US20210238250A9 - Braf-specific tcrs and uses thereof - Google Patents

Braf-specific tcrs and uses thereof Download PDF

Info

Publication number
US20210238250A9
US20210238250A9 US16/638,339 US201816638339A US2021238250A9 US 20210238250 A9 US20210238250 A9 US 20210238250A9 US 201816638339 A US201816638339 A US 201816638339A US 2021238250 A9 US2021238250 A9 US 2021238250A9
Authority
US
United States
Prior art keywords
cell
seq
amino acid
acid sequence
host cell
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
US16/638,339
Other languages
English (en)
Other versions
US20200223899A1 (en
Inventor
Joshua VEATCH
Stanley R. Riddell
Sylvia Lee
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.)
University of Washington
Fred Hutchinson Cancer Center
Original Assignee
University of Washington
Fred Hutchinson Cancer Research Center
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 University of Washington, Fred Hutchinson Cancer Research Center filed Critical University of Washington
Priority to US16/638,339 priority Critical patent/US20210238250A9/en
Publication of US20200223899A1 publication Critical patent/US20200223899A1/en
Publication of US20210238250A9 publication Critical patent/US20210238250A9/en
Assigned to UNIVERSITY OF WASHINGTON reassignment UNIVERSITY OF WASHINGTON ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEE, SYLVIA
Assigned to FRED HUTCHINSON CANCER RESEARCH CENTER reassignment FRED HUTCHINSON CANCER RESEARCH CENTER ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: RIDDELL, STANLEY R., VEATCH, Joshua
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • 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
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4632T-cell receptors [TCR]; antibody T-cell receptor constructs
    • 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
    • A61K39/464454Enzymes
    • A61K39/464462Kinases, e.g. Raf or Src
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • 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
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • A61K2239/57Skin; melanoma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/10041Use of virus, viral particle or viral elements as a vector
    • C12N2740/10043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • T cells Adoptive transfer of tumor-specific T cells is an appealing strategy to eliminate existing tumors and requires the establishment of a robust population of antigen-specific T cells in vivo to eliminate existing tumor and prevent recurrences (Stromnes et al., Immunol. Rev. 257:145, 2014).
  • immune responses to antigens created by mutations in cancer can be recognized by T cells and that these T cells can mediate clinical responses to treatment with adoptive cell therapy and immune checkpoint inhibitors.
  • Antigens that arise from such mutations are particularly appealing targets for immunotherapies due to being completely specific for the cancer relative to normal tissue, and also because they lack central tolerance mechanisms that could limit T cell function against other antigen types.
  • cytotoxic T lymphocytes can mediate direct anti-tumor activity in select patients (Chapuis et al., Cancer Res. 72:LB-136, 2012; Chapuis et al., Sci. Transl. Med. 5:174ra127, 2013; Chapuis et al., Proc. Nat'l. Acad. Sci. U.S.A. 109:4592, 2012) 2-4 , identifying and isolating tumor-reactive T cells with desired characteristics is a laborious and complex endeavor (see Stone and Kranz, Frontiers Immunol.
  • TCR immunotherapies directed against various cancers, such as hairy cell leukemia, malignant melanoma, thyroid, lung, and colon cancers.
  • TCR immunotherapies targeting antigens that are both cancer-specific and widely prevalent in cancers are needed.
  • Presently disclosed embodiments address these needs and provide other related advantages.
  • FIGS. 1A-1I show the identification and characterization of patient-derived CD4 + T cells specific for BRAFV600E.
  • 1 A Positron emission tomography showing recurrent tumor in left iliac region (left) and left thigh (right).
  • 1 B- 1 D Specificity and HLA restriction of BRAF V600E -specific T cells:( 1 B) IFN- ⁇ production by the patient-derived T cell line incubated with autologous B cells pulsed with wildtype and mutant BRAF peptide; ( 1 C) recognition of autologous B cells pulsed with mutant BRAF peptide or transfected with mRNA encoding mutant or wildtype BRAF sequences; ( 1 D) recognition of autologous B cells pulsed with mutant BRAF peptide in the presence or absence of HLA blocking antibodies.
  • FIGS. 2A-2O show the specificity of CD8 + T cells in TIL (tumor infiltrating lymphocytes) and TCR sequencing of T cell clonotypes in blood after adoptive transfer.
  • 2 A IFN- ⁇ production by TIL incubated with autologous CD40L-activated B cells pulsed with 13 pools of peptides encompassing 40 20-mer peptides containing the 20 nonsynonymous mutations present in the patient's melanoma by elispot assay. The final concentration of each peptide in the assay was 10 ⁇ g/ml. Three technical replicates were performed.
  • TCR V ⁇ clonotypes of CD4 + BRAF V600E and CD8 + T cells specific for the specified antigens in pre-treatment and post-treatment blood ( 2 L- 2 O) TCR ⁇ sequencing on the TIL and T cells from TIL incubated with autologous B cells and tiled peptides. Tiled peptides spanned ( 2 L) Tyrosinase, ( 2 M) Mart1, ( 2 N) Mage A3, and ( 2 O) TRP2 and sorted by IFN- ⁇ capture. Antigen-specific TCR ⁇ sequences enriched in the sorted cells are marked with a box.
  • FIGS. 3A-3C show phenotypic analysis for BRAF-specific T cells following treatment with tumor-infiltrating lymphocytes (TILs).
  • TILs tumor-infiltrating lymphocytes
  • 3 A Dump channel gating scheme for excluding monocytes (CD14 + ), B cells (CD19 + ), and dead cells (ViaProbe) from TIL. Viable CD4+ T cells were plotted against tetramer and CD45RA.
  • 3 B CD45RA- memory cells (88.1% of CD4 + ) that were tetramer-positive and -negative were plotted against the indicated cell surface markers. Numbers indicate the percentage of cells in the gated regions or the percentage of tetramer-positive cells for each marker.
  • 3 C Intracellular cytokine staining of activated (CD154+) BRAF-specific T cells.
  • FIGS. 4A-4B show that a synthetic TCR derived from the dominant V ⁇ and V ⁇ sequences of melanoma-responsive patient TILs recognizes cells expressing BRAF V600E .
  • 4 A Frequency of TCRBV ⁇ sequences in peripheral blood mononuclear cells after mock stimulation, BRAF V600E stimulation, or after sorting IFN- ⁇ secretion cells following BRAF V600E peptide re-stimulation.
  • FIG. 5 shows IFN- ⁇ production by CD4 + T cells transduced with one of four TCRs (pJV88-pJV91) made using TCR ⁇ and TCR ⁇ genes identified in the patient TILs. Left, no antigen peptide. Right, antigen peptide.
  • FIGS. 6A and 6B show the effects of CRISPR-mediated knockout of endogenous TCR sequences on expression of a heterologous BRAF-specific TCR in primary human CD4+ T cells.
  • 6 A Stimulated T cells were transfected with Cas9-RNPs targeting TCRA and TCRB and transduced with DNA encoding BRAF V600E -specific TCR. BRAF-specific TCR expression was measured by Vbeta3.1 and TCR expression was measured using anti-CD3.
  • Top panels unmodified cells without (left) or with (right) CRISPR-mediated TCR knockout.
  • Bottom panels transduced cells without (left) or with (right) CRISPR-mediated TCR knockout.
  • T cells modified with BRAF V600E -specific TCR with or without deletion of endogenous TCR were compared to untransduced cells (left-most peaks) and a patient-derived antigen-specific T cell clone (right-most peaks) for binding to tetramer.
  • binding proteins such as T cell receptors (TCRs), that are capable of specifically binding to a BRAF V600E peptide antigen, such as a BRAF V600E peptide antigen associated with a major histocompatibility complex (MHC) (e.g., human leukocyte antigen, HLA).
  • TCRs T cell receptors
  • MHC major histocompatibility complex
  • Binding proteins of this disclosure are useful in, for example, therapies to treat hyperproliferative diseases, such as cancer, characterized by BRAF V600E expression.
  • antigens created by cancer-associated mutations are appealing targets for therapeutic intervention, but are generally unique to an individual patient.
  • antigens caused by essential “driver” mutations of cancer are of interest since they are both specific to cancer cells and occur at high frequencies in patient populations.
  • BRAF protein is involved in cell growth signaling, while mutant BRAF is implicated in a number of cancers (see, e.g., Frasca et al., Endocrine - Related Cancer 15:191(2008)).
  • the substitution mutation V600E (BRAF V600E ) arising in exon 15 of the BRAF gene, activates BRAF to drive a growth signaling pathway that is an early event in carcinogenesis. This mutation is found in all instances of hairy cell leukemia, about half of malignant melanoma cases, and significant numbers of patients with advanced thyroid, lung and colon cancer.
  • compositions and methods described herein will in certain embodiments have therapeutic utility for the treatment of diseases and conditions associated with BRAF V600E expression.
  • diseases include various forms of hyperproliferative disorders, such as hairy cell leukemia, melanoma, thyroid cancers including poorly differentiated thyroid cancer, non-small cell lung cancer, colorectal cancer, papillary cancer, non-Hodgkin lymphoma, glioblastoma, and pilocytic astrocytoma, breast cancer, ovarian cancer, Langerhans cell histiocytosis, and sarcomas (e.g., fibrosarcoma (fibroblastic sarcoma), Dermatofibrosarcoma protuberans (DF SP), osteosarcoma, rhabdomyosarcoma, Ewing's sarcoma, a gastrointestinal stromal tumor, Leiomyosarcoma; angiosarcoma (vascular sarcoma), Kaposi's sarcoma,
  • Non-limiting examples of these and related uses are described herein and include in vitro, ex vivo and in vivo stimulation of BRAF V600E antigen-specific T cell responses, such as by the use of recombinant T cells expressing TCR specific for a BRAF V600E peptide.
  • any concentration range, percentage range, ratio range, or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated.
  • any number range recited herein relating to any physical feature, such as polymer subunits, size or thickness are to be understood to include any integer within the recited range, unless otherwise indicated.
  • the term “about” means ⁇ 20% of the indicated range, value, or structure, unless otherwise indicated. It should be understood that the terms “a” and “an” as used herein refer to “one or more” of the enumerated components.
  • a protein domain, region, or module e.g., a binding domain, hinge region, linker module
  • a protein which may have one or more domains, regions, or modules
  • an “immune system cell” means any cell of the immune system that originates from a hematopoietic stem cell in the bone marrow, which gives rise to two major lineages, a myeloid progenitor cell (which give rise to myeloid cells such as monocytes, macrophages, dendritic cells, megakaryocytes and granulocytes) and a lymphoid progenitor cell (which give rise to lymphoid cells such as T cells, B cells and natural killer (NK) cells, including Natural Killer T (NK-T) cells).
  • a myeloid progenitor cell which give rise to myeloid cells such as monocytes, macrophages, dendritic cells, megakaryocytes and granulocytes
  • lymphoid progenitor cell which give rise to lymphoid cells such as T cells, B cells and natural killer (NK) cells, including Natural Killer T (NK-T) cells.
  • Exemplary immune system cells include a CD4 + T cell, a CD8 + T cell, a CD4 ⁇ CD8 ⁇ double negative T cell, a ⁇ T cell, a regulatory T cell, a natural killer cell, a natural killer T cell, and a dendritic cell.
  • Macrophages and dendritic cells may be referred to as “antigen presenting cells” or “APCs,” which are specialized cells that can activate T cells when a major histocompatibility complex (MEW) receptor on the surface of the APC complexed with a peptide interacts with a TCR on the surface of a T cell.
  • MW major histocompatibility complex
  • MHC Major histocompatibility complex
  • MHC class I molecules are heterodimers having a membrane spanning a chain (with three a domains) and a non-covalently associated ⁇ 2 microglobulin.
  • MEW class II molecules are composed of two transmembrane glycoproteins, ⁇ and ⁇ , both of which span the membrane. Each chain has two domains.
  • MHC class I molecules deliver peptides originating in the cytosol to the cell surface, where a peptide:MHC complex is recognized by CD8 + T cells.
  • HLA class II molecules deliver peptides originating in the vesicular system to the cell surface, where a peptide:MHC complex is recognized by CD4 + T cells.
  • Human MHC is referred to as human leukocyte antigen (HLA).
  • HLA-II types include DP, DM, DOA, DOB, DQ, and DR. Numerous alleles encoding the subunits of the various HLA types are known, including, for example, HLA-DQA1*03, HLA-DQB1*0301, HLA-DQB1*0302, HLA-DQB1*0303.
  • a binding protein according to the present disclosure is capable of recognizing a BRAF V600E peptide complexed with HLA-DQ.
  • the HLA complex comprises HLA-DQB1*0301, *0302, or *0303.
  • the HLA complex comprises HLA-DQB1*0302.
  • the HLA complex comprises HLA-DQA1*03.
  • T cell or “T lymphocyte” is an immune system cell that matures in the thymus and produces T cell receptors (TCRs).
  • T cells can be na ⁇ ve (not exposed to antigen; increased expression of CD62L, CCR7, CD28, CD3, CD127, and CD45RA, and decreased expression of CD45RO as compared to T CM ), memory T cells (T M ) (antigen-experienced and long-lived), and effector cells (antigen-experienced, cytotoxic).
  • T M can be further divided into subsets of: central memory T cells (T CM , increased expression of CD62L, CCR7, CD28, CD127, CD45RO, and CD95, and decreased expression of CD54RA as compared to naive T cells); and effector memory T cells (T EM , decreased expression of CD62L, CCR7, CD28, CD45RA, and increased expression of CD127 as compared to na ⁇ ve T cells or T CM ).
  • T CM central memory T cells
  • T EM effector memory T cells
  • Effector T cells refers to antigen-experienced CD8 + cytotoxic T lymphocytes that have decreased expression of CD62L ,CCR7, CD28, and are positive for granzyme and perforin as compared to T CM .
  • Helper T cells are CD4 + cells that influence the activity of other immune cells by releasing cytokines.
  • CD4 ⁇ T cells can activate and suppress an adaptive immune response, and which of those two functions is induced will depend on presence of other cells and signals.
  • T cells can be collected using known techniques, and the various subpopulations or combinations thereof can be enriched or depleted by known techniques, such as by affinity binding to antibodies, flow cytometry, or immunomagnetic selection.
  • Other exemplary T cells include regulatory T cells, such as CD4 + CD25 + (Foxp3 + ) regulatory T cells and Treg17 cells, as well as Tr1, Th3, CD8 + CD28 ⁇ , and Qa-1 restricted T cells.
  • T cell receptor refers to an immunoglobulin superfamily member (having a variable binding domain, a constant domain, a transmembrane region, and a short cytoplasmic tail; see, e.g., Janeway et al., Immunobiology: The Immune System in Health and Disease, 3 rd Ed., Current Biology Publications, p. 4:33, 1997) capable of specifically binding to an antigen peptide bound to a MHC receptor.
  • a TCR can be found on the surface of a cell or in soluble form and generally is comprised of a heterodimer having ⁇ and ⁇ chains (also known as TCR ⁇ and TCR ⁇ , respectively), or ⁇ and ⁇ chains (also known as TCR ⁇ and TCR ⁇ , respectively).
  • TCR chains e.g., ⁇ -chain, ⁇ -chain
  • the extracellular portion of TCR chains e.g., ⁇ -chain, ⁇ -chain
  • V ⁇ variable domain
  • V ⁇ typically amino acids 1 to 116 based on Kabat numbering
  • variable domains contain complementary determining regions (CDRs) separated by framework regions (FRs) (see, e.g., Jores et al., Proc. Nat'l Acad. Sci. U.S.A.
  • TCR variable domain sequences can be aligned to a numbering scheme (e.g., Kabat, EU, International Immunogenetics Information System (IMGT) and Aho), which can allow equivalent residue positions to be annotated and for different molecules to be compared using Antigen receptor Numbering And Receptor Classification (ANARCI) software tool (2016, Bioinformatics 15:298-300).
  • a numbering scheme provides a standardized delineation of framework regions and CDRs in the TCR variable domains.
  • a TCR is found on the surface of T cells (or T lymphocytes) and associates with the CD3 complex.
  • the source of a TCR as used in the present disclosure may be from various animal species, such as a human, mouse, rat, rabbit or other mammal.
  • CD3 is known in the art as a multi-protein complex of six chains (see, Abbas and Lichtman, 2003; Janeway et al., p. 172 and 178, 1999). In mammals, the complex comprises a CD3 ⁇ chain, a CD3 ⁇ chain, two CD3 ⁇ chains, and a homodimer of CD3 ⁇ chains.
  • the CD3 ⁇ , CD3 ⁇ , and CD3 ⁇ chains are highly related cell surface proteins of the immunoglobulin superfamily containing a single immunoglobulin domain.
  • the transmembrane regions of the CD3 ⁇ , CD3 ⁇ , and CD3 ⁇ chains are negatively charged, which is a characteristic that allows these chains to associate with the positively charged T cell receptor chains.
  • CD3 ⁇ , CD3 ⁇ , and CD3 ⁇ chains each contain a single conserved motif known as an immunoreceptor tyrosine-based activation motif or ITAM, whereas each CD3 ⁇ chain has three ITAMs. Without wishing to be bound by theory, it is believed that the ITAMs are important for the signaling capacity of a TCR complex.
  • CD3 as used in the present disclosure may be from various animal species, including human, mouse, rat, or other mammals.
  • TCR complex refers to a complex formed by the association of CD3 with TCR.
  • a TCR complex can be composed of a CD3 ⁇ chain, a CD3 ⁇ chain, two CD3 ⁇ chains, a homodimer of CD3 ⁇ chains, a TCR ⁇ chain, and a TCR ⁇ chain.
  • a TCR complex can be composed of a CD3 ⁇ chain, a CD3 ⁇ chain, two CD3 ⁇ chains, a homodimer of CD3 ⁇ chains, a TCR ⁇ chain, and a TCR ⁇ chain.
  • a “component of a TCR complex,” as used herein, refers to a TCR chain (i.e., TCR ⁇ , TCR ⁇ , TCR ⁇ or TCR ⁇ ), a CD3 chain (i.e., CD3 ⁇ , CD3 ⁇ , CD3 ⁇ or CD3 ⁇ ), or a complex formed by two or more TCR chains or CD3 chains (e.g., a complex of TCR ⁇ and TCR ⁇ , a complex of TCR ⁇ and TCR ⁇ , a complex of CD3 ⁇ and CD3 ⁇ , a complex of CD3 ⁇ and CD3 ⁇ , or a sub-TCR complex of TCR ⁇ , TCR ⁇ , CD3 ⁇ , CD3 ⁇ , and two CD3 ⁇ chains).
  • CD4 refers to an immunoglobulin co-receptor glycoprotein that assists the TCR in communicating with antigen-presenting cells (see, Campbell & Reece, Biology 909 (Benjamin Cummings, Sixth Ed., 2002); UniProtKB P01730). CD4 is found on the surface of immune cells such as T helper cells, monocytes, macrophages, and dendritic cells, and includes four immunoglobulin domains (D1 to D4) that are expressed at the cell surface. During antigen presentation, CD4 is recruited, along with the TCR complex, to bind to different regions of the MHCII molecule (CD4 binds MHCII ⁇ 2, while the TCR complex binds MHCII ⁇ 1/ ⁇ 1).
  • TAMs immunoreceptor tyrosine activation motifs
  • CD8 co-receptor or “CD8” means the cell surface glycoprotein CD8, either as an alpha-alpha homodimer or an alpha-beta heterodimer.
  • the CD8 co-receptor assists in the function of cytotoxic T cells (CD8 + ) and functions through signaling via its cytoplasmic tyrosine phosphorylation pathway (Gao and Jakobsen, Immunol. Today 21:630-636, 2000; Cole and Gao, Cell. Mol. Immunol. 1:81-88, 2004).
  • CD8 beta chains see UniProtKB identifier P10966
  • a single CD8 alpha chain see UniProtKB identifier P01732
  • variable region refers to the domain of a TCR ⁇ -chain or ⁇ -chain (or ⁇ -chain and ⁇ -chain for ⁇ TCRs) that is involved in binding of the TCR to antigen.
  • the variable domains of the ⁇ -chain and ⁇ -chain (V ⁇ and V ⁇ , respectively) of a native TCR generally have similar structures, with each domain comprising four generally conserved framework regions (FRs) and three CDRs.
  • the V ⁇ domain is encoded by two separate DNA segments, the variable gene segment and the joining gene segment (V-J); the V ⁇ domain is encoded by three separate DNA segments, the variable gene segment, the diversity gene segment, and the joining gene segment (V-D-J).
  • V ⁇ or V ⁇ domain may be sufficient to confer antigen-binding specificity.
