing cells generated that way can have stable CAR expression.
  • In one aspect, CAR-expressing cells (e.g., T cells, NK cells) transiently express CAR vectors for 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 days after transduction. Transient expression of CARs can be effected by RNA CAR vector delivery. In one aspect, the CAR RNA is transduced into the cell by electroporation.
  • A potential issue that can arise in patients being treated using transiently expressing CAR cells, e.g., T cells (particularly with murine scFv bearing CAR-expressing cells (e.g., T cells, NK cells)) is anaphylaxis after multiple treatments. Without being bound by this theory, it is believed that such an anaphylactic response might be caused by a patient developing humoral anti-CAR response, i.e., anti-CAR antibodies having an anti-IgE isotype. It is thought that a patient's antibody producing cells undergo a class switch from IgG isotype (that does not cause anaphylaxis) to IgE isotype when there is a ten to fourteen day break in exposure to antigen.
  • If a patient is at high risk of generating an anti-CAR antibody response during the course of transient CAR therapy (such as those generated by RNA transductions), CAR-expressing cell (e.g., T cell, NK cell) infusion breaks should not last more than ten to fourteen days.
  • In some embodiments of any of the aforesaid methods, the method further includes administering one or more doses of a cell (e.g., an immune cell containing a CAR nucleic acid or CAR polypeptide as described herein), to a mammal (e.g., a mammal having a cancer, e.g., a mammal that is or is identified as being a responder, complete responder, partial responder, non-responder, relapser, or non-relapser according to the methods herein). In some embodiments, the one or more doses of CAR cells (e.g., CD19 CAR cells) comprises at least about 1×106, 5×106, 1×107, 2×107, 5×107, 1×108, 2×108, 5×108, 1×109, 2×109, or 5×109 cells.
  • In one embodiment, up to 10, 9, 8, 7, 6, 5, 4, 3, or 2 doses of cells are administered. In other embodiments, one, two, three, four, five or 6 doses of the cells are administered to the mammal, e.g., in a treatment interval of one, two, three, four or more weeks. In one embodiment, up to 6 doses are administered in two weeks. The doses may the same or different. In one embodiment, a lower dose is administered initially, followed by one or more higher doses. In one exemplary embodiment, the lower dose is about 1×105 to 1×109 cells/kg, or 1×106 to 1×108 cells/kg; and the higher dose is about 2×105 to 2×109 cells/kg or 2×106 to 2×108 cells/kg, followed by 3-6 doses of about 4×105 to 4×109 cells/kg, or 4×106 to 4×108 cells/kg.
  • In one embodiment, the one or more doses of the cells are administered after one or more lymphodepleting therapies, e.g., a lymphodepleting chemotherapy. In one embodiment, the lymphodepleting therapy includes a chemotherapy (e.g., cyclophosphamide).
  • In one embodiment, the one or more doses is followed by a cell transplant, e.g., an allogeneic hematopoietic stem cell transplant. For example, the allogeneic hematopoietic stem cell transplant occurs between about 20 to about 35 days, e.g., between about 23 and 33 days.
  • Biopolymer Delivery Methods
  • In some embodiments, one or more CAR-expressing cells as disclosed herein can be administered or delivered to the subject via a biopolymer scaffold, e.g., a biopolymer implant. Biopolymer scaffolds can support or enhance the delivery, expansion, and/or dispersion of the CAR-expressing cells described herein. A biopolymer scaffold comprises a biocompatible (e.g., does not substantially induce an inflammatory or immune response) and/or a biodegradable polymer that can be naturally occurring or synthetic. Exemplary biopolymers are described, e.g., in paragraphs 1004-1006 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.
  • Exemplary Computer System
  • Various computer systems can be specially configured to leverage information returned on potential cancer responder status indicators (for example, ALL and CLL responder status indicator as described herein such as, e.g., complete responder, partial responder, non-responder). In some embodiments, the computer system can determine and present information on confidence levels associated with various bio-markers and/or indicators for cancer (e.g. a hematological cancer) such as ALL and CLL. For example, the computer systems can evaluate whether a test conducted on a subject indicates a gene signature of a complete responder, partial responder or non-responder, along with a degree of confidence associated with the subject responder classification. In further examples, the system can provide an indication and/or recommendation on increasing the degree of confidence associated with the predicted responder classification. For example, the system can be configured to evaluate any tests and tested biomarkers and/or indicators of cancer that have been performed for a subject against another characteristic identified as independent and/or additive of the existing data. In an embodiment, the system can be configured to evaluate any tests and tested biomarkers and/or cancer indicators (e.g., a hematological cancer such as CLL and ALL) that have been performed for a subject against another characteristic identified as independent and/or additive of the existing data. The system can determine when an additional biomarker and/or indicator (e.g., gene signature) would increase confidence associated with, for example, a change in responder classification. The system can recommend testing of any identified characteristic accordingly.
  • In some embodiments, an interactive system for identification, assessment and/or treatment of a subject having cancer (e.g., a hematological cancer such as ALL and CLL) can be provided. In an embodiment, an interactive system for identification, assessment and/or treatment of a subject having cancer (e.g., a hematological cancer such as ALL and CLL) can be provided. According to one embodiment, the system can be configured to accept user input regarding degree of confidence of a subject assessment. Responsive to the user entered degree of confidence, the system can determine test characteristics to include in an evaluation model. In one example, the system includes specification of independent indicators for disease activity in a subject (e.g., patient) population. The system can be configured to estimate a degree of confidence in a determination of disease activity or a prediction of future disease activity based on what independent indicators are used. The system can be further configured to determined and/or recommend various combinations of the determined independent indicators to improve a degree of confidence in an evaluation.
  • According to another aspect, a computer system can be specially configured to evaluate indicators for cancer (e.g., a hematological cancer such as ALL and CLL). In an embodiment, a computer system can be specially configured to evaluate indicators for ALL and/or CLL. The system can be configured to generate a multivariate model, wherein the multivariate model excludes correlated indicators. In some examples, the system can be configured to identify correlated indicators responsive to evaluating returned test results within a subject (e.g., patient) population having one or more of the indicators. For example, the system can execute regression model analysis to control for various parameters, including, for example, subject age, race, sex, and the presence of other indicators. Responsive to eliminating correlated indicators, the system can generate a model of one or more independent indicators. In some embodiments, the system can be configured to select various combinations of the one or more independent indicators and can further access evaluations (including, for example, evaluating the combination directly) to present information on a confidence level associated with respective selections. The system selected models can be used to generate an expected change in disease activity with the determined confidence level.
  • In an embodiment, the disclosure provides a system for evaluating cancer (e.g., a hematological cancer such as ALL and CLL) in a subject, comprising:
  • at least one processor operatively connected to a memory, the at least one processor when executing is configured to:
  • acquire a value of responder status that comprises a measure of a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene set signature and a combination of one or more of:
  • a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, a CD27 biomarker, a CD45RO biomarker, a PD-1 biomarker, a LAG-3 biomarker, a TIM-3 biomarker, an IL2RA biomarker, an IL21 biomarker, a CD4 biomarker, a CD8 biomarker, a TH1+ helper T cell gene set signature, a TH2+ helper T cell gene set signature, and a memory T cell (e.g., a CD8+ memory T cell, e.g., a naïve T cell (TN), e.g. a memory stem cell (TSCM), e.g. a central memory T cell (TCM), e.g. an effector memory T cell (TEM)) gene set signature; and
  • responsive to a determination of the value of responder status, perform one, two, three, four or more of:
  • identify the subject as a complete responder, partial responder, or non-responder;
  • recommend a CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy as described herein, such as, e.g., CTL019);
  • recommend a selection or alteration of a dosing of a CAR-expressing cell (e.g., T cell, NK cell) therapy; or
  • an alternative therapy, e.g., a standard of care for the particular cancer.
  • In an embodiment, the invention provides a system for evaluating cancer (e.g., a hematological cancer such as ALL and CLL) in a subject, comprising: at least one processor operatively connected to a memory, the at least one processor when executing is configured to: acquire a value of responder status that comprises a measure of a CD19 CAR-expressing cell (e.g., T cell, NK cell) gene set signature and a combination of one or more of: a biomarker listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 15, Table 16 (e.g., CCL20, IL-17a and/or IL-6), Table 17, Table 18, Table 20, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, and KLRG1; and responsive to a determination of the value of responder status, perform one, two, three, four or more of: identify the subject as a complete responder, partial responder, or non-responder; recommend a CAR-expressing cell therapy (e.g., a CD19 CAR-expressing cell therapy as described herein, such as, e.g., CTL019); recommend a selection or alteration of a dosing of a CAR-expressing cell therapy; or an alternative therapy.
  • FIG. 16 is a block diagram of a distributed computer system 200, in which various aspects and functions in accord with the present disclosure may be practiced. The distributed computer system 200 may include one or more computer systems. For example, as illustrated, the distributed computer system 200 includes three computer systems 202, 204 and 206. As shown, the computer systems 202, 204 and 206 are interconnected by, and may exchange data through, a communication network 208. The network 208 may include any communication network through which computer systems may exchange data. To exchange data via the network 208, the computer systems 202, 204, and 206 and the network 208 may use various methods, protocols and standards including, among others, token ring, Ethernet, Wireless Ethernet, Bluetooth, radio signaling, infra-red signaling, TCP/IP, UDP, HTTP, FTP, SNMP, SMS, MMS, SS2, JSON, XML, REST, SOAP, CORBA IIOP, RMI, DCOM and Web Services.
  • According to some embodiments, the functions and operations discussed for identifying, treating or preventing cancer (e.g., a hematological cancer such as ALL and CLL) in a subject can be executed on computer systems 202, 204 and 206 individually and/or in combination. For example, the computer systems 202, 204, and 206 support, for example, participation in a collaborative operations, which may include analyzing treatment data captured on a patient population. In one alternative, a single computer system (e.g., 202) can analyze treatment data captured on a subject (e.g., patient) population to develop characterization models and/or identify independent indicators for disease activity. The computer systems 202, 204 and 206 may include personal computing devices such as cellular telephones, smart phones, tablets, etc., and may also include desktop computers, laptop computers, etc.
  • Various aspects and functions in accord with the present disclosure may be implemented as specialized hardware or software executing in one or more computer systems including the computer system 202 shown in FIG. 16. In one embodiment, computer system 202 is a computing device specially configured to execute the processes and/or operations discussed above. For example, the system can present user interfaces to end-users that present treatment information, diagnostic information, and confidence levels associated with biomarkers and/or genetic indicators, among other options. As depicted, the computer system 202 includes at least one processor 210 (e.g., a single core or a multi-core processor), a memory 212, a bus 214, input/output interfaces (e.g., 216) and storage 218. The processor 210, may include one or more microprocessors or other types of controllers, and can perform a series of instructions that manipulate data (e.g., treatment data, testing data, etc.). As shown, the processor 210 is connected to other system components, including a memory 212, by an interconnection element (e.g., the bus 214).
  • The memory 212 and/or storage 218 may be used for storing programs and data during operation of the computer system 202. For example, the memory 212 may be a relatively high performance, volatile, random access memory such as a dynamic random access memory (DRAM) or static memory (SRAM). In addition, the memory 212 may include any device for storing data, such as a disk drive or other non-volatile storage device, such as flash memory, solid state, or phase-change memory (PCM). In further embodiments, the functions and operations discussed with respect to identifying, treating or preventing cancer (e.g., ALL and/or CLL) in a subject can be embodied in an application that is executed on the computer system 202 from the memory 212 and/or the storage 218.
  • Computer system 202 also includes one or more interfaces 216 such as input devices, output devices, and combination input/output devices. The interfaces 216 may receive input, provide output, or both. The storage 218 may include a computer-readable and computer-writeable nonvolatile storage medium in which instructions are stored that define a program to be executed by the processor. The storage system 218 also may include information that is recorded, on or in, the medium, and this information may be processed by the application. A medium that can be used with various embodiments may include, for example, optical disk, magnetic disk or flash memory, SSD, among others.
  • Further, the invention is not limited to a particular memory system or storage system. Although the computer system 202 is shown by way of example as one type of computer system upon which various functions for identifying, treating or preventing cancer (e.g., a hematological cancer such as ALL and CLL) in a subject may be practiced, aspects of the invention are not limited to being implemented on the computer system, shown in FIG. 16. Various aspects and functions in accord with the present invention may be practiced on one or more computers having different architectures or components than that shown in FIG. 16.
  • In some embodiments, the computer system 202 may include an operating system that manages at least a portion of the hardware components (e.g., input/output devices, touch screens, cameras, etc.) included in computer system 202. One or more processors or controllers, such as processor 210, may execute an operating system which may be, among others, a Windows-based operating system (e.g., Windows NT, ME, XP, Vista, 2, 8, or RT) available from the Microsoft Corporation, an operating system available from Apple Computer (e.g., MAC OS, including System X), one of many Linux-based operating system distributions (for example, the Enterprise Linux operating system available from Red Hat Inc.), a Solaris operating system available from Sun Microsystems, or a UNIX operating systems available from various sources. Many other operating systems may be used, including operating systems designed for personal computing devices (e.g., iOS, Android, etc.) and embodiments are not limited to any particular operating system.
  • According to one embodiment, the processor and operating system together define a computing platform on which applications may be executed. Additionally, various functions for identifying, treating or preventing cancer (e.g., a hematological cancer such as ALL and CLL) in a subject may be implemented in a non-programmed environment (for example, documents created in HTML, XML or other format that, when viewed in a window of a browser program, render aspects of a graphical-user interface or perform other functions). Further, various embodiments in accord with aspects of the present disclosure may be implemented as programmed or non-programmed components, or any combination thereof. Thus, the disclosure is not limited to a specific programming language and any suitable programming language could also be used.
  • EXEMPLIFICATION
  • The invention is further described in detail by reference to the following experimental examples. These examples are provided for purposes of illustration only, and are not intended to be limiting unless otherwise specified. Thus, the invention should in no way be construed as being limited to the following examples, but rather, should be construed to encompass any and all variations which become evident as a result of the teaching provided herein.
  • Without further description, it is believed that one of ordinary skill in the art can, using the preceding description and the following illustrative examples, make and utilize the compounds of the present invention and practice the claimed methods. The following working examples specifically point out various aspects of the present invention, and are not to be construed as limiting in any way the remainder of the disclosure.
  • Example 1: Identification of Novel Transcriptional Gene Signatures that Predict Subject Response to CD19 CAR-Expressing Cell Therapy in Chronic Lymphoid Leukemia (CLL) and Acute Lymphoblastic Leukemia (ALL) Using Whole Genome RNAseq and Unbiased Feature Selection
  • The present Example describes the identification of novel transcriptional gene signatures that predict patient response to CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., CTL019 therapy) in Chronic Lymphoid Leukemia (CLL) and Acute Lymphoblastic Leukemia (ALL), for use in accordance with the present invention.
  • Among other things, the present Example describes novel gene signatures based on mRNA expression levels of selected genes in apheresis and manufactured CD19 CAR-expressing cell (e.g., T cell, NK cell) product samples (e.g., CTL019) prior to re-infusion.
  • In particular, the present Example describes methods of unbiased feature selection to discover novel gene signatures that predict patient response to CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., CTL019) in CLL and ALL, for use in accordance with the present invention.
  • Novel gene signatures based on mRNA expression levels in apheresis and manufactured CD19 CAR-expressing cell product samples prior to re-infusion have been identified that predict patient response to CD19 CAR-expressing cell therapy in Chronic Lymphoid Leukemia (CLL) and Acute Lymphoblastic Leukemia (ALL). The identified signatures were discovered in a whole genome RNAseq study of manufactured product samples which included 7 ALL subject samples and 21 CLL subject samples. ALL subject samples (7 total) were taken from subjects (e.g., patients) with complete response to CD19 CAR-expressing cell therapy. CLL subject samples (21 total) were stratified as follows: biological samples were taken from 2 patients that were complete responders (CRs) to CTL019 therapy, 6 patients that were partial responders (PRs), and 13 non-responders (NRs). The gene signatures were then investigated in a subset of the above patients where samples were collected at apheresis. Several gene signatures discriminating responders from non-responders in manufactured product and apheresis samples were discovered and are described further in Example 2. Healthy donor samples with manufactured product (i.e., reference samples) were acquired and used as a reference level.
  • Novel gene signatures were then discovered using various data analytical approaches: 1) unbiased feature selection; 2) gene set analysis; and 3) differential expression analysis of selected genes of interest. Gene set analysis (2) and differential expression analysis (3) are discussed in further detail in Example 2.
  • Novel gene signatures derived from unbiased feature selection were discovered by determining which genes were differentially expressed between the CRs and NRs and between CRs and PRs. Genes were defined as differentially expressed if their differential expression was statistically significant with a FDR p-value cutoff of 0.1. The gene lists for the CR vs NR comparison (N=128) and CR vs PR comparison (N=34) are tabulated in Table 1A-B.
  • Without wishing to be bound by a particular theory, these data indicate that the differentiation state of T cells in apheresis or CD19 CAR-expressing cell (e.g., T cell, NK cell) product (e.g., CTL019) correlate with subject response (i.e., CR, PR, or NR). Gene signatures for T cells from CR are in a more unstimulated/undifferentiated state. In addition, memory T cell subsets are differentially enriched between CRs versus NRs, with CRs showing similarity to naive T cells (TN) and T memory stem cells (TSCM). An exemplary schematic illustrating the progression of a naïve T cell (TN) through the memory T cell subset stages, into an effector memory T cell (TEM) is shown in FIG. 3).
  • Complete responders to CTL019 therapy have a significantly higher % of CD8+ T cells that express the co-stimulatory molecule CD27 but lack the antigen-experienced T cell marker CD45RO compared to the non-responders. In an embodiment, the threshold for this discrimination was 7% CD27+CD45RO− cells in the CD8+ population. In an embodiment, a complete responder is defined as 7% or greater CD27+CD45RO− cells in the CD8+ population. Without wishing to be bound by a particular theory, the state of memory T cells in CTL019 samples is likely a major component of response (FIG. 3).
  • These data demonstrate that CRs are more like resting TEFF cells, resting TREG cells, naïve CD4 cells, unstimulated memory cells and early memory T cells, whereas NRs are more like activated TEFF cells, activated TREG cells, activated TH1 and TH2 cells, stimulated memory cells, and late T memory cells.
  • TABLE 1A
    Table 1A: Comparison of Complete Responders (CR) vs. Non-responders (NR)
    Gene Unigene Accession No. FDR
    C16orf74 Hs.461655 NM_206967 6.47E−05
    uc021oxm 1.67E−03
    uc021ygq 2.83E−03
    uc021oxp 3.31E−03
    SNED1 Hs.471834 NM_001080437 9.55E−03
    ADAM19 Hs.483944 NM_033274 1.34E−02
    FAIM2 Hs.567424 NM_012306 1.82E−02
    WHAMMP2 1.82E−02
    LOC730091 Hs.659905 1.84E−02
    TCF7 Hs.573153 NM_201633, NM_201632, 1.84E−02
    NM_001134851,
    NM_001134852. NM_213648,
    NM_003202, NM_201634
    TTLL2 Hs.520554 NM_031949 1.84E−02
    LY9 Hs.403857 NM_002348, NM_001033667 2.03E−02
    uc021tnc 2.13E−02
    TRIL Hs.21572 NM_014817 2.28E−02
    uc004crn 2.28E−02
    DPEP2 Hs.372633 NM_022355 2.28E−02
    GZMB Hs.1051 NM_004131 2.35E−02
    FAM102A Hs.568044 NM_203305, NM_001035254 2.35E−02
    ALS2CL Hs.517937 NM_147129 2.35E−02
    EPHA4 Hs.371218 NM_004438 2.35E−02
    IKBIP Hs.252543 NM_201612, NM_201613, 2.88E−02
    NM_153687
    HBEGF Hs.799 NM_001945 2.88E−02
    LHFPL3 Hs.659164 NM_199000 2.88E−02
    RCAN2 Hs.440168 NM_005822 2.97E−02
    MFGE8 Hs.3745 NM_005928, NM_001114614 2.97E−02
    IL24 Hs.723317, NM_006850, NM_181339 2.97E−02
    Hs.58831
    FAIM3 Hs.723317, NM_001142472, 2.97E−02
    Hs.58831 NM_001142473,
    NM_005449
    CDKN1B Hs.238990 NM_004064 2.99E−02
    AQP3 Hs.234642 NM_004925 3.06E−02
    GPR155 Hs.516604 NM_001033045, NM_152529 3.06E−02
    HS6ST2 Hs.385956 NM_147175, NM_001077188 3.25E−02
    SNORD85 3.25E−02
    uc022cci 3.25E−02
    GSTM1 Hs.301961 NM_000561, NM_146421 3.27E−02
    VSIG1 Hs.177164 NM_001170553, NM_182607 3.86E−02
    VIPR1 Hs.348500 NM_004624 3.86E−02
    RCAN3 Hs.656799 NM_013441 4.45E−02
    ADHFE1 Hs.720023 NM_144650 4.49E−02
    HSPH1 Hs.36927 NM_006644 4.62E−02
    ENPP6 Hs.297814 NM_153343 4.74E−02
    RORC Hs.607993, NM_005060, NM_001001523 4.83E−02
    Hs.256022
    TRIB2 Hs.627749, NM_021643 4.96E−02
    Hs.467751
    LRP8 Hs.576154 NM_001018054, NM_004631, 4.96E−02
    NM_033300, NM_017522
    RGS17 Hs.166313 NM_012419 5.05E−02
    TAAR3 Hs.679662 5.18E−02
    C5orf41 Hs.484195 NM_153607, 5.27E−02
    NM_001168394,
    NM_001168393
    MIR3183 5.27E−02
    LTA Hs.36 NM_001159740, NM_000595 5.27E−02
    KLHL24 Hs.407709 NM_017644 5.28E−02
    PIK3IP1 Hs.26670 NM_001135911, NM_052880 5.28E−02
    MAP3K1 Hs.653654 NM_005921 5.29E−02
    VWC2L Hs.534834 NM_001080500 5.29E−02
    IDI2-AS1 5.29E−02
    DUSP4 Hs.417962 NM_001394, NM_057158 5.29E−02
    SKIL Hs.581632 NM_001145098, 5.77E−02
    NM_001145097,
    NM_005414
    uc021oxf 5.86E−02
    AMICA1 Hs.16291 NM_153206, NM_001098526 5.86E−02
    TP53INP1 Hs.492261 NM_001135733, NM_033285 5.86E−02
    GDAP1L1 Hs.517059 NM_024034 6.00E−02
    HK2 Hs.591588, NM_000189 6.43E−02
    Hs.406266
    CBLL1 Hs.592271 NM_024814 6.44E−02
    PSD3 Hs.434255 NM_015310, NM_206909 6.44E−02
    PUS7 Hs.520619 NM_019042 6.44E−02
    MSMO1 6.63E−02
    IDI1 Hs.283652 NM_004508 6.63E−02
    HRH4 Hs.287388 NM_001160166, NM_021624, 6.73E−02
    NM_001143828
    FAM19A1 Hs.655061 NM_213609 6.73E−02
    EHD4 Hs.143703 NM_139265 6.73E−02
    PVR Hs.171844 NM_001135768, NM_006505, 6.74E−02
    NM_001135770, NM_001135769
    MIR1293 6.74E−02
    WDR64 Hs.723441 NM_144625 6.74E−02
    CDKN1A Hs.370771 NM_078467, NM_000389 6.74E−02
    CACNA1I Hs.125116 NM_021096, NM_001003406 6.75E−02
    C21orf63 Hs.208358 NM_058187 6.75E−02
    FLJ41649 Hs.654837 9.72E−02
    MPP7 Hs.499159 NM_173496 9.82E−02
    POP1 Hs.252828 NM_001145861, 9.82E−02
    NM_001145860,
    NM_015029
    CALCOCO1 Hs.156667 NM_020898, NM_001143682 9.82E−02
    COL5A3 Hs.235368 NM_015719 9.82E−02
    LHFP Hs.507798 NM_005780 9.82E−02
    CTSO Hs.75262 NM_001334 9.82E−02
    LEF1 Hs.555947 NM_001166119, 9.82E−02
    NM_001130713,
    NM_001130714,
    NM_016269
    RNASET2 Hs.720966, NM_003730 9.89E−02
    Hs.529989
  • TABLE 1B
    Table 1B: Comparison of Complete responders (CR) vs. Partial responders (PR)
    Gene Unigene Acession No. FDR
    uc021oxm 0.000511026
    uc021oxp 0.000511026
    SPTB Hs.417303 NM_000347, NM_001024858 0.019052239
    ALS2CL Hs.517937 NM_147129, NM_182775 0.025286191
    TCF7 Hs.573153 NM_201633, NM_201632, 0.025286191
    NM_001134851, NM_001134852,
    NM_213648, NM_003202, NM_201634
    TRIL Hs.21572 NM_014817 0.025286191
    WDR86 Hs.647083 NM_198285 0.025286191
    ACSM2B Hs.567879, Hs.298252 NM_182617, NM_001105069 0.044953527
    DUSP4 Hs.417962 NM_001394, NM_057158 0.044953527
    EFHC1 Hs.403171 NM_018100 0.044953527
    HS6ST2 Hs.385956 NM_147175, NM_001077188 0.044953527
    TRIB2 Hs.627749, Hs.467751 NM_021643 0.047492472
    SQLE Hs.71465 NM_003129 0.053797924
    PRR5-ARHGAP8 Hs.720401, Hs.102336 NM_001017530, NM_181333, 0.053797924
    NM_181334, NM_181335, NM_015366,
    NM_001017526, NM_001017529,
    NM_001017528
    C16orf74 Hs.461655 NM_206967 0.056452084
    TMIE Hs.185777 NM_147196 0.056452084
    LOC100131176 Hs.659231 0.056452084
    VSIG1 Hs.177164 NM_001170553, NM_182607 0.056452084
    MIR3194 0.056452084
    RAP1GAP2 Hs.499659 NM_015085, NM_001100398 0.057016164
    FLJ13197 Hs.29725 0.084544923
    TSPEAR 0.084544923
    uc021zdn 0.084544923
    RASA3 Hs.593075 NM_007368 0.084544923
    OLIG3 Hs.195398 NM_175747 0.084544923
    GPR155 Hs.516604 NM_001033045, NM_152529 0.084544923
    uc021ygq 0.084544923
    FAM19A1 Hs.655061 NM_213609 0.084544923
    LY9 Hs.403857 NM_002348, NM_001033667 0.084544923
    ANKRD20A5P 0.084544923
    C21orf15 Hs.580910 0.08962672
    ADHFE1 Hs.720023 NM_144650 0.08962672
    MIR1293 0.098706653
    LOC730091 Hs.659905 0.098706653
  • Example 2: Identification of Novel Transcriptional Gene Signatures which Predict Subject Response to CD19 CAR-Expressing Cell Therapy in Chronic Lymphoid Leukemia (CLL) and Acute Lymphoblastic Leukemia (ALL) Using Gene Set Analysis and Differential Expression Analysis
  • The present Example describes the identification of novel transcriptional gene signatures that predict patient response to CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., CTL019) in CLL and ALL, for use in accordance with the present invention.
  • In particular, the present Example describes methods of Gene Set Analysis to discover novel gene signatures, for use in accordance with the present invention.
  • Among other things, the present Example describes novel gene signatures based on Gene Set Analysis, that are predictive of patent response to CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., CTL019). Gene set analysis was performed on gene sets described in Example 1, and with gene sets from three additional datasets. FIG. 2A depicts an exemplary histogram comparing the number of samples analyzed in a whole genome CTL019 RNAseq analysis. p=product; a=apheresis. Gene sets were sourced from (1) additional experiments were based on gene sets by Szabo et al. (described herein); (2) gene sets published by Abbas et al. in GENOME RESEARCH 2005; and (3) gene sets published by Gattinoni et al. in NATURE MEDICINE 2011. Each of these gene sets are described in further detail below.
  • Szabo et al. gene sets that were used in the Gene Set Analysis are provided in Table 2. Human CD4+ T cells were purified from PBMCs (5 normal donors, males, ages 18-28, with no known allergies or infections). CD4+CD25+(T reg) and CD4+CD25− (T effector) T cells were isolated and anti-CD3/CD28 stimulated for 0 or 16 hours to yield 4 conditions: (1) T reg 0 hours; (2) T reg 16 hours; (3) T effector 0 hours; and (4) T effector 16 hours. Two gene sets were derived for each condition: one set of genes whose expression levels were up-regulated in that condition relative to all others and another set of genes whose expression levels were downregulated in that condition relative to all others. The number of genes in each gene set was determined by a fold change cutoff (see Table 2).
  • TABLE 2
    Table 2: Gene sets comprised of genes up or down regulated
    in TREG and TEFF at resting and activation (Szabo data set)
    Gene Number of Genes
    Downregulated TREG vs TEFF 0 h (FC3 p < 0.05) 120
    Downregulated TREG vs TEFF 16 h (FC3 p < 0.05) 139
    Downregulated TEFF 16 h vs 0 h (FC7 p < 0.05) 246
    Upregulated TREG vs TEFF 16 h (FC4 p < 0.05) 254
    Upregulated TREG vs TEFF 0 h (FC4 p < 0.05) 135
    Upregulated TEFF 16 h vs 0 h (FC9 p < 0.05) 347
    Upregulated TREG 16 h vs 0 h (FC8 p < 0.05) 226
  • Exemplary genes according to Table 2 downregulated in Treg compared with Teff at 0 h include ABCB1, ACSL6, ADAMTS10, ADD2, AIF1, AIF1, AIF1, AIF1, AK5, AKR1E2, ALS2CL, ANK3, ANKRD55, APBA2, AREG, ATHL1, AXIN2, B4GALNT4, BACH2, BCL7A, BEND5, BHLHE40, BPGM, C10orf47, C16orf54, C1orf228, C2orf89, CA6, CACHD1, CACNA1I, CCL5, CELA1, CHD7, CHI3L2, COL18A1, COL6A1, CR2, CYB561, CYSLTR1, D4S234E, DACT1, DENND5A, DHRS3, DLG4, DLL1, DPYSL4, DSC1, EDAR, EMR1, EMR4P, ENC1, EPHA1, FCGBP, FHIT, GADD45G, GIPC3, GIPC3, GPR125, GPR160, H1F0, HDGFRP3, HIPK2, IFITM5, KCNQ1, KLF5, KLHL29, KRT72, KRT73, LASS6, LRRC24, MAN1C1, ME3, MMP28, MTUS1, NBL1, NELL2, NEO1, NKG7, NLRP6, NME4, NOG, NOSIP, NPAS2, NRCAM, OBSCN, OSBPL5, PCSK5, PDZD4, PECAM1, PLLP, PLXDC1, PPFIBP2, PRKAR1B, PTK2, RHOB, RMRP, RNF157, SATB1, SCML4, SDK2, SEC14L2, SEC14L2, SLC15A3, SLC22A17, SLC22A23, SLC40A1, SNTB1, SORBS3, SOX8, ST6GALNAC1, TCF7, TCF7, THEMIS, TMIGD2, VIPR1, WNT10B, WNT7A, ZNF467, ZNF516, and ZNF609.
  • Exemplary genes according to Table 2 downregulated in Treg compared to Teff 16 h include IL2, TNFSF8, NELL2, GOS2, IRF8, IFNG, IGFBP4, GPR125, CD200, GPR81, ADD2, IL21, SNORD86, TMCC2, C1orf228, SLC15A3, IL22, LRRN3, GPR171, FASLG, GZMH, NHS, MCOLN2, BACH2, TAGAP, MPZL2, PRAGMIN, DACT1, CXCL10, SLAMF6, PHGDH, CSF2, PRSS23, UHRF1, PLAC8, ISM1, BTLA, CDC20, GFOD1, HSD11B1, ME3, ZNF704, DHRS3, CXCL13, CCND1, NBL1, CRTAM, MAP6D1, H1F0, CDT1, CCL4, LIF, CD84, TRAT1, MIR155, SLAMF7, AIF1, AIF1, AIF1, AIF1, PRG4, VWCE, CHEK1, SH2D4A, MCM10, RHOU, NPAS2, NFIX, STAP1, DTL, C16orf59, CSDA, GINS2, FAM117B, ABCB1, CLC, PHEX, GDF10, RAB13, BCL7A, MAMLD1, SHF, LPIN2, AHI1, CCND3, HDGFRP3, MIR155HG, PVR, CDCA5, RRAS2, SIPA1L2, RASL10B, GAL, SNORD88C, SNORD18B, CDC6, SRD5A3, ORC6L, B3GNT5, ANK3, MCM2, MIR25, RHOBTB3, TNF, TERT, CSDAP1, CCDC64, CDC25A, ZNF367, MCM7, CASP10, LTA, MCM4, AFF3, FMNL2, TNFRSF21, AXIN2, CHD7, FABP5, XRCC2, CGREF1, CCL4L1, CCL4L2, B4GALNT4, DSCC1, CD97, PTPRK, RAD54L, EPB41L3, MYO1B, ORC1L, CHML, ZWINT, MAD2L1, NDST1, C11orfK, BEGAIN, CD55, and FABP5L3.
  • Exemplary genes according to Table 2 downregulated in Teff at 16 h vs 0 h include ABCA7, ABCG1, ABTB1, ACCS, ADAMTS10, ADD3, AK5, ALS2CL, AMT, ANKRD55, ANXA1, AQP3, AREG, ARL4C, ARRDC2, ARRDC3, BBC3, BCL9L, BIN2, BNIP3L, BTG1, BTN3A1, C10orf110, C11orf21, C11orf35, C14orf181, C16orf54, C16orf74, C17orf108, C1orf162, C1QTNF6, C20orf111, C20orf112, C5orf39, C5orf41, C5orf41, CACNA1I, CAPS, CBX4, CCNL1, CDC14A, CDC42BPG, CECR1, CFP, CHI3L2, CITED4, CLK1, CRIP2, CSGALNACT1, CTSF, CTSW, CXCR4, CYTH4, DCHS1, DDIT3, DDX60L, DISC1, DISC1, DISC1, DISC1, DPEP2, DPYD, DUSP1, DUSP8, EDAR, EMR4P, EPHA4, EPHX1, EPHX2, ERMN, ERP27, EVI2B, FAM13A, FAM13AOS, FAM46C, FAM65B, FBXO32, FHIT, FLT3LG, FOS, FOSB, FRAT1, FYB, GABARAPL1, GABARAPL3, GADD45B, GOLGA7B, GPA33, GPRASP1, GRASP, GSTM2, GZMA, GZMK, HBP1, HERPUD2, HIST1H1C, HIST1H3A, HPCAL4, HSD17B11, ID1, IDUA, IER2, IFI44, IL10RA, IL11RA, IRF2BP2, IRS2, ITGA6, JMY, JUN, JUNB, JUND, KCNQ1, KIAA1370, KIAA1683, KLF2, KLF3, KLF4, KLF5, KLF6, KLHL24, KLHL3, KLRB1, KRT72, KRT73, LIME1, LOC100128071, LOC100289511, LOC282997, LOC283070, LOC338799, LOC728392, LTBP3, MAL, MAP2K6, MDS2, MEGF6, MEGF6, MFGE8, MIR1909, MMP28, MOAP1, MXD4, MYADM, MYLIP, MYO15B, NFKBIZ, NLRC3, NLRP1, NOG, NR1D2, NR1D2, NR3C2, P2RY8, PBXIP1, PCSK5, PDE4D, PDZD4, PERI, PGAM2, PGCP, PHF1, PHF1, PIK3IP1, PIK3R5, PIM1, PION, PLCD1, PLCH2, PLCL1, PLEKHB1, PLK2, PLXDC1, PNRC1, PPP1R15A, ProSAPiPl, RAB37, RAP1GAP2, RARRES3, RASA3, RASGRP2, REM2, RGS1, RGS2, RNF125, SAMD3, SCML4, SEC31B, SIGIRR, SIK1, SLC2A3, SLC2A4RG, SLC2A4RG, SLC9A9, SLFN5, SMAD7, SMPD1, SNORA11, SORL1, SOX4, SULT1B1, SYNE1, SYTL1, TCEA3, TCF7, TCF7, TCP11L2, THEMIS, TMC8, TMEM63A, TMEM71, TMIGD2, TNFAIP3, TNNT3, TP53INP2, TPM2, TRANK1, TRIB2, TSC22D3, TSPAN18, TSPAN18, TSPAN32, TSSK3, TXK, TXNIP, UNC84B, UTRN, VIPR1, VSIG1, VSIG1, WHAMM, WNT10B, WNT7A, XAF1, XYLT1, XYLT1, YPEL2, YPEL3, YPEL5, ZBP1, ZBTB10, ZFP36, ZFP36L2, ZMAT1, ZNF331, and ZNF815.
  • Exemplary genes according to Table 2 upregulated in Treg v Teff at 16 h FC include ZBTB32, LRRC32, STAMBPL1, SNX10, LOC389333, ZNF193, GCNT1, FAS, GK3P, NTRK1, FREQ, IL1R1, CRADD, GNA15, RAB33A, IL18R1, CX3CR1, TNFRSF1B, APOBEC3G, FOXP3, SEPT11, CD70, IL1RL1, NIPA1, PANX2, CHST2, NEDD9, ACOT9, PDGFA, MAST4, TNFRSF8, PHLPP1, IL2RB, CTLA4, SYTL3, ZC3H12C, PTPRJ, UBASH3B, METRNL, PRDM1, SEPT3, TNFRSF18, WNT10A, CCR8, C18orf1, CSF1, CD80, GALNT4, GALNT4, IL1RL2, ADPRH, ZNF282, APOBEC3C, HS3ST3B1, EPAS1, RBKS, KAT2B, C9orf167, TYMP, IL1RAP, C2CD4A, CD68, ABHD4, MICAL2, C6orf145, DUSP16, LRIG1, CASK, EPSTI1, TNFRSF12A, IGSF3, SPATS2L, SPATS2L, MAF, CD58, KLHDC7B, ZBTB38, LAYN, IL1R2, HIP1, ITGB8, ITGB8, IKZF2, LGMN, XIRP1, GPR19, SAMD9L, PRF1, JAKMIP1, MGC29506, ADAMS, HLF, COL9A2, NDRG1, SAMHD1, AKAP5, RNF213, RNF213, APAF1, STX1A, SSH1, SSH1, CCRL2, CCR6, CSF2RB, HAVCR2, KLF5, MX1, ACTA2, OAS3, EMP1, CTNNAL1, MGC12916, CCL17, FOSL2, SAT1, TRPV2, PRIC285, SOCS2, ETV7, TIGIT, RASAL1, OPTN, MGST2, GPR68, MYO1G, PTPLA, TNFRSF11A, ANXA2, IRF5, C14orf139, CAPN2, LFNG, IL12RB1, MYO1E, GLRX, DENND3, ANXA2P2, NQO1, C10orf128, ANTXR2, ANTXR2, SLC26A11, FLVCR2, PREX1, SLC2A8, CDKN2A, TMEM149, SYT11, TOX, TOX2, FUT7, ANXA2P1, FAM129B, DFNB31, TMPRSS6, IL1RN, ISG15, CDKN1B, FAM129A, TST, HDAC9, TMEM110, SMPD1, CDKN1A, C17orf67, ANXA2P3, MPST, IRF7, LMCD1, SNX24, HMOX1, ATP2B4, FCER2, HPGD, RASGRP4, FAM164A, IFI6, FAM110C, XKRX, PBX4, NTNG2, CST7, BASP1, C14orf49, GLIPR1, DHRS2, TWIST1, SPSB1, CYTH4, CADM1, ITIH4, L00541471, CGA, LOC645166, PARP12, NINJ2, MICAL1, OAS1, HLA-DRB4, LGALS3, OASL, CORO2A, HLA-DRB3, KIAA1370, HERC6, STAC, MSC, CCR5, SUOX, RHOC, HLA-DQB2, PDE4A, LOC100302650, XAF1, FCRL3, RTKN2, GLIPR2, HLA-DRB1, IL13, P2RY10, IL10, CXCR6, LSP1, ACP5, SLC1A4, FXYD7, TRIB2, LMNA, HLA-DPA1, MEOX1, LGALS1, HLA-DRB5, IL10RA, HLA-DRA, CARD16, IL5, RGS1, HLA-DQA2, AKR1C3, IL4, HLA-DMA, GPR55, AQP3, MUSTN1, P2RY8, FANK1, IL9, CCNG2, ADAM12, LOC654342, IL17A, PPP2R2B, and FAM46C.
  • Exemplary genes according to Table 2 upregulated Treg vs Teff at 0 h FC4 include C12orf75, SELPLG, SWAP70, RGS1, PRR11, SPATS2L, SPATS2L, TSHR, C14orf145, CASP8, SYT11, ACTN4, ANXA5, GLRX, HLA-DMB, PMCH, RAB11FIP1, IL32, FAM160B1, SHMT2, FRMD4B, CCR3, TNFRSF13B, NTNG2, CLDND1, BARD1, FCER1G, TYMS, ATP1B1, GJB6, FGL2, TK1, SLC2A8, CDKN2A, SKAP2, GPR55, CDCA7, S100A4, GDPD5, PMAIP1, ACOT9, CEP55, SGMS1, ADPRH, AKAP2, HDAC9, IKZF4, CARD17, VAV3, OBFC2A, ITGB1, CIITA, SETD7, HLA-DMA, CCR10, KIAA0101, SLC14A1, PTTG3P, DUSP10, FAM164A, PYHIN1, MYO1F, SLC1A4, MYBL2, PTTG1, RRM2, TP53INP1, CCR5, ST8SIA6, TOX, BFSP2, ITPRIPL1, NCAPH, HLA-DPB2, SYT4, NINJ2, FAM46C, CCR4, GBP5, C15orf53, LMCD1, MKI67, NUSAP1, PDE4A, E2F2, CD58, ARHGEF12, LOC100188949, FAS, HLA-DPB1, SELP, WEE1, HLA-DPA1, FCRL1, ICA1, CNTNAP1, OAS1, METTL7A, CCR6, HLA-DRB4, ANXA2P3, STAM, HLA-DQB2, LGALS1, ANXA2, PI16, DUSP4, LAYN, ANXA2P2, PTPLA, ANXA2P1, ZNF365, LAIR2, L00541471, RASGRP4, BCAS1, UTS2, MIAT, PRDM1, SEMA3G, FAM129A, HPGD, NCF4, LGALS3, CEACAM4, JAKMIP1, TIGIT, HLA-DRA, IKZF2, HLA-DRB1, FANK1, RTKN2, TRIB1, FCRL3, and FOXP3.
  • Exemplary genes according to Table 2 upregulated in Teff at 16 h v 0 h include AARS, ABCF2, ACOT7, ACTL6A, AHSA1, AIM2, AIMP2, ALAS1, ALDH1B1, ANKRD13B, APOL1, ARMCX3, ASPHD2, B3GNT5, B4GALT2, B4GALT5, BATF, BATF3, BCAT2, BCL2L1, BOP1, BTLA, BYSL, C11orf75, C15orf23, C15orf63, C16orf59, C17orf79, C17orf96, C1orf163, C3orf26, C4orf43, C8orf30A, C9orf64, CAD, CBR1, CCDC56, CCDC86, CCL20, CCL3, CCL3L1, CCL3L3, CCL4, CCL4L1, CCL4L2, CCNB1, CCND2, CCT3, CCT5, CCT6A, CCT7, CD109, CD200, CD274, CD3EAP, CD40LG, CD82, CDC20, CDC45L, CDC6, CDK4, CDT1, CENPM, CETN3, CHAC2, CHEK1, CISD1, CISH, CKS1B, COPB2, CORO1C, CSF2, CTNNA1, CTPS, CTTN, DARS2, DCAF12, DCTPP1, DHCR24, DKC1, DTL, E2F1, EBNA1BP2, ECE2, EDARADD, EEF1E1, EGR2, EIF2B3, EIF2S1, EIF5B, EIF6, ENO1, ESPL1, EXOSC3, EXOSC4, F5, F5, FABP5, FABP5L3, FADS1, FAM40B, FARSA, FASLG, FDPS, FKBP4, FKBP4, FOSL1, FREQ, GOS2, G3BP1, GALE, GAR1, GART, GEM, GEMIN6, GEMIN7, GFOD1, GINS1, GINS2, GLRX2, GNG8, GNPDA1, GPATCH4, GPN3, GPR171, GTF2H2D, HIVEP3, HMGCS1, HN1L, HNRNPAB, HSPD1, HSPE1, HYAL2, IARS, IER3, IFNG, IFRD2, IGFBP4, IL12RB2, IL15RA, IL17F, IL2, IL21, IL22, IL2RA, IL3, IRF4, IRF8, ISOC2, KCNK5, KEAP1, KIAA0020, KIAA0664, LAG3, LAPTM4B, LARP4, LIF, LOC286016, LOC344967, LOC442308, LOC728402, LRP8, LSM2, LTA, LYAR, MANF, MATK, MCM10, MCM2, MCM3, MCM4, METTL13, MIR1182, MIR155, MIR155HG, MIR621, MPV17L2, MPZL2, MRM1, MRPL12, MRPL15, MRPL17, MRPL35, MRPL51, MRPS17, MRPS23, MRTO4, MTCH2, MTHFD1L, MTHFD2, MYOF, NAB2, NDFIP2, NDUFAF1, NFE2L3, NFKBIL2, NLN, NME1, NME1-NME2, NOLC1, NOP16, NPTX1, NT5DC2, NUDCD1, NUP43, NUP62, OTUD7B, PACSIN3, PAICS, PAK1IP1, PAM, PDCD1, PDCD2L, PDIA4, PDIA6, PEA15, PFAS, PFDN6, PFDN6, PFKM, PFKP, PGAM1, PGAM4, PHB, PHF6, PKM2, PLAGL2, PNPO, POLD2, POLE2, POLR3K, POP1, PPIL1, PPP1R14B, PRDX1, PRDX3, PRDX4, PRMT1, PRMT5, PRSS23, PSAT1, PSMA3, PSMA5, PSMA6, PSMB3, PSMB5, PSMD1, PSMD11, PSMD14, PTGFRN, PTMS, PTRH1, PTRH2, PUS7, PYCR1, PYCRL, RARS, RBBP8, RCC1, RPF2, RPP25, RRP1, RRP9, RUVBL1, RUVBL2, SAMD4A, SCD, SDC4, SECTM1, SEH1L, SEMA7A, SFT2D1, SFXN1, SH2D2A, SHF, SHMT2, SIPA1L2, SLAMF1, SLC1A5, SLC27A2, SLC27A4, SLC29A1, SLC38A5, SLC39A14, SLC43A3, SLC6A9, SLCO4A1, SNORA18, SNORD17, SORD, SPR, SQLE, SRM, SRXN1, STIP1, STT3A, TALDO1, TAP1, TBKBP1, TBL3, TBX21, TIMM8B, TIMM8B, TIPIN, TMCC2, TMEM165, TMEM194A, TMEM208, TMEM97, TNF, TNFAIP8L2, TNFRSF4, TNFRSF9, TNFSF14, TOMM40, TPI1, TRIP10, TRIP13, TTLL12, TUBA1B, TUBB, TUBB, TUBB, TUBG1, TXN, TXNDC5, UBE2T, UCK2, UGDH, UHRF1, UMPS, UTP6, VDAC1, VDR, WARS, WDR12, WDR18, WDR3, WDR4, WDR77, YIF1A, YWHAG, ZBED2, ZDHHC16, ZNF593, ZNF607, and ZWINT.
  • Exemplary genes according to Table 2 upregulated in Treg 16h vs 0 h Fc8 include AARS, ACOT7, AGRN, AHSA1, AIM2, AIMP2, ALAS1, ALDH1B1, APOL1, APOL2, B4GALT2, BATF, BATF3, BCL2A1, BCL2L1, BOP1, BYSL, C17orf96, C2CD4A, C5orf32, C9orf64, CCDC86, CCL17, CCL20, CCT5, CD3EAP, CD40LG, CD68, CD7, CDK2AP2, CDK4, CHAC2, CHPF, CISD1, CISH, COPB2, CRIM1, CSF1, CTLA4, CTSL1, CTTN, DCTPP1, DHCR24, EBI3, EBNA1BP2, ECE2, EDARADD, EGR2, EMP1, ENO1, EPAS1, EXOSC4, FABP5, FAH, FAM40B, FARSA, FKBP4, FKBP4, FLT1, FLT1, FOSL1, FREQ, G6PD, GALE, GART, GCLM, GEM, GK, GNPDA1, GPR56, HIVEP3, HMGCS1, HMOX1, HN1L, HSPA1A, HSPA1B, HSPD1, HSPE1, HYAL2, IER3, IFRD2, IKBIP, IL10, IL12RB2, IL13, IL15RA, IL17A, IL1R1, IL1R2, IL1RL2, IL1RN, IL2RA, IL3, IL4, IL4I1, IL5, IL9, IRF4, KCNK5, LAG3, LAPTM4B, LIF, LOC344967, LOC389333, LOC442308, LRRC32, LRRC61, LTA, LYAR, MANF, MATK, METRNL, METTL13, MGC29506, MICAL2, MIR1182, MIR155, MIR155HG, MLEC, MRPL12, MRTO4, MTHFD1L, MYOF, NAB2, NDFIP2, NDUFAF1, NKG7, NLN, NME1, NME1-NME2, NOP16, NPM3, NUDCD1, PAICS, PANX2, PDCD1, PDGFA, PDIA4, PDIA6, PFAS, PGAM4, PHB, PNPO, POP1, PPIL1, PPPDE2, PRDX1, PRDX3, PRDX4, PRKAR1B, PRMT1, PRMT5, PSAT1, PSMB5, PSMD1, PSMD11, PTGFRN, PTRH1, PUS7, PYCR1, RASAL1, RBBP8, RCC1, SC4MOL, SCD, SDC4, SECTM1, SEH1L, SEMA7A, SETP11, SERPINE2, SERPINH1, SH2D2A, SLC16A13, SLC16A3, SLC1A5, SLC27A2, SLC27A4, SLC29A1, SLC38A5, SLC39A1, SLC39A14, SLC43A3, SLCO4A1, SOCS1, SPHK1, SPINT1, SQLE, SRM, SRXN1, STIP1, STT3A, TBKBP1, TBX21, TMPRSS6, TNF, TNFRSF11A, TNFRSF12A, TNFRSF18, TNFRSF1B, TNFRSF4, TNFRSF8, TNFRSF9, TNFSF14, TOMM40, TRIP10, TTLL12, TUBB, TUBB, TUBB, TXN, TYMP, UCK2, UGDH, VDR, VTRNA1-3, WARS, WDR12, WDR4, WDR77, XIRP1, YWHAG, ZBED2, ZBTB32, ZDHHC16, and ZNF282.
  • An exemplary list of the TREG genes upregulated at 16 h include AIM2, ALAS1, BATF, C5orf32, CCL17, CD40LG, CHAC2, CSF1, CTSL1, EBNA1BP2, EDARADD, EMP1, EPAS1, FABP5, FAM40B, FKBP4, FOSL1, GCLM, GK, GPR56, HMOX1, HSPD1, HSPE1, IKBIP, IL10, IL13, IL15RA, IL1RN, IL2RA, IL3, IL4, IL5, IL9, KCNK5, LTA, MANF, MIR1182, MIR155, MIR155HG, MYOF, NDUFAF1, NLN, NME1, NME1-NME2, PANX2, PDIA6, PGAM4, PPIL1, PPPDE2, PRDX4, PRKAR1B, PSMD1, PSMD11, PUS7, RBBP8, SLC27A2, SLC39A14, SLC43A3, SRXN1, STIP1, STT3A, TBX21, TNFRSF11A, TNFRSF1B, TNFRSF8, TNFRSF9, TXN, UCK2, VDR, VTRNA1-3, WDR12, YWHAG, ZDHHC16, and ZNF282. The upregulated expression may be determined, e.g., by measuring RNA levels for the indicated genes.
  • An exemplary list of the TEFF genes upregulated at 16 h include AIM2, ALAS1, B4GALT5, BATF, C3orf26, C4orf43, CCL3, CCL4, CCT3, CCT7, CD40LG, CHAC2, CSF2, CTNNA1, EBNA1BP2, EDARADD, EEF1E1, EIF2B3, EIF2S1, FABP5, FAM40B, FKBP4, FOSL1, GFOD1, GLRX2, HSPD1, HSPE1, IFNG, IL15RA, IL21, IL2RA, IL3, KCNK5, KIAA0020, LARP4, LRP8, LTA, MANF, MIR1182, MIR155, MIR155HG, MTCH2, MYOF, NDUFAF1, NLN, NME1, NME1-NME2, OTUD7B, PAM, PDIA6, PEA15, PFKM, PGAM1, PGAM4, PPIL1, PRDX4, PRSS23, PSMD1, PSMD11, PSMD14, PTRH2, PUS7, RBBP8, RPF2, RPP25, SFXN1, SLC27A2, SLC39A14, SLC43A3, SORD, SPR, SRXN1, STIP1, STT3A, TBX21, TMCC2, TMEM165, TNFRSF9, TXN, TXNDC5, UCK2, VDR, WDR12, YWHAG, and ZDHHC16.
  • The Abbas gene sets compared the expression profiles of 17 immune cell types and identified genes uniquely expressed in certain cell types relative to others. The select Abbas gene sets that were included in the Gene Set Analysis are listed in Table 3 and include CD4+ T cells naïve and resting, CD8+ T cells naïve and resting, helper Th1 at 12 hours, helper Th1 at 48 hours, helper Th2 at 12 hours, helper Th2 at 48 hours, memory T resting (naïve) cells, and memory T activated cells.
  • TABLE 3
    Table 3: Gene sets comprised of gene up or down regulated
    resting and activated T-cell subtypes (Abbas data set)
    Gene Set Number of genes
    Downregulated CD8 vs CD4 Naïve T-cells 200
    Downregulated Naïve CD4 vs 12 H activated Th1 200
    Downregulated Naïve CD4 vs 48 H activated Th1 200
    Downregulated Naïve CD4 vs 12 H activated Th2 200
    Downregulated Naïve CD4 vs 48 H activated Th2 200
    Downregulated Th1 vs Th2 12 H activated 200
    Downregulated Th1 vs Th2 48 H activated 200
    Downregulated unstimulated vs stimulated memory 200
    T-cells
    Upregulated CD8 vs CD4 Nave T-cells 200
    Upregulated Naïve CD4 vs 12 H activated Th1 200
    Upregulated Naïve CD4 vs 48 H activated Th1 200
    Upregulated Naïve CD4 vs 12 H activated Th2 200
    Upregulated Naïve CD4 vs 48 H activated Th2 200
    Upregulated Th1 vs Th2 12 H activated 200
    Upregulated Th1 vs Th2 48 H activated 200
    Upregulated unstimulated vs stimulated memory 200
    T-cells
  • The Gattinoni gene sets compared the expression profiles of various CD8+ memory T cell subsets. Specifically, immune cells were isolated from healthy donors and the following CD8+T memory subsets were purified: TN (naïve), TSCM (memory stem cells), TCM (central memory), TEM (effector memory). Gene sets were defined by comparing between all pairs of groups (e.g. TSCM vs. TN) and by identifying those genes that either progressively increased or decreased across the 4 conditions in order from TN→TSCM→TCM→TEM. The select gene sets from Gattinoni et al. that were considered in the Gene Set Analysis are tabulated in Table 4.
  • TABLE 4
    Table 4: Gene sets comprised of gene up or down regulated
    resting and activated T-cell subtypes (Gattinoni data set)
    Gene Set Number of genes
    TCM vs TEM 29
    TN vs TCM 148
    TN vs TEM 212
    TSCM vs TCM 19
    TSCM vs TEM 75
    TSCM vs TN 73
    Progressively down 208
    Progressively up 32
  • Each gene set (e.g., ALL and CLL RNAseq gene sets, Szabo gene sets, Abbas gene sets, and Gattinoni gene sets) was evaluated to determine its association with subject response (i.e., CR, PR, or NR) in the following manner: a meta-gene was calculated for each subject, where the meta-gene score for subject j was defined as

  • m ji=G 1 x ij−μ(x .j)/σ(x .j)
  • where xij is the expression value of gene i in subject j for a given gene set n=1, . . . , G; μ(x.j) is the mean of genes 1, . . . , G in subject j; and σ(x.j) is the standard deviation of genes 1, . . . , G in subject j.
  • A 3-group statistical model was applied to each gene set to determine whether the meta-gene was statistically different between the CLL product CRs, PRs, and NRs. A schematic illustrating this approach is given in FIG. 2B. CRs are more like resting TEFF cells, whereas NR are more like activated TEFF cells. CTL019 NR samples are in a more activated state than CR samples. Gene sets that were found to be significantly altered and predictive of patient response to CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., CTL019) are listed in Table 5.
  • TABLE 5
    Table 5: Gene sets that are enriched in CLL samples
    Prod- Aphe-
    Gene Set Source CRs NRs uct resis
    TEFF 16 h vs 0 h Szabo TEFF oh TEFF 16 h X X
    TREG 16 h vs 0 h Szabo TREG oh TREG 16 h X X
    TREG vs TEFF 16 h Szabo TEFF TREG X X
    Naïve CD4 vs Abbas Naïve Th1 X X
    12 H act Th1 CD4
    Naïve CD4 vs Abbas Naïve Th2 X X
    12 H act Th2 CD4
    Naïve CD4 vs Abbas Naïve Th1 X X
    48 H act Th1 CD4
    Naïve CD4 vs Abbas Naïve Th2 X X
    48 H act Th2 CD4
    Unstim vs stim Abbas Unstim- Stimulated X
    memory ulated
    TSCM vs TEM Gattinoni TSCM TEM X
    TSCM vs TCM Gattinoni TSCM TCM X
    TCM vs TEM Gattinoni TEM TCM X
    Progressively Gattinoni Early Late stage X X
    down stage
  • FIG. 3 depicts an exemplary schematic of memory T cell precursors and subsets. Without wishing to be bound by a particular theory, the state of memory T cells in CTL019 samples is likely a major component of response.
  • For a subset of the patients in the manufactured product study described in Example 1, whole genome RNAseq was also performed on T cells collected by apheresis. The gene sets described above were evaluated in these 14 apheresed samples (2 CRs, 3 PRs, and 9 NRs). Gene sets that were found to be significantly altered and predictive of patient response to CTL019 therapy are tabulated in Table 5.
  • Whole genome RNAseq was performed on 7 ALL manufactured product CR samples and 4 ALL apheresis CR samples. Meta-gene scores for each gene set were calculated for the ALL samples as described above for the CLL samples. Gene sets with meta-gene scores correlating with the expected pattern of response in product and apheresis samples (ALL→CLL CR→CLL PR→CLL NR) are tabulated in Table 6. Gene sets marked with * in Table 6 are also correlated with response in apheresis samples.
  • TABLE 6
    Table 6: Gene sets correlating
    to response over product ALL and CLL
    samples (ALL → CLL CR → CLL PR → CLL NR)
    Gene Set
    Downregulated Treg vs Teff 0 h (FC3 p < 0.05)*
    Downregulated Treg vs Teff 16 h (FC3 p < 0.05)
    Upregulated Treg vs Teff 0 h (FC4 p < 0.05)*
    Upregulated Treg vs Teff 16 h (FC4 p < 0.05)
    TCMvsTEM down gene set
    TCMvsTEM up gene set
    TNvsTCM down gene set*
    TNvsTCM up gene set*
    TNvsTEM down gene set*
    TNvsTEM up gene set*
    TSCMvsTCM down gene set*
    TSCMvsTCM up gene set*
    TSCMvsTEM up gene set*
    TSCMvsTN down gene set*
    TSCMvsTN up gene set*
    Progressively down*
    Progressively up*
    Downregulated CD8 vs CD4 Nave T-cells*
    Upregulated Naïve CD4 vs 12 H activated Th1
    Upregulated Naïve CD4 vs 48 H activated Th1
    Upregulated Naïve CD4 vs 12 H activated Th2
    Upregulated Naïve CD4 vs 48 H activated Th2*
    Downregulated Th1 vs Th2 12 H activated
  • For example, the meta-gene score for the gene set comprised of genes upregulated in TSCM in comparison to TCM is found to be correlated with response in both apheresis and product samples, FIG. 4. The meta-gene scores from healthy donor samples with manufactured product are included in the plot to serve as a reference point. The x-axis is samples by response group where a=apheresis and p=product. The y-axis is normalized meta-gene expression scores. Gene sets enriched in CLL CRs (e.g., CTL019 CRs) are also enriched in acute lymphoblastic leukemias (ALLs). ALL and CLL CRs are enriched in T stem cell (TSCM) subset specific genes, whereas CLL PRs and NRs are enriched in T central memory (TCM) subset genes. The same pattern is seen in apheresis as in product samples. ALL expression patters are most similar to CLL CRs and are even more extreme in the direction of resting/unstimulated/early memory T cells.
  • FIG. 5A depicts an exemplary result from a Principle Component Analysis (PCA) of CTL019 samples. This exemplary PCA result illustrates that CRs, ALL and Normal samples cluster separately from PRs and NRs. FIG. 5B depicts an exemplary result from a PCA of CTL019 and apheresis samples. This exemplary PCA result illustrates that CRs, ALL and Normal samples cluster separately from PRs and NRs and from the apheresis cluster.
  • FIG. 6 depicts an exemplary schematic depicting immunophenotyping of apheresis and product samples.
  • Manufactured CTL019 product (e.g., genetically engineered CAR19-expressing T cells obtained from CLL patients) classified as complete responders (CR), partial responders (PR), non-responders (NR), or pending were assessed for expression of immune checkpoint inhibitor molecules, such as PD-1, LAG3, and TIM3.
  • CD 19 CAR-expressing cells (e.g., T cells, NK cells) from CLL patients (e.g., manufactured product) with different responses to CAR-expressing cell therapy were analyzed by flow cytometery to determine percentages of CD4+ and CD8+ T cells. The CD19 CAR-expressing cells were from: patients that responded to CAR-expressing cell therapy (CR) (n=5); patients that partially responded to CAR-expressing cell therapy (n=8), patients that did not respond to CAR-expressing cell therapy (NR) (n=19); and patients that were pending, e.g., not yet assigned to a group (NA) (n=3). Cells were labeled with antibodies that specifially recognize CD4, CD8, the CAR19 molecule, and immune checkpoint molecules PD-1, LAG3, and TIM3, and secondary antibodies conjugated to fluoresceins, according to standard methods for flow cytometry analysis known in the art. Expression of each marker, e.g., CD4+, CD8+, etc., was deterined by flow cytometry analysis software, and subpopulations (e.g., CD4+ T cells, CD8+T cells, or CAR19-expressing T cells) were further analyzed fro the expression of immune checkpoint molecules PD-1, LAG3, and TIM3.
  • Using the methods and analysis described above, the percentage of CD4-expressing cells and CD8-expressing cells was determined for each patient in each response group. As described above, 36 manufactured CTL019 samples from CLL patients were analyzed, and included 5 CR, 8 PR, 19NR and 3 pending. FIG. 7A depicts an exemplary result illustrating percent CD4+ cells and patient response. Partial responders were shown to have a statistically significant greater percentage of CD4+ cells. FIG. 7B depicts an exemplary result illustrating percent CD8+ cells and patient response. Complete responders were shown to have a statistically significant great percentage of CD8+ cells.
  • An example of the flow cytometry profiles analysis used to determine surface marker expression is shown in FIGS. 8A and 8B. Cells expressing CD4+ were determined using flow cytometry, and were further analyzed for CAR19 and PD-1 expression, such that the x-axis of the profiles indicate CAR19 expression (the top left (Q5) and bottom left (Q8) quadrants show the CAR19-negative CD4+ cells, while the top right (Q6) and bottom right (Q7) quadrants show the CAR19-expressing CD4+ cells) and the y-axis shows PD-1 expression (the bottom left (Q8) and right (Q7) quadrants show the PD-1 negative CD4+ cells and the top left (Q5) and right (Q6) quadrants show the PD-1− expressing CD4+ cells). In the CD4+ population from a CAR-expressing cell (e.g., T cell, NK cell) responder, 44.7% of the CD4+ cells overall expressed PD-1, and about 22.3% of the CAR19-expressing cells were PD-1 positive, while 27.2% of CAR19-expressing cells were PD-1 negative (FIG. 8A). In contrast, in the CD4+ population from a non-responder, there was a significant decrease in CAR19-expressing cells overall (about 15.3% compared to the 49.5% in CR), with 14.7% of the CAR19-expressing cells being PD-1 positive while only 0.64% were PD-1 negative (FIG. 8B). Comparison between the profiles in FIG. 8A and FIG. 8B shows that a much higher percentage of the CD4+ cells from a non-responder express PD-1 (about 92.9%) compared to the CAR-expressing cell responder (about 44.7%).
  • Using the methods and analysis described above, the percentage of PD-1 expressing (PD-1+) cells of the CD4+ population and the CD8+ population was determined for each patient in each response group. Non-responders were shown to have a greater percentage of PD-1+ cells in both the CD4+(FIG. 8C) and CD8+(FIG. 8D) populations compared to those that responded to CAR therapy (CR); the increase of average PD-1 percentage was statistically significant for both CD4+ and CD8+ populations. Partial responders (PR) exhibited higher percentages of PD-1+ cells than responders (CR) in both CD4+(FIG. 8C) and CD8+(FIG. 8D) populations.
  • Further analysis was performed to determine the distribution of cells expressing PD-1, LAG3, and TIM3 from patients with different responses to CAR therapy. Representative cell profile analysis for PD-1, LAG3, and TIM3 expression in the CD4+ population are shown in FIG. 9 and FIG. 10. The cell populations were first analyzed for CAR19+ expression. The CAR19+ population was then analyzed for PD-1 and LAG3 expression (FIG. 9) or PD-1 and TIM-3 expression (FIG. 10). In the LAG3+ population from a CAR-expressing cell (e.g., T cell, NK cell) responder, 36.1% of the CAR19+ cells overall expressed PD-1, and about 7.3% of the LAG3-expressing cells were PD-1 positive, while 5.9% of LAG3-expressing cells were PD-1 negative (FIG. 9). In contrast, in the CAR19+ population from a non-responder, there was a significant increase in LAG3-expressing cells overall (about 69.7% compared to the 13.2% in CR), with 67.3% of the LAG3-expressing cells being CAR19+ positive while only 2.41% were PD-1 negative (FIG. 9). Comparison between the CR and NR flow cytometry profiles in FIG. 9 show that a much higher percentage of the LAG3+ cells from a non-responder express PD-1 (about 67.3%) compared to the CAR-expressing cell (e.g., T cell, NK cell) responder (about 7.3%).
  • Using the methods and analysis described above, the percentage of PD-1 and LAG-3 expressing (PD-1+/LAG-3+) cells of the CAR19+ population was determined for each patient in each response group. Non-responders were shown to have a greater percentage of PD-1+/LAG-3+ cells in the CAR19+ populations compared to those that responded to CAR therapy (CR) (FIG. 9); the increase of average PD-1/LAG-3 percentage was statistically significant for the CAR19+ population. Partial responders (PR) exhibited higher percentages of PD-1+/LAG-3+ cells than responders (CR) in the CAR19+(FIG. 9) population. In an embodiment, NR products exhibit an exhausted phenotype of PD1+CAR+ and co-expression of LAG3.
  • Next, the CAR19+ population was analyzed for PD-1 and TIM-3 expression (FIG. 10). In the TIM+ population from a CAR-expressing cell (e.g., T cell, NK cell) responder, 28.5% of the CAR19+ cells overall expressed PD-1 (FIG. 10). In contrast, in the CAR19+ population from a non-responder, there was a significant increase in TIM3+/PD1+ cells, with 83.3% of the CAR19+-expressing cells being TIM3+/PD1+(FIG. 10).
  • Using the methods and analysis described above, the percentage of PD-1 and TIM-3 expressing (PD-1+/TIM-3+) cells of the CAR19+ population was determined for each patient in each response group. Non-responders were shown to have a greater percentage of PD-1+/TIM-3+ cells in the CAR19+ populations compared to those that responded to CAR therapy (CR) (FIG. 10); the increase of average PD-1/TIM-3 percentage was statistically significant for the CAR19+ population. Partial responders (PR) exhibited higher percentages of PD-1+/TIM-3+ cells than responders (CR) in the CAR19+(FIG. 10) population. In an embodiment, NR products exhibit an exhausted phenotype of PD1+CAR+ and co-expression of TIM3.
  • Cells expressing CD4+ and CD8+ were determined using flow cytometry, and were further analyzed for CD27+ expression. Using the methods and analysis described above, the percentage of CD27 expressing (CD27+) cells of the CD4+ population and the CD8+ population was determined for each patient in each response group. Complete responders (CR) and partial responders (PR) were shown to have a greater percentage of CD27+ cells in both the CD4+(FIG. 11A) and CD8+(FIG. 11B) populations compared to non-responders (NR); the increase of average CD27 percentage was statistically significant for both CD4+ and CD8+ populations. Partial responders (PR) exhibited higher percentages of CD27+ cells than complete responders (CR) in CD4+(FIG. 11A) populations. Complete responders (CR) exhibited higher percentages of CD27+ cells than partial responders (PR) in CD8+(FIG. 11B) populations. In an embodiment, CD27 levels in a CAR product correlate with patient response. In an embodiment, CRs CD8+ cells display higher percentages of CD27+ cells as compared to PRs and NRs.
  • FIG. 12 depicts an exemplary multi-color flow cytometry analysis result identifying correlates of response in apheresis samples. 26 apheresed samples from CLL patients were analyzed. Samples included 4 CR, 6 PR, 14NR and 1 patient was not infused.
  • FIG. 13 depicts an exemplary multi-color flow cytometry analysis result illustrating a correlation between a younger T cell phenotype and response to CTL019 therapy. These data demonstrate that the percentage of CD27+CD45RO− in CD8+ T cells is predictive of which CLL patients will undergo a complete response to CTL019.
  • FIG. 14 depicts an exemplary analysis of apheresis in a human patient prior to CTL019 therapy. Exemplary results illustrate that while patient 1000-00045 presented with very few T cells, 27% of the T cells were CD8+CD27+CD45RO−.
  • FIG. 15 depicts an exemplary result of a patient response (patient 1000-00045) to CTL019 therapy. CD8+CD27+CD45RO− T cells were a positive predictor of the patient response to CTL019 therapy. These exemplary results illustrate that a good prognostic phenotype in apheresis is a high percentage of CD8+CD27+CD45RO− T cells (young phenotype). A poor prognostic phenotype in CTL019 product is a high percentage of PD1+CAR+ and LAG3+ or TIM3+ T cells (exhausted phenotype).
  • Significant gene sets from the analyses above were refined to a subset of genes within the gene sets that are significantly differentially expressed between ALL CRs/CLL CRs and CLL NRs as well as following the expected expression pattern of increasing or decreasing from ALL→CLL CR→CLL PR→CLL NR. An exemplary listing of genes that were significantly differentially expressed are listed in Table 7A. Table 7A is an exemplary list of biomarkers whose expression values predict patient response to CTL019 therapy. Table 7A was further refined to produce a flow cytometry biomarker gene panel by selecting for genes that are cell surface markers. Exemplary cell surface genes that predict patient response to CTL019 therapy are shown in Table 8.
  • TABLE 7A
    Table 7A: Exemplary genes that predict patient response to CTL019 therapy
    Gene Unigene Accession No.
    ABCA7 Hs.134514 NM_019112
    ABTB1 Hs.107812 NM_172027, NM_172028,
    NM_032548
    ACOT9 Hs.298885 NM_001037171, NM_001033583
    ACTA2 Hs.500483 NM_001141945, NM_001613
    ADAMTS10 Hs.657508 NM_030957
    ADD3 Hs.501012 NM_016824, NM_019903, NM_001121
    ADPRH Hs.99884 NM_001125
    AEBP1 Hs.439463 NM_001129
    AES Hs.515053 NM_198970, NM_198969, NM_001130
    AIM2 Hs.281898 NM_004833
    ALAS1 Hs.476308 NM_199166, NM_000688
    ALPK1 Hs.652825 NM_025144, NM_001102406
    ALS2CL Hs.517937 NM_147129, NM_182775
    AMD1 Hs.159118 NM_001634, NM_001033059
    ANKRD55 Hs.436214 NM_024669, NM_001039935
    ANKZF1 Hs.437647 NM_018089, NM_001042410
    ANTXR2 Hs.162963, NM_001145794, NM_058172
    Hs.720941
    ANXA2 Hs.591361, NM_004039, NM_001136015,
    Hs.546235, NM_001002858, NM_001002857
    Hs.511605
    ANXA2P2 Hs.534301
    AP1G2 Hs.343244 NM_003917
    AP1M1 Hs.71040 NM_032493, NM_001130524
    AP2A2 Hs.19121 NM_012305
    APAF1 Hs.552567 NM_181869, NM_181868, NM_013229,
    NM_001160, NM_181861
    APBA2 Hs.721380, NM_001130414, NM_005503
    Hs.618112
    APBB3 Hs.529449 NM_133174, NM_133173,
    NM_133172, NM_006051
    AQP3 Hs.234642 NM_004925
    ARFGAP2 Hs.436204 NM_032389
    ARHGAP33 Hs.515364 NM_052948
    ARHGEF1 Hs.631550 NM_004706, NM_199002, NM_198977
    ARHGEF11 Hs.516954 NM_198236, NM_014784
    ARHGEF18 Hs.465761 NM_001130955, NM_015318
    ARL4C Hs.723194, NM_005737
    Hs.111554
    ARPC5L Hs.132499 NM_030978
    ARRB1 Hs.625320, NM_020251, NM_004041
    Hs.503284
    ARRDC2 Hs.515249 NM_015683, NM_001025604
    ARSB Hs.604199, NM_000046, NM_198709
    Hs.149103
    ATOX1 Hs.125213 NM_004045
    ATP13A3 Hs.529609 NM_024524
    ATP1B3 Hs.477789 NM_001679
    ATP2A2 Hs.506759 NM_001135765, NM_170665,
    NM_001681
    ATP2B4 Hs.343522, NM_001001396, NM_001684
    Hs.511311
    ATP8B4 Hs.511311 NM_024837
    ATXN7L3B Hs.744849 NM_001136262
    AVEN Hs.555966 NM_020371
    B4GALT5 Hs.370487 NM_004776
    BATF Hs.509964 NM_006399
    BCL9L Hs.414740 NM_182557
    BENDS Hs.475348 NM_024603
    BEX4 Hs.184736 NM_001080425, NM_001127688
    BIN1 Hs.193163 NM_139350, NM_139348, NM_139349,
    NM_139343, NM_004305, NM_139345,
    NM_139344, NM_139347, NM_139351,
    NM_139346
    BNIP3L Hs.131226 NM_004331
    BTN3A1 Hs.191510 NM_001145008, NM_007048,
    NM_194441, NM_001145009
    C10orf128 Hs.385493 NM_001010863
    C11orf10 Hs.437779 NM_014206
    C11orf21 Hs.559181 NM_001142946
    C11orf35 Hs.669395 NM_173573
    C12orf5 Hs.504545 NM_020375
    C16orf54 Hs.331095 NM_175900
    C16orf74 Hs.461655 NM_206967
    C17orf48 Hs.47668 NM_020233
    C17orf67 Hs.658949 NM_001085430
    C19orf29 Hs.267446 NM_001080543, NM_021231
    C1QBP Hs.555866 NM_001212
    C20orf11 Hs.353013 NM_017896
    C20orf112 Hs.516978 NM_080616
    C21orf2 Hs.517331 NM_004928
    C2orf67 Hs.591638, NM_152519
    Hs.282260
    C3orf26 NM_001167924, NM_032359
    C4orf43 NM_018352
    C5orf13 Hs.483067, NM_001142475, NM_001142476,
    Hs.36053, NM_004772, NM_001142482,
    Hs.694860 NM_001142477, NM_001142483,
    NM_001142478, NM_001142474,
    NM_001142481, NM_001142479,
    NM_001142480
    C5orf30 Hs.482976 NM_033211
    C5orf32 Hs.529798 NM_032412
    C5orf39 Hs.529385, NM_001014279
    Hs.721020
    CABIN1 Hs.517478 NM_012295
    CACHD1 Hs.443891 NM_020925
    CADM1 Hs.370510 NM_014333, NM_001098517
    CAPG Hs.516155 NM_001747
    CAPS Hs.584744 NM_004058, NM_080590
    CASK Hs.495984 NM_001126054, NM_001126055,
    NM_003688
    CBX4 Hs.405046 NM_003655
    CCDC47 Hs.202011 NM_020198
    CCL17 Hs.546294 NM_002987
    CCL3 Hs.514107 NM_002983
    CCL4 Hs.75703 NM_002984
    CCR1 Hs.301921 NM_001295
    CCT2 Hs.189772 NM_006431
    CCT3 Hs.491494 NM_001008800, NM_005998,
    NM_001008883
    CCT7 Hs.368149 NM_001009570, NM_006429,
    NM_001166284, NM_001166285
    CD248 Hs.195727 NM_020404
    CD40LG Hs.592244 NM_000074
    CD58 Hs.34341 NM_001144822, NM_001779
    CD70 Hs.715224, NM_001252
    Hs.501497
    CD80 Hs.838 NM_005191
    CDC123 Hs.412842 NM_006023
    CDC25B Hs.153752 NM_004358, NM_021872, NM_021873
    CDC42BPG Hs.293590 NM_017525
    CDK7 Hs.184298 NM_001799
    CDKN1A Hs.370771 NM_078467, NM_000389
    CDKN2A Hs.512599 NM_058197, NM_058195, NM_000077
    CERK Hs.200668 NM_022766
    CFP Hs.53155 NM_001145252, NM_002621
    CHAC2 Hs.585944 NM_001008708
    CHI3L2 Hs.514840 NM_001025199, NM_001025197,
    NM_004000
    CHMP7 Hs.5019 NM_152272
    CLDND1 Hs.531371 NM_001040181, NM_001040183,
    NM_001040200, NM_001040199,
    NM_001040182, NM_019895
    CLTC Hs.491351 NM_004859
    CNN3 Hs.483454 NM_001839
    CNOT8 Hs.26703 NM_004779
    CNPY3 Hs.414099 NM_006586
    COQ3 Hs.713623 NM_017421
    CSF1 Hs.591402 NM_000757, NM_172212, NM_172211,
    NM_172210
    CSF2 Hs.1349 NM_000758
    CSNK2A1 Hs.654675, NM_001895, NM_177560, NM_177559
    Hs.644056
    CST7 Hs.143212 NM_003650
    CTC1 Hs.156055 NM_025099
    CTDSP1 Hs.444468 NM_182642, NM_021198
    CTDSP2 Hs.524530 XM_001720210, XM_001722552,
    XM_002344384, XM_001725997,
    NM_005730
    CTNNA1 Hs.656653, NM_001903
    Hs.445981
    CTSL1 Hs.418123 NM_001912, NM_145918
    CUL9 Hs.485434 NM_015089
    CUTA Hs.520070 NM_001014433, NM_001014840,
    NM_015921, NM_001014838,
    NM_001014837
    CYFIP1 Hs.26704 NM_014608, NM_001033028
    CYP2J2 Hs.152096 NM_000775
    DBP Hs.414480, NM_001352
    Hs.528006
    DCAF11 Hs.525251 NM_001163484, NM_181357,
    NM_025230
    DCBLD2 Hs.203691 NM_080927
    DCHS1 Hs.199850 NM_003737
    DCTN6 Hs.158427 NM_006571
    DDX10 Hs.591931 NM_004398
    DENND2D Hs.557850 NM_024901
    DENND5A Hs.501857 NM_015213
    DERL1 Hs.241576 NM_001134671, NM_024295
    DFNB31 Hs.93836 NM_001083885, NM_015404
    DGKD Hs.471675 NM_152879, NM_003648
    DGKZ Hs.502461 NM_001105540, NM_003646,
    NM_201533, NM_201532
    DHRS2 Hs.272499 NM_182908, NM_005794
    DIABLO Hs.169611 NM_138929, NM_019887
    DNAJB6 Hs.490745 NM_005494, NM_058246
    DPEP2 Hs.372633 NM_022355
    DUSP22 Hs.29106 NM_020185, XM_001718070
    E2F6 Hs.603093 NM_198256
    EBNA1BP2 Hs.346868 NM_006824, NM_001159936
    EDARADD Hs.352224 NM_080738, NM_145861
    EED Hs.503510 NM_152991, NM_003797
    EEF1E1 Hs.602353, NM_004280, NM_001135650
    Hs.723203
    EGFL6 Hs.12844 NM_001167890, NM_015507
    EHD1 Hs.523774 NM_006795
    EIF2B3 Hs.533549 NM_001166588, NM_020365
    EIF2S1 Hs.151777 NM_004094
    ELL2 Hs.708710, NM_012081
    Hs.192221
    EMP1 Hs.436298 NM_001423
    EPAS1 Hs.468410 NM_001430
    EPHA4 Hs.371218 NM_004438
    EPHX1 Hs.89649 NM_001136018, NM_000120
    EPPK1 Hs.200412 NM_031308
    ERGIC2 Hs.339453 NM_016570
    ERGIC3 Hs.472558 NM_015966, NM_198398
    ERP29 Hs.75841 NM_001034025, NM_006817
    ETFA Hs.39925 NM_001127716, NM_000126
    ETNK1 Hs.29464 NM_001039481, NM_018638
    ETV7 Hs.272398 NM_016135
    FAAH Hs.720143 NM_001441
    FABP5 Hs.408061 NM_001444
    FAF2 Hs.484242 NM_014613
    FAIM3 Hs.723317, NM_001142472, NM_001142473,
    Hs.58831 NM_005449
    FAM117B Hs.471130 NM_173511
    FAM134B Hs.711125 NM_001034850, NM_019000
    FAM13A Hs.97270 NM_014883, NM_001015045
    FAM193B Hs.484289 NM_019057
    FAM40B Hs.489988 NM_020704, NM_001134336
    FAM63A Hs.723127 NM_018379, NM_001163260,
    NM_001163259, NM_001163258,
    NM_001040217
    FAM65B Hs.559459 NM_014722, NM_015864
    FANCL Hs.720331 NM_001114636, NM_018062
    FANK1 Hs.352591 NM_145235
    FAR2 Hs.298851 NM_018099
    FAU Hs.387208 NM_001997
    FCER1G Hs.433300 NM_004106
    FCER2 Hs.465778 NM_002002
    FCGBP Hs.111732 NM_003890, XM_001717543
    FCHO1 Hs.96485 NM_001161358, NM_001161357,
    NM_001161359, NM_015122
    FCRL3 Hs.292449 NM_052939
    FGD3 Hs.411081 NM_033086, NM_001083536
    FGF9 Hs.111 NM_002010
    FKBP4 Hs.713721, NM_002014
    Hs.524183
    FLOT2 Hs.514038 NM_004475
    FLT3LG Hs.428 NM_001459
    FLVCR2 Hs.615289, NM_017791
    Hs.509966
    FOSL1 Hs.283565 NM_005438
    FOSL2 Hs.596972, NM_005253
    Hs.220971
    FRAT1 Hs.126057 NM_005479
    GAL3ST4 Hs.44856 NM_024637
    GALNT4 Hs.713979, NM_003774
    Hs.25130
    GCLM Hs.315562 NM_002061
    GCNT1 Hs.521568 NM_001490, NM_001097633,
    NM_001097635, NM_001097634,
    NM_001097636
    GFOD1 Hs.484686 NM_018988
    GFPT1 Hs.580300 NM_002056
    GIPC3 Hs.266873 NM_133261
    GK Hs.1466, NM_001128127, NM_000167,
    Hs.654557 NM_203391
    GLRX2 Hs.458283 NM_016066, NM_197962
    GMEB2 Hs.473286 NM_012384
    GNAI1 Hs.134587 NM_002069
    GPA33 Hs.651244 NM_005814
    GPD1L Hs.82432 NM_015141
    GPKOW Hs.503666 NM_015698
    GPR125 Hs.99195 NM_145290
    GPR56 Hs.513633 NM_001145773, NM_001145774,
    NM_001145771, NM_001145772,
    NM_005682, NM_201525,
    NM_001145770, NM_201524
    GPSM3 Hs.520046 NM_022107
    GRAP Hs.567416 NM_006613
    GRASP Hs.407202 NM_181711
    GTF2A2 Hs.512934 NM_004492
    HAVCR1 Hs.129711 NM_001099414, NM_012206
    HBS1L Hs.378532 NM_001145207, NM_001145158,
    NM_006620
    HDAC9 Hs.196054 NM_014707, NM_178423, NM_178425,
    NM_058176, NM_058177
    HIGD1A Hs.711098, NM_014056, NM_001099669,
    Hs.593134, NM_001099668
    Hs.7917
    HIP1 Hs.329266, NM_005338
    Hs.619089
    HLA-DMA Hs.351279 NM_006120
    HLA-DPA1 Hs.347270 NM_033554
    HLA-DQA2 Hs.591798 NM_020056
    HLA-DQB2 Hs.719990 NM_001198858, NM_001300790
    HLA-DRA Hs.520048 NM_019111
    HLA-DRB1 Hs.716081, NM_002124, NM_021983,
    Hs.696211, XM_002346251
    Hs.723344,
    Hs.534322
    HLA-DRB5 Hs.534322 NM_002125
    HLF Hs.196952 NM_002126
    HMOX1 Hs.517581 NM_002133
    HSPD1 Hs.595053, NM_199440, NM_002156
    Hs.723164
    HSPE1 Hs.1197 NM_002157
    HYI Hs.709864 NM_031207
    ICAM3 Hs.654563 NM_002162
    IDUA Hs.89560 NM_000203
    IER2 Hs.501629 NM_004907
    IFNAR2 Hs.708195 NM_207584, NM_207585, NM_000874
    IFNG Hs.856 NM_207585
    IGF1R Hs.643120, NM_000875
    Hs.714012
    IGSF3 Hs.171057 NM_001007237, NM_001542
    IGSF9B Hs.204121 NM_014987
    IKBIP Hs.252543 NM_201612, NM_201613, NM_153687
    IL10 Hs.193717 NM_000572
    IL11RA Hs.591088 NM_004512, NM_147162,
    NM_001142784
    IL13 Hs.845 NM_002188
    IL15RA Hs.524117 NM_002189, NM_172200
    IL1RAP Hs.478673 NM_134470, NM_001167930,
    NM_001167928, NM_001167929,
    NM_002182
    IL1RL1 Hs.66 NM_003856, NM_016232
    IL1RN Hs.81134 NM_000577, NM_173841, NM_173842,
    NM_173843
    IL21 Hs.567559 NM_021803
    IL2RA Hs.231367 NM_000417
    IL2RB Hs.474787 NM_000878
    IL3 Hs.694 NM_000588
    IL4 Hs.73917 NM_000589, NM_172348
    IL5 Hs.2247 NM_000879
    IL6ST Hs.532082 NM_002184, NM_175767
    IL9 Hs.960 NM_000590
    ING4 Hs.524210 NM_001127583, NM_001127582,
    NM_001127586, NM_001127585,
    NM_001127584, NM_016162
    INPP5A Hs.523360, NM_005539
    Hs.715308
    INTS1 Hs.532188 NM_001080453
    IRF2BP2 Hs.350268 NM_001077397, NM_182972
    ISOC1 Hs.483296 NM_016048
    ITGA6 Hs.133397 NM_001079818, NM_000210
    ITPKB Hs.528087, NM_002221
    Hs.659396
    ITPR3 Hs.65758 NM_002224
    JAKMIP1 Hs.479066 NM_144720, NM_001099433
    KAT8 Hs.533803 NM_032188, NM_182958
    KCNK5 Hs.444448 NM_003740
    KCTD12 Hs.644125 NM_138444
    KIAA0020 Hs.493309 NM_014878
    KIAA0141 Hs.210532 NM_014773, NM_001142603
    KIAA0664L3 Hs.715792
    KIAA0748 Hs.33187 NM_001098815, NM_001136030
    KIAA1257 Hs.518247 NM_020741
    KIAA1279 Hs.279580 NM_015634
    KIAA1683 Hs.313471 NM_025249, NM_001145305,
    NM_001145304
    KIAA1797 Hs.136247 NM_017794
    KIF3A Hs.43670 NM_007054
    KIT Hs.479754 NM_001093772, NM_000222,
    XM_001724747, XM_936229
    KLF2 Hs.107740 NM_016270
    KLF3 Hs.298658 NM_016531
    KPNA3 Hs.527919 NM_002267
    KRT72 Hs.662013 NM_080747, NM_001146226,
    NM_001146225
    KRT73 Hs.55410 NM_175068
    LAIR1 Hs.572535 NM_002287, NM_021706
    LARP4 Hs.26613 NM_199188, NM_199190, NM_052879,
    NM_001170808, NM_001170803,
    NM_001170804
    LDLRAP1 Hs.590911 NM_015627
    LEF1 Hs.555947 NM_001166119, NM_001130713,
    NM_001130714, NM_016269
    LGMN Hs.18069 NM_001008530, NM_005606
    LIMA1 Hs.525419 NM_001113547, NM_001113546,
    NM_0016357, NM_017806
    LIME1 Hs.233220 NM_017806
    LMBR1L Hs.272838 NM_018113
    LMNA Hs.594444 NM_005572, NM_170708, NM_170707
    LMO7 Hs.207631 NM_015842, NM_005358
    LOC100289511 Hs.729250 XM_002347442, XM_002343308,
    XM_002344795
    LOC100302650 Hs.729719
    LOC282997 Hs.599931
    LOC283174 Hs.504370
    LOC338799 Hs.524804
    LOC541471 Hs.652166,
    Hs.652426,
    Hs.560805
    LOC728392 Hs.104305 NM_001162371
    LRCH4 Hs.719669, NM_002319
    Hs.125742
    LRP8 Hs.576154 NM_001018054, NM_004631,
    NM_0033300, NM_017522
    LRRN1 Hs.163244 NM_020873
    LSM14B Hs.105379 NM_144703
    LTA Hs.36 NM_001159740, NM_000595
    LTBP3 Hs.289019 NM_001130144, NM_001164266,
    NM_021070
    LYPD3 Hs.631594 NM_014400
    MAF Hs.134859 NM_005360, NM_001031804
    MAL Hs.80395 NM_022438, NM_022439, NM_0002371,
    NM_022440
    MAMLD1 Hs.20136 NM_005491
    MANF Hs.436446 NM_006010
    MAP2K6 Hs.463978 NM_002758
    MAP4K2 Hs.534341 NM_004579
    MARCKSL1 Hs.75061 NM_023009
    MCF2L Hs.170422, NM_001112732, NM_024979
    Hs.597691
    MDS2 Hs.523369
    MED28 Hs.434075, NM_025205
    Hs.644788
    MED6 Hs.497353 NM_005466
    MEGF6 Hs.593645 NM_001409
    MEOX1 Hs.438 NM_001040002, NM_013999,
    NM_004527
    MFGE8 Hs.3745 NM_005928, NM_001114614
    MINPP1 Hs.121260 NM_004897
    MIR1182
    MIR155
    MIR155HG Hs.697120
    MLXIP Hs.721711, NM_014938
    Hs.437153
    MOB1A Hs.602092 NM_018221
    MPI Hs.75694 NM_002435
    MPRIP Hs.462341, NM_201274, NM_015134
    Hs.646854
    MRPL13 Hs.333823 NM_014078
    MRPL22 Hs.483924 NM_014180, NM_001014990
    MRPL33 Hs.515879 NM_145330, NM_004891
    MRPL39 Hs.420696 NM_017446, NM_080794
    MRPL42 Hs.199579 NM_014050, NM_172177, NM_172178
    MRPS28 Hs.521124 NM_014018
    MSC Hs.442619 NM_005098
    MSL1 Hs.532786 NM_001012241
    MTCH2 Hs.269944 NM_014342
    MYADM Hs.380906 NM_001020819, NM_001020818,
    NM_001020821, NM_001020820,
    NM_138373
    MYCBP2 Hs.591221 NM_015057
    MYO15B Hs.390817
    MYOF Hs.602086 NM_013451, NM_133337
    MZF1 Hs.399810 NM_198055, NM_003422
    NAA50 Hs.596074 NM_025146
    NCKAP1 Hs.603732 NM_205842, NM_013436
    NDRG2 Hs.525205 NM_201540, NM_201541, NM_201539,
    NM_201538, NM_201537, NM_201536,
    NM_201535, NM_016250
    NDUFAB1 Hs.189716 NM_005003
    NDUFAF1 Hs.106529 NM_016013
    NDUFV2 Hs.464572 NM_021074
    NEDD9 Hs.37982 NM_182966, NM_001142393,
    NM_006403
    NEK7 Hs.723303, NM_133494
    Hs.24119
    NELL2 Hs.505326 NM_006159, NM_001145110,
    NM_001145108, NM_001145109,
    NM_001145107
    NFATC1 Hs.701518, NM_172388, NM_172387, NM_172389,
    Hs.534074 NM_006162, NM_172390
    NIPA1 Hs.511797 NM_144599, NM_001142275
    NIPAL3 Hs.523442 NM_020448
    NLN Hs.247460 NM_020726
    NME1 Hs.463456 NM_198175, NM_001018138,
    NM_000269, NM_001018139,
    NM_002512, NM_001018137,
    NM_001018136
    NME1-NME2 Hs.463456 NM_198175, NM_001018138,
    NM_001018139, NM_001018137,
    NM_001018136, NM_000269,
    NM_002512
    NME7 Hs.706952 NM_013330, NM_197972
    NPEPPS Hs.443837, NM_006310, XM_001725441,
    Hs.449880 XM_001725426
    NQO1 Hs.406515 NM_001025434, NM_001025433,
    NM_000903
    NRCAM Hs.21422 NM_001037132, NM_001037133,
    NM_005010
    NSDHL Hs.57698 NM_015922, NM_001129765
    NSMAF Hs.372000 NM_003580, NM_001144772
    NT5DC3 Hs.48428 NM_001031701
    NUBP1 Hs.81469 NM_002484
    NUCB2 Hs.654599 NM_005013
    NUMA1 Hs.325978 NM_006185
    NUP153 Hs.601591, NM_005124
    Hs.718703
    OASL Hs.118633 NM_198213, NM_003733
    ODC1 Hs.467701 NM_002539
    OLFM2 Hs.169743 NM_058164
    OSBPL7 Hs.463320 NM_145798
    OTUD7B Hs.98322 NM_020205
    P2RY8 Hs.111377 NM_178129
    P4HA2 Hs.519568 NM_001017974, NM_001017973,
    NM_001142598, NM_001142599,
    NM_004199
    PAM Hs.369430 NM_138766, NM_000919, NM_138822,
    NM_138821
    PAN2 Hs.273397 NM_014871, NM_001166279,
    NM_001127460
    PANX2 Hs.440092 NM_001160300, NM_052839
    PAPD7 Hs.481542 NM_006999, NM_001171806,
    NM_001171805
    PARK7 Hs.419640 NM_007262, NM_001123377
    PBX4 Hs.466257 NM_025245
    PCIF1 Hs.716563 NM_022104
    PCSK5 Hs.368542 NM_006200
    PDE4A Hs.89901 NM_001111308, NM_006202,
    NM_001111307, NM_001111309
    PDIA6 Hs.212102 NM_005742
    PDK1 Hs.470633 NM_002610
    PEA15 Hs.517216 NM_003768
    PFKM Hs.75160 NM_001166688, NM_001166686,
    NM_001166687, NM_000289
    PGAM1 Hs.592599, NM_002629
    Hs.632918
    PGAM2 Hs.632642 NM_000290
    PGAM4 Hs.632822 NM_001029891
    PHKA2 Hs.54941 NM_000292
    PHLPP1 Hs.465337 NM_194449
    PHLPP2 Hs.709458 NM_015020
    PICALM Hs.163893 NM_001008660, NM_007166
    PIK3R5 Hs.278901 NM_001142633, NM_014308
    PIP4K2A Hs.57079 NM_005028
    PITPNM2 Hs.272759 NM_020845
    PLAA Hs.27182 NM_001031689
    PLCG1 Hs.268177 NM_182811, NM_002660
    PLCH2 Hs.170156 NM_014638
    PNISR Hs.520287, NM_015491, NM_032870,
    Hs.644863
    POMP Hs.268742 NM_015932
    PPFIBP2 Hs.655714 NM_003621
    PPIL1 Hs.27693 NM_016059
    PPP2R1B Hs.584790 NM_002716, NM_181699
    PPP2R2B Hs.655213 NM_181676, NM_181675, NM_181674,
    NM_181678, NM_181677, NM_004576,
    NM_001127381
    PPP6R2 Hs.449098, NM_001242898, NM_001242899,
    Hs.733531, NM_001242900, NM_014678
    Hs.740776
    PPPDE2 Hs.570455 NM_015704
    PRDM1 Hs.436023 NM_182907, NM_001198
    PRDX4 Hs.83383 NM_006406
    PREP Hs.436564 NM_002726
    PRKAR1B Hs.520851 NM_001164760, NM_002735,
    NM_001164758, NM_001164759,
    NM_001164762, NM_001164761
    PRKCZ Hs.496255 NM_001033581, NM_002744,
    NM_001033582
    PRR5 Hs.720401, NM_001017530, NM_181333,
    Hs.102336 NM_181334, NM_181335, NM_015366,
    NM_001017526, NM_001017529,
    NM_001017528
    PRSS23 Hs.25338 NM_007173
    PSMA1 Hs.102798 NM_001143937, NM_148976,
    NM_002786
    PSMB1 Hs.352768 NM_002793
    PSMC2 Hs.437366 NM_002803
    PSMD1 Hs.3887 NM_002807
    PSMD11 Hs.655396 NM_002815
    PSMD14 Hs.567410 NM_005805
    PSMD5 Hs.193725 NM_005047
    PTP4A3 Hs.43666 NM_007079, NM_032611
    PTPLA Hs.114062 NM_014241
    PTPN6 Hs.63489 NM_002831, NM_080548, NM_080549
    PTRH2 Hs.12677 NM_016077
    PUS7 Hs.520619 NM_019042
    PYCARD Hs.499094 NM_145182, NM_013258
    R3HDM2 Hs.443673 NM_014925
    RAB1A Hs.310645 NM_004161, NM_015543
    RAB21 Hs.524590 NM_014999
    RAB23 Hs.555016 NM_016277, NM_183227
    RAB33A Hs.654356 NM_004794
    RAB37 Hs.351413 NM_001163990, NM_001163989,
    NM_175738, NM_001006638
    RAB43 Hs.546542, XM_001723593, XM_001720383,
    Hs.723723 XM_001724346, NM_198490,
    XM_002342369
    RABGGTB Hs.78948 NM_004582
    RAD50 Hs.655835 NM_133482, NM_005732
    RAPGEF6 Hs.483329 NM_001164386, NM_001164387,
    NM_001164388, NM_001164389,
    NM_001164390, NM_016340
    RASA3 Hs.593075 NM_007368
    RASGRP2 Hs.99491 NM_153819, NM_001098670,
    NM_001098671
    RBBP8 Hs.546282 NM_002894, NM_203292, NM_203291
    RBKS Hs.11916 NM_022128
    REEP5 Hs.429608 NM_005669
    RGS1 Hs.75256 NM_002922
    RGS14 Hs.9347 NM_006480
    RHOT2 Hs.513242 NM_138769
    RNF19A Hs.292882 NM_015435, NM_183419
    RNF213 Hs.195642 NM_020914, NM_020954,
    NM_002343588
    RNF34 Hs.292804 NM_194271, NM_025126
    RPF2 Hs.372265 NM_032194
    RPP25 Hs.8562 NM_017793
    RYBP Hs.7910 NM_012234
    S1PR1 Hs.154210 NM_001400
    S1PR4 Hs.662006 NM_003775
    SCML4 Hs.486109 NM_198081
    SDHB Hs.465924 NM_003000
    SDK2 Hs.435719 NM_001144952
    SEC24D Hs.189641 NM_014822
    SEC31B Hs.18889 NM_015490
    SELL Hs.728756 NM_000655
    SELP Hs.73800 NM_003005
    SEPT11 Hs.128199 NM_018243
    SEPT3 Hs.120483 NM_019106, NM_145733
    SEPT9 Hs.440932 NM_001113491, NM_001113492,
    NM_001113493, NM_001113494,
    NM_001113495, NM_001113496,
    NM_001293695, NM_001293696,
    NM_001293697, NM_001293698,
    NM_006640,
    SERPINF1 Hs.532768 NM_002615
    SERPINF2 Hs.159509 NM_001165920, NM_001165921,
    NM_000934
    SF1 Hs.502829 NM_004630, NM_201995, NM_201997,
    NM_201998
    SFXN1 Hs.369440 NM_022754
    SH2B1 Hs.723196 NM_001145797, NM_015503,
    NM_001145795, NM_001145796,
    NM_001145812
    SHC1 Hs.433795 NM_001130040, NM_003029,
    NM_001130041, NM_183001
    SIGIRR Hs.501624 NM_021805, NM_001135054,
    NM_001135053
    SIRPG Hs.590883 NM_080816, NM_018556,
    NM_001039508
    SLC16A1 Hs.75231 NM_001166496, NM_003051
    SLC16A10 Hs.591327 NM_018593
    SLC1A4 Hs.654352 NM_003038, NM_001135581
    SLC24A6 Hs.286194 NM_024959
    SLC25A17 Hs.474938 NM_006358
    SLC25A32 Hs.607819 NM_030780
    SLC26A11 Hs.4866 NM_173626, NM_001166348,
    NM_001166347, NM_001166349
    SLC27A2 Hs.720807 NM_003645, NM_001159629
    SLC2A1 Hs.473721 NM_006516
    SLC2A4RG Hs.435126 NM_020062
    SLC2A8 Hs.179522 NM_014580
    SLC35F2 Hs.524014 NM_017515
    SLC39A14 Hs.491232 NM_001128431, NM_015359,
    NM_001135153, NM_001135154
    SLC39A8 Hs.288034 NM_022154, NM_001135148,
    NM_001135147, NM_001135146
    SLC40A1 Hs.643005 NM_014585
    SLC43A3 Hs.99962 NM_199329, NM_017611, NM_014096
    SLIRP Hs.655105 NM_001267863, NM_001267864,
    NM_031210
    SNPH Hs.713451, NM_014723, NM_001136566
    Hs.323833
    SNRK Hs.476052 NM_017719, NM_001100594
    SNRPG Hs.631639, NM_003096, XM_002347904,
    Hs.654528, NM_001146693, XM_002343626,
    Hs.516076, XM_001723258
    Hs.465167
    SNX24 Hs.483200 NM_014035
    SOAT1 Hs.496383 NM_003101
    SORD Hs.878, NM_003104
    Hs.633539
    SOX4 Hs.643910 NM_003107
    SP140L Hs.662198 NM_138402
    SPATS2L Hs.120323 NM_001100424, NM_001100423,
    NM_001100422, NM_015535
    SPG7 Hs.185597 NM_003119, NM_199367
    SPR Hs.301540 NM_003124
    SPSB3 Hs.592080 NM_080861
    SPTBN1 Hs.503178, NM_003128, NM_178313
    Hs.705692
    SRGN Hs.1908 NM_002727
    SRSF5 Hs.632326 NM_001039465, NM_006925
    SRXN1 Hs.719997, NM_080725
    Hs.516830
    SSH1 Hs.199763 NM_001161331, NM_001161330,
    NM_018984
    ST8SIA4 Hs.308628 NM_175052, NM_005668
    STAC Hs.56045 NM_003149
    STAT6 Hs.524518 NM_003153
    STIP1 Hs.337295 NM_006819
    STMN3 Hs.639609 NM_015894
    STRAP Hs.504895 NM_007178
    STT3A Hs.504237 NM_152713
    STX16 Hs.307913 NM_001134772, NM_001134773,
    NM_003763, NM_001001433
    SULT1B1 Hs.129742 NM_014465
    SUN1 Hs.438072 NM_001171944, NM_001171946,
    NM_001171945, NM_001130965,
    NM_025154
    SUN2 Hs.517622 NM_015374
    SVIL Hs.499209 NM_003174, NM_021738
    SYT11 Hs.32984 NM_152280
    SYTL1 Hs.469175 NM_032872
    SYTL3 Hs.436977 NM_001009991
    TACC3 Hs.104019 NM_006342
    TANK Hs.132257 NM_004180, NM_133484
    TBCC Hs.75064 NM_003192
    TBX21 Hs.272409 NM_013351
    TCEA3 Hs.446354 NM_003196
    TCF20 Hs.475018 NM_181492, NM_005650
    TCF7 Hs.573153 NM_201633, NM_201632,
    NM_001134851, NM_001134852,
    NM_213648, NM_003202, NM_201634
    TFRC Hs.529618 NM_001128148, NM_003234
    THNSL1 Hs.645274 NM_024838
    TIGIT Hs.421750 NM_173799
    TIMD4 Hs.334907 NM_001146726, NM_138379
    TJP3 Hs.25527 NM_014428
    TMC6 Hs.632227 NM_001127198, NM_007267
    TMC8 Hs.592102 NM_152468
    TMCC2 Hs.6360 NM_014858
    TMED2 Hs.75914, NM_006815
    Hs.592682
    TMEM110 Hs.556077, NM_198563, NM_205853
    Hs.705605
    TMEM123 Hs.503709 NM_052932
    TMEM165 Hs.479766 NM_018475
    TMEM220 Hs.462230 NM_001004313
    TMEM33 Hs.31082 NM_018126
    TMEM63A Hs.119387 NM_014698
    TMEM66 Hs.521487 NM_016127
    TMEM70 Hs.106650 NM_017866, NM_001040613
    TMEM71 Hs.293842 NM_144649, NM_001145153
    TMIGD2 Hs.263928 NM_001169126, NM_144615
    TNFRSF11A Hs.204044 NM_003839
    TNFRSF1B Hs.256278 NM_001066
    TNFRSF8 Hs.1314 NM_152942, NM_001243
    TNFRSF9 Hs.654459 NM_001561
    TNNT3 Hs.73454 NM_006757, NM_001042782,
    NM_001042780, NM_001042781
    TOP2B Hs.475733 NM_001068
    TPM2 Hs.300772 NM_213674, NM_003289,
    NM_001145822
    TRAPPC6A Hs.466929 NM_024108
    TRIB2 Hs.627749, NM_021643
    Hs.467751
    TRIM22 Hs.501778, NM_006074
    Hs.684559
    TRIP12 Hs.591633 NM_004238
    TRMT5 Hs.380159 NM_020810
    TSC2 Hs.90303 NM_001077183, NM_001114382,
    NM_000548
    TSPAN18 Hs.592575, NM_001031730, NM_130783
    Hs.385634
    TSPAN32 Hs.271954 NM_139022
    TTC4 Hs.720251 NM_004623
    TTC9 Hs.79170 NM_015351
    TTN Hs.134602 NM_133432, NM_133379, NM_133378,
    NM_133437, NM_003319
    TWIST1 Hs.66744 NM_000474
    TXK Hs.479669 NM_003328
    TXN Hs.435136 NM_003329
    TXNDC5 Hs.719272, NM_001145549, NM_201280,
    Hs.150837 NM_030810