  • TCRs that bind a particular antigen may be isolated using a V ⁇ or V ⁇ domain from a TCR that binds the antigen to screen a library of complementary V ⁇ or V ⁇ domains, respectively.
  • CDR complementarity determining region
  • HVR hypervariable region
  • a binding protein of the present disclosure comprises an ⁇ CDR1, an ⁇ CDR2, and/or an ⁇ CDR3 amino acid sequence of a V ⁇ domain as set forth in any one of SEQ ID NOs:1-4. In certain embodiments, a binding protein of the present disclosure comprises a ⁇ CDR1, a ⁇ CDR2, and/or a ⁇ CDR3 amino acid sequence of a V ⁇ domain as set forth in any one of SEQ ID NOs:5-7.
  • Antigen or “Ag” as used herein refers to an immunogenic molecule that provokes an immune response. This immune response may involve antibody production, activation of specific immunologically-competent cells (e.g., T cells), or both.
  • An antigen may be, for example, a peptide, glycopeptide, polypeptide, glycopolypeptide, polynucleotide, polysaccharide, lipid or the like. It is readily apparent that an antigen can be synthesized, produced recombinantly, or derived from a biological sample.
  • Exemplary biological samples that can contain one or more antigens include tissue samples, tumor samples, cells, biological fluids, or combinations thereof.
  • Antigens can be produced by cells that have been modified or genetically engineered to express an antigen.
  • Exemplary antigens include BRAF V600E .
  • epitope includes any molecule, structure, amino acid sequence or protein determinant that is recognized and specifically bound by a cognate binding molecule, such as an immunoglobulin, T cell receptor (TCR), chimeric antigen receptor, or other binding molecule, domain or protein.
  • a cognate binding molecule such as an immunoglobulin, T cell receptor (TCR), chimeric antigen receptor, or other binding molecule, domain or protein.
  • Epitopic determinants generally contain chemically active surface groupings of molecules, such as amino acids or sugar side chains, and can have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • binding domain refers to a molecule or portion thereof (e.g., peptide, oligopeptide, polypeptide, protein) that possesses the ability to specifically and non-covalently associate, unite, or combine with a target (e.g., BRAF V600E ).
  • a binding domain includes any naturally occurring, synthetic, semi-synthetic, or recombinantly produced binding partner for a biological molecule, a molecular complex (i.e., complex comprising two or more biological molecules), or other target of interest.
  • binding domains include single chain immunoglobulin variable regions (e.g., scTCR, scFv), receptor ectodomains, ligands (e.g., cytokines, chemokines), or synthetic polypeptides selected for their specific ability to bind to a biological molecule, a molecular complex or other target of interest.
  • scTCR single chain immunoglobulin variable regions
  • scFv receptor ectodomains
  • ligands e.g., cytokines, chemokines
  • synthetic polypeptides selected for their specific ability to bind to a biological molecule, a molecular complex or other target of interest.
  • binding protein e.g., TCR receptor
  • binding domain or fusion protein thereof
  • K a an affinity or K a (i.e., an equilibrium association constant of a particular binding interaction with units of 1/M) equal to or greater than 10 5 M ⁇ 1 (which equals the ratio of the on-rate [k on ] to the off-rate [k off ] for this association reaction), while not significantly associating or uniting with any other molecules or components in a sample.
  • Binding proteins or binding domains may be classified as “high-affinity” binding proteins or binding domains (or fusion proteins thereof) or as “low-affinity” binding proteins or binding domains (or fusion proteins thereof).
  • “High-affinity” binding proteins or binding domains refer to those binding proteins or binding domains having a K a of at least 10 7 M ⁇ 1 , at least 10 8 M ⁇ 1 , at least 10 9 M ⁇ 1 , at least 10 10 M ⁇ 1 ,at least 10 11 M ⁇ 1 , at least 10 12 M ⁇ 1 , or at least 10 13 M ⁇ 1 .
  • “Low-affinity” binding proteins or binding domains refer to those binding proteins or binding domains having a K a of up to 10 7 M ⁇ 1 , up to 10 6 M ⁇ 1 , up to 10 5 M ⁇ 1 .
  • affinity may be defined as an equilibrium dissociation constant (K d ) of a particular binding interaction with units of M (e.g., 10 ⁇ 5 M to 10 ⁇ 13 M).
  • a receptor or binding domain may have “enhanced affinity,” which refers to selected or engineered receptors or binding domains with stronger binding to a target antigen than a wild type (or parent) binding domain.
  • enhanced affinity may be due to a K a (equilibrium association constant) for the target antigen that is higher than the wild type binding domain, due to a K d (dissociation constant) for the target antigen that is less than that of the wild type binding domain, due to an off-rate (k off ) for the target antigen that is less than that of the wild type binding domain, or a combination thereof.
  • enhanced affinity TCRs may be codon optimized to enhance expression in a particular host cell, such as T cells (Scholten et al., Clin. Immunol. 119:135, 2006).
  • a variety of assays are known for identifying binding domains of the present disclosure that specifically bind a particular target, as well as determining binding domain or fusion protein affinities, such as Western blot, ELISA, analytical ultracentrifugation, spectroscopy and surface plasmon resonance (Biacore®) analysis (see, e.g., Scatchard et al., Ann. N.Y. Acad. Sci. 51:660, 1949; Wilson, Science 295:2103, 2002; Wolff et al., Cancer Res. 53:2560, 1993; and U.S. Pat. Nos. 5,283,173, 5,468,614, or the equivalent). Assays for assessing affinity or apparent affinity or relative affinity are also known.
  • apparent affinity for a TCR is measured by assessing binding to various concentrations of tetramers, for example, by flow cytometry using labeled tetramers.
  • apparent K D of a TCR is measured using 2-fold dilutions of labeled tetramers at a range of concentrations, followed by determination of binding curves by non-linear regression, apparent K D being determined as the concentration of ligand that yielded half-maximal binding.
  • BRAF V600E -specific binding protein refers to a protein or polypeptide that specifically binds to a BRAF V600E peptide antigen or a BRAF V600E peptide antigen:HLA complex, e.g., on a cell surface, and does not bind a HLA complex on a cell surface comprising a BRAF peptide not containing the BRAF V600E mutation.
  • a BRAF V600E -specific binding protein binds a BRAF V600 E-containing peptide:HLA complex (or BRAF V600E -containing peptide:MHC complex) with a K d of less than about 10 ⁇ 8 M, less than about 10 ⁇ 9 M, less than about 10 ⁇ 10 M, less than about 10 ⁇ 11 M, less than about 10 ⁇ 12 M, or less than about 10 ⁇ 13 M, or with an affinity that is about the same as, at least about the same as, or is greater than at or about the affinity exhibited by an exemplary BRAF V600E -specific binding protein provided herein, such as any of the BRAF V600E -specific TCRs provided herein, for example, as measured by the same assay.
  • a BRAF V600 E-specific binding protein comprises a BRAF V600E -specific immunoglobulin superfamily binding protein or binding portion thereof.
  • BRAF V600E binding domain or “BRAF V600E binding fragment” refers to a domain or portion of a BRAF V600E -specific binding protein responsible for the specific BRAF V600E binding.
  • a BRAF V600E -specific binding domain alone i.e., without any other portion of a BRAF V600E -specific binding protein
  • BRAF V600E can be soluble and can bind to BRAF V600E (e.g., in complex with an MHC receptor molecule or functional fragment thereof) with a K d of less than about 10 ⁇ 8 M, less than about 10 ⁇ 9 M, less than about 10 ⁇ 10 M, less than about 10 ⁇ 11 M, less than about 10 ⁇ 12 M, or less than about 10 ⁇ 13 M.
  • Exemplary BRAF V600E -specific binding domains include BRAF V600E -specific scTCR (e.g., single chain ⁇ TCR proteins such as V ⁇ -L-V ⁇ , V ⁇ -L-V ⁇ , V ⁇ -C ⁇ -L-V ⁇ , or V ⁇ -L-V ⁇ -C ⁇ , wherein V ⁇ and V ⁇ are TCR ⁇ and ⁇ variable domains respectively, C ⁇ and C ⁇ are TCR ⁇ and ⁇ constant domains, respectively, and L is a linker) and scFv fragments as described herein, which can be derived from an anti-BRAF V600E TCR or antibody.
  • BRAF V600E -specific scTCR e.g., single chain ⁇ TCR proteins such as V ⁇ -L-V ⁇ , V ⁇ -L-V ⁇ , V ⁇ -C ⁇ -L-V ⁇ , or V ⁇ -L-V ⁇ -C ⁇ , wherein V ⁇ and V ⁇ are TCR ⁇ and ⁇ variable domains respectively, C ⁇ and C ⁇ are TCR
  • APC antigen presenting cells
  • MHC major histocompatibility complex
  • processed antigen peptides originating in the cytosol are generally from about 7 amino acids to about 11 amino acids in length and will associate with class I MEW molecules
  • peptides processed in the vesicular system e.g., bacterial, viral
  • peptides processed in the vesicular system will generally vary in length from about 10 amino acids to about 25 amino acids and associate with class II MHC molecules.
  • BRAF V600 E antigen or “ BRAF V600E peptide antigen” or 'BRAF V600E -containing peptide antigen” refers to a naturally or synthetically produced portion of a BRAF protein ranging in length from about 7 amino acids to about 20 amino acids and comprising the V600E substitution mutation (e.g., a peptide from BRAF 597-603 , or
  • BRAF 590-610 that includes a glutamic acid substituted for a valine at the residue corresponding to position 600 of the full-length wild-type BRAF; see, e.g., Uniprot entry no. P15056 and NCBI Reference identifier NP_004324.2), which can form a complex with a MEW (e.g., HLA) molecule and such a complex can bind with a binding protein specific fora BRAF V600E peptide:MHC (e.g., HLA) complex.
  • exemplary BRAF V600E peptide antigens include those having the amino acid sequence set forth in SEQ ID NO.: 38 or 39.
  • a “linker” refers to an amino acid sequence that connects two proteins, polypeptides, peptides, domains, regions, or motifs and may provide a spacer function compatible with interaction of the two sub-binding domains so that the resulting polypeptide retains a specific binding affinity (e.g., scTCR) to a target molecule or retains signaling activity (e.g., TCR complex).
  • a linker is comprised of about two to about 35 amino acids, for instance, or about four to about 20 amino acids or about eight to about 15 amino acids or about 15 to about 25 amino acids.
  • “Junction amino acids” or “junction amino acid residues” refer to one or more (e.g., about 2-10) amino acid residues between two adjacent motifs, regions or domains of a polypeptide, such as between a binding domain and an adjacent constant domain or between a TCR chain and an adjacent self-cleaving peptide. Junction amino acids may result from the construct design of a fusion protein (e.g., amino acid residues resulting from the use of a restriction enzyme site during the construction of a nucleic acid molecule encoding a fusion protein).
  • altered domain refers to a motif, region, domain, peptide, polypeptide, or protein with a non-identical sequence identity to a wild type motif, region, domain, peptide, polypeptide, or protein (e.g., a wild type TCR ⁇ chain, TCR ⁇ chain, TCR ⁇ constant domain, TCRI ⁇ constant domain) of at least 85% (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%).
  • a wild type motif, region, domain, peptide, polypeptide, or protein e.g., a wild type TCR ⁇ chain, TCR ⁇ chain, TCR ⁇ constant domain, TCRI ⁇ constant domain
  • nucleic acid or “nucleic acid molecule” refers to any of deoxyribonucleic acid (DNA), ribonucleic acid (RNA), oligonucleotides, fragments generated, for example, by the polymerase chain reaction (PCR) or by in vitro translation, and fragments generated by any of ligation, scission, endonuclease action, or exonuclease action.
  • the nucleic acids of the present disclosure are produced by PCR.
  • Nucleic acids may be composed of monomers that are naturally occurring nucleotides (such as deoxyribonucleotides and ribonucleotides), analogs of naturally occurring nucleotides (e.g., ⁇ -enantiomeric forms of naturally-occurring nucleotides), or a combination of both.
  • Modified nucleotides can have modifications in or replacement of sugar moieties, or pyrimidine or purine base moieties.
  • Nucleic acid monomers can be linked by phosphodiester bonds or analogs of such linkages.
  • Analogs of phosphodiester linkages include phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phosphoranilidate, phosphoramidate, and the like. Nucleic acid molecules can be either single stranded or double stranded.
  • isolated means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring).
  • a naturally occurring nucleic acid or polypeptide present in a living animal is not isolated, but the same nucleic acid or polypeptide, separated from some or all of the co-existing materials in the natural system, is isolated.
  • Such nucleic acid could be part of a vector and/or such nucleic acid or polypeptide could be part of a composition (e.g., a cell lysate), and still be isolated in that such vector or composition is not part of the natural environment for the nucleic acid or polypeptide.
  • gene means the segment of DNA involved in producing a polypeptide chain; it includes regions preceding and following the coding region (“leader and trailer”) as well as intervening sequences (introns) between individual coding segments (exons).
  • the term “recombinant” refers to a cell, microorganism, nucleic acid molecule, or vector that has been genetically engineered by human intervention- 13 that is, modified by introduction of a heterologous nucleic acid molecule, or refers to a cell or microorganism that has been altered such that expression of an endogenous nucleic acid molecule or gene is controlled, deregulated, deleted, attenuated, or constitutive.
  • Human generated genetic alterations may include, for example, modifications that introduce nucleic acid molecules (which may include an expression control element, such as a promoter) that encode one or more proteins or enzymes, or other nucleic acid molecule additions, deletions, substitutions, or other functional disruption of or addition to a cell's genetic material. Exemplary modifications include those in coding regions or functional fragments thereof of heterologous or homologous polypeptides from a reference or parent molecule.
  • mutation refers to a change in the sequence of a nucleic acid molecule or polypeptide molecule as compared to a reference or wild-type nucleic acid molecule or polypeptide molecule, respectively.
  • a mutation can result in several different types of change in sequence, including substitution, insertion or deletion of nucleotide(s) or amino acid(s).
  • a mutation is a substitution of one or three codons or amino acids, a deletion of one to about 5 codons or amino acids, or a combination thereof.
  • a “conservative substitution” is recognized in the art as a substitution of one amino acid for another amino acid that has similar properties.
  • Exemplary conservative substitutions are well known in the art (see, e.g., WO 97/09433 at page 10; Lehninger, Biochemistry, 2 nd Edition; Worth Publishers, Inc. NY, N.Y., pp.71-77, 1975; Lewin, Genes IV, Oxford University Press, NY and Cell Press, Cambridge, Ma., p. 8, 1990).
  • construct refers to any polynucleotide that contains a recombinant nucleic acid molecule.
  • a construct may be present in a vector (e.g., a bacterial vector, a viral vector) or may be integrated into a genome.
  • a “vector” is a nucleic acid molecule that is capable of transporting another nucleic acid molecule.
  • Vectors may be, for example, plasmids, cosmids, viruses, a RNA vector or a linear or circular DNA or RNA molecule that may include chromosomal, non-chromosomal, semi-synthetic or synthetic nucleic acid molecules.
  • Exemplary vectors are those capable of autonomous replication (episomal vector) or expression of nucleic acid molecules to which they are linked (expression vectors).
  • Viral vectors include retrovirus, adenovirus, parvovirus (e.g., adeno-associated viruses), coronavirus, negative strand RNA viruses such as ortho-myxovirus (e.g., influenza virus), rhabdovirus (e.g., rabies and vesicular stomatitis virus), paramyxovirus (e.g., measles and Sendai), positive strand RNA viruses such as picornavirus and alphavirus, and double-stranded DNA viruses including adenovirus, herpesvirus (e.g.,
  • Herpes Simplex virus types 1 and 2 Epstein-Barr virus, cytomegalovirus), and poxvirus (e.g., vaccinia, fowlpox and canarypox).
  • Other viruses include Norwalk virus, togavirus, flavivirus, reoviruses, papovavirus, hepadnavirus, and hepatitis virus, for example.
  • retroviruses include avian leukosis-sarcoma, mammalian C-type, B-type viruses, D type viruses, HTLV-BLV group, lentivirus, spumavirus (Coffin, J. M., Retroviridae: The viruses and their replication, In Fundamental Virology, Third Edition, B. N. Fields et al., Eds., Lippincott-Raven Publishers, Philadelphia, 1996).
  • Lentiviral vector means HIV-based lentiviral vectors for gene delivery, which can be integrative or non-integrative, have relatively large packaging capacity, and can transduce a range of different cell types. Lentiviral vectors are usually generated following transient transfection of three (packaging, envelope and transfer) or more plasmids into producer cells. Like HIV, lentiviral vectors enter the target cell through the interaction of viral surface glycoproteins with receptors on the cell surface. On entry, the viral RNA undergoes reverse transcription, which is mediated by the viral reverse transcriptase complex. The product of reverse transcription is a double-stranded linear viral DNA, which is the substrate for viral integration into the DNA of infected cells.
  • operably linked refers to the association of two or more nucleic acid molecules on a single nucleic acid fragment so that the function of one is affected by the other.
  • a promoter is operably linked with a coding sequence when it is capable of affecting the expression of that coding sequence (i.e., the coding sequence is under the transcriptional control of the promoter).
  • Unlinked means that the associated genetic elements are not closely associated with one another and the function of one does not affect the other.
  • expression vector refers to a DNA construct containing a nucleic acid molecule that is operably-linked to a suitable control sequence capable of effecting the expression of the nucleic acid molecule in a suitable host.
  • control sequences include a promoter to effect transcription, an optional operator sequence to control such transcription, a sequence encoding suitable mRNA ribosome binding sites, and sequences which control termination of transcription and translation.
  • the vector may be a plasmid, a phage particle, a virus, or simply a potential genomic insert. Once transformed into a suitable host, the vector may replicate and function independently of the host genome, or may, in some instances, integrate into the genome itself.
  • plasmid,” “expression plasmid,” “virus” and “vector” are often used interchangeably.
  • expression refers to the process by which a polypeptide is produced based on the encoding sequence of a nucleic acid molecule, such as a gene.
  • the process may include transcription, post-transcriptional control, post-transcriptional modification, translation, post-translational control, post-translational modification, or any combination thereof.
  • the term “introduced” in the context of inserting a nucleic acid molecule into a cell means “transfection”, or “transformation” or “transduction” and includes reference to the incorporation of a nucleic acid molecule into a eukaryotic or prokaryotic cell wherein the nucleic acid molecule may be incorporated into the genome of a cell (e.g., chromosome, plasmid, plastid, or mitochondrial DNA), converted into an autonomous replicon, or transiently expressed (e.g., transfected mRNA).
  • a cell e.g., chromosome, plasmid, plastid, or mitochondrial DNA
  • transiently expressed e.g., transfected mRNA
  • heterologous nucleic acid molecule, construct or sequence refers to a nucleic acid molecule or portion of a nucleic acid molecule that is not native to a host cell, but may be homologous to a nucleic acid molecule or portion of a nucleic acid molecule from the host cell.
  • the source of the heterologous nucleic acid molecule, construct or sequence may be from a different genus or species.