    UBASH3B Hs.444075 NM_032873
    UBE2E2 Hs.595802, NM_152653
    Hs.475688
    UBE2Z Hs.514297 NM_023079
    UCHL3 Hs.162241 NM_006002
    UCK2 Hs.458360 NM_012474
    UHRF1BP1L Hs.620701 NM_001006947, NM_015054
    USP19 Hs.255596 NM_006677
    USP53 Hs.595368, NM_019050
    Hs.431081
    UXS1 Hs.469561 NM_025076
    UXT Hs.172791 NM_004182, NM_153477
    VDR Hs.524368 NM_001017535, NM_000376
    VILL Hs.103665 NM_015873
    VIPR1 Hs.348500 NM_004624
    VSIG1 Hs.177164 NM_001170553, NM_182607
    VTRNA1-3
    WDR12 Hs.73291 NM_018256
    WNT7A Hs.72290 NM_004625
    WRB Hs.198308 NM_001146218, NM_004627
    XAF1 Hs.441975 NM_017523, NM_199139
    YPEL3 Hs.513491 NM_031477, NM_001145524
    YWHAE Hs.591239, NM_006761
    Hs.513851
    YWHAG Hs.520974 NM_012479
    ZBTB22 Hs.206770 NM_005453, NM_001145338
    ZBTB38 Hs.723156 NM_001080412
    ZC3H12C Hs.376289 NM_033390
    ZDHHC16 Hs.76662 NM_032327, NM_198046, NM_198045,
    NM_198044, NM_198043
    ZFP36L2 Hs.503093 NM_006887
    ZGPAT Hs.590868 NM_181485, NM_001083113,
    NM_032527
    ZMAT1 Hs.496512 NM_032441, NM_001011657
    ZNF193 Hs.100921 NM_006299
    ZNF238 Hs.69997 NM_205768, NM_006352
    ZNF282 Hs.657701 NM_003575
    ZNF331 Hs.185674 NM_018555, NM_001079907,
    NM_001079906
    ZNF506 Hs.351906 NM_001145404, NM_001099269
    ZNF542 Hs.467326
    ZNF673 Hs.632800 NM_017776, NM_001129900,
    NM_001129898, NM_001129899
    ZNF688 Hs.301463, NM_152458, NM_145271,
    Hs.513509 NM_001024683
    ZNF710 Hs.459311 NM_198526
    ZNF83 Hs.710125, NM_018300, NM_001105550,
    Hs.665751, NM_001105552, NM_001105551,
    Hs.467210, NM_001105554, NM_001105553,
    Hs.659798 NM_001105549
    ZSWIM1 Hs.517075 NM_080603
  • The most significant genes in Table 7A were defined as those with 1) an absolute fold change between ALL CRs/CLL CRs and CLL NRs of greater than 2 and 2) a p-value on the correlation of response and expression of less than 0.01. Thirty-four genes, listed in Table 7B below, met this criteria and the expression of these genes were measured on four additional platforms to compare and validate the findings from RNAseq. The four platforms were OpenArray, Fluidigm, Nanostring, and qPCR. Results from this cross-platform comparison experiment confirmed the results and conclusions described herein. Table 7B also indicates whether each gene is upregulated in complete responders (CR) relative to non-responders, or upregulated in non-responders (NR) relative to complete responders. An exemplary publication disclosing the sequence of each gene is also given in Table 7B, and each publication is incorporated by reference in its entirety, including all nucleic acid and protein sequences therein.
  • TABLE 7B
    Table 7B. Selected genes from Table 7A
    Upregulated
    Gene Unigene Accession No. Exemplary publication in CR or NR
    ALS2CL Hs.517937 NM_147129, Jouan et al., Behav Brain Funct CR
    NM_182775 9, 9 (2013)
    AQP3 Hs.234642 NM_004925 Xie et al., Arch. Dermatol. Res. CR
    305 (5), 397-406 (2013)
    C16orf74 Hs.461655 NM_206967 Kim et al., PLoS ONE 5 (12), CR
    E15260 (2010)
    CCL17 Hs.546294 NM_002987 Lee et al, Pediatr. Res. 74 (5), NR
    545-551 (2013)
    CD248 Hs.195727 NM_020404 Kontsekova et al., Int. J. Oncol. CR
    41 (4), 1365-1372 (2012)
    CSF2 Hs.1349 NM_000758 Sawada et al., J. Exp. Med. 211 NR
    (2), 263-280 (February 2014)
    DHRS2 Hs.272499 NM_182908, Prunotto et al., J Proteomics NR
    NM_005794 82, 193-229 (2013)
    DPEP2 Hs.372633 NM_022355 Willer et al., Nat. Genet. 40 CR
    (2), 161-169 (2008)
    EPAS1 Hs.468410 NM_001430 Mathew et al., Proc. Natl. NR
    Acad. Sci. U.S.A. 111 (1), 291-
    296 (January 2014)
    EPHA4 Hs.371218 NM_004438 Xu et al., Proc. Natl. Acad. Sci. CR
    U.S.A. 110 (36), 14634-14639
    (2013)
    FAIM3 Hs.723317, NM_001142472, Murakami et al., J. Immunol. CR
    Hs.58831 NM_001142473, 189 (2), 587-597 (2012)
    NM_005449
    FAM134B Hs.711125 NM_001034850, Murphy et al., J. Neurol. CR
    NM_019000 Neurosurg. Psychiatr. 83 (1),
    119-120 (2012)
    GPA33 Hs.651244 NM_005814 Deng et al. PLoS ONE 8 (11), CR
    E79629 (2013)
    IL13 Hs.845 NM_002188 Jiang et al., Am. J. Physiol. NR
    Endocrinol. Metab. 305 (11),
    E1359-E1366 (2013)
    IL3 Hs.694 NM_000588 Miyake et al., Cytokine 64 (1), NR
    86-89 (2013)
    IL9 Hs.960 NM_000590 Jabeen et al., J. Clin. Invest. NR
    123 (11), 4641-4653 (2013)
    KRT72 Hs.662013 NM_080747, Principe et al., Proteomics 13 CR
    NM_001146226, (10-11), 1667-1671 (2013)
    NM_001146225
    KRT73 Hs.55410 NM_175068 De Mateo et al., Proteomics 11 CR
    (13), 2714-2726 (2011)
    LTA Hs.36 NM_001159740, Stuart et al., Twin Res Hum NR
    NM_000595 Genet 16 (6), 1079-1086
    (2013)
    MCF2L Hs.170422, NM_001112732, Valdes et al., Ann. Rheum. CR
    Hs.597691 NM_024979 Dis. 71(9), 1537-1540 (2012)
    MDS2 Hs.523369 Meyer et al., PLoS Genet. 6 (8) CR
    (2010)
    MEGF6 Hs.593645 NM_001409 Nakayama et al. Genomics 51 CR
    (1), 27-34 (1998)
    MIR155 Weber et al., FEBS J. 272 (1), NR
    59-73 (2005)
    PPFIBP2 Hs.655714 NM_003621 Bohm et al., Oncol. Rep. 28 CR
    (2), 429-438 (2012)
    SCML4 Hs.486109 NM_198081 Vieira et al., Genet. Med. 10 CR
    (9), 668-674 (2008)
    SDK2 Hs.435719 NM_001144952 Otowa et al., J. Hum. Genet. 54 CR
    (2), 122-126 (2009)
    SPR Hs.301540 NM_003124 Yang et al., J. Biol. Chem. 288 NR
    (26), 19221-19237 (2013)
    SULT1B1 Hs.129742 NM_014465 Ross et al., Nat. Genet. 41 (12), CR
    1345-1349 (2009)
    TCF7 Hs.573153 NM_201633, Nikuseva-Martic et al., Pathol. CR
    NM_201632, Oncol. Res. 19 (3), 545-551
    NM_001134851, (2013)
    NM_001134852,
    NM_213648,
    NM_003202,
    NM_201634
    TNFRSF8 Hs.1314 NM_152942, Yao et al., Am. J. Surg. Pathol. NR
    NM_001243 37 (9), 1407-1412 (2013)
    TSPAN18 Hs.592575, NM_001031730, Yuan et al., PLoS ONE 8 (3), CR
    Hs.385634 NM_130783 E58785 (2013)
    TWIST1 Hs.66744 NM_000474 Zhou et al., J. Exp. Clin. NR
    Cancer Res. 33, 12 (January 2014)
    VIPR1 Hs.348500 NM_004624 Bono et al., Cancer Cell 23 (4), CR
    477-488 (2013)
    VSIG1 Hs.177164 NM_001170553, Chen et al., J Surg Oncol 106 CR
    NM_182607 (3), 286-293 (2012)
  • Cell surface markers differentiating memory T cell subsets that are described in Maus et al. (ANNU. REV. IMMUNOL. 2014) and were not included in the Gattinoni gene sets were also evaluated. Among other things, KLRG1 was identified as a gene whose expression increases in apheresis samples from ALL→CLL CR→CLL PR→CLL NR. KLRG1 expression values predict patient response to CTL019 therapy. At least CD57, CD27, CD122, and CD62L were identified as biomarkers of response in the product samples. Among other things CD57, CD27, CD122, and CD62L expression values predict patient response to CTL019 therapy.
  • In an embodiment, a complete responder (CR) gene signature comprises one or more biomarker profiles described in Table 9.
  • TABLE 9
    Exemplary biomarker profile of
    a complete responder to CAR19 therapy
    Table 9: Exemplary biomarker profile of
    a complete responder to CAR19 therapy
    CD27+ PD1−
    CD8+ LAG3−
    TIM3−
    KLRG1−
    Exemplary CR cell-types
    Resting T effector cells (TEM)
    Resting TREG
    Nave CD4+
    Unstimulated memory T cells (TSCM)
    Early memory T cells
  • In an embodiment, a non-responder (NR) gene signature comprises one or more biomarker profiles described in Table 10.
  • TABLE 10
    Exemplary biomarker profile of
    a non-responder to CAR19 therapy
    Table 10: Exemplary biomarker profile of
    a non-responder to CAR19 therapy
    PD1+ CD27−
    LAG3+
    TIM3+
    KLRG1+
    Exemplary NR cell-types
    Activated T effector cells (TEM)
    Activated TREG
    Activated TH1
    Resting TH2
    Stimulated memory T cells (TSCM)
    Late memory T cells
  • Based on the biological understanding, combinations of genes from unbiased feature selection, gene sets, and selected genes of interest could be used to further differentiate NR's, PR's, and CR's.
  • The previously described work was expanded upon in a study of 35 CLL subject samples. This group of 35 subjects includes the 21 CLL subjects in the previous study, for a total of 5 CRs, 9 PRs, and 21 NRs. In this study manufactured CD19 CAR-expressing cell product samples were collected and cultured overnight with control beads. Novel gene signatures based on mRNA expression levels have been identified that predict patient response. The gene lists for the CR vs NR comparison (N=185) is tabulated in Table 18.
  • TABLE 18
    Gene lists for the CR vs NR comparison
    Gene Unigene Accession No. FDR
    ABCB1 Hs.489033 NM_000927 0.0492
    ACSL1 Hs.406678 NM_001995, NM_001286711, NM_001286708, 0.0362
    NM_001286710, NM_001286712
    ADAM12 NM_003474, NM_001288973, NM_021641, NM_001288974, 0.0089
    NM_001288975
    ADAM23 NM_003812 0.0185
    ADCY1 Hs.192215 NM_001281768, NM_021116 0.0203
    AFAP1 Hs.529369 NM_198595, NM_001134647 0.0884
    AGRN Hs.273330 NM_198576, NM_001305275 0.0212
    ANKRD33B Hs.26039 NM_001164440 0.0097
    APLP2 Hs.370247 NM_001142277, NM_001642, NM_001142278, 0.0429
    NM_001142276, NM_001243299
    AQPEP 0.0615
    ARHGAP32 Hs.440379 NM_001142685, NM_014715 0.0675
    ART3 Hs.731997 NM_001130017, NM_001130016, NM_001130016, 0.0890
    NM_001179
    ATN1 Hs.143766 NM_001007026, NM_001940 0.0675
    ATP9A Hs.649234 NM_006045 0.0029
    B4GALNT1 Hs.159481 NM_001478, NM_001276468, NM_001276469 0.0104
    B4GALT6 NM_004775 0.0965
    C1orf198 Hs.520494 NM_032800, NM_001136494, NM_001136495 0.0047
    C21orf63 0.0029
    C5orf39 0.0047
    C9orf142 NM_183241 0.0870
    CACNB2 Hs.59093 NM_201596, NM_201593, NM_201597, NM_201571, 0.0615
    NM_001167945, NM_201572, NM_000724, NM_201590,
    NM_201570
    CAMK2G NM_001222, NM_172173, NM_172169, NM_172170, 0.0675
    NM_001204492, NM_172171
    CAMSAP2 Hs.23585 NM_203459, NM_001297708, NM_001297707 0.0797
    CCDC74A Hs.351461 NM_138770, NM_001258304, NM_001258306, 0.0492
    NM_001258305
    CCL22 Hs.534347 NM_002990 0.0666
    CCL5 NM_002985, NM_001278736 0.0615
    CD109 Hs.399891 NM_133493, NM_001159588, NM_001159587 0.0423
    CD200 NM_001004196, NM_005944 0.0299
    CD27 Hs.355307 NM_001242 0.0225
    CD52 NM_001803 0.0172
    CDKN1A Hs.370771 NM_000389, NM_001220778, NM_001220777, NM_078467 0.0450
    CERS6 Hs.743222 NM_203463, NM_001256126 0.0384
    CHST2 Hs.8786 NM_004267 0.0063
    CNTNAP2 Hs.655684 NM_014141 0.0872
    CPA5 Hs.567642 NM_001127442, NM_080385, NM_001127441 0.0433
    CPM Hs.654387 NM_001005502, NM_198320, NM_001874, NM_001005502, 0.0466
    NM_198320, NM_001874
    CR1 NM_000573, NM_000651 0.0891
    CTNNA1 Hs.445981, NM_001903, NM_001290310, NM_001290309, 0.0148
    Hs.740112 NM_001290307, NM_001290312
    CXCL9 Hs.77367 NM_002416 0.0299
    CXCR5 NM_001716 0.0939
    DBN1 NM_004395, NM_080881 0.0492
    DEPDC7 Hs.280990 NM_001077242, NM_139160 0.0256
    DIRC3 0.0890
    DLG2 Hs.367656 NM_001364, NM_001142702, NM_001142699, 0.0085
    NM_001142700, NM_001300983, NM_001206769
    DNAJC12 Hs.260720 NM_021800, NM_201262 0.0891
    DRAM1 Hs.525634 NM_018370 0.0376
    DSG2 Hs.412597 NM_001943 0.0384
    DUSP4 NM_001394, NM_057158 0.0148
    EBI3 NM_005755 0.0063
    EEF1A2 Hs.433839 NM_001958 0.0497
    EEF1DP3 0.0939
    EHD4 NM_139265 0.0415
    EMP1 NM_001423 0.0541
    ENPP2 Hs.190977 NM_006209, NM_001130863, NM_001040092 0.0870
    EPAS1 Hs.468410 NM_001430 0.0149
    ERP29 NM_006817, NM_001034025 0.0615
    EVC Hs.646899 NM_001306090, NM_153717, NM_001306092 0.0666
    EVI5 Hs.594434 NM_005665, NM_001308248 0.0373
    FADS2 NM_001281501, NM_001281502, NM_004265 0.0764
    FAM134B Hs.481704 NM_019000, NM_001034850 0.0452
    FAM40B 0.0148
    FAM65C Hs.372578 NM_001290268, NM_080829 0.0615
    FASN Hs.83190 NM_004104 0.0884
    FKBP11 NM_001143781, NM_016594, NM_001143782 0.0407
    FLT1 Hs.594454 NM_002019, NM_001160030, NM_001159920, 0.0699
    NM_001160031
    FLT3LG NM_001204502, NM_001459, NM_001278637, 0.0408
    NM_001278638, NM_001204503
    FOXP1 NM_032682, NM_001244816, NM_001244815, 0.0615
    NM_001244814, NM_001244808, NM_001244812,
    NM_001012505, NM_001244813, NM_001244810
    FSCN1 Hs.118400 NM_003088 0.0694
    GAS8 Hs.431792, NM_001481, NM_001286209, NM_001286205, 0.0811
    Hs.739124 NM_001286208
    GEM Hs.654463 NM_181702, NM_005261 0.0275
    GNA12 Hs.487341 NM_007353, NM_001282441, NM_001282440 0.0360
    GPR56 0.0122
    GZMA NM_006144 0.0805
    HCST NM_014266, NM_001007469 0.0212
    HDC Hs.1481 NM_002112, NM_001306146 0.0890
    HSH2D Hs.631617 NM_032855 0.0243
    IL1A NM_000575 0.0148
    IL1RN NM_173843, NM_173841, NM_000577, NM_173842 0.0615
    IL26 Hs.272350 NM_018402 0.0718
    ILDR2 Hs.133153, NM_199351 0.0860
    Hs.730291
    KLRB1 Hs.169824 NM_002258 0.0074
    KLRC3 Hs.654362 NM_002261, NM_007333 0.0035
    LHFP Hs.507798 NM_005780 0.0053
    LIFR Hs.133421, NM_002310, NM_001127671 0.0362
    Hs.616721
    LINC00476 0.0299
    LMCD1 NM_014583, NM_001278235, NM_001278233, 0.0423
    NM_001278234
    LMNA Hs.594444 NM_001282625, NM_005572, NM_170707, NM_001282626, 0.0677
    NM_001257374, NM_001282624, NM_170708
    LOC347411 0.0407
    LOC619207 0.0407
    LRIG3 NM_001136051, NM_153377 0.0746
    LRP1B Hs.656461 NM_018557 0.0615
    LRRC4C Hs.745123 NM_001258419, NM_020929 0.0733
    LY9 Hs.403857 NM_001261456, NM_001261457, NM_002348, 0.0615
    NM_001033667
    MAST2 Hs.319481 NM_015112 0.0959
    MGAT4A Hs.177576 NM_001160154, NM_012214 0.0085
    MOB1B Hs.691454 NM_173468, NM_001244766 0.0441
    MRPL54 NM_172251 0.0910
    MYOF Hs.602086 NM_133337, NM_013451 0.0615
    NAB2 Hs.159223 NM_005967 0.0373
    NCDN Hs.121870 NM_001014841, NM_001014839, NM_014284 0.0876
    NCKAP1 Hs.603732 NM_013436, NM_205842 0.0299
    NCR3 NM_147130, NM_001145466, NM_001145467 0.0595
    NDUFA12 Hs.674965 NM_018838, NM_001258338 0.0936
    NEDD4L Hs.185677 NM_001243960, NM_001144967, NM_015277, 0.0333
    NM_001144971, NM_001144968, NM_001144969,
    NM_001144970, NM_001144966, NM_001144964,
    NM_001144965
    NEURL3 Hs.149219 NM_001285485, NM_001285486 0.0821
    NINL Hs.631508 NM_025176 0.0709
    NOSIP Hs.7236 NM_001270960, NM_015953 0.0001
    NRP2 Hs.471200 NM_201266, NM_201264, NM_201267, NM_003872, 0.0926
    NM_018534, NM_201279
    OSMR Hs.120658 NM_001168355, NM_003999 0.0432
    PANX2 Hs.440092 NM_001160300, NM_052839 0.0376
    PCBP3 Hs.736936 NM_020528, NM_001130141 0.0089
    PHKA1 Hs.201379 NM_002637, NM_001122670, NM_001172436 0.0130
    PITPNC1 Hs.591185 NM_181671, NM_012417 0.0373
    PLXNB2 Hs.3989 NM_012401 0.0148
    PLXNB3 Hs.632833 NM_005393, NM_001163257 0.0981
    PMCH NM_002674 0.0383
    POU2AF1 Hs.654525, NM_006235 0.0376
    Hs.733573,
    Hs.739353
    PPARG Hs.162646 NM_138712, NM_005037, NM_138711, NM_015869 0.0821
    PPCDC Hs.458922, NM_021823, NM_001301103, NM_001301101, 0.0224
    Hs.640486 NM_001301102, NM_001301104, NM_001301105
    PRDM1 Hs.436023 NM_001198, NM_182907 0.0376
    PRKCDBP NM_145040 0.0615
    PRR5 NM_015366, NM_001198721, NM_181333, NM_001017528, 0.0384
    NM_001017530, NM_001017529
    PSEN2 Hs.25363 NM_000447, NM_012486 0.0299
    PTPN6 Hs.63489 NM_080548, NM_002831, NM_080549 0.0733
    PTPRCAP NM_005608 0.0521
    PTPRD Hs.446083 NM_002839, NM_130391, NM_001171025, NM_130393, 0.0141
    NM_001040712, NM_130392
    PVR NM_006505, NM_001135769, NM_001135768, 0.0149
    NM_001135770
    RABL3 Hs.444360 NM_173825 0.0733
    RBMY1E NM_001006118 0.0089
    RGL1 Hs.497148 NM_001297669, NM_015149, NM_001297670, 0.0891
    NM_001297671, NM_001297672
    RNASE4 NM_001282193, NM_001282192, NM_002937, NM_194431 0.0860
    RORC Hs.256022 NM_001001523, NM_005060 0.0027
    RPS28 NM_001031 0.0763
    S100A4 NM_019554, NM_002961 0.0047
    SCARB1 NM_001082959, NM_005505 0.0327
    SCD Hs.558396 NM_005063 0.0870
    SCML4 NM_001286408, NM_001286409, NM_198081 0.0860
    SDC1 Hs.224607 NM_002997, NM_001006946 0.0299
    SDK2 Hs.435719 NM_001144952 0.0981
    SEPT3 Hs.120483 NM_145733, NM_019106 0.0582
    SEPT5-GP1BB 0.0107
    SGPP2 Hs.591604 NM_152386 0.0347
    SH2B2 Hs.489448 NM_020979 0.0373
    Hs.479116, NM_018986 0.0205
    SH3TC1 Hs.630085
    SKAP1 Hs.316931 NM_003726, NM_001075099 0.0661
    SLC13A3 Hs.655498 NM_022829, NM_001193340, NM_001193339, 0.0254
    NM_001193342, NM_001011554
    SLC22A17 Hs.373498 NM_001289050, NM_016609, NM_020372 0.0944
    SLC27A2 Hs.11729 NM_003645, NM_001159629 0.0811
    SLC29A1 Hs.25450 NM_001304463, NM_001078175, NM_001078177, 0.0150
    NM_001304465, NM_001304466, NM_001304462
    SLC41A2 NM_032148 0.0936
    SLC43A3 Hs.99962 NM_014096, NM_001278201, NM_199329, NM_017611, 0.0423
    NM_001278206
    SLC4A10 Hs.333958 NM_022058, NM_001178015, NM_001178016 0.0376
    SOAT2 Hs.656544 NM_003578 0.0130
    SORCS3 Hs.671950 NM_014978 0.0876
    SPIRE1 Hs.515283 NM_001128626, NM_020148, NM_001128627 0.0615
    SPNS3 NM_182538 0.0001
    SPOCK1 Hs.582184, NM_004598 0.0718
    Hs.596136
    SRCIN1 Hs.448872 NM_025248 0.0661
    SSBP3 Hs.733025 NM_145716, NM_001009955, NM_018070 0.0376
    STX8 NM_004853 0.0937
    SULT2B1 Hs.369331 NM_177973, NM_004605 0.0150
    TERT Hs.492203 NM_198253, NM_001193376, NM_005424 0.0765
    TIE1 Hs.78824 NM_005424, NM_001253357 0.0931
    TLE4 NM_007005, NM_001282760, NM_001282748, 0.0891
    NM_001282749, NM_001282753
    TMOD1 Hs.404289 NM_003275, NM_001166116 0.0308
    TNFRSF19 NM_148957, NM_018647, NM_001204458, NM_001204459 0.0224
    TNFRSF4 Hs.129780 NM_003327 0.0884
    TOB1 Hs.744946 NM_005749, NM_001243885, NM_001243877 0.0945
    TOX2 Hs.26608 NM_001098797, NM_001098796, NM_032883, 0.0347
    NM_001098798
    TRIB2 Hs.467751 NM_021643 0.0666
    TSKU Hs.8361 NM_015516, NM_001258210 0.0027
    TSPAN13 NM_014399 0.0376
    TTBK1 Hs.485436 NM_032538 0.0027
    TTC39C Hs.733420 NM_153211, NM_001243425, NM_001135993, 0.0172
    NM_001292030
    TUBB6 Hs.193491, NM_001303524, NM_032525, NM_001303529, 0.0271
    Hs.744066 NM_001303526, NM_001303525
    uc001acl 0.0212
    uc004aex 0.0299
    uc010eif 0.0243
    uc021oxp 0.0595
    USP44 NM_032147, NM_001042403, NM_001278393 0.0666
    XYLT1 Hs.22907 NM_022166 0.0890
    ZBTB20 NM_001164343, NM_001164347, NM_001164345, 0.0148
    NM_001164342, NM_015642, NM_001164344,
    NM_001164346
    ZBTB32 Hs.99430, NM_014383 0.0384
    Hs.736841
    ZNF219 Hs.250493 NM_016423, NM_001102454, NM_001101672 0.0860
    ZNF683 Hs.353208 NM_001114759, NM_173574, NM_001307925 0.0205
  • Gene set analyses were performed to predict patient response to CD19 CAR-expressing cell therapy (e.g., CTL019). Gene set analysis was performed on gene sets described in Example 1, and with gene sets from three additional datasets described in Example 2 (Szabo et al., Abbas et al., and Gattinoni et al.). Each gene set was evaluated to determine its association with subject response (i.e. CR, PR, or NR) as described in Example 2. Gene sets found to be significantly altered and predictive of patient response to CD19 CAR-expressing cell therapy (e.g., CTL019) are listed in Table 19.
  • TABLE 19
    Gene sets predictive of patient response to CAR therapy
    Gene Set Source CRs NRs
    Treg vs Teff 0 h Szabo Teff 0 h Treg 0 h
    Treg vs Teff 16 h Szabo Teff 16 h Treg 16 h
    Teff 16 h vs 0 h Szabo Teff 0 h Teff 16 h
    Treg 16 h vs 0 h Szabo Treg 0 h Treg 16 h
    Nave CD4 vs 12 h act Th2 Abbas Nave CD4 Th2
    Nave CD4 vs 48 h act Th2 Abbas Nave CD4 Th2
    Nave CD4 vs 12 h act Th1 Abbas Nave CD4 Th1
    Unstim vs stim memory Abbas Unstimulated Stimulated
    Progressively down Gattinoni Early stage Late stage
  • Significant gene sets from the analyses above were refined to a subset of genes within the gene sets that are significantly differentially expressed between the CRs and NRs. An exemplary listing of genes that were significantly differentially expressed are listed in Table 20. Table 20 is an exemplary list of biomarkers whose expression values predict patient response to CTL019 therapy. Table 20 can be further refined into a smaller list of high confidence biomarkers by setting a stricter FDR. For instance, using a FDR of 0.10 will results in a list of 265 genes and a FDR of 0.01 will result in a list of 27 genes.
  • TABLE 20
    Exemplary biomarkers for predicting patient response to CAR therapy
    Gene Unigene Accession No. FDR
    ABCB1 Hs.489033 NM_000927 0.0147
    ABTB1 Hs.107812 NM_172027, NM_032548 0.156
    ACACA NM_198834, NM_198837, NM_198836, 0.226
    NM_198839, NM_198838
    ACSL1 Hs.406678 NM_001995, NM_001286711, NM_001286708, 0.0211
    NM_001286710, NM_001286712
    ACSS2 Hs.517034 NM_001242393, NM_018677, NM_001076552 0.189
    ACTN1 Hs.509765 NM_001102, NM_001130004, NM_001130005 0.137
    ADAM12 NM_003474, NM_001288973, NM_021641, 0.00259
    NM_001288974, NM_001288975
    ADAM7 Hs.116147 NM_003817 0.249
    ADD3 NM_019903, NM_016824, NM_001121 0.243
    ADH7 Hs.389 NM_001166504, NM_000673 0.179
    AES Hs.515053 NM_198969, NM_001130, NM_198970 0.0307
    AGRN Hs.273330 NM_198576, NM_001305275 0.00795
    AHCYL1 Hs.743973 NM_006621, NM_001242675, NM_001242676, 0.24
    NM_001242673, NM_001242674
    AHI1 NM_001134830, NM_017651, NM_001134831, 0.106
    NM_001134832
    AIM2 NM_004833 0.224
    AK5 Hs.559718 NM_174858, NM_012093 0.21
    AKR1C3 NM_003739, NM_001253908 0.212
    ALDH18A1 Hs.500645 NM_002860, NM_001017423 0.234
    ALG5 NM_013338, NM_001142364 0.171
    ALOX5AP NM_001629, NM_001204406 0.0878
    ALPP Hs.284255 NM_001632 0.218
    ANAPC13 Hs.106909 NM_001242374, NM_015391, NM_001242375 0.246
    ANKRD10 NM_017664, NM_001286721 0.229
    ANKRD6 NM_001242813, NM_001242809, NM_014942, 0.0842
    NM_001242811, NM_001242814
    ANXA2P3 0.219
    AP1G2 Hs.343244, NM_001282475, NM_001282474, NM_003917 0.0756
    Hs.740123
    APOA2 NM_001643 0.0265
    APOBEC3C NM_014508 0.11
    APP Hs.434980 NM_000484, NM_001136129, NM_201414, 0.129
    NM_001204303, NM_201413, NM_001204302,
    NM_001136016, NM_001136130, NM_001204301,
    NM_001136131
    AQP3 Hs.234642 NM_004925 0.0645
    ARFRP1 Hs.389277, NM_001267549, NM_001134758, NM_001267544, 0.112
    Hs.661969 NM_001267546, NM_001267545, NM_001267547,
    NM_003224, NM_001267548
    ARHGEF18 Hs.465761 NM_015318, NM_001130955 0.207
    ARID5A NM_212481 0.184
    ARL4C Hs.111554, NM_001282431, NM_005737 0.091
    Hs.730678
    ARNTL2 NM_001248003, NM_001248005, NM_001248002, 0.0709
    NM_001248004, NM_020183
    ARRDC2 Hs.515249 NM_001025604, NM_001286826, NM_015683 0.248
    ATF7IP2 Hs.513343, NM_024997, NM_001256160 0.0661
    Hs.742019
    ATP6V1B2 Hs.295917 NM_001693 0.171
    ATP8A2 NM_016529 0.222
    AURKAIP1 NM_017900, NM_001127230, NM_001127229 0.207
    AUTS2 Hs.21631 NM_001127231, NM_015570, NM_001127232 0.158
    B4GALT7 Hs.455109 NM_007255 0.115
    BACH2 Hs.269764 NM_021813, NM_001170794 0.115
    BANP Hs.461705, NM_001173541, NM_079837, NM_001173540, 0.236
    Hs.690969 NM_017869, NM_001173543, NM_001173539,
    NM_001173542
    BARD1 NM_000465, NM_001282549, NM_001282543, 0.232
    NM_001282548, NM_001282545
    BASP1 Hs.201641 NM_006317, NM_001271606 0.137
    BCL11B Hs.709690 NM_001282237, NM_138576, NM_001282238, 0.134
    NM_022898
    BCOR NM_017745, NM_001123384, NM_001123385, 0.22
    NM_001123383
    BEX4 Hs.184736 NM_001080425, NM_001127688 0.218
    BFSP1 Hs.129702 NM_001195, NM_001161705, NM_001278607, 0.186
    NM_001278606, NM_001278608
    BHLHE40 NM_003670 0.224
    BIN1 Hs.193163 NM_139344, NM_139348, NM_139351, 0.192
    NM_139346, NM_139347, NM_139349,
    NM_139345, NM_004305, NM_139343,
    NM_139350
    BIN2 Hs.14770 NM_001290008, NM_001290009, NM_016293, 0.094
    NM_001290007
    BIRC3 NM_182962, NM_001165 0.127
    BUB1 Hs.469649 NM_001278617, NM_004336, NM_001278616 0.141
    C11orf21 Hs.559181 NM_001142946 0.0679
    C11orf48 0.103
    C11orf67 0.227
    C11orf82 0.196
    C14orf49 0.217
    C16orf45 Hs.738182 NM_033201, NM_001142469 0.181
    C16orf74 NM_206967 0.197
    C17orf53 Hs.437059 NM_024032, NM_001171251 0.185
    C17orf66 0.229
    C1orf162 Hs.288010 NM_174896, NM_001300834 0.135
    C1orf54 NM_001301040, NM_001301039, NM_024579, 0.212
    NM_001301042
    C20orf111 0.0568
    C20orf112 0.102
    C2orf28 0.24
    C2orf89 0.159
    C5orf30 Hs.482976 NM_033211 0.207
    C5orf39 0.00226
    C7orf10 0.162
    C7orf59 0.0455
    C9orf23 0.0943
    CA6 NM_001215, NM_001270500, NM_001270501 0.0798
    CAMK1 NM_003656 0.215
    CAMK2G NM_001222, NM_172173, NM_172169, 0.0175
    NM_172170, NM_001204492, NM_172171
    CAMK4 NM_001744 0.0524
    CAPG Hs.687978 NM_001747, NM_001256140, NM_001256139 0.0735
    CAPS NM_080590, NM_004058 0.0524
    CARM1 NM_199141 0.162
    CBLB Hs.430589 NM_170662 0.061
    CCDC47 Hs.202011 NM_020198 0.104
    CCDC56 0.0674
    CCL20 Hs.75498 NM_001130046, NM_004591 0.249
    CCL4L1 0.0434
    CCL5 NM_002985, NM_001278736 0.015
    CCNB1 Hs.23960 NM_031966 0.195
    CCND3 NM_001760, NM_001287427, NM_001136126, 0.0618
    NM_001136017, NM_001136125, NM_001287434
    CCR6 NM_031409, NM_004367 0.0767
    CD109 Hs.399891 NM_133493, NM_001159588, NM_001159587 0.013
    CD200 NM_001004196, NM_005944 0.00952
    CD22 Hs.579691, NM_001185099, NM_001771, NM_001185100, 0.124
    Hs.716252 NM_001278417, NM_001185101
    CD244 Hs.157872 NM_016382, NM_001166663, NM_001166664 0.0605
    CD248 Hs.195727 NM_020404 0.0548
    CD3D NM_000732, NM_001040651 0.0455
    CD4 NM_000616 0.166
    CD5 Hs.58685 NM_014207 0.0926
    CD55 NM_000574, NM_001300903, NM_001300904, 0.139
    NM_001300902, NM_001114752
    CD68 Hs.647419 NM_001251, NM_001040059 0.144
    CD80 Hs.838 NM_005191 0.061
    CDC14A Hs.127411 NM_033312, NM_033313, NM_003672 0.149
    CDC25B Hs.153752 NM_001287519, NM_001287520, NM_021873, 0.136
    NM_004358, NM_001287522, NM_021872,
    NM_001287518, NM_001287516, NM_001287517
    CDC42BPB Hs.654634 NM_006035 0.0835
    CDC42EP3 Hs.369574, NM_006449, NM_001270437, NM_001270438, 0.126
    Hs.689535 NM_001270436
    CDC6 NM_001254 0.201
    CDKN1A Hs.370771 NM_000389, NM_001220778, NM_001220777, 0.013
    NM_078467
    CDKN2D Hs.435051 NM_079421, NM_001800 0.218
    CDT1 NM_030928 0.195
    CECR1 Hs.170310 NM_177405, NM_001282228, NM_001282227, 0.147
    NM_001282229, NM_001282226, NM_001282225
    CEMP1 NM_001048212 0.179
    CEP55 Hs.14559 NM_001127182, NM_018131 0.241
    CFH Hs.363396 NM_000186, NM_001014975 0.158
    CFHR2 NM_005666 0.074
    CGREF1 Hs.159525 NM_001166240, NM_006569, NM_001166239 0.201
    CHEK1 Hs.24529 NM_001114121, NM_001274, NM_001244846, 0.136
    NM_001114122
    CHL1 Hs.148909, NM_006614, NM_001253387, NM_001253388 0.226
    Hs.731409
    CHMP7 Hs.5019 NM_152272 0.0594
    CHST11 Hs.17569 NM_001173982, NM_018413 0.149
    CHST12 Hs.744987 NM_001243794, NM_001243795, NM_018641 0.192
    CHST2 Hs.8786 NM_004267 0.00226
    CHSY1 Hs.110488, NM_014918 0.11
    Hs.734921
    CLCA2 Hs.241551 NM_006536 0.172
    CMAHP 0.0642
    CNPY3 NM_006586 0.0605
    COL18A1 Hs.517356 NM_130445, NM_030582, NM_130444 0.189
    COL6A1 Hs.474053 NM_001848 0.201
    CORO1C Hs.330384 NM_014325, NM_001276471, NM_001105237 0.0566
    COX4I1 NM_001861 0.222
    CRADD Hs.591016, NM_003805 0.144
    Hs.719191
    CRKL Hs.5613 NM_005207 0.201
    CSGALNACT1 Hs.613729 NM_001130518, NM_018371 0.0589
    CSNK2A1 NM_001895, NM_177559, NM_177560 0.171
    CSTB NM_000100 0.0477
    CSTF2 NM_001325, NM_001306206 0.158
    CTDSP1 Hs.444468 NM_001206878, NM_182642, NM_021198 0.195
    CTNNA1 Hs.445981, NM_001903, NM_001290310, NM_001290309, 0.00562
    Hs.740112 NM_001290307, NM_001290312
    CTNNA2 Hs.167368 NM_004389, NM_001282598, NM_001164883, 0.145
    NM_001282597, NM_001282600, NM_001282599
    CTNNAL1 NM_001286974, NM_003798 0.124
    CTNNBIP1 Hs.463759 NM_020248, NM_001012329 0.091
    CTNND2 Hs.314543 NM_001332, NM_001288717, NM_001288715, 0.226
    NM_001288716
    CTSF Hs.11590 NM_003793 0.224
    CTSW Hs.416848 NM_001335 0.0929
    CTTN Hs.596164 NM_138565, NM_005231, NM_001184740 0.0642
    CUX1 Hs.191482 NM_181500, NM_001202546, NM_001202544, 0.137
    NM_001202543, NM_001202545, NM_181552,
    NM_001913
    CXCL13 Hs.100431 NM_006419 0.192
    CYB561 Hs.355264 NM_001017917, NM_001915, NM_001017916 0.208
    CYFIP1 Hs.26704 NM_014608, NM_001033028, NM_001287810 0.192
    D4S234E 0.224
    DAB1 Hs.477370 NM_021080 0.0455
    DAXX NM_001141969, NM_001254717, NM_001141970, 0.192
    NM_001350
    DBN1 NM_004395, NM_080881 0.0127
    DENND2D NM_024901, NM_001271833 0.0882
    DENND3 Hs.18166 NM_014957 0.223
    DEPDC7 Hs.280990 NM_001077242, NM_139160 0.00865
    DGKD Hs.471675 NM_152879, NM_003648 0.0524
    DGKI Hs.737768 NM_004717 0.0596
    DHCR24 Hs.498727 NM_014762 0.0843
    DIXDC1 Hs.655626 NM_001278542, NM_001037954, NM_033425 0.224
    DLG2 Hs.367656 NM_001364, NM_001142702, NM_001142699, 0.00226
    NM_001142700, NM_001300983, NM_001206769
    DMRT1 Hs.98586 NM_021951 0.0589
    DNAJB5 Hs.237506 NM_001135004, NM_012266, NM_001135005 0.0843
    DNAJC6 Hs.647643 NM_014787, NM_001256864, NM_001256865 0.219
    DNM1 Hs.522413 NM_004408, NM_001005336, NM_001288737, 0.244
    NM_001288738, NM_001288739
    DOCK7 Hs.744927 NM_033407, NM_001272000, NM_001272002, 0.061
    NM_001271999, NM_001272001
    DONSON NM_017613 0.0754
    DPEP2 Hs.372633 NM_022355 0.11
    DPP7 NM_013379 0.236
    DPYD Hs.335034 NM_000110, NM_001160301 0.133
    DPYSL2 Hs.593187 NM_001197293, NM_001386, NM_001244604 0.106
    DSN1 Hs.632268 NM_001145318, NM_001145315, NM_001145317, 0.0767
    NM_001145316, NM_024918
    DTL Hs.656473 NM_016448, NM_001286229, NM_001286230 0.234
    DUSP10 Hs.497822 NM_007207 0.162
    DUSP16 NM_030640 0.238
    DUSP22 NM_020185, NM_001286555 0.0735
    DUSP4 NM_001394, NM_057158 0.00463
    DVL2 Hs.118640 NM_004422 0.0847
    DYNLL1 Hs.5120 NM_001037494, NM_001037495, NM_003746 0.195
    EAPP Hs.433269 NM_018453 0.246
    EBI3 NM_005755 0.00226
    EBI3 0.00226
    EED NM_003797, NM_152991, NM_001308007 0.0798
    EEF1D NM_001195203, NM_032378, NM_001130056, 0.101
    NM_001289950, NM_001960, NM_001130053,
    NM_001130055, NM_001130057
    EFS Hs.24587 NM_005864, NM_032459, NM_001277174 0.0594
    EGFL6 Hs.12844 NM_015507, NM_001167890 0.229
    EIF2C4 0.158
    ELL2 Hs.192221 NM_012081 0.0455
    EMB Hs.561411 NM_198449 0.24
    EMP1 NM_001423 0.0144
    ENPP2 Hs.190977 NM_006209, NM_001130863, NM_001040092 0.0275
    EPAS1 Hs.468410 NM_001430 0.00636
    EPB41L4B Hs.591901 NM_019114, NM_018424 0.172
    EPHA4 Hs.371218 NM_001304536, NM_001304537, NM_004438 0.157
    ERGIC3 NM_015966, NM_198398 0.207
    ERI2 Hs.248437 NM_080663, NM_001142725 0.0552
    ERP29 NM_006817, NM_001034025 0.0113
    ESPL1 Hs.153479 NM_012291 0.236
    ESR1 Hs.208124 NM_001291230, NM_001122741, NM_001291241, 0.192
    NM_000125, NM_001122742, NM_001122740
    ESRRG NM_001134285, NM_001243511, NM_001243518, 0.195
    NM_001438, NM_001243514, NM_001243513,
    NM_206595, NM_001243506, NM_001243510,
    NM_001243507, NM_001243512, NM_001243509,
    NM_001243515, NM_001243519, NM_206594
    ETV3 NM_001145312, NM_005240 0.208
    EVI5 Hs.594434 NM_005665, NM_001308248 0.0127
    FADS1 Hs.503546, NM_013402 0.16
    Hs.739285
    FADS2 NM_001281501, NM_001281502, NM_004265 0.0243
    FAH NM_000137 0.131
    FAIM2 Hs.567424 NM_012306 0.215
    FAIM3 0.128
    FAM125B 0.0735
    FAM134B Hs.481704 NM_019000, NM_001034850 0.0128
    FAM134C Hs.632262 NM_178126 0.107
    FAM40B 0.00562
    FAM46C Hs.356216 NM_017709 0.241
    FAM65B Hs.559459 NM_014722, NM_015864, NM_001286447, 0.0965
    NM_001286445, NM_001286446
    FANCI Hs.513126 NM_018193, NM_001113378 0.133
    FAU NM_001997 0.137
    FDPS NM_001135822, NM_001242824, NM_002004, 0.0852
    NM_001135821, NM_001242825
    FGD3 Hs.411081 NM_001083536, NM_001286993, NM_033086 0.125
    FGF21 Hs.283015 NM_019113 0.249
    FHIT Hs.655995 NM_002012, NM_001166243 0.0505
    FKBP11 NM_001143781, NM_016594, NM_001143782 0.0128
    FLI1 Hs.504281 NM_002017, NM_001167681, NM_001271012, 0.0965
    NM_001271010
    FLT1 Hs.594454 NM_002019, NM_001160030, NM_001159920, 0.0243
    NM_001160031
    FLT3LG NM_001204502, NM_001459, NM_001278637, 0.0124
    NM_001278638, NM_001204503
    FLVCR2 Hs.509966 NM_017791, NM_001195283 0.149
    FMNL2 Hs.654630 NM_052905 0.171
    FNBP1 NM_015033 0.142
    FOXN3 NM_001085471, NM_005197 0.207
    FOXP1 NM_032682, NM_001244816, NM_001244815, 0.015
    NM_001244814, NM_001244808, NM_001244812,
    NM_001012505, NM_001244813, NM_001244810
    FXYD5 NM_014164, NM_144779, NM_001164605 0.0852
    FXYD7 NM_022006 0.189
    G0S2 NM_015714 0.0594
    GAB2 Hs.429434 NM_012296, NM_080491 0.0532
    GAB3 Hs.496982 NM_080612, NM_001282283, NM_001081573 0.136
    GABARAPL1 Hs.524250 NM_031412 0.162
    GAD2 NM_001134366, NM_000818 0.129
    GARS Hs.404321 NM_002047 0.109
    GATM Hs.75335 NM_001482 0.124
    GBP5 Hs.513726 NM_052942, NM_001134486 0.168
    GCET2 0.243
    GEM Hs.654463 NM_181702, NM_005261 0.00795
    GK Hs.1466 NM_203391, NM_001128127, NM_000167, 0.0432
    NM_001205019
    GLCCI1 Hs.131673 NM_138426 0.211
    GLIPR2 Hs.493819 NM_001287010, NM_001287013, NM_022343, 0.0466
    NM_001287011, NM_001287014, NM_001287012
    GMNN NM_001251990, NM_001251989, NM_015895, 0.215
    NM_001251991
    GNG4 Hs.159711 NM_004485, NM_001098722, NM_001098721 0.101
    GNLY NM_006433, NM_001302758, NM_012483 0.184
    GPC1 NM_002081 0.186
    GPD1L Hs.82432 NM_015141 0.099
    GPKOW NM_015698 0.167
    GPR114 0.17
    GPR56 0.00463
    GPRC5C Hs.446438 NM_022036, NM_018653 0.158
    GPRIN3 NM_198281 0.215
    GPSM3 Hs.520046 NM_001276501, NM_022107 0.122
    GRAMD3 Hs.363558, NM_001146319, NM_023927, NM_001146322, 0.0594
    Hs.664026 NM_001146321, NM_001146320
    GRAP NM_006613 0.158
    GTF3C4 NM_012204 0.231
    GTPBP1 Hs.276925 NM_004286 0.0665
    GYG1 Hs.477892, NM_004130, NM_001184720, NM_001184721 0.124
    Hs.727448
    GZMA NM_006144 0.0211
    GZMH NM_001270780, NM_033423, NM_001270781 0.159
    H19 Hs.533566 0.151
    H1F0 Hs.745024 NM_005318 0.0432
    HAO2 Hs.659767 NM_001005783, NM_016527 0.17
    HBS1L Hs.378532 NM_006620, NM_001145158, NM_001145207 0.0528
    HERPUD2 NM_022373 0.236
    HKDC1 Hs.522988 NM_025130 0.236
    HLA-DPA1 Hs.347270 NM_033554, NM_001242524, NM_001242525 0.158
    HLA-DQA2 NM_020056, NM_002122 0.179
    HLA-DQB2 Hs.731563 NM_001198858, NM_001300790 0.226
    HLA-DRB5 NM_002125 0.0645
    HMGCR Hs.628096 NM_000859, NM_001130996 0.0466
    HMGCS1 NM_001098272, NM_002130 0.0666
    HNRPLL 0.0566
    HOXC8 NM_022658 0.142
    HPGD Hs.596913 NM_000860, NM_001145816, NM_001256301, 0.0276
    NM_001256306, NM_001256307, NM_001256305
    HPS5 Hs.437599 NM_007216, NM_181507, NM_181508 0.167
    HSD11B1 NM_005525, NM_001206741, NM_181755 0.0502
    HSD17B11 Hs.594923 NM_016245 0.156
    HSD17B12 Hs.132513 NM_016142 0.143
    HSPA1L Hs.690634 NM_005527 0.193
    HSPD1 NM_002156, NM_199440 0.139
    ICAM1 Hs.643447 NM_000201 0.132
    ICAM2 NM_000873, NM_001099789, NM_001099788, 0.0847
    NM_001099786, NM_001099787
    ICAM3 Hs.654563 NM_002162 0.229
    IER2 Hs.501629 NM_004907 0.158
    IER3 NM_003897 0.206
    IFI44 NM_006417 0.223
    IFIH1 Hs.163173 NM_022168 0.0524
    IGBP1 Hs.496267 NM_001551 0.151
    IGSF3 Hs.171057 NM_001007237, NM_001542 0.0466
    IL17RA Hs.48353 NM_014339, NM_001289905 0.135
    IL1A NM_000575 0.00562
    IL1RAP Hs.478673 NM_001167929, NM_001167928, NM_002182, 0.0524
    NM_134470, NM_001167930, NM_001167931
    IL1RAPL1 Hs.658912 NM_014271 0.167
    IL1RN NM_173843, NM_173841, NM_000577, NM_173842 0.0168
    IL21 NM_021803, NM_001207006 0.167
    IL32 NM_001012718, NM_004221, NM_001012633, 0.177
    NM_001012631, NM_001012635, NM_001012634,
    NM_001012632, NM_001012636
    IL8 0.234
    INPP4B NM_003866, NM_001101669 0.0965
    IRF4 NM_002460, NM_001195286 0.142
    IRF6 Hs.591415 NM_006147, NM_001206696 0.0699
    IRF8 NM_002163 0.114
    ISG20 Hs.459265 NM_001303234, NM_001303233, NM_002201, 0.0354
    NM_001303237
    ITGA6 Hs.133397 NM_000210, NM_001079818 0.0441
    ITGAE Hs.513867 NM_002208 0.192
    ITPA NM_033453, NM_181493, NM_001267623 0.16
    ITPK1 NM_001142594, NM_001142593, NM_014216 0.12
    JUN Hs.696684 NM_002228 0.141
    JUNB Hs.25292 NM_002229 0.0455
    KAZALD1 Hs.733496 NM_030929 0.214
    KCNK1 NM_002245 0.104
    KCNK5 Hs.444448 NM_003740 0.246
    KCNQ1 NM_000218, NM_181798 0.205
    KIFC1 Hs.436912 NM_002263 0.234
    KIT Hs.479754 NM_001093772, NM_000222 0.0466
    KLF2 Hs.744182 NM_016270 0.166
    KLF3 Hs.298658 NM_016531 0.24
    KLF4 Hs.376206 NM_004235 0.136
    KLF7 Hs.59908 NM_003709, NM_001270943, NM_001270942, 0.203
    NM_001270944
    KLRB1 Hs.169824 NM_002258 0.00226
    KLRC1 Hs.512576 NM_001304448, NM_002259, NM_213657, 0.0455
    NM_007328, NM_213658
    KLRD1 Hs.562457, NM_002262, NM_007334, NM_001114396 0.158
    Hs.668357
    KRT72 Hs.662013 NM_001146225, NM_001146226, NM_080747 0.212
    LAIR1 NM_001289026, NM_001289027, NM_002287, 0.238
    NM_021706, NM_001289025, NM_001289023
    LAMB3 Hs.497636 NM_000228, NM_001127641, NM_001017402 0.0642
    LCLAT1 Hs.468048 NM_001304445, NM_001002257, NM_182551 0.234
    LIF Hs.2250 NM_002309, NM_001257135 0.0837
    LIMA1 Hs.525419 NM_001243775, NM_001113546, NM_016357, 0.0774
    NM_001113547
    LITAF Hs.459940 NM_001136473, NM_004862, NM_001136472 0.0747
    LMCD1 NM_014583, NM_001278235, NM_001278233, 0.0127
    NM_001278234
    LMNA Hs.594444 NM_001282625, NM_005572, NM_170707, 0.0166
    NM_001282626, NM_001257374, NM_001282624,
    NM_170708
    LMNB2 Hs.538286 NM_032737 0.144
    LOC282997 0.198
    LOC728392 0.12
    LOC728855 0.0567
    LRRC16A Hs.649550 NM_001173977, NM_017640 0.0756
    LSAMP Hs.26409 NM_002338 0.223
    LTA Hs.36 NM_001159740, NM_000595 0.135
    LYAR Hs.425427 NM_017816, NM_001145725 0.0882
    MAP2K5 NM_145160, NM_002757, NM_001206804 0.107
    MAP2K6 Hs.463978 NM_002758 0.0594
    MATK Hs.631845 NM_139354, NM_139355, NM_002378 0.177
    MBP Hs.551713 NM_001025081, NM_001025090, NM_001025092, 0.192
    NM_002385, NM_001025100, NM_001025101
    MCAM NM_006500 0.245
    MCM10 Hs.198363 NM_018518, NM_182751 0.232
    MCM2 Hs.477481 NM_004526 0.119
    MCM4 NM_182746, NM_005914 0.144
    MCTP2 Hs.33368 NM_001159643, NM_018349, NM_001159644 0.147
    ME3 Hs.199743 NM_001014811, NM_001161586, NM_006680 0.136
    MEST NM_177524, NM_001253901, NM_001253902, 0.192
    NM_001253900, NM_002402, NM_177525
    METTL13 Hs.494705 NM_014955, NM_001007239, NM_015935 0.207
    METTL7A Hs.744021 NM_014033 0.0605
    MFNG Hs.517603 NM_002405, NM_001166343 0.061
    MGAT4C Hs.589093, NM_013244 0.0544
    Hs.739389
    MICAL2 Hs.501928, NM_014632, NM_001282663, NM_001282665, 0.158
    Hs.735627 NM_001282666, NM_001282667, NM_001282664
    MID1IP1 Hs.522605 NM_001098790, NM_021242, NM_001098791 0.229
    MIR155HG Hs.697120 0.0386
    MIS18A Hs.190518 NM_018944 0.0906
    MLEC Hs.701392, NM_001303627, NM_014730, NM_001303628 0.244
    Hs.744910
    MLH1 Hs.195364 NM_000249, NM_001258271, NM_001258274, 0.159
    NM_001167618, NM_001167617, NM_001167619,
    NM_001258273
    MMP19 NM_002429, NM_001272101 0.165
    MPP1 Hs.496984 NM_001166460, NM_002436, NM_001166461, 0.212
    NM_001166462
    MRC2 Hs.7835 NM_006039 0.215
    MRPL39 NM_017446, NM_080794 0.151
    MRPS17 Hs.44298 NM_015969 0.0524
    MT1G NM_005950, NM_001301267 0.214
    MTHFD1 Hs.652308 NM_005956 0.0524
    MTHFD2 NM_006636 0.0924
    MTMR4 Hs.514373 NM_004687 0.171
    MYB Hs.606320, NM_001130173, NM_001130172, NM_005375, 0.0528
    Hs.626299 NM_001161657, NM_001161656, NM_001161658,
    NM_001161659, NM_001161660
    MYL6 NM_021019, NM_079423 0.137
    MYO1C Hs.286226 NM_001080779, NM_001080950, NM_033375 0.128
    MYO1F Hs.465818 NM_012335 0.0965
    MYOF Hs.602086 NM_133337, NM_013451 0.0243
    NAB2 Hs.159223 NM_005967 0.0128
    NCAPD2 Hs.5719 NM_014865 0.233
    NCAPD3 NM_015261 0.236
    NCAPH NM_015341, NM_001281710, NM_001281711, 0.17
    NM_001281712
    NCKAP1 Hs.603732 NM_013436, NM_205842 0.0111
    NDFIP2 Hs.525093 NM_001161407, NM_019080 0.0528
    NELL2 Hs.505326 NM_001145108, NM_006159, NM_001145110, 0.0558
    NM_001145107, NM_001145109
    NFKBIZ Hs.319171 NM_001005474, NM_031419 0.181
    NHSL2 Hs.397836, NM_001013627 0.192
    Hs.660859
    NINJ2 NM_016533, NM_001294345, NM_001294346 0.061
    NKG7 NM_005601 0.123
    NKIRAS1 Hs.173202 NM_020345 0.201
    NMT2 NM_004808, NM_001308295 0.0842
    NOG Hs.248201 NM_005450 0.171
    NOSIP Hs.7236 NM_001270960, NM_015953 3.65E−05
    NPC2 Hs.433222 NM_006432 0.156
    NPY NM_000905 0.0466
    NR1D2 Hs.37288 NM_001145425, NM_005126 0.139
    NR2E1 Hs.157688 NM_003269, NM_001286102 0.0871
    NR3C2 Hs.163924 NM_000901, NM_001166104 0.0645
    NR4A3 Hs.279522 NM_173199, NM_006981, NM_173200 0.171
    NSMCE1 Hs.284295 NM_145080 0.0711
    NUP205 Hs.743250 NM_015135 0.162
    OASL NM_003733, NM_198213, NM_001261825 0.192
    ODC1 NM_002539, NM_001287188, NM_001287190, 0.139
    NM_001287189
    OLFM2 NM_058164, NM_001304348, NM_001304347 0.229
    ORC6 NM_014321 0.0798
    OSBPL8 Hs.430849 NM_020841, NM_001003712 0.227
    OTUD7B NM_020205 0.0487
    P2RX4 NM_002560, NM_001256796, NM_001261397, 0.0671
    NM_001261398
    P4HA2 Hs.519568 NM_001142598, NM_001142599, NM_001017974, 0.107
    NM_001017973, NM_004199
    PACSIN3 Hs.334639 NM_016223, NM_001184974, NM_001184975 0.226
    PAICS Hs.518774 NM_001079525, NM_001079524, NM_006452 0.167
    PAM Hs.369430, NM_001177306, NM_138822, NM_138821, 0.218
    Hs.738567 NM_000919, NM_138766
    PANX2 Hs.440092 NM_001160300, NM_052839 0.0128
    PARP8 Hs.369581 NM_001178055, NM_024615, NM_001178056 0.122
    PBX4 Hs.466257 NM_025245 0.211
    PCSK5 Hs.368542 NM_006200, NM_001190482 0.0473
    PDCD1 Hs.158297 NM_005018 0.0747
    PDE10A Hs.348762, NM_006661, NM_001130690 0.215
    Hs.638546
    PDGFRB Hs.509067 NM_002609 0.0628
    PECAM1 Hs.376675 NM_000442 0.0666
    PELP1 Hs.744899 NM_014389, NM_001278241 0.192
    PER1 Hs.445534 NM_002616 0.0674
    PEX16 NM_057174, NM_004813 0.0666
    PFDN5 NM_002624, NM_145897 0.135
    PFKM Hs.75160 NM_001166686, NM_000289, NM_001166688, 0.0961
    NM_001166687
    PGAP1 Hs.229988 NM_024989 0.158
    PGCP 0.186
    PHEX NM_001282754, NM_000444 0.149
    PHF6 Hs.356501 NM_032458, NM_001015877, NM_032335 0.202
    PHLPP1 Hs.465337 NM_194449 0.0853
    PIK3C2G Hs.22500 NM_001288772, NM_001288774, NM_004570 0.0629
    PINK1 Hs.389171 NM_032409 0.24
    PION 0.187
    PITPNC1 Hs.591185 NM_181671, NM_012417 0.00827
    PKMYT1 Hs.732385, NM_182687, NM_004203, NM_001258451, 0.246
    Hs.734466 NM_001258450
    PLAC8 Hs.546392 NM_016619, NM_001130716, NM_001130715 0.0653
    PLAGL2 Hs.154104 NM_002657 0.174
    PLCG2 NM_002661 0.118
    PLCL1 NM_006226 0.107
    PLCL2 Hs.202010, NM_001144382, NM_015184 0.141
    Hs.741267
    PLIN2 NM_001122 0.0747
    PLK2 Hs.398157 NM_006622, NM_001252226 0.205
    PLXNB2 Hs.3989 NM_012401 0.0064
    PLXND1 Hs.301685 NM_015103 0.229
    PMAIP1 Hs.96 NM_021127 0.0316
    PMCH NM_002674 0.0126
    PNLIPRP1 Hs.73923 NM_006229, NM_001303135 0.137
    PNMA1 Hs.194709 NM_006029 0.11
    POU2AF1 Hs.654525, NM_006235 0.0127
    Hs.733573
    POU6F1 Hs.555886 NM_002702 0.229
    PPCDC Hs.458922, NM_021823, NM_001301103, NM_001301101, 0.00748
    Hs.640486 NM_001301102, NM_001301104, NM_001301105
    PPFIBP2 Hs.655714, NM_003621, NM_001256568, NM_001256569 0.0544
    Hs.739217
    PPP1R15A Hs.631593 NM_014330 0.0645
    PPP2R2B Hs.739387 NM_181678, NM_181674, NM_181675, NM_181676, 0.061
    NM_001271899, NM_181677, NM_001271900,
    NM_001271948
    PPP2R3C Hs.530712 NM_017917, NM_001305156, NM_001305155 0.167
    PPP2R5C Hs.368264, NM_001161726, NM_178586, NM_178587, 0.158
    Hs.679341 NM_002719, NM_001161725
    PPP3CA Hs.435512 NM_000944, NM_001130692, NM_001130691 0.0679
    PQBP1 NM_001167990, NM_005710, NM_001032384, 0.141
    NM_144495, NM_001032381, NM_001167989,
    NM_001032382, NM_001032383
    PRC1 Hs.366401 NM_003981, NM_199413, NM_001267580 0.0642
    PRDM1 Hs.436023 NM_001198, NM_182907 0.0127
    PRF1 Hs.2200 NM_001083116, NM_005041 0.192
    PRKAR1B Hs.520851 NM_001164761, NM_001164758, NM_002735, 0.167
    NM_001164760, NM_001164759, NM_001164762
    PRKCDBP NM_145040 0.0181
    PRKCH Hs.333907, NM_006255 0.0889
    Hs.630857
    PRKCQ Hs.498570 NM_006257, NM_001282644, NM_001242413, 0.115
    NM_001282645
    PRKD3 Hs.660757 NM_005813 0.195
    PRKG2 Hs.570833 NM_001282485, NM_006259, NM_001282483, 0.205
    NM_001282482, NM_001282481, NM_001282480
    PRNP Hs.472010 NM_001080121, NM_001271561, NM_001080122, 0.236
    NM_000311, NM_183079, NM_001080123
    PRR5 NM_015366, NM_001198721, NM_181333, 0.0135
    NM_001017528, NM_001017530, NM_001017529
    PRSS23 Hs.25338, NM_001293180, NM_001293178, NM_007173, 0.158
    Hs.729257 NM_001293179
    PSMA5 Hs.485246 NM_002790, NM_001199773, NM_001199774, 0.213
    NM_001199772
    PSMB9 NM_002800 0.234
    PSMC2 Hs.437366 NM_001204453, NM_002803 0.246
    PSMD11 NM_001270482, NM_002815 0.206
    PSMD14 Hs.740477 NM_005805 0.131
    PTGFRN Hs.418093 NM_020440 0.215
    PTPN12 Hs.61812 NM_002835, NM_001131008, NM_001131009 0.205
    PTPN14 Hs.193557, NM_005401 0.124
    Hs.688910
    PTPN3 Hs.436429, NM_001145368, NM_002829, NM_001145369, 0.135
    Hs.698275 NM_001145370
    PTPN4 Hs.469809 NM_002830 0.187
    PTPN6 Hs.63489 NM_080548, NM_002831, NM_080549 0.0174
    PTTG1 NM_004219, NM_001282383, NM_001282382 0.235
    PUS7 Hs.520619 NM_019042 0.127
    PVR NM_006505, NM_001135769, NM_001135768, 0.0064
    NM_001135770
    PYCARD NM_013258, NM_145182 0.0843
    PYCR1 NM_006907, NM_153824, NM_001282279, 0.235
    NM_001282281, NM_001282280
    RAB37 NM_175738, NM_001163989, NM_001006638, 0.192
    NM_001163990
    RACGAP1 Hs.505469 NM_013277, NM_001126104, NM_001126103 0.167
    RAD51 NM_001164270, NM_002875, NM_133487, 0.246
    NM_001164269
    RAP1GAP2 Hs.499659, NM_015085, NM_001100398 0.167
    Hs.685132
    RARRES3 NM_004585 0.116
    RASAL1 Hs.528693 NM_001193520, NM_004658, NM_001193521, 0.125
    NM_001301202
    RASGEF1A Hs.125293 NM_001282862, NM_145313 0.0831
    RASGRP2 Hs.99491 NM_001098671, NM_001098670, NM_153819 0.115
    RASGRP4 Hs.130434 NM_001146202, NM_001146204, NM_001146205, 0.129
    NM_170604, NM_001146207, NM_001146203,
    NM_001146206
    RBMS1 Hs.470412, NM_016836, NM_002897 0.0466
    Hs.654231
    REEP2 NM_001271803, NM_016606 0.135
    REPIN1 Hs.647086 NM_001099695, NM_014374, NM_013400, 0.243
    NM_001099696
    RGS1 Hs.75256 NM_002922 0.149
    RGS12 NM_198229, NM_002926, NM_198227 0.192
    RGS9 Hs.664380 NM_001165933, NM_001081955, NM_003835 0.222
    RHOB Hs.502876 NM_004040 0.0842
    RNF125 Hs.633703 NM_017831 0.135
    RNF19A Hs.292882, NM_015435, NM_183419, NM_001280539 0.0544
    Hs.735657
    RPLP2 NM_001004 0.111
    RPS20 NM_001146227, NM_001023 0.189
    RPS27 NM_001030 0.0505
    RPS28 NM_001031 0.0135
    RPS6KA3 Hs.445387 NM_004586 0.189
    RSAD2 Hs.17518 NM_080657 0.229
    RTF1 Hs.511096 NM_015138 0.219
    RYBP NM_012234 0.156
    S100A4 NM_019554, NM_002961 0.00133
    S1PR1 Hs.154210 NM_001400 0.151
    S1PR4 Hs.662006, NM_003775 0.0505
    Hs.688059
    SALL2 Hs.416358, NM_005407, NM_001291446, NM_001291447 0.0685
    Hs.745364
    SCD Hs.558396 NM_005063 0.0265
    SCGB1A1 NM_003357 0.152
    SCML4 NM_001286408, NM_001286409, NM_198081 0.0243
    SDC4 Hs.632267 NM_002999 0.156
    SDK2 Hs.435719 NM_001144952 0.0307
    SECTM1 Hs.558009 NM_003004 0.0735
    SELPLG NM_003006, NM_001206609 0.245
    SEMA3B Hs.82222 NM_001290060, NM_001005914, NM_001290061, 0.0477
    NM_001290063, NM_001290062, NM_004636
    SEPT3 Hs.120483 NM_145733, NM_019106 0.0175
    SERPINF1 NM_002615 0.11
    SERPINF2 Hs.159509 NM_001165921, NM_001165920, NM_000934 0.21
    SFMBT2 Hs.407983 NM_001018039, NM_001029880 0.0871
    SFXN1 NM_022754 0.174
    SGCB Hs.438953 NM_000232 0.12
    SGTB Hs.482301 NM_019072 0.246
    SH2B2 Hs.489448 NM_020979 0.0127
    SHF Hs.310399 NM_001301169, NM_001301168, NM_138356, 0.0567
    NM_001301170, NM_001301171
    SIGIRR Hs.501624 NM_001135054, NM_021805, NM_001135053 0.143
    SIGLEC9 Hs.245828 NM_014441, NM_001198558 0.246
    SIPA1L1 Hs.654657 NM_001284245, NM_015556, NM_001284247, 0.215
    NM_001284246
    SKAP1 Hs.316931 NM_003726, NM_001075099 0.0127
    SLA2 Hs.713578 NM_032214, NM_175077 0.227
    SLAMF1 NM_003037 0.246
    SLC11A1 Hs.591607 NM_000578 0.144
    SLC14A1 NM_001308278, NM_015865, NM_001146036, 0.243
    NM_001128588, NM_001308279, NM_001146037
    SLC16A1 Hs.75231 NM_003051, NM_001166496 0.18
    SLC1A4 Hs.654352 NM_001193493, NM_003038 0.246
    SLC1A5 Hs.631582 NM_005628, NM_001145145, NM_001145144 0.218
    SLC22A17 Hs.373498 NM_001289050, NM_016609, NM_020372 0.0366
    SLC25A20 Hs.13845 NM_000387 0.22
    SLC27A2 Hs.11729 NM_003645, NM_001159629 0.0274
    SLC29A1 Hs.25450 NM_001304463, NM_001078175, NM_001078177, 0.00562
    NM_001304465, NM_001304466, NM_001304462
    SLC2A1 Hs.473721 NM_006516 0.0507
    SLC2A3 Hs.419240 NM_006931 0.192
    SLC35F2 Hs.524014 NM_017515 0.0567
    SLC39A1 Hs.7854 NM_001271958, NM_001271957, NM_014437, 0.144
    NM_001271959, NM_001271960, NM_001271961
    SLC39A14 Hs.491232 NM_015359, NM_001135154, NM_001128431, 0.156
    NM_001135153
    SLC43A1 Hs.591952 NM_003627, NM_001198810 0.192
    SLC43A3 Hs.99962 NM_014096, NM_001278201, NM_199329, 0.015
    NM_017611, NM_001278206
    SLC46A3 NM_181785, NM_001135919 0.0767
    SLCO3A1 Hs.311187 NM_013272, NM_001145044 0.151
    SMAP2 Hs.15200 NM_022733, NM_001198978, NM_001198980, 0.164
    NM_001198979
    SMTN NM_134270, NM_006932, NM_134269, 0.162
    NM_001207018, NM_001207017
    SMYD5 Hs.631882 NM_006062 0.246
    SNAI2 Hs.360174 NM_003068 0.0502
    SNTB1 Hs.46701 NM_021021 0.162
    SNTG1 NM_001287813, NM_018967, NM_001287814 0.0585
    SORD NM_003104 0.0455
    SPATA7 NM_018418, NM_001040428 0.0965
    SPATS2L Hs.120323, NM_001282735, NM_015535, NM_001100422, 0.061
    Hs.734045 NM_001282743, NM_001100424, NM_001100423,
    NM_001282744
    SPINK2 NM_021114, NM_001271718, NM_001271720, 0.0642
    NM_001271722, NM_001271721
    SPINT1 Hs.233950 NM_003710, NM_001032367, NM_181642 0.246
    SPSB3 Hs.592080 NM_080861 0.122
    SQLE Hs.71465 NM_003129 0.0502
    SREK1IP1 Hs.69504 NM_173829 0.163
    SSBP3 Hs.733025 NM_145716, NM_001009955, NM_018070 0.0112
    SSH1 NM_018984, NM_001161330, NM_001161331 0.163
    SSR2 NM_003145 0.0544
    ST6GALNAC2 Hs.592105 NM_006456 0.224
    ST8SIA1 Hs.408614 NM_003034 0.0699
    STAMBP Hs.469018, NM_213622, NM_006463, NM_201647 0.11
    Hs.732857
    STAP1 Hs.435579 NM_012108 0.0685
    STAT6 Hs.524518 NM_003153, NM_001178078, NM_001178081, 0.158
    NM_001178079, NM_001178080
    STIL Hs.525198, NM_001282936, NM_003035, NM_001048166, 0.205
    Hs.673209 NM_001282937, NM_001282939, NM_001282938
    STIP1 Hs.337295, NM_001282652, NM_006819, NM_001282653 0.223
    Hs.618350
    STK38 Hs.409578 NM_001305102, NM_007271 0.0924
    STMN3 NM_015894, NM_001276310 0.0735
    STOM NM_004099, NM_001270526, NM_198194, 0.112
    NM_001270527
    STX1A Hs.647024 NM_004603, NM_001165903 0.222
    STX6 Hs.518417 NM_005819, NM_001286210 0.236
    SV2A Hs.516153 NM_014849 0.216
    SVIL Hs.499209 NM_003174, NM_021738 0.243
    SYT1 Hs.310545 NM_001135805, NM_005639, NM_001291901, 0.0645
    NM_001135806
    SYTL1 NM_032872, NM_001193308 0.23
    SYTL2 Hs.369520 NM_206929, NM_206930, NM_001162951, 0.0502
    NM_001162952, NM_001289610, NM_001289608,
    NM_032943, NM_001162953
    SYTL3 NM_001242395, NM_001009991, NM_001242384, 0.129
    NM_001242394
    TAF7 Hs.438838 NM_005642 0.0836
    TARP 0.22
    TARS NM_001258437, NM_152295, NM_001258438 0.158
    TBCC NM_003192 0.246
    TBX15 Hs.146196 NM_152380 0.00865
    TCEAL4 NM_001300901, NM_024863, NM_001006935, 0.0594
    NM_001006937
    TCF7 NM_003202, NM_201634, NM_001134851, 0.114
    NM_213648, NM_201632
    TERT Hs.492203 NM_198253, NM_001193376 0.0311
    TGFB1 Hs.645227 NM_000660 0.12
    TGFBR3 NM_001195684, NM_001195683, NM_003243 0.188
    TIGIT Hs.421750 NM_173799 0.234
    TJP3 Hs.25527 NM_001267560, NM_001267561 0.248
    TLE4 NM_007005, NM_001282760, NM_001282748, 0.0302
    NM_001282749, NM_001282753
    TMEM194A 0.106
    TMEM212 Hs.642307 NM_001164436 0.102
    TMEM48 0.171
    TMEM5 Hs.216386 NM_014254, NM_001278237 0.178
    TMEM71 Hs.293842 NM_144649, NM_001145153 0.234
    TMEM80 NM_001042463, NM_174940, NM_001276274, 0.246
    NM_001276253
    TMEM9B NM_001286094, NM_020644, NM_001286095 0.192
    TMPRSS6 Hs.370885 NM_153609, NM_001289001, NM_001289000 0.0585
    TNFAIP1 Hs.76090 NM_021137 0.167
    TNFRSF11A NM_001270951, NM_003839, NM_001278268, 0.0605
    NM_001270949, NM_001270950
    TNFRSF18 Hs.212680 NM_148901, NM_004195, NM_148902 0.0366
    TNFRSF25 Hs.462529 NM_148967, NM_148970, NM_148966, 0.163
    NM_003790, NM_148965
    TNFRSF4 Hs.129780 NM_003327 0.0337
    TNFRSF8 Hs.1314 NM_001243, NM_001281430 0.106
    TNFRSF9 Hs.86447 NM_001561 0.0524
    TOX2 Hs.26608 NM_001098797, NM_001098796, NM_032883, 0.0107
    NM_001098798
    TP53INP2 Hs.516994 NM_021202 0.139
    TPCN1 Hs.524763 NM_017901, NM_001143819, NM_001301214 0.181
    TPK1 Hs.660232 NM_022445, NM_001042482 0.24
    TPMT Hs.444319 NM_000367 0.232
    TRAPPC6A NM_024108, NM_001270893, NM_001270891, 0.0638
    NM_001270892
    TRIB1 Hs.444947 NM_025195, NM_001282985 0.074
    TRIB2 Hs.467751 NM_021643 0.0181
    TRIM25 Hs.528952 NM_005082 0.143
    TRIP10 Hs.515094 NM_004240, NM_001288962, NM_001288963 0.149
    TRPM3 NM_001007471, NM_020952, NM_206946, 0.195
    NM_206945, NM_001007470, NM_206948,
    NM_024971, NM_206944, NM_206947
    TSPAN18 Hs.385634 NM_130783 0.129
    TSPAN32 NM_139022 0.0502
    TTC21B Hs.310672 NM_024753 0.243
    TTC39C Hs.733420 NM_153211, NM_001243425, NM_001135993, 0.00494
    NM_001292030
    TTLL4 Hs.471405 NM_014640 0.158
    TUBB Hs.636480 NM_001293213, NM_178014, NM_001293215, 0.236
    NM_001293216, NM_001293212
    TUBB2B Hs.300701 NM_178012 0.0386
    TUBB6 Hs.193491, NM_001303524, NM_032525, NM_001303529, 0.00952
    Hs.744066 NM_001303526, NM_001303525
    UBA52 Hs.5308 NM_003333, NM_001033930 0.0902
    UBASH3B Hs.444075 NM_032873 0.0666
    UBL3 Hs.145575 NM_007106 0.0929
    UHRF1BP1L Hs.620701 NM_015054, NM_001006947 0.0666
    UNC119 Hs.410455 NM_005148, NM_054035 0.168
    URGCP Hs.663312 NM_017920, NM_001077664, NM_001290075, 0.141
    NM_001290076, NM_001077663
    USP22 Hs.462492 NM_015276 0.0745
    USP25 NM_013396, NM_001283042, NM_001283041 0.232
    USP51 Hs.40061 NM_201286 0.186
    UXS1 Hs.730756 NM_001253875, NM_025076, NM_001253876 0.167
    UXT NM_004182, NM_153477 0.0826
    VDAC3 NM_001135694, NM_005662 0.158
    VDR Hs.524368 NM_001017535, NM_001017536, NM_000376 0.124
    VIPR1 Hs.348500, NM_001251882, NM_001251885, NM_004624, 0.0642
    Hs.683175 NM_001251883, NM_001251884
    VNN2 Hs.293130, NM_004665, NM_078488, NM_001242350 0.129
    Hs.740120
    VSIG1 Hs.177164 NM_182607, NM_001170553 0.0709
    WASF2 Hs.469244 NM_006990, NM_001201404 0.137
    WEE1 NM_003390, NM_001143976 0.0594
    WIPI1 Hs.463964 NM_017983 0.171
    WNT10A Hs.121540 NM_025216 0.0502
    XYLT1 Hs.22907 NM_022166 0.0278
    YPEL1 NM_013313 0.195
    YWHAG Hs.744840 NM_012479 0.0783
    ZBP1 Hs.302123 NM_001160417, NM_030776, NM_001160418, 0.218
    NM_001160419
    ZBTB20 NM_001164343, NM_001164347, 0.00562
    NM_001164345, NM_001164342, NM_015642,
    NM_001164344, NM_001164346
    ZBTB32 Hs.99430, NM_014383 0.0128
    Hs.736841
    ZC3H12A Hs.656294 NM_025079 0.198
    ZC3H12C Hs.376289 NM_033390 0.0628
    ZC3H12D NM_207360 0.0709
    ZEB2 Hs.34871 NM_014795, NM_001171653 0.0594
    ZFP161 0.0965
    ZFP36L2 Hs.503093 NM_006887 0.218
    ZHX2 Hs.377090 NM_014943 0.144
    ZNF267 NM_003414 0.127
    ZNF282 Hs.729056 NM_003575, NM_001303481 0.121
    ZNF506 NM_001099269, NM_001145404 0.195
    ZNF587 Hs.744891 NM_032828, NM_001204817 0.236
    ZNF652 NM_014897, NM_001145365 0.0429
    ZNF688 NM_145271, NM_001024683 0.229
    ZNF704 Hs.434957, NM_001033723 0.215
    Hs.730558
    ZNRF1 Hs.427284 NM_032268 0.164
  • Example 3: Prognostic Flow Cytometry-Based Assays
  • Prognostic flow cytometry-based assays are developed to screen subjects with cancer (e.g., patients with a hematological cancer such as ALL and CLL) for CAR-expressing cell (e.g., T cell, NK cell) therapy, e.g., CD19 CAR-expressing cell therapy as described herein such as, e.g., CTL019 therapy. In some embodiments, subjects are participating in clinical trials.
  • A sample (e.g., a blood sample) is isolated from a patient and a fluorescent flow cytometry-based assay is performed screening for one or more cell surface or secreted biomarkers described in Examples 1 and 2. An exemplary list of markers that are measured, e.g., by flow cytometry if cell surface-expressed, or by ELISA if secreted, and whose expression values predict patient response to CAR-expressing cell (e.g., T cell, NK cell) therapy, e.g., CD19 CAR-expressing cell therapy as described herein such as, e.g., CTL019 therapy includes, but is not limited to, genes listed in Table 8.
  • TABLE 8
    Exemplary markers measured that predict patient response to CAR-expressing cell therapy
    Gene Unigene Accession No.
    ATP1B3 Hs.477789 NM_001679
    CCL17 Hs.546294 NM_002987
    CCL3 Hs.514107 NM_002983
    CCL4 Hs.75703 NM_002984
    CCR1 Hs.301921 NM_001295
    CD40LG Hs.592244 NM_000074
    CD58 Hs.34341 NM_001144822, NM_001779
    CD70 Hs.715224, Hs.501497 NM_001252
    CD80 Hs.838 NM_005191
    CSF1 Hs.591402 NM_000757, NM_172212, NM_172211, NM_172210
    FCER2 Hs.465778 NM_002002
    GPR56 Hs.513633 NM_001145773, NM_001145774, NM_001145771,
    NM_001145772, NM_005682, NM_201525,
    NM_001145770
    HAVCR1 Hs.129711 NM_001099414, NM_012206
    HLA-DMA Hs.351279 NM_006120
    HLA-DPA1 Hs.347270 NM_033554
    HLA-DRA Hs.520048 NM_019111
    HLA-DRB1 Hs.716081, Hs.696211, NM_002124, NM_021983, XM_002346251
    Hs.723344, Hs.534322
    HLA-DRB5 Hs.534322 NM_002125
    ICAM3 Hs.654563 NM_002162
    IFNAR2 Hs.708195 NM_207584, NM_207585, NM_000874
    IFNG Hs.856 NM_207585
    IGF1R Hs.643120, Hs.714012 NM_000875
    IL10 Hs.193717 NM_000572
    IL13 Hs.845 NM_002188
    IL15RA Hs.524117 NM_002189, NM_172200
    IL21 Hs.567559 NM_021803
    IL2RA Hs.231367 NM_000417
    IL2RB Hs.474787 NM_000878
    IL3 Hs.694 NM_000588
    IL4 Hs.73917 NM_000589, NM_172348
    IL5 Hs.2247 NM_000879
    IL6ST Hs.532082 NM_002184, NM_175767
    IL9 Hs.960 NM_000590
    ITGA6 Hs.133397 NM_000210, NM_001079818
    KIT Hs.479754 NM_000222, NM_001093772
    LAIR1 Hs.572535 NM_001289023, NM_001289025, NM_001289026,
    NM_001289027, NM_002287, NM_021706
    NFATC1 Hs.534074, Hs.701518 NM_001278669, NM_001278670, NM_001278672,
    NM_001278673, NM_001278675, NM_006162,
    NM_172387, NM_172388, NM_172389, NM_172390
    SELL Hs.728756 NM_000655
    SELP Hs.73800 NM_003005
    SIRPG Hs.590883 NM_001039508, NM_018556, NM_080816
    STAT6 Hs.524518 NM_001178078, NM_001178079, NM_001178080,
    NM_001178081, NM_003153
    TFRC Hs.529618 NM_001128148, NM_003234
    TIMD4 Hs.334907 NM_001146726, NM_138379
    TNFRSF1B Hs.256278 NM_001066
    TNFRSF9 Hs.86447, Hs.738942 NM_001561
  • Example 4: Classifiers to Predict Class Membership
  • Based on the biological understanding, combinations of genes from unbiased feature selection, gene sets, and selected genes of interest are used to further differentiate complete responders from partial responders and non-responders. In an embodiment, combinations of genes from unbiased feature selection, gene sets, and selected genes of interest are used to further differentiate relapsers from non-relapsers. In an embodiment, classifiers are built based on all genes to predict class membership. In an embodiment, predictions of class membership further differentiate NR's, PR's, and CR's. In an embodiment, predictions of class membership further differentiate relapsers from non-relapsers. Alternatively or additionally, a classifier is built which uses a subset of predetermined significant features. Significant features include, but are not limited to, enriched meta-gene, a subset of significantly differentially expressed genes in the meta-genes, and combinations thereof.
  • Example 5: Cytokine Expression Signatures Predictive of CAR-Expressing Cell Potency
  • The present example describes the identification of exemplary cytokine expression signatures that predict patient response to CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., a CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy, e.g., a CTL019 therapy) in Chronic Lymphoid Leukemia (CLL) and Acute Lymphoblastic Leukemia (ALL), for use in accordance with the present invention.
  • Among other things, the present Example describes novel cytokine expression signatures that predict the potency of manufactured CAR-expressing cell (e.g., T cell, NK cell) cell products based on secreted cytokine profiles following activation in vitro.
  • In an embodiment, novel cytokine expression signatures described herein predict the potency of manufactured CAR-expressing cell (e.g., T cell, NK cell) products to kill target tumor cells.
  • In an embodiment, novel cytokine expression signatures described herein are correlated with patient response to CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g. CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy, e.g., a CTL019 CAR-expressing cell therapy) in CLL to improve the CAR-expressing cell product prior to infusion in patients.
  • In an embodiment, novel cytokine expression signatures described herein are used to assess manufactured CAR-expressing cell (e.g., T cell, NK cell) products (e.g., CD19 CAR-expressing cell products, e.g., CTL019 product). In an embodiment, novel cytokine expression signatures described herein provide an endpoint in manufacturing process optimiziation.
  • Novel cytokine expression signatures based on cytokine protein expression levels in manufactured CD19 CAR-expressing cell (e.g., T cell, NK cell) product samples prior to re-infusion have been identified that predict patient response to CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy in Chronic Lymphoid Leukemia (CLL). The identified signatures were discovered in a cytokine protein expression study of manufactured product samples prepared from 21 CLL subject samples. CLL subject samples (21 total) were stratified as follows: CTL019 manufactured product was derived from 6 patients that were complete responders (CRs) to CTL019 therapy, 5 patients that were partial responders (PRs), and 10 non-responders (NRs). Several cytokine expression signatures discriminating responders from non-responders in manfactured product were discovered using a Luminex® panel of 13 cytokines.
  • The potency of CTL019 manufactured products from 21 CLL patients were assessed in a tumor cell killing assay. Briefly, manufactured CTL019 products were “activated” in vitro by CD19-expressing K562 (K562-19 cells). Without wishing to be bound by a particular theory, CD19 expressing K562 cells (e.g., K562-19 cells) mimic leukemic CD19 expressing B-cells in CLL patients. CTL019 cells are engineered to identify and kill cells that express CD19 antigen on their cell surface and CTL019-mediated killing of K562-19 cells serves as a proxy for asessing potency of CTL019-mediated killing of tumor cells.
  • Following CTL019 product activation, cytokine protein expression profiles were measured in the co-cultured media using a Luminex® panel of cytokines. The expression profiles of exemplary cytokines were measured and the potency of CTL019 cell products was correlated with the expression of different cytokines. Exemplary cytokines considered in this analysis are provided in Table 14.
  • TABLE 14
    Exemplary cytokines
    Cytokine Entrez ID Official Gene Symbol
    CCL-20/MIP-3a 6364 CCL20
    GM-CSF 1437 CSF2
    IFNγ 3458 IFNG
    IL-10 3586 IL10
    IL-13 3596 IL13
    IL-17a 3605 IL17A
    IL-2 3558 IL2
    IL-21 59067 IL21
    IL-4 3565 IL4
    IL-5 3567 IL5
    IL-6 3569 IL6
    IL-9 3578 IL9
    TNFα 7124 TNF
  • Novel cytokine expression profiles were then discovered using various data analytical approaches including 1) bi-clustering analysis; and 2) univariate analysis.
  • Cytokine expression data derived from the Luminex® assay were log-normalized and subjected to bi-clustering analyses (hierarchical clustering was performed using the complete linkage method). Bi-clustering analyses of cytokine expression in stimulated CTL019 products and CLL patients yield four major clusters (a cut-off distance ≤1.0, resulted in 4 clusters as shown in FIG. 17) and distinct subgroups of CRs/PRs and NRs were identified. An exemplary heatmap of bi-clustering of cytokine expression in stimulated CTL019 products and CLL patients is shown in FIG. 17. Surprisingly, two clusters (Cluster 1 and Cluster 3) were almost exclusively comprised of CRs and PRs, whereas the other two clusters (Cluster 2 and Cluster 4) contained predominantly NRs. On average, cytokine expression levels were higher in CRs/PRs versus NRs (FIG. 17).
  • Next, a 3-group univariate analysis using ANOVA (analysis of variance) was performed that compared CRs versus PRs versus NRs. Statistical significance was determined using a p-value cut-off of 0.05. Statistical significance (e.g., p-values) of different cytokines to distinguish CRs, PRs and NRs are listed in Table 15.
  • TABLE 15
    Statistical significance of different cytokines to distinguish
    CRs, PRs and NRs in a 3-group univariate analysis using ANOVA
    Cytokine p-value
    CCL20/MIP3a 0.001838
    IL-17a 0.001857
    IL-6 0.006017
    TNFα 0.013499
    IL-2 0.034397
    IL-21 0.055684
    IL-5 0.075396
    IL-10 0.08935
    IL-9 0.098761
    IFNγ 0.137263
    GM-CSF 0.191839
    IL-4 0.197774
    IL-13 0.222134
  • The 3-group model of univariate analysis identified 5 cytokines, e.g., IL-17a, CCL-20/MIP3a, IL-6, IL-2, and TNFα, as statistically significant markers of response to CTL019 therapy in CLL patients (FIG. 18 and Table 15). Exemplary results of log-normalized expressions of statistically significant cytokines that distinguish CRs, PRs and NRs in CLL patients are shown in FIG. 18.
  • Manufactured CTL019 product was evaluted by flow cytometry to determine percentages of CAR+ cells. The 5 cytokines identified in the 3-group model of univariate analysis were further correlated to the percentage of CAR+ cells in each of the manufactured CTL019 products. Exemplary correlation coefficients and corresponding p-values of cytokine expression (derived from Luminex® panel discussed above) and percentages of CAR+ cells (determined by flow cytometery) are provided in Table 16.
  • TABLE 16
    Correlation coefficients and corresponding p-values
    of cytokine expression and percentage of CAR+ cells
    Cytokine Correlation coefficient p. value
    IL-17a 0.278349 0.221794
    IL-10 0.390318 0.08024
    CCL20/MIP3a 0.395273 0.076147
    IL-5 0.494758 0.022598
    IL-4 0.525982 0.014321
    TNFα 0.539276 0.011642
    GM-CSF 0.588262 0.005032
    IL-6 0.631841 0.002122
    IFNγ 0.660738 0.001112
    IL-2 0.661608 0.001089
    IL-21 0.674378 0.0008
    IL-9 0.70858 0.000324
    IL-13 7.53E−01 8.19E−05
  • The percentage of CAR+ cells in CTL019 product represents transduction efficiency. In CLL, the percent of CAR+ cells at pre-harvest levels distinguish responders (e.g., complete responders and partial responders) from non-responders (NR). FIG. 21 depicts an exemplary scatter plot showing percent of CAR+ cells (i.e., transduction rate) at pre-harvest for complete responders (CR) in red, partial responders (PR) in blue and non-responders (NR) in red. Transduction efficiencies were measured pre-harvest and correlated with subject response (e.g., CR, PR, or NR). The solid line represents a 15% transduction efficiency that separates the majority of non-responders from responders. Without wishing to be bound to a particular theory, these data indicate that pre-harvest CAR transduction efficiency is a marker of response to CAR-expressing cell (e.g., T cell, NK cell) therapy in CLL.
  • Correlation analysis of IL17a with CCL20 cytokine expression was performed, and their association with clinical response was evaluated by scatter plot analyses. FIG. 19A depicts an exemplary scatter plot showing log-normalized correlation of IL17A (y-axis) and CCL20 (x-axis) expression with a correlation coefficient of 0.928 and corresponding p-value of 1.36e-09. Dashed lines represent the classification boundary for separating NRs from CRs/PRs. Each dot in FIG. 19A represents a CLL patient, and the cross-hatch (NR), black (PR) and white (CR) represent the clinical response. Classification boundary in FIG. 19A demonstrates that the combination of IL-17a and CCL20 separates almost all NRs from CRs/PRs, and PRs in turn are clustered separately from CRs. Among other things, these data demonstrate CAR+ cell expression of one or more cytokines listed in Table 16 predict clinical response.
  • Surprisingly, IL-17a and CCL-20 expression levels were not correlated with the percentage of CAR+ cells in the CTL019 product (representing transduction efficiency). Without wishing to be bound by a particular theory, these data indicate IL-17a and CCL-20 cytokine expression levels are informative (e.g., predictive of response) with regard to potency of a manufactured CAR-expressing cell (e.g., T cell, NK cell) product, e.g., a manufactured CD19 CAR-expressing cell product, in several ways. First, cytokine signatures are correlated with patient response to CTL019 CAR-expressing cell therapy in CLL. Therefore, cytokine signatures described herein can be used to improve and/or modify CAR-expressing cell (e.g., T cell, NK cell) product (e.g., a CD19 CAR-expressing cell product such as, e.g., CTL019) prior to infusion in patients, for greater clinical efficacy. Second, cytokine signatures described herein can be used to assess manufactured CAR-expressing cell (e.g., T cell, NK cell) products thereby providing, among other things, an end point in manufacturing process optimization.
  • In an embodiment, cytokine signatures described herein define the potency of a CAR-expressing cell (e.g., T cell, NK cell) product. In an embodiment, cytokine signatures described herein are markers of response to a CAR-expressing cell (e.g., T cell, NK cell) product in a hematological cancer (e.g., CLL or ALL).
  • In an embodiment, cytokine signatures described herein predict subject response to a CAR-expressing cell (e.g., T cell, NK cell) product.
  • In an embodiment, cytokine signatures described in Table 16 predict subject response to a CAR-expressing cell (e.g., T cell, NK cell) product.
  • In an embodiment, IL-17a and CCL-20 expression levels predict subject response to a CAR-expressing cell (e.g., T cell, NK cell) product.
  • Example 6: Identification of Factors that Predict Subject Relapse to CD19 CAR-Expressing Cell Therapy in B-cell Acute Lymphocytic Leukemia (B-ALL)
  • The present Example describes, among other things, the identification of novel transcriptional gene signatures that predict patient relapse to CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., CTL019 therapy) in B cell Acute Lymphocytic Leukemia (B-ALL), for use in accordance with the present invention.
  • Among other things, the present Example describes novel gene signatures based on mRNA expression levels of selected genes in the patient prior to CD19 CAR-expressing cell (e.g., T cell, NK cell) treatment (e.g., CTL019) (apheresis or bone marrow) or in manufactured CD19 CAR-expressing cell (e.g., T cell, NK cell) product samples (e.g., CTL019) prior to re-infusion. In an embodiment, the present example describes novel gene signatures that discriminate relapsers to CTL019 therapy in B-ALL from non-relapsers to CTL019 therapy in B-ALL.
  • The present Example describes methods of unbiased feature selection to discover novel gene signatures that predict subject relapse to CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., CTL019) in B-ALL, for use in accordance with the present invention.
  • The present Example also describes methods of Gene Set Analysis to discover novel gene signatures, for use in accordance with the present invention.
  • Novel gene signatures based on mRNA expression levels in manufactured CD19 CAR-expressing cell (e.g., T cell, NK cell) product samples prior to re-infusion were identified that predict subject relapse to CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy in B cell Acute Lymphocytic Leukemia (B-ALL). The identified signatures were discovered in a whole genome RNAseq study of manufactured product samples which included 7 B-ALL subject samples. B-ALL subject samples (7 total) were stratified as follows: biological samples were taken from 4 subjects who did not relapse (“non-relapsers”) following CTL019 therapy, and 3 subjects who did relapse (“relapsers”) following CTL019 therapy. Several gene signatures discriminating responders from non-responders, and relapsers from non-relapsers, in manufactured product samples were discovered and are described further in detail below.
  • Novel gene signatures were then discovered using various data analytical approaches: 1) unbiased feature selection; 2) gene set analysis; and 3) differential expression analysis of selected genes of interest.
  • Novel gene signatures derived from unbiased feature selection were discovered by determining which genes were differentially expressed between the 2-group comparison of relapsers and non-relapsers which compared the 3 relapsers to the 4 non-relapsers. Genes were defined as differentially expressed if their differential expression was statistically significant in the 2-group comparison with a FDR p-value cutoff of 0.25. The gene list for the relapser versus non-relapser comparison (N=17) is tabulated in Table 17. 2-group statistical models were applied to determine whether the meta-gene was statistically different between the groups, similar to the approach illustrated in FIG. 2B. FIG. 2B depicts an exemplary heat map of genes upregulated in activated TEFF versus resting TEFF cells for complete responders (CR), partial responders (PR), and non-responders (NR).
  • Without wishing to be bound by a particular theory, these data indicate that the differentiation state of T cells in CD19 CAR-expressing cell (e.g., T cell, NK cell) product (e.g., CTL019) correlate with subject response (i.e., CR, PR, or NR) and predict subject relapse to CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., CTL019 therapy) in B-ALL. As described in Example 1, complete responders gene signatures are more like resting TREG and TEFF cells. Among other things, gene signatures for relapsers (e.g., a complete responder that relapses to CTL019 therapy) contain genes upregulated in TREG versus TEFF cells at resting. Without wishing to be bound by a particular theory, these data indicate that relapsers to CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., CTL019) in B-ALL have higher levels of TREG compared to non-relapsers to CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., CTL019). FIG. 21 depicts exemplary results illustrating that TREG are differentially enriched in relapsers (R) versus non-relapsers, e.g., relapsers express high levels of TREG genes compared to complete responders (CR) (e.g., non-relapsers). An exemplary publication or sequence number disclosing the sequence of each gene is also given in Table 17, and each publication is incorporated by reference in its entirety, including all nucleic acid and protein sequences therein.
  • TABLE 17
    Table 17: Exemplary Genes that Predict Patient Relapse to CTL019 Therapy
    Exemplary
    Gene miRBase Unigene Accession No. FDR Publication
    MIR199A1 MI0000242 NR_029586.1 2.11E−05 Landgraf et al., Cell
    129 (7), 1401-1414
    (2007)
    PPIAL4D Hs.730589 NM_001164261.1 3.94E−05 SEQ ID NO: 102
    MIR1203 MI0006335 NR_031607.1 4.63E−03 Marton et al.,
    Leukemia 22 (2), 330-
    338 (2008)
    uc021ovp 6.73E−03 SEQ ID NO: 103
    ITM2C Hs.111577 NM_001012514.2 1.17E−01 Yoshida et al., Int. J.
    NM_001012516.2 Mol. Med. 25 (4), 649-
    NM_001287240.1 656 (2010)
    NM_001287241.1
    NM_030926.5
    HLA-DQB1 Hs.409934 NM_001243961.1 1.17E−01 Pankuweit et al., Gene
    Hs.534322 NM_001243962.1 531 (2), 180-183
    NM_002123.4 (2013)
    TTTY10 Hs.461175 NR_001542.1 1.25E−01 Derrien et al., Genome
    Res. 22 (9), 1775-1789
    (2012)
    TXLNG2P Hs.522863 NR_045128.1 2.27E−01 Prakash et al., PLoS
    NR_045129.1 ONE 5 (10), E13284
    (2010)
    MIR4650-1 MI0017277 NR_039793.1 2.27E−01 Persson et al., Cancer
    Res. 71 (1), 78-86
    (2011)
    KDM5D Hs.80358 NM_001146705.1 2.27E−01 Kim et al., J. Am. Soc.
    NM_001146706.1 Nephrol. 20 (9), 2025-
    NM_004653.4 2033 (2009)
    USP9Y Hs.598540 NM_004654.3 2.27E−01 Luddi et al., N. Engl. J.
    Med. 360 (9), 881-885
    (2009)
    PRKY Hs.584730 NR_028062.1 2.27E−01 Hogan et al., Clin Med
    Res 7 (3), 69-84
    (2009)
    RPS4Y2 Hs.367761 NM_001039567.2 2.27E−01 Ye et al., BMC
    Bioinformatics 13, 134
    (2012)
    RPS4Y1 Hs.282376 NM_001008.3 2.27E−01 Eljaafari et al., J.
    Immunol. 190 (1),
    184-194 (2013)
    NCRNA00185 Hs.138453 NR_001543.3 2.28E−01 Prakash et al., PLoS
    Hs.729534 NR_125733.1 ONE 5 (10), E13284
    Hs.734681 NR_125734.1 (2010)
    NR_125735.1
    NR_125736.1
    NR_125737.1
    SULT1E1 Hs.479898 NM_005420.2 2.33E−01 Xu et al., Mol. Cell.
    Endocrinol. 369 (1-2),
    140-149 (2013)
    EIF1AY Hs.461178 NM_001278612.1 2.38E−01 Luna et al.,
    NM_004681.3 Biochemistry 52 (52),
    9510-9518 (2013)
  • Gene set analysis yielded a number of gene signatures predictive of subject relapse to CTL019 therapy in B-ALL. The following genes showed increased levels in relapsers and decreased levels in non relapsers: MIR199A1, MIR1203, uc021ovp, ITM2C, and HLA-DQB1. The following genes showed decreased levels in relapsers and increased levels in non relapsers: PPIAL4D, TTTY10, TXLNG2P, MIR4650-1, KDM5D, USP9Y, PRKY, RPS4Y2, RPS4Y1, NCRNA00185, SULT1E1, and EIF1AY.
  • In particular, the present Example describes methods of Gene Set Analysis to discover novel gene signatures, for use in accordance with the present invention.
  • Among other things, the present Example describes novel gene signatures based on Gene Set Analysis, that are predictive of patent relapse to CD19 CAR-expressing cell (e.g., T cell, NK cell) therapy (e.g., CTL019) in B-ALL. Gene set analysis was performed on gene sets described in Table 17, and with gene sets described in Example 2, e.g., gene sets were sourced from (1) additional experiments were based on gene sets by Szabo et al., (disclosed herein); (2) gene sets published by Abbas et al. in Genome Research 2005; and (3) gene sets published by Gattinoni et al. in Nature Medicine 2011. Each of Szabo, Abbas and Gattinoni gene sets are described in detail in Example 2. The gene sets defined by Szabo and considered in this analysis are tabulated in Table 2 of Example 2. The gene sets defined by Abbas and considered in this analysis are tabulated in Table 3 of Example 2. The gene sets defined by Gattinoni and considered in this analysis are tabulated in Table 4 of Example 2.
  • Each gene set (e.g., B-ALL RNAseq gene sets, Szabo gene sets, Abbas gene sets, and Gattinoni gene sets) was evaluated to determine its association with subject response (i.e., relapser or non-relapser) in the following manner: a meta-gene was calculated for each subject, where the meta-gene score for subject j was defined as