  • a heterologous nucleic acid molecule is added (i.e., is not endogenous or native) to a host cell or host genome by, for example, conjugation, transformation, transfection, electroporation, or the like, wherein the added molecule may integrate into the host genome or exist as extra-chromosomal genetic material (e.g., as a plasmid or other form of self-replicating vector), and may be present in multiple copies.
  • heterologous refers to a non-native enzyme, protein or other activity encoded by a heterologous polynucleotide introduced into the host cell, even if the host cell encodes a homologous protein or activity.
  • heterologous nucleic acid molecule can be introduced into a host cell as separate nucleic acid molecules, as a plurality of individually controlled genes, as a polycistronic nucleic acid molecule, as a single nucleic acid molecule encoding a fusion protein, or any combination thereof.
  • a host cell can be modified to express two or more heterologous nucleic acid molecules encoding desired binding proteins specific for a BRAF V600E antigen peptide (e.g., TCR ⁇ and TCR ⁇ ).
  • heterologous nucleic acid molecules When two or more heterologous nucleic acid molecules are introduced into a host cell, it is understood that the two or more heterologous nucleic acid molecules can be introduced as a single nucleic acid molecule (e.g., on a single vector), on separate vectors, integrated into the host chromosome at a single site or multiple sites, or any combination thereof.
  • the number of referenced heterologous nucleic acid molecules or protein activities refers to the number of encoding nucleic acid molecules or the number of protein activities, not the number of separate nucleic acid molecules introduced into a host cell.
  • the term “endogenous” or “native” refers to a gene, protein, or activity that is normally present in a host cell. Moreover, a gene, protein or activity that is mutated, overexpressed, shuffled, duplicated or otherwise altered as compared to a parent gene, protein or activity is still considered to be endogenous or native to that particular host cell.
  • an endogenous control sequence from a first gene e.g., promoter, translational attenuation sequences
  • heterologous refers to a molecule or activity found in or derived from a host cell, species or strain.
  • a heterologous polynucleotide may be homologous to a native host cell gene, and may optionally have an altered expression level, a different sequence, an altered activity, or any combination thereof.
  • Sequence identity refers to the percentage of amino acid residues in one sequence that are identical with the amino acid residues in another reference polypeptide sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity.
  • the percentage sequence identity values can be generated using the NCBI BLAST2.0 software as defined by Altschul et al. (1997) “Gapped BLAST and PSI-BLAST: a new generation of protein database search programs”, Nucleic Acids Res. 25:3389-3402, with the parameters set to default values.
  • hematopoietic progenitor cell is a cell that can be derived from hematopoietic stem cells or fetal tissue and is capable of further differentiation into mature cells types (e.g., immune system cells).
  • exemplary hematopoietic progenitor cells include those with a CD24 Lo Lin ⁇ CD117 + phenotype or those found in the thymus (referred to as progenitor thymocytes).
  • a host refers to a cell (e.g., T cell) or microorganism targeted for genetic modification with a heterologous nucleic acid molecule to produce a polypeptide of interest (e.g., an anti-BRAF V600E TCR).
  • a host cell may optionally already possess or be modified to include other genetic modifications that confer desired properties related or unrelated to, e.g., biosynthesis of the heterologous protein (e.g., inclusion of a detectable marker; deleted, altered or truncated endogenous TCR; or increased co-stimulatory factor expression).
  • a host cell is a human hematopoietic progenitor cell transduced with a heterologous nucleic acid molecule encoding a TCR ⁇ chain specific for a BRAF V600E antigen peptide.
  • hyperproliferative disorder refers to excessive growth or proliferation as compared to a normal or undiseased cell.
  • exemplary hyperproliferative disorders include tumors, cancers, neoplastic tissue, carcinoma, sarcoma, malignant cells, pre-malignant cells, as well as non-neoplastic or non-malignant hyperproliferative disorders (e.g., adenoma, fibroma, lipoma, leiomyoma, hemangioma, fibrosis, restenosis, as well as autoimmune diseases such as rheumatoid arthritis, osteoarthritis, psoriasis, inflammatory bowel disease, or the like).
  • BRAF also known as B-RAF1, BRAF1, NS7, RAFB1, B-Raf, B-Raf proto-oncogene, and serine/threonine kinase refers to a 766-amino acid protein encoded by the BRAF gene.
  • the transcript sequence for human wild-type BRAF is set forth in NCBI Reference identifier NM_004333.4 (SEQ ID NO:78), and the protein sequence is set forth in NCBI Reference identifier NP_004324.2 (SEQ ID NO:36).
  • BRAF is a member of the Raf kinase family of growth signal transduction protein kinases, and plays a role in regulating the MAP kinase/ERKs signaling pathway, which affects cell division, differentiation, and secretion.
  • BRAF is composed of three conserved domains characteristic of Raf kinases: a Ras-GTP-binding self-regulatory domain; a serine-rich hinge region; and a catalytic kinase domain that phosphorylates a consensus sequence on protein substrates (CR1, CR2, and CR3, respectively). Active B-Raf forms dimers.
  • a mutant form of BRAF kinase comprising a V600E mutation is an oncogenic driver present in numerous neoplastic conditions, including 40% of melanoma cases, 10% of colorectal cancer cases, and 1% of non-small cell lung cancer cases, and confers constitutive signaling that promotes tumor cell growth and survival.
  • Small molecule BRAF inhibitors have some efficacy in melanoma, but resistance evolves by recruitment of alternative signaling pathways without loss of expression of BRAF V600E protein. See Shi et al., Cancer Disc. 4(1):80 (2014).
  • the present disclosure provides a binding protein comprising: (a) a T cell receptor (TCR) ⁇ chain variable (V ⁇ ) domain having a CDR3 amino acid sequence set forth in any one of SEQ ID NOS:29-32, or a CDR3 amino acid sequence set forth in any one of SEQ ID NOS:29-32 with up to five amino acid substitutions, insertions, and/or deletions, and a TCR ⁇ chain variable (V ⁇ ) domain; (b) a V ⁇ domain, and a V ⁇ domain having a CDR3 amino acid sequence as set forth in any one of SEQ ID NOS:33-35, or a CDR3 amino acid sequence set forth in any one of SEQ ID NOS:33-35 with up to five amino acid substitutions, insertions, and/or deletions; or (c) a V ⁇ domain of (a) and a V ⁇ domain of (b), wherein the binding protein is capable of specifically binding to a HLA complex on a cell surface comprising a BRAF peptid
  • V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:29 and the V ⁇ domain comprises the CDR3 amino acid sequence of
  • V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO: 30 and the V ⁇ P domain comprises the CDR3 amino acid sequence of SEQ ID NO:34
  • V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:31 and the V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:34
  • V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:32 and the
  • V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:35
  • the V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:29 and the V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:34
  • the V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:29 and the V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:35
  • the V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:30 and the V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:33
  • the V ⁇ domain comprises the CDR3 amino acid of SEQ ID NO:30 and the V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:35
  • the V ⁇ domain comprises the CDR3 amino acid of SEQ ID NO:31 and the V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:33
  • the V ⁇ domain comprises the CDR3 amino acid sequence of SEQ ID NO:31 and the V ⁇ domain comprises the CDR3 amino acid
  • V ⁇ exons are assembled from different variable and joining gene segments (V-J), and V ⁇ exons are assembled from different variable, diversity, and joining gene segments (V-D-J).
  • V-J variable and joining gene segments
  • V-D-J variable, diversity, and joining gene segments
  • the TCR ⁇ chromosomal locus has 70-80 variable gene segments and 61 joining gene segments.
  • the TCR ⁇ chromosomal locus has 52 variable gene segments, and two separate clusters of each containing a single diversity gene segment, together with six or seven joining gene segments.
  • Functional V ⁇ and V ⁇ gene exons are generated by the recombination of a variable gene segment with a joining gene segment for V ⁇ , and a variable gene segment with a diversity gene segment and a joining gene segment for V ⁇ .
  • V ⁇ and V ⁇ domains each comprise three hypervariable loops, also referred to as complementary determining regions (CDRs) that contact the peptide-MHC complex.
  • CDR1 and CDR2 are encoded within the variable gene segment, whereas CDR3 is encoded by the region spanning the variable and joining segments for V ⁇ , or the region spanning variable, diversity, and joining segments for V ⁇ .
  • CDR3 is significantly more diverse because of the addition and loss of nucleotides during the recombination process.
  • TCR variable domain sequences can be aligned to a numbering scheme (Kabat, Chothia, Enhanced Chothia, and Aho), allowing equivalent residue positions to be annotated and for different molecules to be compared using ANARCI software tool (2016, Bioinformatics 15:298-300).
  • a numbering scheme provides a standardized delineation of framework regions and CDRs in the TCR variable domains.
  • V ⁇ CDR3 amino acid sequence comprises the amino acid sequence set forth in SEQ ID NO:29 and the V ⁇ domain further comprises a CDR1 amino acid sequence and CDR2 amino acid sequence encoded by the polynucleotide sequence set forth in SEQ ID NO:11
  • the V ⁇ CDR3 amino acid sequence comprises the amino acid sequence set forth in SEQ ID NO:30 and the V ⁇ domain further comprises a CDR1 amino acid sequence and CDR2 amino acid sequence encoded by the polynucleotide sequence set forth in SEQ ID NO:12
  • the V ⁇ CDR3 amino acid sequence comprises the amino acid sequence set forth in SEQ ID NO:31 and the V ⁇ domain further comprises a CDR1 amino acid sequence and CDR2 amino acid sequence encoded by the
  • the VP CDR3 amino acid sequence comprises the amino acid sequence set forth in any one of SEQ ID NOS:33-35 and the V ⁇ domain further comprises a CDR1 amino acid sequence and CDR2 amino acid sequence encoded by the polynucleotide sequence set forth in SEQ ID NO:8.
  • Methods of identifying binding pairs of TCR V ⁇ and V ⁇ domains include, for example, those described in PCT Patent Publication No. WO 2016/161273; Redmond et al., 2016, Genome Med. 8: 80; Munson et al., 2016, Proc. Natl. Acad. Sci. 113:8272-7; Kim et al., 2012, PLoS ONE 7:e37338 (each of the methods from which are incorporated by reference in its entirety). Accordingly, a V ⁇ domain for the BRAF V600 E-specific V ⁇ domains described herein (e.g., a V ⁇ domain comprising CDR3 as set forth in any one of SEQ ID NOS:33-35), or vice versa, may be identified.
  • a BRAF V600E -specific binding protein described herein may possess one or more amino acid substitutions, deletions, or insertions relative to a naturally occurring binding protein (e.g., TCR).
  • a naturally occurring binding protein e.g., TCR
  • Conservative substitutions of amino acids are known and may occur naturally or may be introduced when the binding protein or TCR is recombinantly produced.
  • Amino acid substitutions, deletions, and insertions may be introduced into a protein using mutagenesis methods (see, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, 3d ed., Cold Spring Harbor Laboratory Press, NY, 2001).
  • Oligonucleotide-directed site-specific (or segment specific) mutagenesis procedures may be employed to provide an altered polynucleotide that has particular codons altered according to the substitution, deletion, or insertion desired.
  • random or saturation mutagenesis techniques such as alanine scanning mutagenesis, error prone polymerase chain reaction mutagenesis, and oligonucleotide-directed mutagenesis may be used to prepare immunogen polypeptide variants (see, e.g., Sambrook et al., supra).
  • amino acid that is substituted at a particular position in a peptide or polypeptide is conservative (or similar).
  • a similar amino acid or a conservative amino acid substitution is one in which an amino acid residue is replaced with an amino acid residue having a similar side chain.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • amino acids with acidic side chains e.g., aspartic acid, glutamic acid
  • amino acids with uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, histidine
  • amino acids with nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • amino acids with beta-branched side chains e.g., threonine, valine, isoleucine
  • amino acids with aromatic side chains e.g., tyrosine, phenylalanine, tryptophan
  • Proline which is considered more difficult to classify, shares properties with amino acids that have aliphatic side chains (e.g., leucine, valine, isoleucine, and alanine).
  • substitution of glutamine for glutamic acid or asparagine for aspartic acid may be considered a similar substitution in that glutamine and asparagine are amide derivatives of glutamic acid and aspartic acid, respectively.
  • similarity between two polypeptides is determined by comparing the amino acid sequence and conserved amino acid substitutes thereto of the polypeptide to the sequence of a second polypeptide (e.g., using GENEWORKS, Align, the BLAST algorithm, or other algorithms described herein and practiced in the art).
  • the binding protein of the instant disclosure comprises a V ⁇ domain that is at least about 90% (e.g., at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or at least about 99.9%) identical to the amino acid sequence set forth in any one of SEQ ID NOS:1-4, and comprises a VP domain that is at least about 90% (e.g., at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or at least about 99.9%) identical to the amino acid sequence set forth in any one of SEQ ID NOS:5-7, provided that (a) at least three or four of the CDRs have no mutations
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in any one of SEQ ID NOS:1-4. In certain embodiments, the V ⁇ domain comprises or consists of the amino acid sequence set forth in any one of SEQ ID NOS:5-7.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:1 and the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:5.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:1 and the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:6.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:1 and the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:7.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:2 and the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:5.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:2 and the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:6.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:2 and the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:7.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:3 and the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:5.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:3 and the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:6.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:3 and the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:7.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:4 and the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:5.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:4 and the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:6.
  • the V ⁇ domain comprises or consists of the amino acid sequence set forth in SEQ ID NO:4 and the V ⁇ domain comprises or consists of of the amino acid sequence set forth in SEQ ID NO:7.
  • a BRAF v600E -specific binding protein is a TCR, an antigen-binding fragment of a TCR, or a chimeric antigen receptor.
  • a “chimeric antigen receptor” (also called a CAR) is a fusion protein comprising an antigen binding domain (e.g., obtained or derived from an immunoglobulin or immunoglobulin-like molecule, such as an scFv derived from an antibody or TCR specific for a cancer antigen, or an antigen-binding domain obtained or derived from a killer immunoreceptor from an NK cell) linked to a transmembrane domain and one or more intracellular signaling domains (optionally containing co-stimulatory domain(s)) (see, e.g., Sadelain et al., Cancer Discov., 3(4): 388-398, 2013; see also Harris and Kranz, Trends Pharmacol.
  • an antigen binding domain e.g., obtained or derived from an immunoglobulin or immunoglobulin-like molecule, such as an scFv derived from an antibody or TCR specific for a cancer antigen, or an antigen-binding domain obtained or
  • a binding protein comprises a CAR comprising a BRAF V600E -specific TCR binding domain (see, e.g., Walseng et al., Scientific Reports 7:10713, 2017; the TCR CAR constructs and methods of which are hereby incorporated by reference in their entirety).
  • Methods of making CARs are described, for example, in U.S. Pat. Nos. 6,410,319; 7,446,191; U.S. Patent Publication No. 2010/065818; U.S. Pat. No. 8,822,647; PCT Publication No. WO 2014/031687; U.S. Pat. No. 7,514,537; and Brentjens et al., Clin. Cancer Res. 13:5426, 2007.
  • the antigen-binding fragment of the TCR comprises a single chain TCR (scTCR), which comprises both the TCR V ⁇ and V ⁇ domains TCR, but only a single TCR constant domain (C ⁇ or C ⁇ .
  • scTCR single chain TCR
  • the antigen-binding fragment of the TCR, or chimeric antigen receptor is chimeric (e.g., comprises amino acid residues or motifs from more than one donor or species), humanized (e.g., comprises residues from a non-human organism that are altered or substituted so as to reduce the risk of immunogenicity in a human), or human.
  • Binding proteins according to the present disclosure may further comprise a TCR constant domain, e.g., joined to the C-terminus of a V ⁇ domain, a V ⁇ domain, or both.
  • a TCR ⁇ -chain constant domain may be encoded by a TRBC1 gene or TRBC2 gene, and a TCR ⁇ -chain may be encoded by a TRAC gene.
  • the TCR comprises an a chain constant (C ⁇ ) domain having at least 90% (e.g., at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or at least about 99.9%) sequence identity to the amino acid sequence set forth in SEQ ID NO:25.
  • the C ⁇ domain comprises the amino acid sequence set forth in SEQ ID NO:25.
  • the TCR comprises a ⁇ chain (C ⁇ ) constant domain having at least 90% (e.g., at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or at least about 99.9%) sequence identity to the amino acid sequence set forth in SEQ ID NO:26.
  • C ⁇ domain comprises the amino acid sequence set forth in SEQ ID NO:26.
  • a binding protein comprises a a-chain (comprised of a V ⁇ domain and a C ⁇ domain) comprising the amino acid sequence set forth in any one of SEQ ID NOS:55-58. In certain embodiments, a binding protein comprises a (3-chain (comprised of a V ⁇ domain and a C ⁇ domain) comprising the amino acid sequence set forth in any one of SEQ ID NOS:59-61.
  • Methods useful for isolating and purifying recombinantly produced soluble binding proteins may include obtaining supernatants from suitable host cell/vector systems that secrete the recombinant soluble TCR into culture media and then concentrating the media using a commercially available filter. Following concentration, the concentrate may be applied to a single suitable purification matrix or to a series of suitable matrices, such as an affinity matrix or an ion exchange resin. One or more reverse phase HPLC steps may be employed to further purify a recombinant polypeptide. These purification methods may also be employed when isolating an immunogen from its natural environment.
  • Methods for large scale production of one or more of the isolated/recombinant soluble TCR described herein include batch cell culture, which is monitored and controlled to maintain appropriate culture conditions. Purification of the soluble TCR may be performed according to methods described herein and known in the art and that comport with laws and guidelines of domestic and foreign regulatory agencies.
  • nucleic acid molecules encoding a binding protein e.g., a TCR
  • a BRAF V600E peptide:HLA complex are used to transfect/transduce a host cell (e.g., a T cell) for use in adoptive transfer therapy.
  • Advances in TCR sequencing have been described (e.g., Robins et al., Blood 114:4099, 2009; Robins et al., Sci. Translat. Med. 2:47ra64, 2010; Robins et al., (September 10) J. Imm. Meth. Epub ahead of print, 2011; Warren et al., Genome Res.
  • T cells 21:790, 2011 may be employed in the course of practicing embodiments according to the present disclosure.
  • methods for transfecting/transducing T cells with desired nucleic acids have been described (e.g., U.S. Patent Application Pub. No. US 2004/0087025) as have adoptive transfer procedures using T cells of desired antigen-specificity (e.g., Schmitt et al., Hum. Gen. 20:1240, 2009; Dossett et al., Mol. Ther. 17:742, 2009; Till et al., Blood 112:2261, 2008; Wang et al., Hum. Gene Ther.
  • the BRAF V600E -specific binding proteins or domains as described herein may be functionally characterized according to any of a large number of art-accepted methodologies for assaying T cell activity, including determination of T cell binding, activation or induction and also including determination of T cell responses that are antigen-specific. Examples include determination of T cell proliferation, T cell cytokine release, antigen-specific T cell stimulation, MHC restricted T cell stimulation, CTL activity (e.g., by detecting 51 Cr release from pre-loaded target cells), changes in T cell phenotypic marker expression, and other measures of T-cell functions. Procedures for performing these and similar assays are may be found, for example, in Lefkovits ( Immunology Methods Manual: The Comprehensive Sourcebook of Techniques, 1998).
  • MHC-peptide tetramer staining refers to an assay used to detect antigen-specific T cells, which features a tetramer of MHC molecules, each comprising an identical peptide having an amino acid sequence that is cognate (e.g., identical or related to) at least one antigen (e.g., BRAF V600E ), wherein the complex is capable of binding T cell receptors specific for the cognate antigen.
  • Each of the MHC molecules may be tagged with a biotin molecule. Biotinylated MHC/peptides are tetramerized by the addition of streptavidin, which can be fluorescently labeled. The tetramer may be detected by flow cytometry via the fluorescent label.
  • an MHC-peptide tetramer assay is used to detect or select enhanced affinity TCRs of the instant disclosure.
  • cytokines may be determined according to methods described herein and practiced in the art, including for example, ELISA, ELISPOT, intracellular cytokine staining, and flow cytometry and combinations thereof (e.g., intracellular cytokine staining and flow cytometry).
  • Immune cell proliferation and clonal expansion resulting from an antigen-specific elicitation or stimulation of an immune response may be determined by isolating lymphocytes, such as circulating lymphocytes in samples of peripheral blood cells or cells from lymph nodes, stimulating the cells with antigen, and measuring cytokine production, cell proliferation and/or cell viability, such as by incorporation of tritiated thymidine or non-radioactive assays, such as MTT assays and the like.
  • lymphocytes such as circulating lymphocytes in samples of peripheral blood cells or cells from lymph nodes
  • stimulating the cells with antigen and measuring cytokine production, cell proliferation and/or cell viability, such as by incorporation of tritiated thymidine or non-radioactive assays, such as MTT assays and the like.
  • Thl cytokines such as IFN- ⁇ , IL-12, IL-2, and TNF- ⁇
  • Type 2 cytokines such as IL-4, IL-5, IL-9, IL-10, and IL-13.
  • compositions comprising a binding protein according to the present disclosure and a pharmaceutically acceptable carrier, diluent, or excipient.
  • pharmaceutically acceptable excipients are biologically compatible vehicles, e.g., physiological saline, which are described in greater detail herein, that are suitable for administration to a human or other non-human mammalian subject.
  • Antigen presentation by immune cells is determined in part by the HLA complexes present on the cells.
  • HLA proteins encoded by different HLA alleles can vary in their ability to present particular antigen peptides and interact with immune cell proteins (e.g., TCRs).
  • a given antigen peptide may be presented by HLA-DQ complexes, but not HLA-DR complexes, or vice versa.
  • a binding protein according to the present disclosure is capable of recognizing a BRAFV 600E peptide complexed with HLA-DQ.
  • the HLA complex comprises HLA-DQB1*0301, *0302, or *0303. In certain embodiments, the HLA complex comprises HLA-DQB1*0302. In further embodiments, the HLA complex comprises HLA-DQA1*03.
  • a BRAF V600E peptide specifically bound by a binding protein of the present disclosure comprises from about 7 to about 27 amino acids, from about 10 to about 25 amino acids, or from about 12 to about 20 amino acids, or from about 15 to about 19 amino acids.
  • the BRAF V600E peptide comprises the amino acid sequence set forth in SEQ ID NO:38 or 39.
  • isolated polynucleotides and expression vectors that encode binding proteins according to the present disclosure are provided. Construction of an expression vector that is used for genetically engineering and producing a binding protein or TCR specific for a BRAF V600E peptide of interest can be accomplished by using any suitable molecular biology engineering techniques known in the art. To obtain efficient transcription and translation, a polynucleotide in each recombinant expression construct includes at least one appropriate expression control sequence (also called a regulatory sequence), such as a leader sequence and particularly a promoter operably (i.e., operatively) linked to the nucleotide sequence encoding the immunogen.
  • a regulatory sequence also called a regulatory sequence
  • binding proteins e.g., TCRs or CARs
  • a nucleic acid may be a single- or a double-stranded DNA, cDNA or RNA in any form, and may include a positive and a negative strand of the nucleic acid which complement each other, including anti-sense DNA, cDNA and RNA.
  • a polynucleotide of the present disclosure can vary (i.e., comprise a different nucleotide sequence) as compared to a reference polynucleotide sequence disclosed herein and still encode a same amino acid or polypeptide, due to, for example, the degeneracy of the genetic code.
  • a polynucleotide encoding for example, a binding protein or a portion thereof, a self-cleaving peptide, a linker peptide; or a binding protein-encoding construct, may have at least about 80% (e.g., at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or at least about 99.9%) identity to a polynucleotide according to any one of SEQ ID NOs:8-24; 27; 28; 44-48; 62-68; and 73-77.
  • a polynucleotide encoding a binding protein of the present disclosure is codon optimized for efficient expression in a target host cell.
  • An exemplary vector may comprise a polynucleotide capable of transporting another polynucleotide to which it has been linked, or which is capable of replication in a host organism.
  • Some examples of vectors include plasmids, viral vectors, cosmids, and others.
  • Some vectors may be capable of autonomous replication in a host cell into which they are introduced (e.g.
  • bacterial vectors having a bacterial origin of replication and episomal mammalian vectors
  • other vectors may be integrated into the genome of a host cell or promote integration of the polynucleotide insert upon introduction into the host cell and thereby replicate along with the host genome (e.g., lentiviral vector, retroviral vector).
  • some vectors are capable of directing the expression of genes to which they are operatively linked (these vectors may be referred to as “expression vectors”).
  • agents e.g., polynucleotides encoding binding proteins or recombinant TCRs specific for BRAF V600E , or variants thereof, as described herein
  • each agent may reside in separate or the same vectors, and multiple vectors (each containing a different agent or the same agent) may be introduced to a cell or cell population or administered to a subject.
  • polynucleotides encoding binding proteins specific for a BRAF V600E peptide:HLA complex may be operatively linked to certain elements of a vector.
  • polynucleotide sequences that are needed to effect the expression and processing of coding sequences to which they are ligated may be operatively linked.
  • Expression control sequences may include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequences);
  • polynucleotides encoding binding proteins of the instant disclosure are contained in an expression vector that is a viral vector, such as a lentiviral vector or a ⁇ -retroviral vector.
  • expression vectors comprising a polynucleotide encoding a binding protein of the present disclosure, wherein the polynucleotide is operably linked to an expression control sequence (e.g., a promoter).
  • the vector is capable of delivering the polynucleotide to a host cell.
  • the host cell is a hematopoietic progenitor cell or a human immune system cell.
  • the immune system cell is a CD4+T cell, a CD8+ T cell, a CD4 ⁇ CD8 ⁇ double negative T cell, a ⁇ T cell, a natural killer cell, a dendritic cell, or any combination thereof.
  • the immune system cell is a CD4+ T cell.
  • the T cell is a naive T cell, a central memory T cell, an effector memory T cell, or any combination thereof.
  • the vector is a viral vector.
  • the viral vector is a lentiviral vector or a y-retroviral vector.
  • host cells comprise a heterologous polynucleotide according to the present disclosure, wherein the host cell expresses on its cell surface a binding protein encoded by the heterologous polynucleotide (i.e., expresses a binding protein according to the present disclosure).
  • an expression vector is delivered to an appropriate cell, for example, a T cell or an antigen-presenting cell, i.e., a cell that displays a peptide/MHC complex on its cell surface (e.g., a dendritic cell).
  • a host cell e.g., a T cell, NK cell, or NK-T cell
  • the encoded binding protein is capable of binding a BRAF V600E antigen:HLA complex in the absence of a CD4 or CD8 co-receptor.
  • the host cell is a hematopoietic progenitor cell or a human immune system cell.
  • the immune system cell can be a CD4 + T cell, a CD8 + T cell, a CD4 ⁇ CD8 ⁇ double negative T cell, a ⁇ T cell, a natural killer cell, a natural killer T cell, a dendritic cell, or any combination thereof.
  • the encoded binding protein comprises a MHCII-restricted TCR binding domain and the host cell (e.g., a CD8 + T cell) comprises a polynucleotide encoding a heterologous CD4 + co-receptor.
  • the T cell can be na ⁇ ve, a central memory T cell, an effector memory T cell, a stem cell memory T cell, or any combination thereof.
  • the T cell is a CD4 + T cell, a CD8 + T cell, or both.
  • the expression vectors introduced into the host cells may also include, for example, lymphoid tissue-specific transcriptional regulatory elements (TREs), such as a B lymphocyte, T lymphocyte, or dendritic cell specific TREs. Lymphoid tissue specific TREs are known in the art (see, e.g., Thompson et al., Mol. Cell. Biol. 12:1043, 1992); Todd et al., J. Exp. Med. 177:1663, 1993); Penix et al., J. Exp. Med. 178:1483, 1993).
  • TREs lymphoid tissue-specific transcriptional regulatory elements
  • a host cell may include any individual cell or cell culture which may receive a vector or the incorporation of nucleic acids or express proteins. The term also encompasses progeny of the host cell, whether genetically or phenotypically the same or different. Suitable host cells may depend on the vector and may include mammalian cells, animal cells, human cells, simian cells, insect cells, yeast cells, and bacterial cells. These cells may be induced to incorporate the vector or other material by use of a viral vector, transformation via calcium phosphate precipitation, DEAE-dextran, electroporation, microinjection, or other methods. See, for example, Sambrook et al., Molecular Cloning: A Laboratory Manual 2d ed. (Cold Spring Harbor Laboratory, 1989).
  • a host cell that comprises a heterologous polynucleotide or an expression vector according to the present disclosure, wherein the host cell expresses on its cell surface a binding protein encoded by the heterologous polynucleotide.
  • the portion of the heterologous polynucleotide that encodes the V ⁇ domain is at least about 80% identical to the polynucleotide sequence set forth in any one of SEQ ID NOS:18-21.
  • the portion of the heterologous polynucleotide that encodes the V ⁇ domain is at least about 80% identical to the polynucleotide sequence set forth in any one of SEQ ID NOS:22-24.
  • the portion of the heterologous polynucleotide that encodes the V ⁇ domain comprises or consists of the polynucleotide sequence set forth in any one of SEQ ID NOS:18-21. In certain embodiments, the portion of the heterologous polynucleotide that encodes the V ⁇ domain comprises or consists of the polynucleotide sequence set forth in any one of SEQ ID NOS:22-24.
  • the portion of the heterologous polynucleotide that encodes the V ⁇ domain comprises or consists of the polynucleotide sequence set forth in any one of SEQ ID NOS:18-21, and the portion of the heterologous polynucleotide that encodes the V ⁇ domain comprises or consists of the polynucleotide sequence set forth in any one of SEQ ID NOS:22-24.
  • the portion of the heterologous polynucleotide that encodes the V ⁇ domain is linked to a portion that encodes a TCR ⁇ -chain constant domain, wherein the portion that encodes the a-chain constant domain comprises or consists of a sequence that is at least about 80% identical to the polynucleotide sequence set forth in SEQ ID NO:27.
  • the portion of the heterologous polynucleotide that encodes the V ⁇ domain is linked to a portion that encodes a TCR ( ⁇ -chain constant domain, wherein the portion that encodes the ( ⁇ -chain constant domain comprises or consists of a sequence that is at least about 80% identical to the polynucleotide sequence set forth in SEQ ID NO:28.
  • the portion of the polynucleotide that encodes the V ⁇ domain comprises or consists of SEQ ID NO:18 and the portion that encodes the V ⁇ domain comprises or consists of SEQ ID NO:22.
  • the portion of the polynucleotide that encodes the V ⁇ domain comprises or consists of SEQ ID NO:19 and the portion that encodes the V ⁇ domain comprises or consists of SEQ ID NO:23.
  • the portion of the polynucleotide that encodes the V ⁇ domain comprises or consists of SEQ ID NO:20 and the portion that encodes the V ⁇ domain comprises or consists of SEQ ID NO:23.
  • the portion of the polynucleotide that encodes the V ⁇ domain comprises or consists of SEQ ID NO:21 and the portion that encodes the V ⁇ domain comprises or consists of SEQ ID NO:24.
  • a host cell e.g., a T cell
  • a BRAF V600E -specific binding protein of the present disclosure is capable of producing an interferon when co-cultured with an antigen-presenting cell presenting or expressing a BRAF V600E -containing antigen.
  • the produced interferon comprises interferon-gamma (IFN- ⁇ ).
  • the target cell has been pulsed with a peptide or polypeptide comprising or consisting of the BRAF V600E -containing antigen.
  • the target cell has been transfected with a polynucleotide (e.g., DNA, cDNA, or mRNA) encoding a polypeptide or peptide comprising or consisting of the BRAF V600E -containing antigen.
  • a polynucleotide e.g., DNA, cDNA, or mRNA
  • a host cell of the present disclosure produces IFN- ⁇ when cultured with an antigen-presenting cell that has been pulsed with a BRAF V600E -containing antigen at a concentration of about 0.005 ⁇ g/mL to about 10 ⁇ g/mL antigen. In further embodiments, the host cell produces at least about 1,000 pg/mL IFN- ⁇ when cultured with an antigen-presenting cell that has been pulsed with a BRAF V600E -containing antigen at a concentration of at least about 0.1 ⁇ g/mL antigen.
  • the host cell produces at least about 1,000 pg/mL IFN- ⁇ when cultured with an antigen-presenting cell pulsed with a BRAF V600E -containing antigen at a concentration of at least about 0.1, 0.2, 0.3, 0.4, or 0.5 ⁇ g/mL antigen. In some embodiments, the host cell produces from about 1,000 pg/mL to about 10,000 pg/mL IFN- ⁇ when cultured with an antigen-presenting cell pulsed with a BRAF V600E -containing antigen at a concentration of about 0.5 ⁇ g/mL antigen to about 10 ⁇ g/mL antigen.
  • a target cell comprises a B cell.
  • the B cell expresses an HLA-DQ allele.
  • the B cell is of B-LCL line 1331.
  • a portion of the polynucleotide encodes a self-cleaving peptide and is disposed between a TCR ⁇ -chain-encoding portion and a TCR ⁇ -chain-encoding portion.
  • Self-cleaving peptides useful for expression of separable polypeptides by a single vector include, for example, Porcine teschovirus-1 2A (P2A) peptide, Thoseaasigna virus 2A (T2A) peptide, Equine rhinitis A virus (ERAV) 2A (E2A) peptide, and Foot-and-Mouth disease virus 2A (F2A) peptide.
  • the portion of the heterologous polynucleotide that encodes the self-cleaving peptide comprises or consists of the polynucleotide sequence set forth in any one of SEQ ID NOS:44-48.
  • the encoded self-cleaving peptide comprises or consists of the amino acid sequence set forth in any one of SEQ ID NOS:49-52.
  • the host cell is a hematopoietic progenitor cell or a human immune system cell.
  • the immune system cell is a CD4 + T cell, a CD8 + T cell, a CD4 ⁇ CD8 ⁇ double negative T cell, a ⁇ T cell, a natural killer cell, a dendritic cell, or any combination thereof.
  • the immune system cell is a T cell.
  • T cell is a na ⁇ ve T cell, a central memory T cell, an effector memory T cell, or any combination thereof.
  • the T cell is a CD4+ T cell.
  • the binding protein or TCR expressed by the T cell is capable of more efficiently associating with a CD3 protein, a CD4 protein, or both, as compared to endogenous TCR. In certain embodiments, the binding protein or TCR higher surface expression on a T cell as compared to endogenous TCR.
  • a host cell that comprises a heterologous polynucleotide encoding a BRAF V600E -specific binding protein is an immune cell which is modified to reduce or eliminate expression of one or more endogenous genes that encode a polypeptide product selected from PD-1, LAG-3, CTLA4, TIM3, TIGIT, an HLA molecule, a TCR molecule, or any component or combination thereof.
  • certain endogenously expressed immune cell proteins may downregulate the immune activity of a modified immune host cell (e.g., PD-1, LAG-3, CTLA4, TIGIT), or may compete with a heterologous binding protein of the present disclosure for expression by the host cell, association with TCR complex components (e.g., CD3 proteins), or may interfere with the binding activity of a heterologously expressed binding protein of the present disclosure (e.g., an endogenous TCR that binds to a non-BRAF V600E antigen or a non-BRAF V600E antigen:HLA complex and interferes with binding of a presently disclosed binding protein to a BRAF V600E antigen) and interferes with the immune host cell binding a target cell that expresses BRAF V600E antigen), or any combination thereof.
  • a modified immune host cell e.g., PD-1, LAG-3, CTLA4, TIGIT
  • TCR complex components e.g., CD3 proteins
  • endogenous proteins e.g., immune host cell proteins, such as an HLA
  • endogenous proteins expressed on a donor immune cell to be used in a cell transfer therapy may be recognized as foreign by an allogeneic recipient, which may result in elimination or suppression of the donor immune cell by the allogeneic recipient.
  • a modified host immune cell is a donor cell (e.g., allogeneic) or an autologous cell.
  • a modified immune host cell of this disclosure comprises a chromosomal gene knockout of one or more of a gene that encodes PD-1, LAG-3, CTLA4, TIM3, TIGIT, an HLA component (e.g., a gene that encodes an ⁇ 1 macroglobulin, an ⁇ 2 macroglobulin, an ⁇ 3 macroglobulin, a ⁇ 1 microglobulin, or a ⁇ 2 microglobulin), or a TCR component (e.g., a gene that encodes a TCR variable region or a TCR constant region) (see, e.g., Torikai et al., Nature Sci. Rep.
  • HLA component e.g., a gene that encodes an ⁇ 1 macroglobulin, an ⁇ 2 macroglobulin, an ⁇ 3 macroglobulin, a ⁇ 1 microglobulin, or a ⁇ 2 microglobulin
  • TCR component e.g., a gene that encodes a TCR variable
  • chromosomal gene knockout refers to a genetic alteration in a host cell that prevents production, by the host cell, of a functionally active endogenous polypeptide product.
  • Alterations resulting in a chromosomal gene knockout can include, for example, introduced nonsense mutations (including the formation of premature stop codons), missense mutations, gene deletion, and strand breaks, as well as the heterologous expression of inhibitory nucleic acid molecules that inhibit endogenous gene expression in the host cell.
  • a chromosomal gene knock-out or gene knock-in is made by chromosomal editing of a host cell. Chromosomal editing can be performed using, for example, endonucleases.
  • endonucleases As used herein “endonuclease” refers to an enzyme capable of catalyzing cleavage of a phosphodiester bond within a polynucleotide chain.
  • an endonuclease is capable of cleaving a targeted gene thereby inactivating or “knocking out” the targeted gene.
  • An endonuclease may be a naturally occurring, recombinant, genetically modified, or fusion endonuclease.
  • the nucleic acid strand breaks caused by the endonuclease are commonly repaired through the distinct mechanisms of homologous recombination or non-homologous end joining (NHEJ).
  • NHEJ non-homologous end joining
  • a donor nucleic acid molecule may be used for a donor gene “knock-in”, for target gene “knock-out”, and optionally to inactivate a target gene through a donor gene knock in or target gene knock out event.
  • NHEJ is an error-prone repair process that often results in changes to the DNA sequence at the site of the cleavage, e.g., a substitution, deletion, or addition of at least one nucleotide. NHEJ may be used to “knock-out” a target gene.
  • endonucleases include zinc finger nucleases, TALE-nucleases, CRISPR-Cas nucleases, meganucleases, and megaTALs.
  • a “zinc finger nuclease” refers to a fusion protein comprising a zinc finger DNA-binding domain fused to a non-specific DNA cleavage domain, such as a Fokl endonuclease.
  • ZFN zinc finger nuclease
  • Each zinc finger motif of about 30 amino acids binds to about 3 base pairs of DNA, and amino acids at certain residues can be changed to alter triplet sequence specificity (see, e.g., Desjarlais et al., Proc. Natl. Acad. Sci.
  • ZFNs mediate genome editing by catalyzing the formation of a site-specific DNA double strand break (DSB) in the genome, and targeted integration of a transgene comprising flanking sequences homologous to the genome at the site of DSB is facilitated by homology directed repair.
  • DSB DNA double strand break
  • a DSB generated by a ZFN can result in knock out of target gene via repair by non-homologous end joining (NHEJ), which is an error-prone cellular repair pathway that results in the insertion or deletion of nucleotides at the cleavage site.
  • NHEJ non-homologous end joining
  • a gene knockout comprises an insertion, a deletion, a mutation or a combination thereof, made using a ZFN molecule.
  • TALEN transcription activator-like effector nuclease
  • TALE is composed of one or more TALE repeat domains/units, each generally having a highly conserved 33-35 amino acid sequence with divergent 12th and 13th amino acids.
  • the TALE repeat domains are involved in binding of the TALE to a target DNA sequence.
  • the divergent amino acid residues referred to as the Repeat Variable Diresidue (RVD), correlate with specific nucleotide recognition.
  • RVD Repeat Variable Diresidue
  • the natural (canonical) code for DNA recognition of these TALEs has been determined such that an HD (histine-aspartic acid) sequence at positions 12 and 13 of the TALE leads to the TALE binding to cytosine (C), NG (asparagine-glycine) binds to a T nucleotide, NI (asparagine-isoleucine) to A, NN (asparagine-asparagine) binds to a G or A nucleotide, and NG (asparagine-glycine) binds to a T nucleotide.
  • Non-canonical (atypical) RVDs are also known (see, e.g., U.S. Patent Publication No.
  • TALENs can be used to direct site-specific double-strand breaks (DSB) in the genome of T cells.
  • Non-homologous end joining (NHEJ) ligates DNA from both sides of a double-strand break in which there is little or no sequence overlap for annealing, thereby introducing errors that knock out gene expression.
  • homology directed repair can introduce a transgene at the site of DSB providing homologous flanking sequences are present in the transgene.
  • a gene knockout comprises an insertion, a deletion, a mutation or a combination thereof, and made using a TALEN molecule.
  • CRISPR/Cas nuclease system refers to a system that employs a CRISPR RNA (crRNA)-guided Cas nuclease to recognize target sites within a genome (known as protospacers) via base-pairing complementarity and then to cleave the DNA if a short, conserved protospacer associated motif (PAM) immediately follows 3′ of the complementary target sequence.
  • CRISPR/Cas systems are classified into three types (i.e., type I, type II, and type III) based on the sequence and structure of the Cas nucleases.
  • the crRNA-guided surveillance complexes in types I and III need multiple Cas subunits.
  • Type II system the most studied, comprises at least three components: an RNA-guided Cas9 nuclease, a crRNA, and a trans-acting crRNA (tracrRNA).
  • the tracrRNA comprises a duplex forming region.
  • a crRNA and a tracrRNA form a duplex that is capable of interacting with a Cas9 nuclease and guiding the Cas9/crRNA:tracrRNA complex to a specific site on the target DNA via Watson-Crick base-pairing between the spacer on the crRNA and the protospacer on the target DNA upstream from a PAM.
  • Cas9 nuclease cleaves a double-stranded break within a region defined by the crRNA spacer. Repair by NHEJ results in insertions and/or deletions which disrupt expression of the targeted locus.
  • a transgene with homologous flanking sequences can be introduced at the site of DSB via homology directed repair.
  • the crRNA and tracrRNA can be engineered into a single guide RNA (sgRNA or gRNA) (see, e.g., Jinek et al., Science 337:816-21, 2012).
  • a gene knockout comprises an insertion, a deletion, a mutation or a combination thereof, and made using a CRISPR/Cas nuclease system.
  • Exemplary gRNA sequences and methods of using the same to knock out endogenous genes that encode immune cell proteins include those described in Ren et al., Clin. Cancer Res. 23(9):2255-2266 (2017), the gRNAs, CAS9 DNAs, vectors, and gene knockout techniques of which are hereby incorporated by reference in their entirety.
  • a gene knockout comprises a CRISPR-mediated gene knockout of a TCR ⁇ -chain constant region locus (C ⁇ , a TCR ⁇ -chain constant region locus (C ⁇ ), or both.
  • a gRNA sequence targeting a TCR C ⁇ locus comprises the nucleotide sequence AGAGTCTCTCAGCTGGTACA (SEQ ID NO:136).
  • a gRNA sequence targeting a TCR C ⁇ locus comprises the nucleotide sequence TGTGCTAGACATGAGGTCTA (SEQ ID NO:137).
  • a gRNA sequence targeting a TCR C ⁇ locus comprises the nucleotide sequence GCAGTATCTGGAGTCATTGA (SEQ ID NO:138). In certain embodiments, a gRNA sequence targeting a TCR C ⁇ locus comprises the nucleotide sequence GGAGAATGACGAGTGGACCC (SEQ ID NO:139). In some embodiments, a gene knockout comprises a CRISPR-mediated gene knockout of a human ⁇ 2M locus. In certain embodiments, a gRNA sequence targeting a human ⁇ 2M comprises the nucleotide sequence CGCGAGCACAGCTAAGGCCA (SEQ ID NO:140).
  • a gene knockout comprises a CRISPR-mediated gene knockout of a PD-1 locus.
  • a gRNA sequence targeting a PD-1 comprises the nucleotide sequence GGCCAGGATGGTTCTTAGGT (SEQ ID NO:141)
  • Exemplary meganucleases include I-SceI, I-CeuI, PI-Pspl, PI-Sce, I-SceIV, I-Csml, I-PanI, I-SceII, I-Ppol, I-SceIII, I-CreI, I-TevI, I-TevII and I-TevIII, whose recognition sequences are known (see, e.g., U.S. Pat. Nos. 5,420,032 and 6,833,252; Belfort et al., Nucleic Acids Res.
  • naturally-occurring meganucleases may be used to promote site-specific genome modification of a target selected from PD-1, LAG3, TIM3, CTLA4, TIGIT, an HLA-encoding gene, or a TCR component-encoding gene.
  • a target selected from PD-1, LAG3, TIM3, CTLA4, TIGIT, an HLA-encoding gene, or a TCR component-encoding gene.
  • an engineered meganuclease having a novel binding specificity for a target gene is used for site-specific genome modification (see, e.g., Porteus et al., Nat. Biotechnol. 23:967-73, 2005; Sussman et al., J. Mol. Biol. 342:31-41, 2004; Epinat et al., Nucleic Acids Res.
  • a chromosomal gene knockout is generated using a homing endonuclease that has been modified with modular DNA binding domains of TALENs to make a fusion protein known as a megaTAL.
  • MegaTALs can be utilized to not only knock-out one or more target genes, but to also introduce (knock in) heterologous or exogenous polynucleotides when used in combination with an exogenous donor template encoding a polypeptide of interest, such as a TCR ⁇ chain, TCR ⁇ chain or both, wherein the knocked-in TCR produced by the cell is specific for a BRAF V600E antigen or peptide.
  • a chromosomal gene knockout comprises an inhibitory nucleic acid molecule that is introduced into a host cell (e.g., an immune cell) comprising a heterologous polynucleotide encoding an antigen-specific receptor that specifically binds to a tumor associated antigen, wherein the inhibitory nucleic acid molecule encodes a target-specific inhibitor and wherein the encoded target-specific inhibitor inhibits endogenous gene expression (i.e., of PD-1, TIM3, LAG3, CTLA4, TIGIT, an HLA component, or a TCR component, or any combination thereof) in the host immune cell.
  • a host cell e.g., an immune cell
  • a heterologous polynucleotide encoding an antigen-specific receptor that specifically binds to a tumor associated antigen
  • the inhibitory nucleic acid molecule encodes a target-specific inhibitor and wherein the encoded target-specific inhibitor inhibits endogenous gene expression (i.e., of PD-1,
  • Chromosomal gene knockout can be confirmed directly by DNA sequencing of the host immune cell following use of the knockout procedure or agent. Chromosomal gene knockouts can also be inferred from the absence of gene expression (e.g., the absence of an mRNA or polypeptide product encoded by the gene) following the knockout.
  • methods of the instant disclosure are for treating a hyperproliferative disorder, wherein the methods comprise administering to human subject in need thereof a composition comprising a binding protein specific for human BRAF V600E or a host cell according to the present disclosure.
  • the instant disclosure is directed to methods for treating a hyperproliferative disorder or a condition characterized by BRAF V600E expression by administering to human subject in need thereof a composition comprising a binding protein or a host cell expressing a binding protein specific for a BRAF V600E peptide:HLA complex according to any the aforementioned binding proteins.
  • the presence of a hyperproliferative disorder or malignant condition in a subject refers to the presence of dysplastic, cancerous and/or transformed cells in the subject, including, for example neoplastic, tumor, non-contact inhibited or oncogenically transformed cells, or the like (e.g., solid cancers; hematologic cancers including lymphomas and leukemias, such as acute myeloid leukemia, chronic myeloid leukemia, etc.
  • BRAF V600E expression including hairy cell leukemia, melanoma, thyroid cancer such as poorly differentiated thyroid cancer, non-small cell lung cancer, colorectal cancer, papillary cancer, non-Hodgkin lymphoma, adenocarcinoma of the lung, and brain tumors including glioblastoma and pilocytic astrocytomas.
  • thyroid cancer such as poorly differentiated thyroid cancer, non-small cell lung cancer, colorectal cancer, papillary cancer, non-Hodgkin lymphoma, adenocarcinoma of the lung, and brain tumors including glioblastoma and pilocytic astrocytomas.
  • an appropriate dose and treatment regimen provide one or more of a binding protein or a BRAF V600E peptide:HLA complex or a host cell expressing the same, and optionally an adjunctive therapy (e.g., a cytokine such as IL-2, IL-15, IL-21 or any combination thereof), in an amount sufficient to provide therapeutic or prophylactic benefit.
  • an adjunctive therapy e.g., a cytokine such as IL-2, IL-15, IL-21 or any combination thereof
  • Therapeutic or prophylactic benefit resulting from therapeutic treatment or prophylactic or preventative methods include, for example an improved clinical outcome, wherein the object is to prevent or retard or otherwise reduce (e.g., decrease in a statistically significant manner relative to an untreated control) an undesired physiological change or disorder, or to prevent, retard or otherwise reduce the expansion or severity of such a disease or disorder.
  • Beneficial or desired clinical results from treating a subject include abatement, lessening, or alleviation of symptoms that result from or are associated the disease or disorder to be treated; decreased occurrence of symptoms; improved quality of life; longer disease-free status (i.e., decreasing the likelihood or the propensity that a subject will present symptoms on the basis of which a diagnosis of a disease is made); diminishment of extent of disease; stabilized (i.e., not worsening) state of disease; delay or slowing of disease progression; amelioration or palliation of the disease state; and remission (whether partial or total), whether detectable or undetectable; or overall survival.
  • Treatment can also mean prolonging survival when compared to expected survival if a subject were not receiving treatment.
  • Subjects in need of the methods and compositions described herein include those who already have the disease or disorder, as well as subjects prone to have or at risk of developing the disease or disorder.
  • Subjects in need of prophylactic treatment include subjects in whom the disease, condition, or disorder is to be prevented (i.e., decreasing the likelihood of occurrence or recurrence of the disease or disorder).
  • the clinical benefit provided by the compositions (and preparations comprising the compositions) and methods described herein can be evaluated by design and execution of in vitro assays, preclinical studies, and clinical studies in subjects to whom administration of the compositions is intended to benefit, as described in the examples.
  • Cells expressing the binding protein as described herein may be administered to a subject in a pharmaceutically or physiologically acceptable or suitable excipient or carrier.
  • Pharmaceutically acceptable excipients are biologically compatible vehicles, e.g., physiological saline, which are described in greater detail herein, that are suitable for administration to a human or other non-human mammalian subject.
  • a therapeutically effective dose in the context of adoptive cell therapy, is an amount of host cells (expressing a binding protein according to the present disclosure) used in adoptive transfer that is capable of producing a clinically desirable result (i.e., a sufficient amount to induce or enhance a specific T cell immune response against cells 600E expressing BRAF v (e.g., a cytotoxic T cell response) in a statistically significant manner) in a treated human or non-human mammal.
  • a clinically desirable result i.e., a sufficient amount to induce or enhance a specific T cell immune response against cells 600E expressing BRAF v (e.g., a cytotoxic T cell response) in a statistically significant manner
  • the dosage for any one patient depends upon many factors, including the patient's size, weight, body surface area, age, the particular therapy to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently.
  • a preferred dose for administration of a host cell comprising a recombinant expression vector as described herein is about 10 7 cells/m 2 , about 5 ⁇ 10 7 cells/m 2 , about 10 8 cells/m 2 , about 5 ⁇ 10 8 cells/m 2 , about 10 9 cells/m 2 , about 5 ⁇ 10 9 cells/m 2 , about 10 10 cells/m 2 , about 5 ⁇ 10 10 cells/m 2 , or about 10 11 cell s/m 2 .
  • a unit dose can comprise T cells, e.g., CD4 + T cells, CD8 + T cells, or both, wherein the T cells can comprise bulk T cells, naive T cells, stem cell memory T cells, central memory T cells, or effector memory T cells.
  • a unit dose comprises BRAF V600E -specific CD4 + T cells and does not comprise CD8 + T cells.
  • a unit dose comprises BRAF V600E -specific CD8 + T cells, which may be engineered to express a heterologous CD4 + co-receptor, and optionally does not comprise CD4 + T cells.
  • a unit dose comprises BRAF V600E -specific CD4 + T host cells of the present disclosure and further comprises CD4 + T cells or CD8 + T cells (e.g., allogeneic or autologous, modified (e.g., to express a heterologous protein such as a TCR, a CAR, a CD4 co-receptor, a CD8 co-receptor, or any combination thereof) or unmodified) that have binding specificity for one or more other antigens or antigen-HLA complexes, such as, for example: a BRAF V600E antigen, a BRAF antigen that does not comprise a V600E mutation; a BRAF V600E -containing antigen that associates with a different HLA than does the binding protein of a presently disclosed host cell in the unit dose; or a different antigen that is associated with hyperproliferative disease or disorder; e.g., NY-ESO-1, SSX-2, Tyrosinase, TMG1-4
  • a unit dose comprises (i) a composition comprising at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, or at least about 95% engineered CD4 + T cells, combined with (ii) a composition comprising at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, or at least about 95% engineered CD8 + T cells, in about a 1:1 ratio, wherein the unit dose contains a reduced amount or substantially no naive T cells (i.e., has less than about 50%, less than about 40%, less than about 30%, less than about 20%, less than about 10%, less than about 5%, or less then about 1% the population of na ⁇ ve T cells present in a unit dose as compared to a patient sample having a comparable number of PBMCs).
  • a unit dose comprises (i) a composition comprising at least about 50% engineered CD4 + T cells, combined with (ii) a composition comprising at least about 50% engineered CD8 + T cells, in about a 1:1 ratio, wherein the unit dose contains a reduced amount or substantially no na ⁇ ve T cells.
  • a unit dose comprises (i) a composition comprising at least about 60% engineered CD4 + T cells, combined with (ii) a composition comprising at least about 60% engineered CD8 + T cells, in about a 1:1 ratio, wherein the unit dose contains a reduced amount or substantially no na ⁇ ve T cells.
  • a unit dose comprises (i) a composition comprising at least about 70% engineered CD4 + T cells, combined with (ii) a composition comprising at least about 70% engineered CD8 + T cells, in about a 1:1 ratio, wherein the unit dose contains a reduced amount or substantially no na ⁇ ve T cells.
  • a unit dose comprises (i) a composition comprising at least about 80% engineered CD4 + T cells, combined with (ii) a composition comprising at least about 80% engineered CD8 + T cells, in about a 1:1 ratio, wherein the unit dose contains a reduced amount or substantially no na ⁇ ve T cells.
  • a unit dose comprises (i) a composition comprising at least about 85% engineered CD4 + T cells, combined with (ii) a composition comprising at least about 85% engineered CD8 + T cells, in about a 1:1 ratio, wherein the unit dose contains a reduced amount or substantially no na ⁇ ve T cells.
  • a unit dose comprises (i) a composition comprising at least about 90% engineered CD4 + T cells, combined with (ii) a composition comprising at least about 90% engineered CD8 + T cells, in about a 1:1 ratio, wherein the unit dose contains a reduced amount or substantially no na ⁇ ve T cells.
  • a unit dose comprises equal, or approximately equal numbers of engineered CD45RA ⁇ CD3 + CD8 + and engineered CD45RA ⁇ CD3 + CD4 + T M cells.
  • compositions may be administered in a manner appropriate to the disease or condition to be treated (or prevented) as determined by persons skilled in the medical art.
  • An appropriate dose and a suitable duration and frequency of administration of the compositions will be determined by such factors as the health condition of the patient, size of the patient (i.e., weight, mass, or body area), the type and severity of the patient's disease, the particular form of the active ingredient, and the method of administration.
  • an appropriate dose and treatment regimen provide the composition(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit (such as described herein, including an improved clinical outcome, such as more frequent complete or partial remissions, or longer disease-free and/or overall survival, or a lessening of symptom severity).
  • a dose should be sufficient to prevent, delay the onset of, or diminish the severity of a disease associated with disease or disorder.
  • Prophylactic benefit of the immunogenic compositions administered according to the methods described herein can be determined by performing pre-clinical (including in vitro and in vivo animal studies) and clinical studies and analyzing data obtained therefrom by appropriate statistical, biological, and clinical methods and techniques, all of which can readily be practiced by a person skilled in the art.
  • a condition associated with BRAF V600E expression includes any disorder or condition in which a BRAF V600E -driven cellular or molecular event is present, and typically manifests in overgrowth of diseased cells relative to normal cells.
  • Some conditions associated with BRAF V600E expression may include acute as well as chronic disorders and diseases, such as those pathological conditions that predispose the subject to a particular disorder.
  • conditions associated with BRAF V600E expression include hyperproliferative disorders, which refer to states of activated and/or proliferating cells (which may also be transcriptionally overactive) in a subject including tumors, neoplasms, cancer, malignancy, etc.
  • the hyperproliferative disorder may also include an aberration or dysregulation of cell death processes, whether by necrosis or apoptosis.
  • Such aberration of cell death processes may be associated with a variety of conditions, including cancer (including primary, secondary malignancies as well as metastasis), or other conditions.
  • a cancer that is characterized by BRAF V600E expression may be treated through the use of compositions and methods disclosed herein.
  • cancer may refer to any accelerated proliferation of cells, including solid tumors, ascites tumors, blood or lymph or other malignancies; connective tissue malignancies; metastatic disease; minimal residual disease following transplantation of organs or stem cells; multi-drug resistant cancers, primary or secondary malignancies, angiogenesis related to malignancy, or other forms of cancer.
  • cancer may refer to any accelerated proliferation of cells, including solid tumors, ascites tumors, blood or lymph or other malignancies; connective tissue malignancies; metastatic disease; minimal residual disease following transplantation of organs or stem cells; multi-drug resistant cancers, primary or secondary malignancies, angiogenesis related to malignancy, or other forms of cancer.
  • specific embodiments wherein only one of the above types of disease is included, or where specific conditions may be excluded regardless of whether or not they are characterized by BRAF V600E expression.
  • Certain methods of treatment or prevention contemplated herein include administering a host cell (which may be autologous, allogeneic or syngeneic) comprising a desired polynucleotide as described herein that is stably integrated into the chromosome of the cell.
  • a host cell which may be autologous, allogeneic or syngeneic
  • a cellular composition may be generated ex vivo using autologous, allogeneic or syngeneic immune system cells (e.g., T cells, antigen-presenting cells, natural killer cells) in order to administer a desired, BRAF V600E -targeted T-cell composition to a subject as an adoptive immunotherapy.
  • the host cell is a hematopoietic progenitor cell or a human immune cell.
  • the immune system cell is a CD4 + T cell, a CD8 + T cell, a CD4 ⁇ CD8 ⁇ double-negative T cell, a ⁇ T cell, a natural killer cell, a natural killer T cell, a dendritic cell, or any combination thereof.
  • the immune system cell is a naive T cell, a central memory T cell, a stem cell memory T cell, an effector memory T cell, or any combination thereof.
  • the cell is a CD4 + T cell.
  • administration of a composition or therapy refers to delivering the same to a subject, regardless of the route or mode of delivery. Administration may be effected continuously or intermittently, and parenterally. Administration may be for treating a subject already confirmed as having a recognized condition, disease or disease state, or for treating a subject susceptible to or at risk of developing such a condition, disease or disease state.
  • Co-administration with an adjunctive therapy may include simultaneous and/or sequential delivery of multiple agents in any order and on any dosing schedule (e.g., BRAF V600E -specific recombinant (i.e., engineered) host cells with one or more cytokines; immunosuppressive therapy such as calcineurin inhibitors, corticosteroids, microtubule inhibitors, low dose of a mycophenolic acid prodrug, or any combination thereof).
  • cytokines e.g., cytokines
  • immunosuppressive therapy such as calcineurin inhibitors, corticosteroids, microtubule inhibitors, low dose of a mycophenolic acid prodrug, or any combination thereof.
  • a plurality of doses of a recombinant host cell as described herein is administered to the subject, which may be administered at intervals between administrations of about two to about four weeks.
  • a cytokine e.g., IL-2, IL-15, IL-21
  • IL-2, IL-15, IL-21 is administered sequentially, provided that the subject was administered the recombinant host cell at least three or four times before cytokine administration.
  • the cytokine is administered concurrently with the host cell.
  • the cytokine is administered subcutaneously.
  • the subject being treated is further receiving immunosuppressive therapy, such as calcineurin inhibitors, corticosteroids, microtubule inhibitors, low dose of a mycophenolic acid prodrug, or any combination thereof.
  • immunosuppressive therapy such as calcineurin inhibitors, corticosteroids, microtubule inhibitors, low dose of a mycophenolic acid prodrug, or any combination thereof.
  • the subject being treated has received a non-myeloablative or a myeloablative hematopoietic cell transplant, wherein the treatment may be administered at least two to at least three months after the non-myeloablative hematopoietic cell transplant.
  • An effective amount of a therapeutic or pharmaceutical composition refers to an amount sufficient, at dosages and for periods of time needed, to achieve the desired clinical results or beneficial treatment, as described herein.
  • An effective amount may be delivered in one or more administrations. If the administration is to a subject already known or confirmed to have a disease or disease-state, the term “therapeutic amount” may be used in reference to treatment, whereas “prophylactically effective amount” may be used to describe administrating an effective amount to a subject that is susceptible or at risk of developing a disease or disease-state (e.g., recurrence) as a preventative course.
  • a disease or disease-state e.g., recurrence
  • the level of a CTL immune response may be determined by any one of numerous immunological methods described herein and routinely practiced in the art.
  • the level of a CTL immune response may be determined prior to and following administration of any one of the herein described BRAF V600E -specific binding proteins expressed by, for example, a T cell.
  • Cytotoxicity assays for determining CTL activity may be performed using any one of several techniques and methods routinely practiced in the art (see, e.g., Henkart et al., “Cytotoxic T-Lymphocytes” in Fundamental Immunology, Paul (ed.) (2003 Lippincott Williams & Wilkins, Philadelphia, Pa.), pages 1127-50, and references cited therein).
  • Antigen-specific T cell responses are typically determined by comparisons of observed T cell responses according to any of the herein described T cell functional parameters (e.g., proliferation, cytokine release, CTL activity, altered cell surface marker phenotype, etc.) that may be made between T cells that are exposed to a cognate antigen in an appropriate context (e.g., the antigen used to prime or activate the T cells, when presented by immunocompatible antigen-presenting cells) and T cells from the same source population that are exposed instead to a structurally distinct or irrelevant control antigen.
  • a cognate antigen e.g., the antigen used to prime or activate the T cells, when presented by immunocompatible antigen-presenting cells
  • a response to the cognate antigen that is greater, with statistical significance, than the response to the control antigen signifies antigen-specificity.
  • a biological sample may be obtained from a subject for determining the presence and level of an immune response to a BRAF V600E -containing antigen peptide as described herein.
  • a “biological sample” as used herein may be a blood sample (from which serum or plasma may be prepared), biopsy specimen, body fluids (e.g., lung lavage, ascites, mucosal washings, synovial fluid), bone marrow, lymph nodes, tissue explant, organ culture, or any other tissue or cell preparation from the subject or a biological source.
  • Biological samples may also be obtained from the subject prior to receiving any immunogenic composition, which biological sample is useful as a control for establishing baseline (i.e., pre-immunization) data.
  • compositions described herein may be presented in unit-dose or multi-dose containers, such as sealed ampoules or vials. Such containers may be frozen to preserve the stability of the formulation until.
  • a unit dose comprises a recombinant host cell as described herein at a dose of about 10 7 cells/m 2 to about 10 11 cells/m 2 .
  • suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens, including e.g., parenteral or intravenous administration or formulation. If the subject composition is administered parenterally, the composition may also include sterile aqueous or oleaginous solution or suspension.
  • Suitable non-toxic parenterally acceptable diluents or solvents include water, Ringer's solution, isotonic salt solution, 1,3-butanediol, ethanol, propylene glycol or polythethylene glycols in mixtures with water.
  • Aqueous solutions or suspensions may further comprise one or more buffering agents, such as sodium acetate, sodium citrate, sodium borate or sodium tartrate.
  • buffering agents such as sodium acetate, sodium citrate, sodium borate or sodium tartrate.
  • any material used in preparing any dosage unit formulation should be pharmaceutically pure and substantially non-toxic in the amounts employed.
  • the active compounds may be incorporated into sustained-release preparation and formulations.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit may contain a predetermined quantity of recombinant cells or active compound calculated to produce the desired therapeutic effect in association with an appropriate pharmaceutical carrier.
  • an appropriate dosage and treatment regimen provides the active molecules or cells in an amount sufficient to provide therapeutic or prophylactic benefit.
  • a response can be monitored by establishing an improved clinical outcome (e.g., more frequent remissions, complete or partial, or longer disease-free survival) in treated subjects as compared to non-treated subjects.
  • Increases in preexisting immune responses to a tumor protein generally correlate with an improved clinical outcome.
  • Such immune responses may generally be evaluated using standard proliferation, cytotoxicity or cytokine assays, which are routine in the art and may be performed using samples obtained from a subject before and after treatment.
  • unit dose forms comprising host cells according to the present disclosure are provided.
  • Methods according to this disclosure may further include administering one or more additional agents to treat the disease or disorder in a combination therapy.
  • a combination therapy comprises administering a BRAF V600E -specific binding protein (or an engineered host cell expressing the same) with (concurrently, simultaneously, or sequentially) an immune checkpoint inhibitor.
  • a combination therapy comprises administering a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) with an agonist of a stimulatory immune checkpoint agent.
  • a combination therapy comprises administering a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) with a secondary therapy, such as chemotherapeutic agent, a radiation therapy, a surgery, an antibody, or any combination thereof.
  • a secondary therapy such as chemotherapeutic agent, a radiation therapy, a surgery, an antibody, or any combination thereof.
  • immune suppression agent refers to one or more cells, proteins, molecules, compounds or complexes providing inhibitory signals to assist in controlling or suppressing an immune response.
  • immune suppression agents include those molecules that partially or totally block immune stimulation; decrease, prevent or delay immune activation; or increase, activate, or up regulate immune suppression.
  • immunosuppression agents to target include PD-1, PD-L1, PD-L2, LAG3, CTLA4, B7-H3, B7-H4, CD244/2B4, HVEM, BTLA, CD160, TIM3, GAL9, KIR, PVR1G (CD112R), PVRL2, adenosine, A2aR, immunosuppressive cytokines (e.g., IL-10, IL-4, IL-1RA, IL-35), IDO, arginase, VISTA, TIGIT, LAIR1, CEACAM-1, CEACAM-3, CEACAM-5, Treg cells, or any combination thereof.
  • immunosuppression agents to target include PD-1, PD-L1, PD-L2, LAG3, CTLA4, B7-H3, B7-H4, CD244/2B4, HVEM, BTLA, CD160, TIM3, GAL9, KIR, PVR1G (CD112R), PVRL2, adenosine, A2
  • An immune suppression agent inhibitor may be a compound, an antibody, an antibody fragment or fusion polypeptide (e.g., Fc fusion, such as CTLA4-Fc or LAG3-Fc), an antisense molecule, a ribozyme or RNAi molecule, or a low molecular weight organic molecule.
  • a method may comprise administering a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) with one or more inhibitor of any one of the following immune suppression components, singly or in any combination.
  • a BRAF V 600E -specific binding protein is used in combination with a PD-1 inhibitor, for example a PD-1-specific antibody or binding fragment thereof, such as pidilizumab, nivolumab (Keytruda, formerly MDX-1106), pembrolizumab (Opdivo, formerly MK-3475), MEDI0680 (formerly AMP-514), AMP-224, BMS-936558 or any combination thereof.
  • a PD-1 inhibitor for example a PD-1-specific antibody or binding fragment thereof, such as pidilizumab, nivolumab (Keytruda, formerly MDX-1106), pembrolizumab (Opdivo, formerly MK-3475), MEDI0680 (formerly AMP-514), AMP-224, BMS-936558 or any combination thereof.
  • a BRAF V600E - specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with a PD-L1 specific antibody or binding fragment thereof, such as BMS-936559, durvalumab (MEDI4736), atezolizumab (RG7446), avelumab (MSB0010718C), MPDL3280A, or any combination thereof.
  • a BRAF V600E -specific binding protein of the present disclosure is used in combination with a LAG3 inhibitor, such as LAG525, IMP321, IMP701, 9H12, BMS-986016, or any combination thereof.
  • a BRAF V 600E -specific binding protein is used in combination with an inhibitor of CTLA4.
  • a BRAF V600E - specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with a CTLA4 specific antibody or binding fragment thereof, such as ipilimumab, tremelimumab, CTLA4-Ig fusion proteins (e.g., abatacept, belatacept), or any combination thereof.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with a B7-H3 specific antibody or binding fragment thereof, such as enoblituzumab (MGA271), 376.96, or both.
  • a B7-H4 antibody binding fragment may be a scFv or fusion protein thereof, as described in, for example, Dangaj et al., Cancer Res. 73:4820, 2013, as well as those described in U.S. Pat. No. 9,574,000 and PCT Patent Publication Nos. WO/201740724A1 and WO 2013/025779A1.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of CD244.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of BLTA, HVEM, CD160, or any combination thereof.
  • Anti CD-160 antibodies are described in, for example, PCT Publication No. WO 2010/084158.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of TIM3.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of Ga19.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of adenosine signaling, such as a decoy adenosine receptor.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of A2aR.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of KIR, such as lirilumab (BMS-986015).
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of an inhibitory cytokine (typically, a cytokine other than TGF ⁇ or Treg development or activity.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an IDO inhibitor, such as levo-1-methyl tryptophan, epacadostat (INCB024360; Liu et al., Blood 115:3520-30, 2010), ebselen (Terentis et al., Biochem. 49:591-600, 2010), indoximod, NLG919 (Mautino et al., American Association for Cancer Research 104th Annual Meeting 2013; Apr 6-10, 2013), 1-methyl-tryptophan (1-MT)-tira-pazamine, or any combination thereof.
  • an IDO inhibitor such as levo-1-methyl tryptophan, epacadostat (INCB024360; Liu et al., Blood 115:3520-30, 2010), ebselen (Terentis et al., Biochem. 49:591-600, 2010), indox
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an arginase inhibitor, such as N(omega)-Nitro-L-arginine methyl ester (L-NAME), N-omega-hydroxy-nor-1-arginine (nor-NOHA), L-NOHA, 2(S)-amino-6-boronohexanoic acid (ABH), S-(2-boronoethyl)-L-cysteine (BEC), or any combination thereof.
  • an arginase inhibitor such as N(omega)-Nitro-L-arginine methyl ester (L-NAME), N-omega-hydroxy-nor-1-arginine (nor-NOHA), L-NOHA, 2(S)-amino-6-boronohexanoic acid (ABH), S-(2-boronoethyl)-L-cysteine (
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of VISTA, such as CA-170 (Curis, Lexington, Mass.).
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of TIGIT such as, for example, COM902 (Compugen, Toronto, Ontario Canada), an inhibitor of CD155, such as, for example, COM701 (Compugen), or both.
  • an inhibitor of TIGIT such as, for example, COM902 (Compugen, Toronto, Ontario Canada)
  • CD155 such as, for example, COM701 (Compugen)
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of PVRIG, PVRL2, or both.
  • Anti-PVRIG antibodies are described in, for example, PCT Publication No. WO 2016/134333.
  • Anti-PVRL2 antibodies are described in, for example, PCT Publication No. WO 2017/021526.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with a LAIR1 inhibitor.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an inhibitor of CEACAM-1, CEACAM-3, CEACAM-5, or any combination thereof.
  • a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) is used in combination with an agent that increases the activity (i.e., is an agonist) of a stimulatory immune checkpoint molecule.
  • a BRAF V600E -specific binding protein of the present disclosure can be used in combination with a CD137 (4-1BB) agonist (such as, for example, urelumab), a CD134 (OX-40) agonist (such as, for example, MEDI6469, MEDI6383, or MEDI0562), lenalidomide, pomalidomide, a CD27 agonist (such as, for example, CDX-1127), a CD28 agonist (such as, for example, TGN1412, CD80, or CD86), a CD40 agonist (such as, for example, CP-870,893, rhuCD40L, or SGN-40), a CD122 agonist (such as, for example, IL-2) an agonist of GITR (such as, for example, humanized monoclonal antibodies described in PCT Patent Publication No.
  • a CD137 (4-1BB) agonist such as, for example, urelumab
  • a CD134 (OX-40) agonist such as
  • a method may comprise administering a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) with one or more agonist of a stimulatory immune checkpoint molecule, including any of the foregoing, singly or in any combination.
  • a combination therapy comprises a BRAF V600E -specific binding protein of the present disclosure (or an engineered host cell expressing the same) and a secondary therapy comprising one or more of: an antibody or antigen binding-fragment thereof that is specific for a cancer antigen expressed by the non-inflamed solid tumor, a radiation treatment, a surgery, a chemotherapeutic agent, a cytokine, RNAi, or any combination thereof.
  • a combination therapy method comprises administering a BRAF V600E -specific binding protein and further administering a radiation treatment or a surgery.
  • Radiation therapy is well-known in the art and includes X-ray therapies, such as gamma-irradiation, and radiopharmaceutical therapies.
  • Surgeries and surgical techniques appropriate to treating a given cancer or non-inflamed solid tumor in a subject are well-known to those of ordinary skill in the art.
  • a combination therapy method comprises administering a BRAF V600F -specific binding protein of the present disclosure (or an engineered host cell expressing the same) and further administering a chemotherapeutic agent.
  • a chemotherapeutic agent includes, but is not limited to, an inhibitor of chromatin function, a topoisomerase inhibitor, a microtubule inhibiting drug, a DNA damaging agent, an antimetabolite (such as folate antagonists, pyrimidine analogs, purine analogs, and sugar-modified analogs), a DNA synthesis inhibitor, a DNA interactive agent (such as an intercalating agent), and a DNA repair inhibitor.
  • Illustrative chemotherapeutic agents include, without limitation, the following groups: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) and purine analogs, folate antagonists and related inhibitors (mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine (cladribine)); antiproliferative/antimitotic agents including natural products such as vinca alkaloids (vinblastine, vincristine, and vinorelbine), microtubule disruptors such as taxane (paclitaxel, docetaxel), vincristin, vinblastin, nocodazole, epothilones and navelbine, epidipodophyllotoxins (etoposide, teniposide), DNA damaging agents (actinomycin, amsacrine, anthracyclines, bleomycin, bus
  • Cytokines are increasingly used to manipulate host immune response towards anticancer activity. See, e.g., Floros & Tarhini, Semin. Oncol. 42(4):539-548, 2015. Cytokines useful for promoting immune anticancer or antitumor response include, for example, IFN- ⁇ , IL-2, IL-3, IL-4, IL-10, IL-12, IL-13, IL-15, IL-16, IL-17, IL-18, IL-21, IL-24, and GM-CSF, singly or in any combination with the binding proteins or cells expressing the same of this disclosure.
  • RNA interference and in particular the use of microRNAs (miRNAs) and small inhibitory RNAs (siRNAs) provides an approach for knocking down expression of cancer genes (see, e.g., Larsson et al., Cancer Treat. Rev. 16(55):128-135, 2017), which can be used in combination with the binding proteins or cells expressing the same of this disclosure.
  • miRNAs microRNAs
  • siRNAs small inhibitory RNAs
  • any of the therapeutic agents may be administered once or more than once to the subject over the course of a treatment, and, in combinations, may be administered to the subject in any order or any combination.
  • An appropriate dose, suitable duration, and frequency of administration of a therapeutic agent will be determined by such factors as a condition of the patient; size, type, spread, growth, and severity of the tumor or cancer; particular form of the active ingredient; and the method of administration.
  • VAF variant allele frequency
  • T cell responses to these potential neoantigens were evaluated by stimulating peripheral blood mononuclear cells (PBMC) obtained from the patient after TIL therapy with a pool of peptides flanking each of the 20 mutations. No CD8 + T cell responses to the candidate neoantigens were detected; however, a CD4 + T cell response specific for 20-mer peptides encompassing BRAF V600E was identified.
  • the BRAF V600E -reactive cells were purified by IFN- ⁇ capture and shown to recognize autologous B cells pulsed with mutant but not wildtype BRAF peptide, confirming specificity for the mutant peptide ( FIG. 1B ).
  • BRAF V600E is processed and presented by class II MHC + APC
  • autologous B cells were transfected with mRNA encoding wildtype or mutant BRAF sequences targeted to the endosome.
  • the T cells recognized B cells expressing mutant, but not wildtype BRAF ( FIG. 1C ).
  • HLA-DQ Recognition was blocked by anti-HLA-DQ but not anti-class I or anti HLA-DR antibodies, identifying HLA-DQ as the likely restricting allele ( FIG. 1D ).
  • Analysis of multiple B cell lines of known genotype suggested restriction by HLA-DQA1*03 paired with HLA-DQB1*03, with weak recognition of DQB1*0301 and stronger recognition of DQB1*0302 and DQB1*0303 ( FIG. 1F and Table 2).
  • BRAF V600E Mean IFN-G Cell line Peptide pg/ml HLA DRB1 HLA DQB1 1331 ⁇ 10 404 302 + 41791 CFS ⁇ 10 0401, 0101 0301, 0501 + 1547 DEM ⁇ 23 0401, 1602 0302, 0502 + 29873 DEU ⁇ 6 401 301 + 10359 FAL ⁇ 9 0403, 0801 03BG, 0402 + 31620 BM14 ⁇ 536 401 302 + 42832 DMB ⁇ 38 0101, 1501 0501, 0602 + 7 DLM ⁇ 26 0403, 0801 03BG, 0402 + 39388 AMM ⁇ 6 0802, 1501 0402, 06WG + 9 CLC ⁇ 9 0301, 1104 02AB, 0301 +
  • the complete patient HLA typing was as follows: A*11:01:01/A*24:02:01; B*15:01:01/B*40:01:02; C*03:03:01/C*03:04:01; DPA1*01:03:01/DPA1*01:03:01; DPB1*04:01:01/DPB1*04:01:01; DQA1*03:01:01/DQA1*03:02; DQB1*03:02:01/DQB1*03:03:02; DRB1*04:03:01/DRB1*09:01:02; DRB4*01:03:01/DRB4*01:03:02.
  • Tumor-specific CD4 + T cells can have anti-tumor activity through direct cell killing and cytokine release (see, e.g., Quezada et al., J. Exp. Med. 207(3):637-650 (2010); Manici et al., J. Exp. Med.
  • CD8 + T cells were the prevalent population in TIL (Table 3). Briefly, the final TIL product infused into the patient was analyzed by flow cytometry for phenotype and, following stimulation with PMA/Ionomycin, was stained intracellularly for cytokines. Percentages shown in Table 3 are percentages of CD45 + cells (top) or CD4 or CD8 cells (bottom)
  • the patient TIL contained BRAF V600E -specific CD4 + T cells and a diverse CD8 + T cell response to self-antigens.
  • the patient was enrolled for TIL generation under an FDA-approved IND and a clinical protocol approved by the Institutional Review Board of Fred Hutchinson
  • TIL tumor fragments in 6,000 IU/ml recombinant IL-2 (Proleukin; Novartis), using methodologies developed at the Surgery Branch of the
  • TIL cultures were selected based on cell growth and autologous tumor reactivity as determined by IFN- ⁇ secretion following co-culture with autologous tumor cells.
  • the TIL were cryopreserved until needed for use, then thawed and further expanded using a rapid expansion protocol, as previously-described (Riddell and Greenberg, J. Immunological Methods 128(2):189-201 (1990)).
  • the expanded TIL were administered to the patient following a lymphodepleting chemotherapy regimen of cyclophosphamide 60mg/kg/day ⁇ 2 days, then fludarabine 25 mg/m 2 /day ⁇ 5 days.
  • the patient received high-dose IL-2 at 600,000 IU/kg IV every 8 hours, for a total of 9 doses.
  • Tumor responses were assessed using RECIST version 1.1 with CT and MRI at weeks 6, 12, and 24, then every 3-6 months, at the discretion of the primary provider.
  • Post-treatment blood was used to isolate non-tumor DNA.
  • a single-cell suspension derived from the iliac nodal tumor recurrence was flow sorted (propidium iodide negative and CD45 negative) to deplete abundant infiltrating lymphocytes and enrich for neoplastic cells.
  • Normal tissue and sorted tumor cells were processed with the Qiagen DNA/RNA AllPrep Micro kit to isolate DNA for exome capture, with RNA reserved for subsequent RNA-seq profiling. Genomic DNA concentration was quantified on an Invitrogen Qubit® 2.0 Fluorometer (Life Technologies-Invitrogen, Carlsbad, Calif., USA) and Trinean DropSense96 spectrophotometer (Caliper Life Sciences, Hopkinton, Mass.).
  • Exome sequencing libraries were prepared using the Agilent SureSelectXT Reagent Kit and exon targets isolated using the Agilent All Human Exon v6 (Agilent Technologies, Santa Clara, Calif., USA). 200 ng of genomic DNA was fragmented using a Covaris LE220 focused-ultrasonicator (Covaris, Inc., Woburn, Mass., USA) and libraries prepared and captured on a Sciclone NGSx Workstation (PerkinElmer, Waltham, Ma., USA). Library size distributions were validated using an Agilent 2200
  • the resulting libraries were sequenced on an Illumina HiSeq 2500 using a paired-end 100bp (PE100) strategy.
  • Image analysis and base calling was performed using Illumina's Real Time Analysis v1.18 software, followed by “demultiplexing” of indexed reads and generation of FASTQ files using Illumina's bcl2fastq Conversion Software v1.8.4 (http://supportillumina.com/downloads/bc12fastq_conversion_software_184. html).
  • Read pairs passing standard Illumina quality filters were retained for further analysis, yielding 77M read pairs for the tumor and 89M read pairs for the normal.
  • Paired reads were aligned to the human genome reference (GRCh37/hg19) with the BWA-MEM short-read aligner (see, e.g., Li, H., arXiv preprint arXiv:1303.3997 (2013); Li and Rudbin, Bioinfomatics 25(14):1754-1760).
  • the resulting alignment files in standard BAM format, were processed by Picard 2.0.1 and GATK 3.5[37] for quality score recalibration, indel realignment, and duplicate removal according to recommended best practices (see, Auwera et al., Current Protocols in Bioinformatics pp. 11.10.1-11.10.33 (2013)).
  • RNA-seq was performed on flow-sorted tumor cells from the same single cell suspension.
  • RNA-seq libraries were prepared from total RNA using the TruSeq RNA Sample Prep v2 Kit (Illumina, Inc., San Diego, Calif., USA) and a Sciclone NGSx Workstation (PerkinElmer, Waltham, Mass., USA). Library size distributions were validated using an Agilent 2200 TapeStation (Agilent Technologies, Santa Clara, Calif., USA). Additional library QC, blending of pooled indexed libraries, and cluster optimization was performed using Life Technologies' Invitrogen Qubit® 2.0 Fluorometer (Life Technologies-Invitrogen, Carlsbad, Calif., USA).
  • the library was sequenced on an Illumina HiSeq 2500 to generate 133M 50nt paired reads (PESO). Reads were aligned to a RefSeq derived reference transcriptome with RSEM 1.2.19 (see Li and Dewer, BMC Bioinformatics 12(1):323 (2011)). Gene-level expression values from RSEM, in TPM units, were added to the summary of missense somatic variants.
  • PBMC peripheral blood mononuclear cells
  • CTL RPMI media with L-glutamine and HEPES (Gibco) supplemented with 10% human serum (produced in house), 50 ⁇ M beta-mercaptoethanol, penicillin and streptomycin, 4 mM L-glutamine and 2 ng/ml recombinant human IL-7 (Peprotech).
  • PBMC peripheral blood mononuclear cells
  • Recombinant IL-2 (Peprotech) was added to a final concentration of 10 U/ml on day +3, and half media changes with supplemental IL-2 were performed on days +3, +6, and +9. On day +13, cells were used in an ELISA and cytokine staining assays.
  • Antigen-specific T cell enrichment was carried out by staining live cells for secreted IFN- ⁇ using the IFN- ⁇ secretion assay APC (Milltenyi) following the manufacturer's instructions, and using autologous B cells as antigen presenting cells pulsed with 10 ⁇ g/m121-mer BRAF mutant peptide.
  • CD4 + IFN- ⁇ secreting cells were sorted on a FACS Aria2. Sorted cells were rested in CTL supplemented with 10 ng/ml human IL-15 for 5 days, then expanded using a rapid expansion protocol described previously (Riddell and Greenberg, supra) .
  • Antigen-specific T cells were further enriched by sorting for V ⁇ 3.1 positive, CD4 + cells by staining with anti-V ⁇ 3.1 (Thermo Scientific, cat. no. TCR2740), expanded, and cryopreserved at day 13 or 14 after expansion. Cryopreserved cells were thawed and rested overnight in CTL supplemented with 10 U/ml IL-2 prior to assays.
  • Autologous B cells were isolated from fresh or thawed PBMC using magnetic beads coated with antibodies recognizing CD19 (Miltenyi, cat. no. 130-050-301) and magnetic positive selection according to the manufacturer's instructions (Miltenyi, cat. no. 130-042-401).
  • Primary B cells were incubated in a 1:1 ratio with NIH 3T3 cells expressing humanCD40L for 7 days in B cell medium supplemented with 200U/ml human IL-4 (Peprotech) as described (see Tran et al., Science 344(3184):641-645 (2014)). B cells were subsequently harvested and restimulated with 3T3 CD40L and fresh medium every 3 days. B cells were used in assays at day +3 of stimulation 2 or 3.
  • ELISA assays 50,000 effector T cells were incubated in 96 well round bottom plates with 100,000 B cells or B-LCL lines and 10 ⁇ gl or specific concentrations of peptides in RPMI (Gibco) supplemented with 5% heat inactivated fetal bovine serum. IFN- ⁇ in supernatants was quantitated using the ready set go human IFN- ⁇ ELISA kit (ebiosciences) in technical triplicate. HLA blocking experiments were carried out with 20 ⁇ gl antibody anti class I (Biolegend, cat. no. 311411) anti-HLA DR (clone L243, cat. no. 307611) and HLA-DQ (Abcam, clone spv-13, cat. no.
  • LCL cell lines 1331, DUCAF, VAVY, BM14, DEM and DEU were utilized.
  • LCL cell lines were pulsed with 10 ⁇ g/ml of BRAF mutant peptide or DMSO control for 4 hours and then washed 3 times with PBS prior to ELISA assay.
  • codon optimized linear DNA fragments encoding HLA-DRB1 0404 protein or the HLA-DQB1 0302 protein linked by a T2A skip sequence to HLA-DQA1 0301 protein were synthesized using GenestringsTM (Life Sciences) and cloned into the vector MP71 (Engels et al., Hum. Gene Ther.
  • Retroviral transduction was performed as described by Sommermeyer et al., ( Leukemia, 2015))into the VAVY cell line homozygous for HLA DRB1 0301, DQA1 0501, and DQB1 0201 (Research cell bank). Cells positive for DRB1 0404 were sorted on a FACSAria2 sorter using the antibody DRB1-PE (Biolegend, cat. no.
  • DQB1 03 DQA1 03 were sorted using the anti DQ antibody clone HLADQ1-FITC (Biolegend, cat. no. 318104). Assays were performed with and without pulsing with BRAF V600E peptide. ELISA experiments were performed in technical duplicate or triplicate as indicated and are representative of two independent experiments.
  • Deep sequencing was performed to identify TCR gene usage in BRAF V600E -specific T cells and other T cells in the TIL.
  • Three TCR clonotypes showed marked expansion after stimulation of post-treatment PBMC with BRAF V600E peptide, and these sequences were further enriched after IFN- ⁇ capture, indicating their specificity for the mutant peptide ( FIG. 2G ).
  • TCR V ⁇ sequencing of tumor, TIL, and PBMC obtained prior to TIL infusion identified all 3 TCR V ⁇ clones in the tumor, and 2 of 3 in TIL. All 3 TCR Vb sequences were below the level of detection in pre-treatment PBMC indicating enrichment at the tumor site ( FIG. 2H ).
  • FIG. 2I A total of 34 common V ⁇ sequences collectively made up >50% of the TIL product ( FIG. 2I ). Only 5 of these 34 clones were detected in the pretreatment blood, with 4 at very low frequency ( FIG. 2J ).
  • IFN- ⁇ capture was used to sort these cells from TIL and assess TCR V ⁇ usage. Seven different V ⁇ sequences in the sorted cells were identified and represented 4.7% of the T cells in the TIL product ( FIGS. 2L-2O ). These 7 clonotypes and one of the BRAF-specific clones were detected in blood obtained 10 and 24 months post-treatment ( FIGS. 2J and 2K ).
  • RNA expression targeted to the endosome was carried out using the method described by Kreiter et al. (I Immunol. 180(1):309-318 (2008)) where antigens are targeted to the endosome by fusion of the antigen to class I MHC sorting signals.
  • the mRNA expression construct pJV57 was constructed by gene synthesis (Geneart, Life Sciences), which contained a T7 promoter fused to the N terminal 25 amino acids of the human HLA-B gene, followed by a BamHI restriction site, the coding sequence of enhanced GFP, an Agel restriction site, the C terminal 55 amino acids of the human HLA-B gene, followed by the human beta globin untranslated region followed by a 30 nucleotide poly A tail followed by a Sapl restriction site directing cleavage in the poly A tail.
  • Construct pJV84 was cloned by ligating the following into AgeI/BamHI digested pJV57: annealed oligonucleotides (Ultramers, Integrated DNA Technologies) encoding BRAF amino acids 575-624 flanked by a 5′ Agel and 3′ BamHI site containing the E600 substitution.
  • Construct pJV85 was made by ligating annealed oligonucleotides (Ultramers, Integrated DNA Technologies) encoding BRAF amino acids 575-624 flanked by a 5′ AgeI and 3′ BamHI site containing the wildtype V600 amino acid.
  • pJV84 and pJV85 were then linearized with SapI (Thermo Fisher) and mRNA was in vitro transcribed using the Highscribe T7 ARCA mRNA kit (New England Biolabs) and purified by lithium precipitation according to the manufacturer's instructions. mRNA was electroporated into CD40L stimulated B cells 16 hours prior to co-culture experiments as described by Tran et al. (supra).
  • BRAF V600E -specific CD4 + T cells showed an effector memory phenotype (CD45RA ⁇ CCR7 ⁇ CD27 ⁇ KLRG1 + ) and expressed low levels of PD-1 ( FIGS. 3A, 3B ).
  • the majority of BRAF V600E -specific cells expressed CXCR3 and CCR4.
  • a fraction of the cells also expressed the skin-homing marker cutaneous lymphocyte-associated antigen (CLA).
  • BRAF V600E peptide-activated cells produced IFN- ⁇ , TNF- ⁇ , IL-4, and IL-21 ( FIG. 3C ), sometimes in combination (data not shown).
  • the HLA-DQA1*03/DQB1*03 restricting allele for the BRAF V600E -specific CD4 + T cells is present in 29% of individuals in the International Histocompatibility Workgroup database (Petersdorf, E., personal communication, International Histocompatibility Working Group in Hematopoietic Cell Transplantation. 2017), and isolation of the BRAF V600E -specific TCR genes from this patient can facilitate adoptive therapy for patients with BRAF mutant tumors with TCR engineered T cells.
  • TCR V ⁇ sequencing on samples with varying levels of BRAF-reactive clones identified four (4) TCR V ⁇ sequences that correlated in frequency with the three (3) TCR V ⁇ sequences ( FIG. 2G ).
  • TCRs from the three (3) TCRB alleles and four (4) TRCA alleles identified were constructed and tested for antigen-specific response ( FIGS. 4A, 4B , and 5).
  • the dominant TCR from the patient (pJV88) was expressed in CD4 + T cells from two healthy donors and conferred specificity to cells expressing BRAF V600E but not wildtype BRAF sequences ( FIG. 2H ).
  • the TCR pJV90 which was made from the second most dominant V ⁇ clone from the patient and one of the possible alpha chains, showed some activity but also possibly nonspecific baseline activation.
  • TCRB sequencing was carried out using the human TCRB sequencing kit (Adaptive Biotechnology) following the manufacturer's instructions and sequenced using a MiSeq (Fred Hutchinson Cancer Research Center Genomics core) with data analysis carried out by Adaptive biotechnology software.
  • TCRA sequencing was carried out using the human TCRA sequencing service (Adaptive Biotechnology).
  • TCR construction was in the vector PRRL (Jones et al., Hum. Gene Ther. 20(6):630-640 (2009)), which was further modified by introducing six point mutations into the start codon and putative promoter region of the woodchuck hepatitis virus X protein as in Lim and Brown, RNA Biology 13(9):743-747 (2016)) with the beta chain followed by a P2A translational skip sequence followed by the alpha chain with cysteines introduced to facilitate pairing (see Kuball et al., Blood 109(6):2331-2338 (2007)).
  • a codon-optimized DNA fragment containing the TRBV28 and CDR3 and TRBJ1-3 sequences followed by TCRB1 sequence with a cysteine substituted at residue 57 followed by a P2A skip sequence and the TRAV21 and CDR3 sequences followed by TRAJ43 and TRAC sequences was synthesized as a genestring (Life Sciences) and cloned using the NEBuilder cloning kit (New England Biolabs) into the vector PRRL-SIN linearized with Pstl and Ascl (Thermo Fisher) and sequence verified.
  • One week after transduction cells were sorted based on Vbeta3.1 expression using antibody clone 8F10 (Thermo Scientific, cat. no. TCR2740) and expanded via rapid expansion as described above. T cells were used in assays or cryopreserved on day 14 of the rapid expansion.
  • the transferred TCR alpha and beta chains may compete with endogenous TCR subunits for expression and signaling machinery.
  • stimulated T cells were first transfected with CRISPR-Cas9 ribonucleoproteins with guide RNA sequences directing cleavage of the endogenous TCR alpha (guide ma AGAGTCTCTCAGCTGGTACA; SEQ ID NO:136) and TCR beta (guide ma: GGAGAATGACGAGTGGACCC; SEQ ID NO:139) constant region genes (from Ren et al., Clin. Cancer Res. 23(9):2255-2266 (2017)).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Medicinal Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Hematology (AREA)
  • Mycology (AREA)
  • Virology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
US16/638,339 2017-08-11 2018-08-10 Braf-specific tcrs and uses thereof Pending US20210238250A9 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/638,339 US20210238250A9 (en) 2017-08-11 2018-08-10 Braf-specific tcrs and uses thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762544695P 2017-08-11 2017-08-11
PCT/US2018/046350 WO2019033057A1 (fr) 2017-08-11 2018-08-10 Tcr de braf spécifiques et utilisations associées
US16/638,339 US20210238250A9 (en) 2017-08-11 2018-08-10 Braf-specific tcrs and uses thereof

Publications (2)

Publication Number Publication Date
US20200223899A1 US20200223899A1 (en) 2020-07-16
US20210238250A9 true US20210238250A9 (en) 2021-08-05

Family

ID=65272504

Family Applications (1)

Application Number Title Priority Date Filing Date
US16/638,339 Pending US20210238250A9 (en) 2017-08-11 2018-08-10 Braf-specific tcrs and uses thereof

Country Status (9)

Country Link
US (1) US20210238250A9 (fr)
EP (1) EP3665191A4 (fr)
JP (2) JP2020533962A (fr)
KR (1) KR20200035104A (fr)
CN (1) CN111212848A (fr)
AU (1) AU2018314242A1 (fr)
CA (1) CA3070855A1 (fr)
MX (1) MX2020001287A (fr)
WO (1) WO2019033057A1 (fr)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109679917B (zh) * 2018-09-30 2020-09-25 北京鼎成肽源生物技术有限公司 一种lrfft2细胞
CN111057690A (zh) * 2019-12-23 2020-04-24 维塔恩(广州)医药有限公司 肿瘤相关基因braf突变相关抗原短肽及其应用
WO2021243695A1 (fr) * 2020-06-05 2021-12-09 Guangdong Tcrcure Biopharma Technology Co., Ltd. Thérapie cellulaire tcr-t ciblant le virus d'epstein-barr
WO2024097637A1 (fr) * 2022-10-31 2024-05-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Récepteurs de lymphocytes t restreints par hla-a3 dirigés contre braf ayant une mutation v600e

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7811993B2 (en) * 2005-06-29 2010-10-12 The Wistar Institute Of Anatomy And Biology Methods and compositions for treating melanoma
WO2013170066A1 (fr) * 2012-05-09 2013-11-14 H. Lee Moffitt Cancer Center & Research Institute, Inc. Peptides pour le traitement du cancer
BR112016014156A8 (pt) * 2013-12-20 2023-04-11 Hutchinson Fred Cancer Res Moléculas efetoras quiméricas etiquetadas e receptores das mesmas
US10202640B2 (en) * 2014-05-07 2019-02-12 The Board Of Trustees Of The Leland Stanford Junior University Single cell analysis of T cells using high-throughput multiplex amplification and deep sequencing
EP3212223B1 (fr) * 2014-10-27 2021-08-18 Fred Hutchinson Cancer Research Center Compositions et procédés pour intensifier l'efficacité de l'immunothérapie cellulaire adoptive
WO2016161273A1 (fr) * 2015-04-01 2016-10-06 Adaptive Biotechnologies Corp. Procédé d'identification des récepteurs de lymphocytes t spécifiques à compatibilité humaine pour une cible antigénique
MA43377A (fr) * 2015-12-04 2021-05-19 Hutchinson Fred Cancer Res Méthodes et compositions liées à la toxicité associée à la thérapie cellulaire
WO2017112944A1 (fr) * 2015-12-23 2017-06-29 Fred Hutchinson Cancer Research Center Récepteurs des cellules t à forte affinité et leurs utilisations

Also Published As

Publication number Publication date
WO2019033057A1 (fr) 2019-02-14
EP3665191A1 (fr) 2020-06-17
CA3070855A1 (fr) 2019-02-14
JP2020533962A (ja) 2020-11-26
AU2018314242A1 (en) 2020-02-13
JP2023105045A (ja) 2023-07-28
US20200223899A1 (en) 2020-07-16
CN111212848A (zh) 2020-05-29
KR20200035104A (ko) 2020-04-01
MX2020001287A (es) 2020-08-20
EP3665191A4 (fr) 2021-05-05

Similar Documents

Publication Publication Date Title
US11034748B2 (en) High affinity MAGE-A1-specific TCRs and uses thereof
US20240165232A1 (en) Chimeric receptor proteins and uses thereof
US20210238250A9 (en) Braf-specific tcrs and uses thereof
US20210340201A1 (en) Immunotherapy targeting kras or her2 antigens
US20220160764A1 (en) High avidity wt1 t cell receptors and uses thereof
US20220267403A1 (en) Binding proteins specific for 5t4 and uses thereof
CN113286815B (zh) 间皮素特异性t细胞受体及其在免疫治疗中的应用
US20210252057A1 (en) Merkel cell polyomavirus t antigen-specific tcrs and uses thereof
US20220409661A1 (en) T-cell immunotherapy specific for wt-1
JPWO2020041501A5 (fr)
WO2022076353A1 (fr) Compositions et procédés de traitement de la maladie associée à l'expression de mage-a1
WO2023230014A1 (fr) Protéines de liaison et cellules modifiées spécifiques de néoantigènes et leurs utilisations

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

AS Assignment

Owner name: UNIVERSITY OF WASHINGTON, WASHINGTON

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LEE, SYLVIA;REEL/FRAME:059068/0157

Effective date: 20200319

Owner name: FRED HUTCHINSON CANCER RESEARCH CENTER, WASHINGTON

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VEATCH, JOSHUA;RIDDELL, STANLEY R.;REEL/FRAME:059068/0129

Effective date: 20201113

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