  • m ji=G 1 x ij−μ(x .j)/σ(x .j)
  • where xij is the expression value of gene i in subject j for a given gene set n=1, . . . , G; μ(x.j) is the mean of genes 1, . . . , G in subject j; and σ(x.j) is the standard deviation of genes 1, . . . , G in subject j.
  • A 2-group statistical model was applied to each gene set to determine whether the meta-gene was statistically different between the manufactured CTL019 product of relapsers and non-relapsers. A schematic illustrating this approach is given in FIG. 2B. Of the Szabo, Abbas, and Gattinoni gene sets, there was one gene set that was significantly differentially enriched between relapsers and non-relapsers. This gene set was from the Szabo collection and contains genes upregulated in TREG versus TEFF cells at resting, and correlated with patient relapse to CTL019 therapy. Specifically, this gene set was found to be enriched in relapsers, indicating that relapsers have higher levels of TREGS compared to non-relapsers. For example, the meta-gene score for the gene set comprised of genes upregulated in TREG in comparison to TEFF cells is found to be correlated with patient relapse in product samples (see FIG. 20). FIG. 20 depicts exemplary results (p=0.000215) illustrating that TREG genes have high expression levels in relapsers (R) compared to non-relapser, complete responders (CR). The x-axis is samples by response group where CR=complete responder and R=relapser. The y-axis is normalized meta-gene expression scores.
  • In an embodiment, gene signatures described herein are used to enable manufactured product improvements, thereby reducing the likelihood of patient relapse. In an embodiment, gene signatures described herein are used to modify therapeutic application of manufactured product, thereby reducing the likelihood of patient relapse.
  • In an embodiment, gene signatures described herein are identified in a subject prior to CAR-expressing cell (e.g., T cell, NK cell) treatment (e.g., a CD19 CAR-expressing cell treatment, e.g., CTL019 therapy) that predict relapse to CAR-expressing cell (e.g., T cell, NK cell) treatment. In an embodiment, gene signatures described herein are identified in an apheresis sample. In an embodiment, gene signatures described herein are identified in a bone marrow sample. In an embodiment, gene signatures described herein are identified in a manufactured CAR-expressing cell (e.g., T cell, NK cell) product (e.g., a CD19 CAR-expressing cell product, e.g., CTL019) prior to infusion.
  • Without wishing to be bound by a particular theory, these data indicate that decreasing the TREG signature in the patient prior to apheresis or during manufacturing of the CAR-expressing cell (e.g., T cell, NK cell) product significantly reduces the risk of patient relapse.
  • Example 7: Quantity of CD27+PD1− CART Cells Infused into Patient Predicts Response to Therapy
  • The number of CD27+PD1− cells in the CTL019 infusion product was determined for 29 CLL patients (8 complete responders and 21 non-responders). The relationship between number of CD27+PD1− CART cells infused and response to therapy is shown as a bar graph in FIG. 22. and as a scatter plot in FIG. 23. A threshold was set at 1×107 CART cells per patient. A statistically significant difference (p<0.0001) was observed between the complete responders and non-responders. This experiment shows that complete remission of CLL patients to CART19 immunotherapy is associated with higher numbers of infused CD27+PD1− CART cells.
  • EQUIVALENTS
  • Other embodiments of the invention will be apparent to those skilled in the art from a consideration of the specification or practice of the invention disclosed herein. While this invention has been disclosed with reference to specific aspects, it is apparent that other aspects and variations of this invention may be devised by others skilled in the art without departing from the true spirit and scope of the invention. The appended claims are intended to be construed to include all such aspects and equivalent variations.

Claims (64)

1. A method of evaluating a subject, having a cancer, comprising:
acquiring a value of responder or relapser status to a therapy comprising a CAR-expressing cell population for the subject, wherein said value of responder or relapser status comprises a measure of one, two, three, four, five, six, seven, or more of the following:
(i) the level or activity of CD27 and/or CD45RO− immune effector cells in a sample;
(ii) the level or activity of one, two, three, or more of resting TEFF cells, resting TREG cells, younger T cells, or early memory T cells, or a combination thereof, in a sample;
(iii) the level or activity of one, two, three, or more of activated TEFF cells, activated TREG cells, older T cells, or late memory T cells, or a combination thereof, in a sample;
(iv) the level or activity of an immune cell exhaustion marker in a sample;
(v) the level or activity of one, two, three, four, five, ten, twenty or more of the biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 16, Table 17, Table 18, Table 20, FIG. 2B, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature;
(vi) a cytokine level or activity in a CAR-expressing cell product sample, wherein the cytokine is chosen from one, two, three, four, five or more of the cytokines listed in Table 16;
(vii) a transduction efficiency of a CAR-expressing cell in a manufactured CAR-expressing cell product sample; or
(viii) a quantity of CD27+PD-1− cells in a sample,
wherein said value is indicative of the subject's responsiveness or relapsing status to the CAR-expressing cell therapy, thereby evaluating the subject.
2-3. (canceled)
4. A method for treating a subject having a cancer, comprising:
administering to the subject a therapeutically effective dose of a CAR-expressing cell therapy, if the subject is identified as being responsive to a therapy comprising a CAR-expressing cell population, wherein said identifying comprises a measure of one, two, three, four, five, six, seven, or more of the following:
(i) the level or activity of CD27 and/or CD45RO− immune effector cells in a sample;
(ii) the level or activity of one, two, three, or more of resting TEFF cells, resting TREG cells, younger T cells, or early memory T cells, or a combination thereof, in a sample;
(iii) the level or activity of one, two, three, or more of activated TEFF cells, activated TREG cells, older T cells, or late memory T cells, or a combination thereof, in a sample;
(iv) the level or activity of an immune cell exhaustion marker in a sample;
(v) the level or activity of one, two, three, four, five, ten, twenty or more of the biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 16, Table 17, Table 18, Table 20, FIG. 2B, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature;
(vi) a cytokine level or activity in a CAR-expressing cell product sample, wherein the cytokine is chosen from one, two, three, four, five or more of the cytokines listed in Table 16; or
(vii) a transduction efficiency of a CAR-expressing cell in a manufactured CAR-expressing cell product sample; or
(viii) a quantity of CD27+PD-1− cells in a sample,
thereby treating the subject.
5. The method of claim 4
wherein responsive to said value, performing one, two, three, four, five, six, seven, or more of:
identifying the subject as a complete responder, partial responder or non-responder, or a relapser or a non-relapser;
administering a CAR-expressing cell therapy;
administering an altered dosing of a CAR-expressing cell therapy;
altering the schedule or time course of a CAR-expressing cell therapy;
administering an additional agent in combination with a CAR-expressing cell therapy;
administering to a non-responder or partial responder a therapy that increases the number of younger T cells or naïve T cells in the subject prior to treatment with a CAR-expressing cell therapy;
modifying a manufacturing process of a CAR-expressing cell therapy;
administering an alternative therapy; or
if the subject is, or is identified as, a non-responder or a relapser, decreasing the TREG cell population and/or TREG gene signature.
6. The method of claim 4, wherein the CAR-expressing cell therapy comprises a plurality of CAR-expressing immune effector cells.
7. The method of claim 4, wherein the CAR-expressing cell therapy is a CAR19 therapy.
8. The method of claim 4, wherein the measure of one or more of (i)-(viii) is obtained from:
(a) an apheresis sample acquired from the subject, wherein optionally the apheresis sample; or
(b) a manufactured CAR-expressing cell product sample.
9. (canceled)
10. The method of claim 4, wherein the subject is evaluated prior to, during, or after receiving the CAR-expressing cell therapy.
11. The method of claim 4, wherein the cancer is a hematological cancer.
12. The method of claim 4, wherein the hematological cancer is an ALL or a CLL.
13. The method of claim 4, wherein the subject is a human patient.
14. The method of claim 4, further comprises identifying the subject as a responder, a non-responder, a relapser or a non-relapser, based on a measure of one or more of (i)-(viii).
15. The method of claim 4, wherein the measure of one or more of (i)-(viii) evaluates a profile for one or more of gene expression, flow cytometry or protein expression.
16. The method of claim 4, wherein the level or activity of a CD8+ T cell is evaluated using a profile or signature indicative of the percentage of CD8+ T cell in the sample.
17. The method of claim 4, wherein the level or activity of CD27+CD45RO− immune effector cells is evaluated using a profile or signature indicative of the percentage of CD27+CD45RO− immune effector cells in the sample.
18. The method of claim 4, wherein the level or activity is evaluated using a profile or gene signature according to one, two, three, four, five, ten, twenty, fifty, sixty, seventy, one hundred or more of a biomarker or gene set listed in Tables 1A, 1B, 3, 4, 5, 6, 7A, 7B, or FIG. 2B.
19. The method of claim 4, wherein the biomarker is a secreted or a cell surface biomarker listed in Table 8.
20. The method of claim 4, wherein a responder has, or is identified as having, a greater level or activity of one, two, or more of GZMK, PPF1BP2, or naïve T cells as compared to a non-responder.
21. The method of claim 4, wherein a non-responder has, or is identified as having, a greater level or activity of one, two, three, four, five, six, seven, or more of IL22, IL-2RA, IL-21, IRF8, IL8, CCL17, CCL22, effector T cells, or regulatory T cells, as compared to a responder.
22. The method of claim 4, wherein a complete responder has, or is identified as having:
(a) a greater percentage of CD8+ T cells compared to a reference value;
(b) a greater percentage of CD27+CD45RO− immune effector cells compared to a reference value;
(c) a greater percentage of CD4+ T cells compared to a reference value; or
(d) a greater percentage of one, two, three, or more of resting TEFF cells, resting TREG cells, younger T cells, or early memory T cells, or a combination thereof, compared to a reference value.
23-25. (canceled)
26. The method of claim 4, wherein a non-responder has, or is identified as having:
(a) a greater percentage of one, two, three, or more of activated TEFF cells, activated TREG cells, older T cells, or late memory T cells, or a combination thereof, compared to a reference value, e.g., a responder number of activated TEFF cells, activated TREG cells, older T cells, or late memory T cells; or
(b) a greater percentage of PD-1 or LAG-3 expressing immune effector cells compared to the percentage of PD-1 or LAG-3 expressing immune effector cells from a responder, or a greater percentage of PD-1+/LAG-3+ cells in the CAR-expressing cell population compared to a responder to the CAR-expressing cell therapy,
an exhausted phenotype of PD1+CAR+ and co-expression of LAGS in the CAR-expressing cell population, or
a greater percentage of PD-1+/TIM-3+ cells in the CAR-expressing cell compared to the responder.
27. (canceled)
28. The method of claim 4, wherein a partial responder has, or is identified as having, a higher percentage of PD-1+/LAG-3+ cells than responders in the CAR-expressing cell population, or a higher percentage of PD-1+/TIM-3+ cells than responders in the CAR-expressing cell population.
29. The method of claim 4, wherein the presence of CD8+CD27+CD45RO− T cells in an apheresis sample is a positive predictor of the subject response to a CAR-expressing cell therapy.
30. The of claim 4, wherein a high percentage of PD1+CAR+ and LAG3+ or TIM3+ T cells in an apheresis sample is a poor prognostic predictor of the subject response to a CAR-expressing cell therapy.
31. The method of claim 4, wherein a responder to a CAR19 therapy has, or is identified as having, the biomarker profile of Table 9.
32. The method of claim 4, wherein a non-responder to a CAR19 therapy has, or is identified as having:
(a) the biomarker comprising one or more of PD-1+ immune effector cells, TIM-3+ immune effector cells, LAG-3+ immune effector cells, KLRG1+ immune effector cells, CD27-immune effector cells, activated TEFF cells, activated TREG cells, activated TH1, activated TH2 cells, stimulated memory cells, or late T memory cells, or a combination thereof; or
(b) the biomarker profile of Table 10.
33. (canceled)
34. The method of claim 4, wherein gene expression of one, two, three, four or more of KLRG1, CD57, CD27, CD122, or CD62L is predictive of patient response to CTL019 therapy.
35. The method of claim 4, wherein a relapser has, or is identified as having, elevated levels of one, two, three, four, five, six, seven, eight, nine, ten or more of C5orf32, CCL17, CSF1, CTSL1, EMP1, EPAS1, GCLM, GK, GPR56, HMOX1, IKBIP, IL10, IL13, IL1RN, IL4, IL5, IL9, MIR155, PANX2, PGAM4, PRKAR1B, TNFRSF11A, TNFRSF1B, TNFRSF8, VTRNA1-3, or ZNF282.
36. The method of claim 4, wherein the responder has one, two, three or more of the following profile:
(i) has a greater number of CD27+ immune effector cells compared to a reference value;
(ii) has a greater number of CD8+ T cells compared to a reference value;
(iii) has a lower number of cells expressing one or more checkpoint inhibitors compared to a reference value; or
(iv) has a greater number of one, two, three, or more of resting TEFF cells, resting TREG cells, younger cells, or early memory T cells, or a combination thereof, compared to a reference value.
37. The method of claim 4, wherein the cytokine level or activity of (vi) is chosen from one, two, three, four, five, six, seven, eight, or more of cytokine CCL20/MIP3a, IL17A, IL6, GM-CSF, IFNγ, IL10, IL13, IL2, IL21, IL4, IL5, IL9 or TNFα, or a combination thereof.
38. The method of claim 4, wherein a transduction efficiency of 15% or higher in (vii) is indicative of increased responsiveness or decreased relapse.
39. The method of claim 4, wherein said value of responder or relapser status comprises a measure of the level or activity of one, two, three, four, five, ten, twenty or more of the biomarkers having a FDR p-value of below 0.1 or 0.01 listed in Table 1A, 1B, 17, 18, or 20.
40. A method of evaluating the potency of a CAR-expressing cell product, said method comprising:
acquiring a value for one, two, three, four, five, six, seven, eight, or more of:
(i) the level or activity of CD27 and/or CD45RO− immune effector cells in the CAR-expressing cell product;
(ii) the level or activity of one, two, three, or more of resting TEFF cells, resting TREG cells, younger T cells, or early memory T cells, or a combination thereof, in the CAR-expressing cell product;
(iii) the level or activity of one, two, three, or more of activated TEFF cells, activated TREG cells, older T cells, or late memory T cells, or a combination thereof, in the CAR-expressing cell product;
(iv) the level or activity of an immune cell exhaustion marker;
(v) the level or activity of one, two, three, four, five, ten, twenty or more of the biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 16, Table 17, Table 18, Table 20, FIG. 2B, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature;
(vi) a cytokine level or activity in the CAR-expressing cell product sample wherein the cytokine is chosen from one, two, three, four, five or more of the cytokines listed in Table 16;
(vii) a transduction efficiency of CAR-expressing cells in the product;
(viii) a quantity of CD27+PD-1− cells in a sample; or
(ix) the level or activity of a TREG cell or cell population;
wherein:
(a) an increase in (i), (ii), (vi), (vii), (viii), or any combination thereof, is indicative of increased potency of the CAR-expressing cell product, and
wherein an increase in (iii), (iv), (ix), or any combination thereof, is indicative of decreased potency of the CAR-expressing cell product;
(b) the cytokine is chosen from one, two, three, four, five, six, seven, eight, or more (or all) of CCL20/MIP3a, IL17A, IL6, GM-CSF, IFNγ, IL10, IL13, IL2, IL21, IL4, IL5, IL9 or TNFα, or a combination thereof; or
(c) a transduction efficiency of 15% or higher is indicative of increased potency.
41-42. (canceled)
43. A method for optimizing manufacturing of a CAR-expressing cell product comprising:
(1) acquiring a sample comprising CAR-expressing cell;
(2) activating the CAR-expressing cell in vitro;
(3) evaluating the potency of the potency of the activated CAR-expressing cell by determining one, two, three, four, five, six, seven, eight, or more of:
(i) the level or activity of CD27 and/or CD45RO− immune effector cells in the CAR-expressing cell product;
(ii) the level or activity of one, two, three, or more of resting TEFF cells, resting TREG cells, younger T cells, or early memory T cells, or a combination thereof, in the CAR-expressing cell product;
(iii) the level or activity of one, two, three, or more of activated TEFF cells, activated TREG cells, older T cells, or late memory T cells, or a combination thereof, in the CAR-expressing cell product;
(iv) the level or activity of an immune cell exhaustion marker in the CAR-expressing cell product;
(v) the level or activity of one, two, three, four, five, ten, twenty or more of the biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 16, Table 17, Table 18, Table 20, FIG. 2B, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature;
(vi) a cytokine level or activity in a CAR-expressing cell product sample, wherein the cytokine is chosen from one, two, three, four, five or more of the cytokines listed in Table 16;
(vii) a transduction efficiency of CAR-expressing cells in the product;
(viii) a quantity of CD27+PD-1− cells in a sample; or
(ix) the level or activity of a TREG cell or cell population,
wherein an increase in (i), (ii), (vi), (vii), (viii), or any combination thereof, is indicative of increased potency of the CAR-expressing cell product, and
wherein an increase in (iii), (iv), (ix), or any combination thereof, is indicative of decreased potency of the CAR-expressing cell product.
thereby optimizing manufacturing of the product.
44. The method of claim 43, further comprising a step of enriching for cells having an increase in any of (i), (ii), (vi), (vii), (viii), or any combination thereof, or a decrease in any of (iii), (iv), (ix), or any combination thereof; or
wherein the cytokine level or activity is chosen from one or more of cytokine CCL20/MIP3a, IL17A, IL6, GM-CSF, IFNγ, IL10, IL13, IL2, IL21, IL4, IL5, IL9 or TNFα, or a combination thereof.
45. (canceled)
46. The method of claim 43, which comprises a further step that depletes TREG cells.
47. The method of claim 46, wherein TREG cells are depleted via CD25-depletion, GITR depletion, mTOR inhibition, or a combination thereof.
48. The method of claim 43, wherein (i), (ii), (iii), (iv), (v), (vi), (vii), (viii), (ix), or any combination thereof (e.g., all) are evaluated following activation in vitro, and/or wherein the CAR-expressing cell therapy comprises CTL019.
49. (canceled)
50. The method of claim 4, wherein the subject receives a treatment prior to the initiation of a CAR-expressing cell therapy or a treatment post-CAR-expressing cell therapy.
51. The method of claim 4, wherein the cancer is associated with CD19 expression.
52. The method of claim 4, wherein the cancer is a hematological cancer.
53. The method of claim 4, wherein the cancer or hematological cancer is selected from the group consisting of B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), B cell promyelocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma, marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, and Waldenstrom macroglobulinemia.
54. The method of claim 4, where the CD19 CAR-expressing cell gene signature comprises a value for expression of at least 5, 6, 7, 8, 9 or 10 genes comprising a CD19 CAR-expressing cell gene signature.
55. A kit for providing a prognosis for success rate of a CAR-expressing cell therapy in a subject having cancer, said kit comprising:
a set of reagents that specifically detects the level or activity of one or more genes listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 17, Table 18, Table 20, FIG. 2B, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, and/or a CD19 CAR-expressing cell gene set signature; and
instructions for using said kit;
wherein said instructions for use provide that if one or more of the detected expression levels is different from the subject is more likely to respond positively to a CAR-expressing cell therapy; and
wherein the set of reagent detects the expression of mRNA expressed from said set of genes; and/or
wherein the set of reagents detects the expression of polypeptides encoded by said set of genes.
56-57. (canceled)
58. A system for evaluating cancer in a subject, comprising:
at least one processor operatively connected to a memory, the at least one processor when executing is configured to:
acquire a value of responder or relapser status that comprises a measure of one, two, three, four, five, six, seven, or more of the following:
(i) the level or activity of CD27 and/or CD45RO− immune effector cells;
(ii) the level or activity of one, two, three, or more of resting TEFF cells, resting TREG cells, younger T cells, or early memory T cells, or a combination thereof, in a sample;
(iii) the level or activity of one, two, three, or more of activated TEFF cells, activated TREG cells, older T cells, or late memory T cells, or a combination thereof, in a sample;
(iv) the level or activity of an immune cell exhaustion marker;
(v) the level or activity of one, two, three, four, five, ten, twenty or more of the biomarkers listed in Table 1A, Table 1B, Table 7A, Table 7B, Table 8, Table 9, Table 10, Table 14, Table 16, Table 17, Table 18, Table 20, FIG. 2B, PD-1, LAG-3, TIM-3, CD57, CD27, CD122, CD62L, KLRG1, or a CD19 CAR-expressing cell gene set signature;
(vi) a cytokine level or activity in a CAR-expressing cell product sample, wherein the cytokine is chosen from one, two, three, four, five or more of the cytokines listed in Table 16;
(vii) a transduction efficiency of a CAR-expressing cell in a manufactured CAR-expressing cell product sample; or
(viii) a quantity of CD27+PD-1− cells in a sample,
responsive to a determination of the value of responder status, perform one, two, three, four, five, six, seven, or more of:
identify the subject as a complete responder, partial responder, non-responder, relapser or non-relapser;
recommend administering a CAR-expressing cell therapy;
recommend a selection or alteration of a dosing of a CAR-expressing cell therapy;
recommend a selection or alteration of a schedule or time course of a CAR-expressing cell therapy;
recommend administering an additional agent in combination with a CAR-expressing cell therapy;
recommend administering to a non-responder or partial responder a therapy that increases the number of naïve T cells in the subject prior to treatment with a CAR-expressing cell therapy;
recommend modifying a manufacturing process of a CAR-expressing cell therapy;
recommend modifying the CAR-expressing cell product prior to infusion into the patient;
recommend adjusting the CAR-expressing cell infusion dose to achieve clinical efficacy;
recommend administering an alternative therapy;
recommend a selection of an alternative therapy; or
if the subject is, or is identified as, a non-responder or a relapser, recommend decreasing the TREG cell population and/or TREG gene signature administration of cyclophosphamide, an anti-GITR antibody, an mTOR inhibitor, or a combination thereof.
59-64. (canceled)
65. The method of claim 4, wherein the CAR19 therapy is CTL019.
66. The method of claim 4, wherein the immune effector cells are in a CD4+ or a CD8+ T cell population.
67. The method of claim 4, wherein the younger T cells are younger CD4 or CD8 cells, or gamma/delta cells.
68. The method of claim 4, wherein the immune cell exhaustion marker is one, two or more immune checkpoint inhibitors.
69. The method of claim 68, wherein the immune checkpoint inhibitor is PD-1, TIM-3 and/or LAG-3.
70. The method of claim 4, wherein the biomarker is CCL20, IL-17a and/or IL-6.
71. The method of claim 4, wherein the cytokine level or activity is a quality of cytokine repertoire.
72. The method of claim 4, wherein the quantity of CD27+PD-1− cells in a sample is a quantity greater than or equal to 1×107 cells.
73. The method of claim 8, wherein the apheresis sample or the manufactures CAR-expressing cell product is evaluated prior to infusion or re-infusion.
74. The method of claim 22, wherein the reference value is a value of a non-responder percentage of CD8+ T cells.
US16/986,943 2014-10-08 2020-08-06 Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof Pending US20210172020A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/986,943 US20210172020A1 (en) 2014-10-08 2020-08-06 Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201462061553P 2014-10-08 2014-10-08
US201562144682P 2015-04-08 2015-04-08
PCT/US2015/054542 WO2016057705A1 (en) 2014-10-08 2015-10-07 Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
US201715517597A 2017-04-07 2017-04-07
US16/986,943 US20210172020A1 (en) 2014-10-08 2020-08-06 Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2015/054542 Continuation WO2016057705A1 (en) 2014-10-08 2015-10-07 Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
US15/517,597 Continuation US10774388B2 (en) 2014-10-08 2015-10-07 Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof

Publications (1)

Publication Number Publication Date
US20210172020A1 true US20210172020A1 (en) 2021-06-10

Family

ID=54478204

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/517,597 Active 2036-02-25 US10774388B2 (en) 2014-10-08 2015-10-07 Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
US16/986,943 Pending US20210172020A1 (en) 2014-10-08 2020-08-06 Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/517,597 Active 2036-02-25 US10774388B2 (en) 2014-10-08 2015-10-07 Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof

Country Status (11)

Country Link
US (2) US10774388B2 (en)
EP (1) EP3204777A1 (en)
JP (2) JP6815992B2 (en)
KR (1) KR20170068504A (en)
CN (2) CN114107424A (en)
AU (2) AU2015330898B2 (en)
CA (1) CA2963935A1 (en)
IL (2) IL251525A0 (en)
RU (1) RU2743657C2 (en)
SG (1) SG11201702895SA (en)
WO (1) WO2016057705A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11535662B2 (en) 2017-01-26 2022-12-27 Novartis Ag CD28 compositions and methods for chimeric antigen receptor therapy
US11549099B2 (en) 2016-03-23 2023-01-10 Novartis Ag Cell secreted minibodies and uses thereof
US11578130B2 (en) 2013-12-20 2023-02-14 Novartis Ag Regulatable chimeric antigen receptor
US11591404B2 (en) 2014-08-19 2023-02-28 Novartis Ag Treatment of cancer using a CD123 chimeric antigen receptor
US11608382B2 (en) 2018-06-13 2023-03-21 Novartis Ag BCMA chimeric antigen receptors and uses thereof
WO2023019204A3 (en) * 2021-08-11 2023-03-30 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating and/or characterizing hematological malignancies and precursor conditions
WO2023081386A1 (en) * 2021-11-04 2023-05-11 The Regents Of The University Of Michigan Prevention and treatment of cytokine release syndrome and neurotoxicity associated with car-t cell therapy
US11667691B2 (en) 2015-08-07 2023-06-06 Novartis Ag Treatment of cancer using chimeric CD3 receptor proteins
US11747346B2 (en) 2015-09-03 2023-09-05 Novartis Ag Biomarkers predictive of cytokine release syndrome
US11865167B2 (en) 2013-02-20 2024-01-09 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
US11872249B2 (en) 2016-10-07 2024-01-16 Novartis Ag Method of treating cancer by administering immune effector cells expressing a chimeric antigen receptor comprising a CD20 binding domain
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
US11919946B2 (en) 2013-03-15 2024-03-05 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy

Families Citing this family (76)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108267581A (en) 2012-08-24 2018-07-10 耶鲁大学 For the systems, devices and methods of high-throughput multi detection
WO2014130635A1 (en) 2013-02-20 2014-08-28 Novartis Ag Effective targeting of primary human leukemia using anti-cd123 chimeric antigen receptor engineered t cells
UY35468A (en) 2013-03-16 2014-10-31 Novartis Ag CANCER TREATMENT USING AN ANTI-CD19 CHEMERIC ANTIGEN RECEIVER
ES2923942T3 (en) 2013-11-06 2022-10-03 Us Health Procedure for the subtyping of lymphoma types by means of expression profiles
CA2931684C (en) 2013-12-19 2024-02-20 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
WO2015112626A1 (en) 2014-01-21 2015-07-30 June Carl H Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
PL3129470T3 (en) 2014-04-07 2021-11-29 Novartis Ag Treatment of cancer using anti-cd19 chimeric antigen receptor
MX2017001013A (en) 2014-07-21 2018-02-21 Novartis Ag Treatment of cancer using a cll-1 chimeric antigen receptor.
KR102594343B1 (en) 2014-07-21 2023-10-26 노파르티스 아게 Treatment of cancer using a cd33 chimeric antigen receptor
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
TWI750110B (en) 2014-07-21 2021-12-21 瑞士商諾華公司 Treatment of cancer using humanized anti- bcma chimeric antigen receptor
AU2015317608B2 (en) 2014-09-17 2021-03-11 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US10774388B2 (en) 2014-10-08 2020-09-15 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
EP3227684B1 (en) 2014-12-03 2019-10-02 Isoplexis Corporation Analysis and screening of cell secretion profiles
KR20170093254A (en) 2014-12-29 2017-08-14 노파르티스 아게 Methods for producing chimeric antigen receptor-expressing cells
US11459390B2 (en) 2015-01-16 2022-10-04 Novartis Ag Phosphoglycerate kinase 1 (PGK) promoters and methods of use for expressing chimeric antigen receptor
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
US11353448B2 (en) 2015-02-13 2022-06-07 California Institute Of Technology Methods and compositions for quantifying metabolites and proteins from single cells
IL303972A (en) 2015-04-08 2023-08-01 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
AU2016297014B2 (en) 2015-07-21 2021-06-17 Novartis Ag Methods for improving the efficacy and expansion of immune cells
EP3699294A1 (en) * 2015-12-04 2020-08-26 Agency for Science, Technology and Research Clinical correlates of immunotherapy efficacy
JP7082055B2 (en) 2015-12-22 2022-06-07 ノバルティス アーゲー Antibodies to Mesothelin Chimeric Antigen Receptor (CAR) and PD-L1 Inhibitors for Combined Use in Anticancer Treatment
US11446398B2 (en) 2016-04-11 2022-09-20 Obsidian Therapeutics, Inc. Regulated biocircuit systems
ES2859423T3 (en) 2016-04-20 2021-10-04 Us Health Mantle cell lymphoma evaluation and related procedures
WO2017210617A2 (en) * 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
EP3491016A1 (en) * 2016-07-28 2019-06-05 Novartis AG Combination therapies of chimeric antigen receptors and pd-1 inhibitors
JP7160482B2 (en) 2016-09-02 2022-10-25 レンティジェン・テクノロジー・インコーポレイテッド Compositions and methods for treating cancer with DUOCAR
EP3510398A1 (en) * 2016-09-12 2019-07-17 Isoplexis Corporation System and methods for multiplexed analysis of cellular and other immunotherapeutics
DK3538891T3 (en) 2016-11-11 2022-03-28 Isoplexis Corp COMPOSITIONS AND PROCEDURES FOR CONTEMPORARY GENOMIC, TRANSCRIPTOMIC AND PROTEOMIC ANALYSIS OF SINGLE CELLS
US10918398B2 (en) 2016-11-18 2021-02-16 Stryker Corporation Method and apparatus for treating a joint, including the treatment of cam-type femoroacetabular impingement in a hip joint and pincer-type femoroacetabular impingement in a hip joint
EP3545284A4 (en) 2016-11-22 2020-07-01 Isoplexis Corporation Systems, devices and methods for cell capture and methods of manufacture thereof
CN110248678A (en) * 2016-12-03 2019-09-17 朱诺治疗学股份有限公司 The method for adjusting CAR-T cell
EP3568414A1 (en) 2017-01-10 2019-11-20 Juno Therapeutics, Inc. Epigenetic analysis of cell therapy and related methods
WO2018132571A1 (en) * 2017-01-13 2018-07-19 Mayo Foundation For Medical Education And Research Materials and methods for treating cancer
CA3049445A1 (en) * 2017-01-17 2018-10-11 The University Of Chicago Dysfunctional antigen-specific cd8+ t cells in the tumor microenvironment
JP2018130113A (en) * 2017-02-15 2018-08-23 国立大学法人 東京大学 Detection method and detection kit for chronic myelogenous leukemia (cml), method for isolating tyrosine kinase inhibitor (tki) resistant cml, reducing agent for tki resistance in cml and screening method therefor
KR20190126812A (en) * 2017-02-23 2019-11-12 더 카운실 오브 더 퀸즐랜드 인스티튜트 오브 메디컬 리서치 Biomarkers for Disease Diagnosis
EA201992232A1 (en) 2017-03-22 2020-05-14 Новартис Аг COMPOSITIONS AND METHODS FOR IMMUNOUNCOLOGY
US11913075B2 (en) 2017-04-01 2024-02-27 The Broad Institute, Inc. Methods and compositions for detecting and modulating an immunotherapy resistance gene signature in cancer
US11851649B2 (en) * 2017-05-17 2023-12-26 Seattle Children's Hospital Generating mammalian T cell activation inducible synthetic promoters (SYN+PRO) to improve T cell therapy
CA3064597A1 (en) 2017-06-02 2018-12-06 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
KR20200044899A (en) 2017-08-28 2020-04-29 브리스톨-마이어스 스큅 컴퍼니 TIM-3 antagonist for the treatment and diagnosis of cancer
MA50057A (en) * 2017-09-01 2020-07-08 Juno Therapeutics Inc GENE EXPRESSION AND ASSESSMENT OF A RISK OF DEVELOPING TOXICITY FOLLOWING CELL THERAPY
WO2019068087A1 (en) * 2017-09-29 2019-04-04 University Of Maryland, College Park System of prediction of response to cancer therapy and methods of using the same
EP3695408A4 (en) * 2017-10-02 2021-12-15 The Broad Institute, Inc. Methods and compositions for detecting and modulating an immunotherapy resistance gene signature in cancer
WO2019089858A2 (en) * 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
TW201925782A (en) * 2017-11-30 2019-07-01 瑞士商諾華公司 BCMA-targeting chimeric antigen receptor, and uses thereof
US11464569B2 (en) 2018-01-29 2022-10-11 Stryker Corporation Systems and methods for pre-operative visualization of a joint
KR102080384B1 (en) * 2018-02-28 2020-02-21 주식회사 마크로젠 Cancer stem cell-specific biomarkers
WO2019183610A1 (en) * 2018-03-23 2019-09-26 La Jolla Institute For Allergy And Immunology Tissue resident memory cell profiles, and uses thereof
IL277977B1 (en) * 2018-04-12 2024-03-01 Kite Pharma Inc Chimeric receptor t cell treatment using characteristics of the tumor microenvironment
WO2019213282A1 (en) 2018-05-01 2019-11-07 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
CN113227792A (en) 2018-10-13 2021-08-06 纪念斯隆-凯特琳癌症中心 Methods and compositions for identifying and treating subjects at risk of poor response to CAR T cell therapy
SG11202104882WA (en) * 2018-11-15 2021-06-29 Ampel Biosolutions Llc Machine learning disease prediction and treatment prioritization
WO2020102726A1 (en) * 2018-11-16 2020-05-22 Rapa Therapeutics, Llc Method for manufacturing of human hybrid regulatory t/th2 cells (hybrid treg/th2 cells) from de-differentiated t cells
WO2020124083A1 (en) * 2018-12-14 2020-06-18 The Brigham And Women's Hospital, Inc. Biomarker panels for on-treatment prediction of response to immuno-oncology drugs
KR102179381B1 (en) * 2018-12-28 2020-11-16 주식회사 네오젠티씨 Composition for improving immunity reinforcement or anti-cancer activity comprising MAL expressed stem cell like memory T cell
WO2020172591A1 (en) * 2019-02-22 2020-08-27 Mitra Rxdx, Inc. A method to predict a patient's response to an anti-cancer drug from an expression level of a set of genes
CN113853443A (en) * 2019-03-08 2021-12-28 株式会社Neogentc Marker for predicting tumor reactivity of lymphocytes and use thereof
CN111850114A (en) * 2019-04-25 2020-10-30 上海交通大学医学院附属瑞金医院 Gene group for evaluating molecular typing of diffuse large B cell lymphoma, kit and analysis method thereof
CN110412281B (en) * 2019-06-26 2022-09-09 四川大学华西医院 Application of BEGAIN autoantibody detection reagent in preparation of lung cancer screening kit
US20230002833A1 (en) * 2019-11-29 2023-01-05 Sungkwang Medical Foundation Biomarkers for predicting therapeutic responsiveness to immune therapeutic agent
WO2021150925A1 (en) * 2020-01-24 2021-07-29 Dana-Farber Cancer Institute, Inc. Uses of biomarkers for improving immunotherapy
US20230107770A1 (en) * 2020-02-20 2023-04-06 H. Lee Moffitt Cancer Center And Research Institute, Inc. Method of enhancing immunotherapy using er stress pathway inhibitors
US20230119171A1 (en) * 2020-03-31 2023-04-20 Cedars-Sinai Medical Center Biomarker panels for stratification of response to immune checkpoint blockade in cancer
GB202005599D0 (en) * 2020-04-17 2020-06-03 Univ London Modulation of t cell cytotoxicity and related therapy
WO2022011129A1 (en) * 2020-07-09 2022-01-13 The Board Of Trustees Of The Leland Stanford University Multi-parallel analysis of t-cell therapies
WO2022054796A1 (en) * 2020-09-08 2022-03-17 学校法人 埼玉医科大学 Biomarker for predicting response to cancer treatment
CN113416253B (en) * 2021-05-24 2022-12-13 复旦大学 Isolated antigen ITPRIPL1 binding proteins and uses thereof
CN112321721B (en) * 2020-11-05 2021-07-06 山东仁济生物科技有限公司 Chimeric antigen receptor, immune cell modified by chimeric antigen receptor and application of immune cell in treatment of advanced pancreatic cancer
WO2022109607A2 (en) * 2020-11-19 2022-05-27 Tempus Labs, Inc. Determination of cytotoxic gene signature and associated systems and methods for response prediction and treatment
JP2024502094A (en) 2020-12-30 2024-01-17 アラウノス セラピューティクス インコーポレイテッド Recombinant vectors containing polycistronic expression cassettes and methods for their use
CN112562867A (en) * 2021-02-22 2021-03-26 天津迈德新医药科技有限公司 Device, storage medium and electronic device for predicting very early HIV infection risk
WO2022215843A1 (en) * 2021-04-06 2022-10-13 (주)에스엠티바이오 Method for predicting therapeutic response of cancer patient to anticancer immunotherapy using natural killer cells
GB202107091D0 (en) 2021-05-18 2021-06-30 Kinomica Ltd Acute myeloid leukaemia midostaurin biomarkers
WO2023141551A1 (en) * 2022-01-21 2023-07-27 Partner Therapeutics, Inc. Granulocyte-macrophage colony-stimulating factor-based dermatological pathology treatments

Family Cites Families (238)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ZA737247B (en) 1972-09-29 1975-04-30 Ayerst Mckenna & Harrison Rapamycin and process of preparation
US4376110A (en) 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US4517288A (en) 1981-01-23 1985-05-14 American Hospital Supply Corp. Solid phase system for ligand assay
GB2116183B (en) 1982-03-03 1985-06-05 Genentech Inc Human antithrombin iii dna sequences therefore expression vehicles and cloning vectors containing such sequences and cell cultures transformed thereby a process for expressing human antithrombin iii and pharmaceutical compositions comprising it
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
CA1291031C (en) 1985-12-23 1991-10-22 Nikolaas C.J. De Jaeger Method for the detection of specific binding agents and their correspondingbindable substances
US4868103A (en) 1986-02-19 1989-09-19 Enzo Biochem, Inc. Analyte detection by means of energy transfer
US4843155A (en) 1987-11-19 1989-06-27 Piotr Chomczynski Product and process for isolating RNA
US5906936A (en) 1988-05-04 1999-05-25 Yeda Research And Development Co. Ltd. Endowing lymphocytes with antibody specificity
US6303121B1 (en) 1992-07-30 2001-10-16 Advanced Research And Technology Method of using human receptor protein 4-1BB
US6352694B1 (en) 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US5858358A (en) 1992-04-07 1999-01-12 The United States Of America As Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of T cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
WO1992010591A1 (en) 1990-12-14 1992-06-25 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
US6319494B1 (en) 1990-12-14 2001-11-20 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
US6004811A (en) 1991-03-07 1999-12-21 The Massachussetts General Hospital Redirection of cellular immunity by protein tyrosine kinase chimeras
US7049136B2 (en) 1991-03-07 2006-05-23 The General Hospital Corporation Redirection of cellular immunity by receptor chimeras
NZ241855A (en) 1991-03-07 1994-04-27 Gen Hospital Corp Use of therapeutic cells to obtain cellular response to infection, tumours or autoimmune-generated cells, cells with chimaeric receptors (with binding component and destruction signal), dna encoding the receptor, vectors and antibodies to receptor
GB9125768D0 (en) 1991-12-04 1992-02-05 Hale Geoffrey Therapeutic method
DE69326967T2 (en) 1992-01-17 2000-06-15 Lakowicz Joseph R Phase modulation energy transfer fluoroimmunoassay
IL104570A0 (en) 1992-03-18 1993-05-13 Yeda Res & Dev Chimeric genes and cells transformed therewith
US8211422B2 (en) 1992-03-18 2012-07-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric receptor genes and cells transformed therewith
GB9221220D0 (en) 1992-10-09 1992-11-25 Sandoz Ag Organic componds
US7211259B1 (en) 1993-05-07 2007-05-01 Immunex Corporation 4-1BB polypeptides and DNA encoding 4-1BB polypeptides
JP4105761B2 (en) 1993-11-19 2008-06-25 アボット・ラボラトリーズ Semi-synthetic analog immunomodulator of rapamycin (macrolide)
NZ277498A (en) 1993-12-17 1998-03-25 Novartis Ag Rapamycin derivatives
US5362718A (en) 1994-04-18 1994-11-08 American Home Products Corporation Rapamycin hydroxyesters
AU694222B2 (en) 1994-05-02 1998-07-16 Bernd Groner Bifunctional protein, preparation and use
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
US5712149A (en) 1995-02-03 1998-01-27 Cell Genesys, Inc. Chimeric receptor molecules for delivery of co-stimulatory signals
US6103521A (en) 1995-02-06 2000-08-15 Cell Genesys, Inc. Multispecific chimeric receptors
CZ295428B6 (en) 1995-02-24 2005-08-17 The General Hospital Corporation Therapeutic cell for redirecting of cellular immunity by receptor chimeras
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
US6692964B1 (en) 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
WO1996041807A1 (en) 1995-06-09 1996-12-27 Novartis Ag Rapamycin derivatives
BE1009856A5 (en) 1995-07-14 1997-10-07 Sandoz Sa Pharmaceutical composition in the form of a solid release including macrolide and a vehicle.
US5854033A (en) 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
GB9526131D0 (en) 1995-12-21 1996-02-21 Celltech Therapeutics Ltd Recombinant chimeric receptors
AU731826B2 (en) 1996-02-28 2001-04-05 Ariad Pharmaceuticals, Inc. Synthetic Multimerizing Agents
US5874240A (en) 1996-03-15 1999-02-23 Human Genome Sciences, Inc. Human 4-1BB receptor splicing variant
EP0937082A2 (en) 1996-07-12 1999-08-25 Ariad Pharmaceuticals, Inc. Materials and method for treating or preventing pathogenic fungal infection
AU4055697A (en) 1996-08-16 1998-03-06 Schering Corporation Mammalian cell surface antigens; related reagents
US6111090A (en) 1996-08-16 2000-08-29 Schering Corporation Mammalian cell surface antigens; related reagents
JP2002512502A (en) 1996-10-25 2002-04-23 セル・ジェネシス・インコーポレイテッド Target cell lysis of cancer cells
US6673901B2 (en) 1997-06-12 2004-01-06 Research Corporation Technologies, Inc. Artificial antibody polypeptides
GB9713473D0 (en) 1997-06-25 1997-09-03 Celltech Therapeutics Ltd Biological products
US20030060444A1 (en) 1997-06-25 2003-03-27 Celltech Therapeutics, Ltd. Cell activation process and reagents therefor
US6015815A (en) 1997-09-26 2000-01-18 Abbott Laboratories Tetrazole-containing rapamycin analogs with shortened half-lives
TW557297B (en) 1997-09-26 2003-10-11 Abbott Lab Rapamycin analogs having immunomodulatory activity, and pharmaceutical compositions containing same
JP2001520039A (en) 1997-10-21 2001-10-30 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド Human tumor necrosis factor receptor-like proteins, TR11, TR11SV1 and TR11SV2
AU2591599A (en) 1998-02-09 1999-08-23 Genentech Inc. Novel tumor necrosis factor receptor homolog and nucleic acids encoding the same
US20040040047A1 (en) 1998-03-30 2004-02-26 Spencer David M. Regulated apoptosis using chemically induced dimerization of apoptosis factors
ATE298248T1 (en) 1998-04-15 2005-07-15 Brigham & Womens Hospital T-CELL INHIBITING RECEPTOR COMPOSITIONS AND USE THEREOF
GB9809658D0 (en) 1998-05-06 1998-07-01 Celltech Therapeutics Ltd Biological products
JP2002524081A (en) 1998-09-04 2002-08-06 スローン − ケッタリング インスティチュート フォー キャンサー リサーチ Fusion receptor specific for prostate-specific membrane antigen and uses thereof
WO2000023573A2 (en) 1998-10-20 2000-04-27 City Of Hope Cd20-specific redirected t cells and their use in cellular immunotherapy of cd20+ malignancies
AU4418900A (en) 1999-04-16 2000-11-02 Celltech Therapeutics Limited Synthetic transmembrane components
PT1196186E (en) 1999-07-12 2008-02-14 Genentech Inc Promotion or inhibition of angiogenesis and cardiovascularization by tumor necrosis factor ligand/receptor homologs
ATE264863T1 (en) 1999-08-24 2004-05-15 Ariad Gene Therapeutics Inc 28-EPIRAPALOGUE
JP2003516124A (en) 1999-10-15 2003-05-13 ユニバーシティー オブ マサチューセッツ RNA interference pathway genes as a means of targeted genetic interference
GB9925848D0 (en) 1999-11-01 1999-12-29 Celltech Therapeutics Ltd Biological products
US6326193B1 (en) 1999-11-05 2001-12-04 Cambria Biosciences, Llc Insect control agent
KR20030032922A (en) 2000-02-24 2003-04-26 싸이트 테라피스 인코포레이티드 Simultaneous stimulation and concentration of cells
US7572631B2 (en) 2000-02-24 2009-08-11 Invitrogen Corporation Activation and expansion of T cells
US6867041B2 (en) 2000-02-24 2005-03-15 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US6797514B2 (en) 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
IL136511A0 (en) 2000-06-01 2001-06-14 Gavish Galilee Bio Appl Ltd Genetically engineered mhc molecules
AU2001275474A1 (en) 2000-06-12 2001-12-24 Akkadix Corporation Materials and methods for the control of nematodes
GB0025307D0 (en) 2000-10-16 2000-11-29 Celltech Chiroscience Ltd Biological products
ATE338124T1 (en) 2000-11-07 2006-09-15 Hope City CD19-SPECIFIC TARGETED IMMUNE CELLS
US7070995B2 (en) 2001-04-11 2006-07-04 City Of Hope CE7-specific redirected immune cells
US20090257994A1 (en) 2001-04-30 2009-10-15 City Of Hope Chimeric immunoreceptor useful in treating human cancers
AU2002256390B2 (en) 2001-04-30 2007-08-30 City Of Hope Chimeric immunoreceptor useful in treating human cancers
US7514537B2 (en) 2001-04-30 2009-04-07 City Of Hope Chimeric immunoreceptor useful in treating human gliomas
US20030148982A1 (en) 2001-11-13 2003-08-07 Brenner Malcolm K. Bi-spcific chimeric T cells
US7745140B2 (en) 2002-01-03 2010-06-29 The Trustees Of The University Of Pennsylvania Activation and expansion of T-cells using an engineered multivalent signaling platform as a research tool
WO2003057171A2 (en) 2002-01-03 2003-07-17 The Trustees Of The University Of Pennsylvania Activation and expansion of t-cells using an engineered multivalent signaling platform
US7638326B2 (en) 2002-01-03 2009-12-29 The Trustees Of The University Of Pennsylvania Activation and expansion of T-cells using an engineered multivalent signaling platform
EP1478648B1 (en) 2002-02-01 2014-04-30 ARIAD Pharmaceuticals, Inc. Phosphorus-containing compounds and uses thereof
GB0208104D0 (en) 2002-04-09 2002-05-22 Univ Dundee Method
TWI275390B (en) 2002-04-30 2007-03-11 Wyeth Corp Process for the preparation of 7-substituted-3- quinolinecarbonitriles
US7446190B2 (en) 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
US20040047858A1 (en) 2002-09-11 2004-03-11 Blumberg Richard S. Therapeutic anti-BGP(C-CAM1) antibodies and uses thereof
CN1753912B (en) 2002-12-23 2011-11-02 惠氏公司 Antibodies against PD-1 and uses therefor
US20050129671A1 (en) 2003-03-11 2005-06-16 City Of Hope Mammalian antigen-presenting T cells and bi-specific T cells
KR20060052681A (en) 2003-05-23 2006-05-19 와이어쓰 Gitr ligand and gitr ligand-related molecules and antibody and uses thereof
EP1660126A1 (en) 2003-07-11 2006-05-31 Schering Corporation Agonists or antagonists of the clucocorticoid-induced tumour necrosis factor receptor (gitr) or its ligand for the treatment of immune disorders, infections and cancer
US8198020B2 (en) 2003-08-22 2012-06-12 Potentia Pharmaceuticals, Inc. Compositions and methods for enhancing phagocytosis or phagocyte activity
US7435596B2 (en) 2004-11-04 2008-10-14 St. Jude Children's Research Hospital, Inc. Modified cell line and method for expansion of NK cell
US20050113564A1 (en) 2003-11-05 2005-05-26 St. Jude Children's Research Hospital Chimeric receptors with 4-1BB stimulatory signaling domain
US7220755B2 (en) 2003-11-12 2007-05-22 Biosensors International Group, Ltd. 42-O-alkoxyalkyl rapamycin derivatives and compositions comprising same
WO2005055808A2 (en) 2003-12-02 2005-06-23 Genzyme Corporation Compositions and methods to diagnose and treat lung cancer
US20050216685A1 (en) 2004-02-03 2005-09-29 Heden Donald G Intelligent media storage system
GB0409799D0 (en) 2004-04-30 2004-06-09 Isis Innovation Method of generating improved immune response
US7754482B2 (en) 2004-05-27 2010-07-13 The Trustees Of The University Of Pennsylvania Artificial antigen presenting cells and uses therefor
WO2006083289A2 (en) 2004-06-04 2006-08-10 Duke University Methods and compositions for enhancement of immunity by in vivo depletion of immunosuppressive cell activity
WO2006036445A2 (en) 2004-09-24 2006-04-06 Trustees Of Dartmouth College Chimeric nk receptor and methods for treating cancer
EP3530321A1 (en) 2004-12-10 2019-08-28 Peter MacCallum Cancer Institute Methods and compositions for adoptive immunotherapy
DK1866339T3 (en) 2005-03-25 2013-09-02 Gitr Inc GTR-binding molecules and their applications
CN109485727A (en) 2005-05-09 2019-03-19 小野药品工业株式会社 The human monoclonal antibodies of programmed death-1 (PD-1) and the method for carrying out treating cancer using anti-PD-1 antibody
GB0510390D0 (en) 2005-05-20 2005-06-29 Novartis Ag Organic compounds
DK1907424T3 (en) 2005-07-01 2015-11-09 Squibb & Sons Llc HUMAN MONOCLONAL ANTIBODIES TO PROGRAMMED death ligand 1 (PD-L1)
US20070036773A1 (en) 2005-08-09 2007-02-15 City Of Hope Generation and application of universal T cells for B-ALL
ES2513165T3 (en) 2005-10-07 2014-10-24 Exelixis, Inc. N- (3-amino-quinoxalin-2-yl) -sulfonamide derivatives and their use as phosphatidylinositol-3-kinase inhibitors
US20110212086A1 (en) 2006-01-19 2011-09-01 Genzyme Corporation GITR Antibodies For The Treatment of Cancer
US20100105136A1 (en) 2006-10-09 2010-04-29 The General Hospital Corporation Chimeric t-cell receptors and t-cells targeting egfrviii on tumors
US20080131415A1 (en) 2006-11-30 2008-06-05 Riddell Stanley R Adoptive transfer of cd8 + t cell clones derived from central memory cells
HUE038506T2 (en) 2007-03-30 2018-10-29 Memorial Sloan Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred T lymphocytes
ES2437327T3 (en) 2007-06-18 2014-01-10 Merck Sharp & Dohme B.V. Antibodies for the human programmed PD-1 receptor of programmed death
ES2591281T3 (en) 2007-07-12 2016-11-25 Gitr, Inc. Combination therapies that employ GITR binding molecules
JP2011500730A (en) 2007-10-26 2011-01-06 ガバニング カウンセル オブ ザ ユニバーシティ オブ トロント Treatment and diagnostic method using Tim-3
WO2009091826A2 (en) 2008-01-14 2009-07-23 The Board Of Regents Of The University Of Texas System Compositions and methods related to a human cd19-specific chimeric antigen receptor (h-car)
WO2009097140A1 (en) 2008-01-30 2009-08-06 Memorial Sloan-Kettering Cancer Center Methods for off -the -shelf tumor immunotherapy using allogeneic t-cell precursors
US8747847B2 (en) 2008-02-11 2014-06-10 Curetech Ltd. Monoclonal antibodies for tumor treatment
CN102014912A (en) 2008-02-21 2011-04-13 阿斯利康(瑞典)有限公司 Combination therapy 238
US8379824B2 (en) 2008-03-06 2013-02-19 At&T Intellectual Property I, Lp Methods and apparatus to provide a network-based caller identification service in a voice over internet protocol network
EP2262837A4 (en) 2008-03-12 2011-04-06 Merck Sharp & Dohme Pd-1 binding proteins
US20110177070A1 (en) 2008-07-02 2011-07-21 Emergent Product Development Seatlle, LLC TGF-Beta Antagonist Multi-Target Binding Proteins
AR072999A1 (en) 2008-08-11 2010-10-06 Medarex Inc HUMAN ANTIBODIES THAT JOIN GEN 3 OF LYMPHOCYTARY ACTIVATION (LAG-3) AND THE USES OF THESE
US20110223188A1 (en) 2008-08-25 2011-09-15 Solomon Langermann Targeted costimulatory polypeptides and methods of use to treat cancer
PE20110435A1 (en) 2008-08-25 2011-07-20 Amplimmune Inc ANTAGONIST COMPOSITIONS OF PD-1
PL3006459T3 (en) 2008-08-26 2022-01-17 City Of Hope Method and compositions for enhanced anti-tumor effector functioning of t cells
WO2010030002A1 (en) 2008-09-12 2010-03-18 国立大学法人三重大学 Cell capable of expressing exogenous gitr ligand
US8476431B2 (en) 2008-11-03 2013-07-02 Itellikine LLC Benzoxazole kinase inhibitors and methods of use
BRPI0917592B1 (en) 2008-12-09 2021-08-17 Genentech, Inc ANTI-PD-L1 ANTIBODY, COMPOSITION, MANUFACTURED ARTICLES AND USES OF A COMPOSITION
US9206440B2 (en) 2009-01-23 2015-12-08 Roger Williams Hospital Viral vectors encoding multiple highly homologus non-viral polypeptides and the use of same
EP3192811A1 (en) 2009-02-09 2017-07-19 Université d'Aix-Marseille Pd-1 antibodies and pd-l1 antibodies and uses thereof
CA2950529C (en) 2009-04-03 2019-04-30 Verastem, Inc. Pyrimidine substituted purine compounds as kinase (s) inhibitors
KR20180077322A (en) 2009-04-30 2018-07-06 텔 하쇼머 메디컬 리서치 인프라스트럭쳐 앤드 서비시스 리미티드. Anti ceacam1 antibodies and methods of using same
KR20170119746A (en) 2009-09-03 2017-10-27 머크 샤프 앤드 돔 코포레이션 Anti-gitr antibodies
WO2011041093A1 (en) 2009-10-01 2011-04-07 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-vascular endothelial growth factor receptor-2 chimeric antigen receptors and use of same for the treatment of cancer
US9181527B2 (en) 2009-10-29 2015-11-10 The Trustees Of Dartmouth College T cell receptor-deficient T cell compositions
GB0919054D0 (en) 2009-10-30 2009-12-16 Isis Innovation Treatment of obesity
PL2496698T3 (en) 2009-11-03 2019-07-31 City Of Hope TRUNCATED EPIDERIMAL GROWTH FACTOR RECEPTOR (EGFRt) FOR TRANSDUCED T CELL SELECTION
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
JP5856073B2 (en) 2009-12-29 2016-02-09 エマージェント プロダクト デベロップメント シアトル, エルエルシー RON binding construct and method of use thereof
ES2724451T3 (en) 2010-02-04 2019-09-11 Univ Pennsylvania ICOS fundamentally regulates the expansion and function of inflammatory human Th17 lymphocytes
WO2011146862A1 (en) 2010-05-21 2011-11-24 Bellicum Pharmaceuticals, Inc. Methods for inducing selective apoptosis
US9242014B2 (en) 2010-06-15 2016-01-26 The Regents Of The University Of California Receptor tyrosine kinase-like orphan receptor 1 (ROR1) single chain Fv antibody fragment conjugates and methods of use thereof
JP2013532153A (en) 2010-06-18 2013-08-15 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッド Bispecific antibodies against TIM-3 and PD-1 for immunotherapy against chronic immune disease
KR20130098301A (en) 2010-07-09 2013-09-04 엑셀리시스, 인코포레이티드 Combinations of kinase inhibitors for the treatment of cancer
CA2805608A1 (en) 2010-07-16 2012-01-19 Piramal Enterprises Limited Substituted imidazoquinoline derivatives as kinase inhibitors
CN103501816A (en) 2010-10-27 2014-01-08 贝勒医学院 Chimeric CD27 receptors for redirecting t cells to CD70-positive malignancies
BR122021026173B1 (en) 2010-12-09 2023-12-05 The Trustees Of The University Of Pennsylvania PHARMACEUTICAL COMPOSITION
WO2012082841A2 (en) 2010-12-14 2012-06-21 University Of Maryland, Baltimore Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer
EP2665521A4 (en) 2011-01-18 2014-09-03 Univ Pennsylvania Compositions and methods for treating cancer
AU2012230780B2 (en) 2011-03-23 2016-10-27 Fred Hutchinson Cancer Center Method and compositions for cellular immunotherapy
WO2012127464A2 (en) 2011-03-23 2012-09-27 Gavish-Galilee Bio Applications Ltd Constitutively activated t cells for use in adoptive cell therapy
SG193956A1 (en) 2011-04-01 2013-11-29 Sloan Kettering Inst Cancer T cell receptor-like antibodies specific for a wt1 peptide presented by hla-a2
CN103596981B (en) 2011-04-08 2017-06-16 美国卫生和人力服务部 Anti-epidermal growth factor receptor variant III Chimeric antigen receptors and its purposes for treating cancer
JP6066991B2 (en) 2011-04-08 2017-01-25 ベイラー カレッジ オブ メディスンBaylor College Of Medicine Methods for reversing the effects of tumor microenvironment using chimeric cytokine receptors
US20130071414A1 (en) 2011-04-27 2013-03-21 Gianpietro Dotti Engineered cd19-specific t lymphocytes that coexpress il-15 and an inducible caspase-9 based suicide gene for the treatment of b-cell malignancies
WO2013006490A2 (en) 2011-07-01 2013-01-10 Cellerant Therapeutics, Inc. Antibodies that specifically bind to tim3
EP2736539B1 (en) 2011-07-25 2017-08-23 Nationwide Children's Hospital, Inc. Recombinant virus products and methods for inhibition of expression of dux4
CN114835823A (en) 2011-07-29 2022-08-02 宾夕法尼亚大学董事会 Transducible co-stimulatory receptors
CA2844742A1 (en) 2011-08-11 2013-02-14 Intellikine, Llc Kinase inhibitor polymorphs
WO2013033626A2 (en) 2011-08-31 2013-03-07 Trustees Of Dartmouth College Nkp30 receptor targeted therapeutics
US20130108641A1 (en) 2011-09-14 2013-05-02 Sanofi Anti-gitr antibodies
WO2013040557A2 (en) 2011-09-16 2013-03-21 The Trustees Of The University Of Pennsylvania Rna engineered t cells for the treatment of cancer
EP3692794A1 (en) 2011-09-16 2020-08-12 Baylor College of Medicine Targeting the tumor microenvironment using manipulated nkt cells
WO2013054320A1 (en) 2011-10-11 2013-04-18 Tel Hashomer Medical Research Infrastructure And Services Ltd. Antibodies to carcinoembryonic antigen-related cell adhesion molecule (ceacam)
BR112014008849A2 (en) 2011-10-20 2017-09-12 Us Health chimeric anti-cd22 antigen receptors
JP6385277B2 (en) 2011-12-01 2018-09-05 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッドThe Brigham and Women’s Hospital, Inc. Anti-CEACAM1 recombinant antibody for cancer treatment
CA3205751A1 (en) 2012-02-22 2013-08-29 The Trustees Of The University Of Pennsylvania Use of icos-based cars to enhance antitumor activity and car persistence
BR112014020502A2 (en) 2012-02-22 2019-09-24 Univ Pennsylvania isolated nucleic acid sequence, isolated antigen chimeric receptor, cell, vector, and methods for stimulating an immune response, for providing antitumor immunity, for treating a mammal, for generating and expanding a population of t-cells, and to modulate the amount of cytokine secreted by a t-cell
WO2013126712A1 (en) 2012-02-22 2013-08-29 The Trustees Of The University Of Pennsylvania Compositions and methods for generating a persisting population of t cells useful for the treatment of cancer
EP2872617A4 (en) 2012-07-13 2015-12-09 Univ Pennsylvania Epitope spreading associated with car t-cells
HUE053556T2 (en) * 2012-07-13 2021-07-28 Univ Pennsylvania Toxicity management for anti-tumor activity of cars
EA034644B1 (en) 2012-07-13 2020-03-02 Дзе Трастиз Оф Дзе Юниверсити Оф Пенсильвания Use of cart19 to deplete normal b cells to induce tolerance
BR112015000505A2 (en) 2012-07-13 2017-06-27 Univ Pennsylvania method of analyzing a genetically modified t-cell to detect a contaminant
US20150140019A1 (en) 2012-07-13 2015-05-21 The Trustees Of The University Of Pennsylvania Compositions and Methods for Regulating CAR T Cells
ES2786263T3 (en) 2012-07-13 2020-10-09 Univ Pennsylvania Enhancement of T-lymphocyte CAR activity by co-introduction of a bispecific antibody
JP2015525781A (en) 2012-07-31 2015-09-07 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッドThe Brigham and Women’s Hospital, Inc. Modulating the immune response
LT2884999T (en) 2012-08-20 2021-04-12 Fred Hutchinson Cancer Research Center Method and compositions for cellular immunotherapy
US9937205B2 (en) 2012-09-04 2018-04-10 The Trustees Of The University Of Pennsylvania Inhibition of diacylglycerol kinase to augment adoptive T cell transfer
WO2014039044A1 (en) * 2012-09-06 2014-03-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of producing t memory stem cell populations
EP2904106A4 (en) 2012-10-01 2016-05-11 Univ Pennsylvania Compositions and methods for targeting stromal cells for the treatment of cancer
WO2014055657A1 (en) 2012-10-05 2014-04-10 The Trustees Of The University Of Pennsylvania Use of a trans-signaling approach in chimeric antigen receptors
KR102229873B1 (en) 2012-10-12 2021-03-19 더 브리검 앤드 우먼즈 하스피털, 인크. Enhancement of the immune response
LT2956175T (en) 2013-02-15 2017-12-11 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
WO2014130635A1 (en) 2013-02-20 2014-08-28 Novartis Ag Effective targeting of primary human leukemia using anti-cd123 chimeric antigen receptor engineered t cells
EP3626741A1 (en) 2013-02-20 2020-03-25 The Trustees Of The University Of Pennsylvania Treatment of cancer using humanized anti-egfrviii chimeric antigen receptor
US9434935B2 (en) 2013-03-10 2016-09-06 Bellicum Pharmaceuticals, Inc. Modified caspase polypeptides and uses thereof
US9944690B2 (en) 2013-03-14 2018-04-17 Bellicum Pharmaceuticals, Inc. Methods for controlling T cell proliferation
EP3623380A1 (en) 2013-03-15 2020-03-18 Michael C. Milone Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
UY35468A (en) 2013-03-16 2014-10-31 Novartis Ag CANCER TREATMENT USING AN ANTI-CD19 CHEMERIC ANTIGEN RECEIVER
SG11201509609SA (en) 2013-05-24 2015-12-30 Univ Texas Chimeric antigen receptor-targeting monoclonal antibodies
WO2014197638A2 (en) 2013-06-05 2014-12-11 Bellicum Pharmaceuticals, Inc. Methods for inducing partial apoptosis using caspase polypeptides
CA2929181A1 (en) 2013-11-13 2015-05-21 Novartis Ag Mtor inhibitors for enhancing the immune response
US9512084B2 (en) 2013-11-29 2016-12-06 Novartis Ag Amino pyrimidine derivatives
CA2931684C (en) 2013-12-19 2024-02-20 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
JP6793902B2 (en) 2013-12-20 2020-12-02 ノバルティス アーゲー Adjustable chimeric antigen receptor
WO2015112626A1 (en) 2014-01-21 2015-07-30 June Carl H Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
JOP20200096A1 (en) 2014-01-31 2017-06-16 Children’S Medical Center Corp Antibody molecules to tim-3 and uses thereof
WO2015142661A1 (en) 2014-03-15 2015-09-24 Novartis Ag Regulatable chimeric antigen receptor
EP3593812A3 (en) 2014-03-15 2020-05-27 Novartis AG Treatment of cancer using chimeric antigen receptor
PL3129470T3 (en) 2014-04-07 2021-11-29 Novartis Ag Treatment of cancer using anti-cd19 chimeric antigen receptor
WO2015193740A2 (en) 2014-06-17 2015-12-23 Acerta Pharma B.V. Therapeutic combinations of a btk inhibitor, a pi3k inhibitor and/or a jak-2 inhibitor
TWI750110B (en) 2014-07-21 2021-12-21 瑞士商諾華公司 Treatment of cancer using humanized anti- bcma chimeric antigen receptor
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
MX2017001013A (en) 2014-07-21 2018-02-21 Novartis Ag Treatment of cancer using a cll-1 chimeric antigen receptor.
US20170274014A1 (en) 2014-07-21 2017-09-28 Jennifer Brogdon Combinations of low, immune enhancing, doses of mtor inhibitors and cars
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
US20170226495A1 (en) 2014-07-21 2017-08-10 Novartis Ag Sortase molecules and uses thereof
KR102594343B1 (en) 2014-07-21 2023-10-26 노파르티스 아게 Treatment of cancer using a cd33 chimeric antigen receptor
ES2781175T3 (en) 2014-07-31 2020-08-31 Novartis Ag Optimized subset of T cells containing a chimeric antigen receptor
CA2958200A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using a gfr alpha-4 chimeric antigen receptor
PT3183268T (en) 2014-08-19 2020-05-15 Novartis Ag Anti-cd123 chimeric antigen receptor (car) for use in cancer treatment
AU2015317608B2 (en) 2014-09-17 2021-03-11 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US10774388B2 (en) 2014-10-08 2020-09-15 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
WO2016061368A1 (en) 2014-10-15 2016-04-21 The Children's Hospital Of Philadelphia Compositions and methods for treating b-lymphoid malignancies
US20180334490A1 (en) 2014-12-03 2018-11-22 Qilong H. Wu Methods for b cell preconditioning in car therapy
KR20170093254A (en) 2014-12-29 2017-08-14 노파르티스 아게 Methods for producing chimeric antigen receptor-expressing cells
US11459390B2 (en) 2015-01-16 2022-10-04 Novartis Ag Phosphoglycerate kinase 1 (PGK) promoters and methods of use for expressing chimeric antigen receptor
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
ES2876974T3 (en) 2015-04-07 2021-11-15 Novartis Ag Combination therapy with chimeric antigen receptor and amino pyrimidine derivatives
IL303972A (en) 2015-04-08 2023-08-01 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
WO2016168595A1 (en) 2015-04-17 2016-10-20 Barrett David Maxwell Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
EP3286211A1 (en) 2015-04-23 2018-02-28 Novartis AG Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
AU2016297014B2 (en) 2015-07-21 2021-06-17 Novartis Ag Methods for improving the efficacy and expansion of immune cells
WO2017027392A1 (en) 2015-08-07 2017-02-16 Novartis Ag Treatment of cancer using chimeric cd3 receptor proteins
WO2017040930A2 (en) 2015-09-03 2017-03-09 The Trustees Of The University Of Pennsylvania Biomarkers predictive of cytokine release syndrome
BR112018005295A2 (en) 2015-09-17 2018-12-11 Novartis Ag T-cell therapies with increased efficacy
EP4012415A3 (en) 2015-12-04 2022-12-07 Juno Therapeutics, Inc. Methods and compositions related to toxicity associated with cell therapy
JP7082055B2 (en) 2015-12-22 2022-06-07 ノバルティス アーゲー Antibodies to Mesothelin Chimeric Antigen Receptor (CAR) and PD-L1 Inhibitors for Combined Use in Anticancer Treatment
ES2944597T3 (en) 2015-12-30 2023-06-22 Novartis Ag Enhanced Efficacy Immune Effector Cell Therapies
WO2018013918A2 (en) 2016-07-15 2018-01-18 Novartis Ag Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
EP3491016A1 (en) 2016-07-28 2019-06-05 Novartis AG Combination therapies of chimeric antigen receptors and pd-1 inhibitors
KR20190036551A (en) 2016-08-01 2019-04-04 노파르티스 아게 Treatment of Cancer Using Chimeric Antigen Receptors in Combination with Inhibitors of PRO-M2 Macrophage Molecules
WO2018059549A1 (en) 2016-09-30 2018-04-05 Novartis Ag Immune effector cell therapies with enhanced efficacy
CN110225927B (en) 2016-10-07 2024-01-12 诺华股份有限公司 Chimeric antigen receptor for the treatment of cancer
EP3577134A1 (en) 2017-01-31 2019-12-11 Novartis AG Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
CN112203725A (en) 2018-06-13 2021-01-08 诺华股份有限公司 BCMA chimeric antigen receptors and uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Affymetrix GeneChip® Human Genome U133 Set Package insert (2009) *
Del Rio et al (Journal of Clinical Oncology, 2007, 25(7): 773-780) *
Kuss et al (British Journal of Cancer, 2003, 88: 223-230) *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11865167B2 (en) 2013-02-20 2024-01-09 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
US11919946B2 (en) 2013-03-15 2024-03-05 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US11578130B2 (en) 2013-12-20 2023-02-14 Novartis Ag Regulatable chimeric antigen receptor
US11591404B2 (en) 2014-08-19 2023-02-28 Novartis Ag Treatment of cancer using a CD123 chimeric antigen receptor
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
US11667691B2 (en) 2015-08-07 2023-06-06 Novartis Ag Treatment of cancer using chimeric CD3 receptor proteins
US11747346B2 (en) 2015-09-03 2023-09-05 Novartis Ag Biomarkers predictive of cytokine release syndrome
US11549099B2 (en) 2016-03-23 2023-01-10 Novartis Ag Cell secreted minibodies and uses thereof
USRE49847E1 (en) 2016-10-07 2024-02-27 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
US11872249B2 (en) 2016-10-07 2024-01-16 Novartis Ag Method of treating cancer by administering immune effector cells expressing a chimeric antigen receptor comprising a CD20 binding domain
US11535662B2 (en) 2017-01-26 2022-12-27 Novartis Ag CD28 compositions and methods for chimeric antigen receptor therapy
US11608382B2 (en) 2018-06-13 2023-03-21 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11952428B2 (en) 2018-06-13 2024-04-09 Novartis Ag BCMA chimeric antigen receptors and uses thereof
WO2023019204A3 (en) * 2021-08-11 2023-03-30 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating and/or characterizing hematological malignancies and precursor conditions
WO2023081386A1 (en) * 2021-11-04 2023-05-11 The Regents Of The University Of Michigan Prevention and treatment of cytokine release syndrome and neurotoxicity associated with car-t cell therapy

Also Published As

Publication number Publication date
US20170306416A1 (en) 2017-10-26
AU2015330898B2 (en) 2022-03-10
WO2016057705A1 (en) 2016-04-14
JP2018501463A (en) 2018-01-18
CA2963935A1 (en) 2016-04-14
EP3204777A1 (en) 2017-08-16
IL279420A (en) 2021-01-31
SG11201702895SA (en) 2017-05-30
JP6815992B2 (en) 2021-01-20
JP2021073440A (en) 2021-05-13
AU2015330898A1 (en) 2017-04-27
RU2017115413A (en) 2018-11-15
CN106973568B (en) 2021-07-23
IL251525A0 (en) 2017-05-29
US10774388B2 (en) 2020-09-15
RU2017115413A3 (en) 2019-05-20
CN106973568A (en) 2017-07-21
CN114107424A (en) 2022-03-01
RU2743657C2 (en) 2021-02-20
AU2022203932A1 (en) 2022-06-23
KR20170068504A (en) 2017-06-19

Similar Documents

Publication Publication Date Title
US20210172020A1 (en) Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
JP6905163B2 (en) Biomarkers that predict cytokine release syndrome
US20200061113A1 (en) Chimeric antigen receptor therapy characterization assays and uses thereof
US20210220404A1 (en) Chimeric antigen receptors and uses thereof
US20210396739A1 (en) Biomarkers for evaluating car-t cells to predict clinical outcome
JP2021534783A (en) Method for producing chimeric antigen receptor-expressing cells
KR20220146530A (en) Methods of Making Chimeric Antigen Receptor-Expressing Cells
EP3704229B1 (en) Process for producing a t cell composition
US20220298222A1 (en) Combination therapy of a t cell therapy and an enhancer of zeste homolog 2 (ezh2) inhibitor and related methods
WO2023081894A2 (en) Pre-effector car-t cell gene signatures
WO2024056809A1 (en) Treatment of autoimmune disorders using chimeric antigen receptor therapy
AU2022330406A1 (en) Methods of making chimeric antigen receptor–expressing cells

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

AS Assignment

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA;REEL/FRAME:066546/0954

Effective date: 20240223

Owner name: THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA;REEL/FRAME:066546/0954

Effective date: 20240223

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS AG;REEL/FRAME:066545/0835

Effective date: 20240222

Owner name: THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS AG;REEL/FRAME:066545/0835

Effective date: 20240222

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION