WO2015066262A1 - Méthodes de prévention de toxicité de la thérapie cellulaire adoptive - Google Patents

Méthodes de prévention de toxicité de la thérapie cellulaire adoptive Download PDF

Info

Publication number
WO2015066262A1
WO2015066262A1 PCT/US2014/063037 US2014063037W WO2015066262A1 WO 2015066262 A1 WO2015066262 A1 WO 2015066262A1 US 2014063037 W US2014063037 W US 2014063037W WO 2015066262 A1 WO2015066262 A1 WO 2015066262A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
csf
perforin
cell therapy
expression
Prior art date
Application number
PCT/US2014/063037
Other languages
English (en)
Inventor
Charles L. Sentman
Original Assignee
Trustees Of Dartmouth College
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 Trustees Of Dartmouth College filed Critical Trustees Of Dartmouth College
Publication of WO2015066262A1 publication Critical patent/WO2015066262A1/fr

Links

Classifications

    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/22Colony stimulating factors (G-CSF, GM-CSF)
    • 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
    • C12N2510/00Genetically modified cells

Definitions

  • T cells play important roles in anti-tumor immunity. Genetic modification of T cells with tumor- targeting chimeric antigen receptors (CARs) and adoptive transfer of CAR-modified T cells have emerged as a promising way of treating cancers (Rossig & Brenner (2004) Mol . Ther . 10:5- 18; Ho, et al . (2003) Cancer Cell 3:431-437; Porter, et al . (2011) N. Engl. J. Med. 365:725-733; Kalos, et al . (2011) Sci. Tranal. Med. 3:95ra73; Park, et al . (2011) Trends Biotechnol. 29:550-7) .
  • CARs tumor- targeting chimeric antigen receptors
  • One advantage of this strategy over other approaches is the ability to expand large number of tumor-specific T cells (>10 10 cells) in a relatively short time ( ⁇ 4 weeks) (Sadelain, et al . (2009) Curr. Opin. Immunol. 21:215-223; Sadelain, et al . (2003) Nat. Rev. Cancer 3:35-45) .
  • the functional activities of CAR-modified T cells such as production of Thl cytokines, cytotoxicity and in vivo persistence can be enhanced by integrating the signaling domains of co-stimulatory molecules (such as CD28, 4-1BB and OX40) to CARs (Porter, et al. (2011) supra; Park, et al . (2011) supra) .
  • CARs have been derived from single chain antibody fragment (scFv) against antigens on tumor cells, such as CD19 in B cell lymphoma (Porter, et al . (2011) supra; Carpenito, et al . (2009) Proc. Natl. Acad. Sci . USA 106:3360-65).
  • scFv single chain antibody fragment
  • NK cells attack tumor and virally- infected cells in the absence of major histocompatibility complex (MHC) restriction, utilizing a combination of signals from activating and inhibitory receptors (Lanier (2005) Annu. Rev. Immunol. 23:225-74) .
  • One group of activating NK cell receptors are natural cytotoxicity receptors (NCRs) , which includes NKp46 (NCR1 and CD335) , NKp44 (NCR2 and CD336) and NKp30 (NCR3 and CD337) (Lanier (2005) supra; Moretta, et al . (2006) Semin. Immunol. 18:151-8; Vivier, et al . (2011) Science 331:44-49).
  • NKp30 is a type I transmembrane protein that contains a single Ig-like extracellular domain followed by a short stalk region connected to a transmembrane segment and intracellular domain (Pende, et al. (1999) J. Exp. Med. 190:1505-16). In mice, NKp30 is not expressed since it is a pseudogene (Hollyoake, et al . (2005) Mol. Biol. Evol . 22 :1661-72) .
  • NKp30 receptor When expressed on NK cells, NKp30 receptor as a monomer associates with 0 ⁇ 3 ⁇ and FcRy for signal transduction (Pende, et al . (1999) supra; Delahaye, et al . (2011) Nat. Med. 17:700-707). Two cellular ligands for NKp30 receptor have been identified: BAT3 and B7-H6 (Brandt, et al . (2009) J. Exp. Med. 206:1495-1503; Pogge von Strandmann, et al . (2007) Immunity 27:965-74).
  • BA 3 is a nuclear protein, which is involved in the interaction with P53 and induction of apoptosis after stress such as DNA damage (Pogge von Strandmann, et al . (2007) supra; Sasaki, et al . (2007) Genes Dev. 21:848-861).
  • BAT3 can also be released by immature dendritic cells (iDCs) on the surfaces of exosomes to stimulate NK cells (Simhadri, et al . (2008) PLoS One 3:e3377).
  • iDCs immature dendritic cells
  • B7-H6 is a newly identified B7 family member. Unlike BAT3 , B7-H6 is expressed on the surface of tumor cells, but not most normal cells (Brandt, et al .
  • NKp30 uses both front and back ⁇ -sheets to engage the Ig- like V region of B7-H6 via predominantly hydrophobic interactions (Joyce, et al . (2011) supra; Li, et al . (2011) supra) .
  • NKp30 receptor has been shown to be important in mediating anti -tumor effects in gastrointestinal stromal tumors and lymphoid leukemia (Correia, et al . (2011) supra; Delahaye, et al . (2011) supra).
  • CARs based upon NKp30 which contain the CD28 and/or 003 ⁇ signaling domains, have been shown to be effective in adoptive immunotherapy against B7-H6 + tumor cells in vivo (Zhang, et al . (2012) J. Immunol. 189:2290-99).
  • the present invention provides methods for preventing toxicity of adoptive cell therapy by administering to a subject in need of adoptive cell therapy, a population of cells deficient in the expression or activity of GM-CSF or perforin.
  • toxicity includes a cytokine storm or cytotoxicity toward healthy cells.
  • the adoptive cell therapy is chimeric antigen receptor-bearing lymphocyte therapy, T cell therapy, natural killer cell therapy, gamma/delta T cell therapy, or natural killer T cell therapy.
  • said population of cells can be the result a GM-CSF gene deletion or GM-CSF gene disruption; contact with a microRNA, siRNA, shRNA, or antisense molecule that inhibits the expression of GM-CSF; or alternatively, contact with an antibody that blocks that activity of GM-CSF or GM-CSF receptor.
  • a population of perforin-deficient cells can be the result of a perforin gene deletion or perforin gene disruption; or contact with a microRNA, siRNA, shRNA, or antisense molecule that inhibits the expression of perforin. Kits including reagents for adoptive cell therapy and the preparation of a population of cells deficient in GM-CSF or perforin are also provided.
  • Figures 1A-1B provide data showing the changes in serum cytokines 18 hours after infusion of CAR cells from B6 mice (CH) or from GM-CSF-deficient mice. Control T cells that do not cause illness are shown (WT) . The GM-CSF- deficient CAR cells had many reduced cytokines.
  • Figures 2A-2B provide data showing the changes in serum cytokines 18 hours after infusion of CAR cells from B6 mice (CH) or from perforin-deficient mice (pfp) . Control T cells that do not cause illness are shown (WT) . The perforin-deficient CAR cells had many reduced cytokines.
  • Figure 3 provides data showing the acute weight changes associated with illness.
  • CAR chimeric
  • WT wild-type control
  • Figure 4 provides data showing the "health score” of mice treated with control T cells (WT) or chNKG2D T cells from B6 (CH) , IFNy-deficient , GM-CSF-deficien , or perforin-deficient mice. A score of 1 is healthy and a score of 4 is dead. This analysis indicated that when the chNKG2D T cells were from mice deficient in either GM-CSF or perforin, there was no adverse affect on health score.
  • WT control T cells
  • CH B6
  • Figure 5 illustrates the structure of the LentiCRISPR vector and target sgRNAs (LGH-1, SEQ ID NO : 1 and 2; LGH-2, SEQ ID NO : 3 and 4; LGH-3, SEQ ID NO : 5 and 6; LGH-4, SEQ ID NO : 7 and 8) used to eliminate human GM-CSF.
  • LGH-1 SEQ ID NO : 1 and 2
  • LGH-2 SEQ ID NO : 3 and 4
  • LGH-3 SEQ ID NO : 5 and 6
  • LGH-4 SEQ ID NO : 7 and 8
  • LTR long-terminal repeat
  • psi+ packaging signal
  • RRE rev response element
  • cPPT central polypurine tract
  • sgRNA single guide RNA
  • EFS elongation factor- la short promoter
  • SpCas9 Streptococcus pyogenes Cas9
  • P2A 2A self - cleaving peptide
  • Puro puromycin selection marker
  • WPRE posttranscriptional regulatory element.
  • the present invention provides methods for preventing or reducing toxicity of adoptive cell therapy by administering to a subject in need of adoptive cell therapy a population of cells deficient in the expression or activity of G -CSF or perforin.
  • toxicity is intended to include a cytokine storm or cytotoxicity toward healthy cells, which results from administration of immune cells during adoptive cell therapy.
  • a cytokine storm, cytokine cascade or hypercytokinemia is an immune reaction, which results in hyperrelease of inflammatory mediators, in particular cytokines, in response to stimulation of T cells and macrophages.
  • cytokines inflammatory mediators
  • cells deficient in the expression or activity of GM-CSF can result in a measurable decrease (e.g., a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90% decrease) in the level of cytokines produced in response to adoptive cell therapy.
  • cells deficient in the expression or activity of perforin can result in a measurable decrease (e.g., a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90% decrease) in the number of healthy cells killed in response to adoptive cell therapy.
  • a measurable decrease e.g., a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90% decrease
  • Adoptive immunotherapy refers to the autologous, syngeneic or allogeneic transfer of immune cells capable of mediating an immune response.
  • the present invention has a wide range of applications, including pre-exposure vaccination of individuals with both in vivo- and in vitro- primed T cells, treatment of cancer subjects using tumor- targeted T cell immunotherapy, treatment of bone marrow transplant subjects (for whom opportunistic infections, such as CMV, are problematic and yet amenable to treatment with targeted T cells such as CMV-targeted cytotoxic lymphocytes) , enhancement of conventional vaccination efficacy through T cell adjuvant therapy, treatment of outbreaks of emergent or re-emergent pathogens, etc.
  • the source of the immune cells that are deficient in or lack functional GM-CSF or perforin may be of any kind.
  • immune cells are obtained from a bank of umbilical cord blood, peripheral blood, human embryonic stem cells, draining lymph nodes, or induced pluripotent stem cells, for example.
  • Immune cells of the invention include, but are not limited to T- lymphocytes (T-cells) , naive T cells (T N ) , memory T cells (for example, central memory T cells (TC M ) , effector memory cells (T EM ) / natural killer cells, hematopoietic stem cells and/or pluripotent embryonic/induced stem cells capable of giving rise to therapeutically relevant progeny.
  • individual immune cells of the invention may be CD4+/CD8-, CD4-/CD8+, CD4-/CD8- or CD4+/CD8+.
  • the immune cells may be a mixed population of CD4+/CD8- and CD4-/CD8+ cells or a population of a single clone.
  • CD4+ immune cells of the invention may produce IL-2, ⁇ , TNFa and other effector cytokines when co-cultured in vitro with cells expressing the target antigens (for example CD20+ and/or CD 19+ tumor cells) .
  • CD8 + immune cells of the invention may lyse antigen- specific target cells when co-cultured in vitro with the target cells.
  • immune cells may be any one or more of CD45 A + CD62L + naive cells, CD45RO CD62I7 central memory cells, CD62LT effector memory cells or a combination thereof (Berger, et al . (2009) Curr. Opin. Immunol. 21(2)224-232).
  • immune cells of the invention are deficient in GM-CSF or perforin expression or activity, or lack functional GM-CSF or perforin proteins.
  • An immune cell deficient in GM-CSF or perforin expression or activity or lacking functional GM-CSF or perforin protein refers to a cell that does not express or expresses significantly reduced levels ⁇ e.g., less than 30, 20, 10, or 5% of normal levels) of G -CSP or perforin; a cell in which the activity of GM-CSF or perforin protein has been eliminated or significantly reduced (e.g., less than 30, 20, 10, or 5% of normal activity) ; or a cell in which the expression or activity of the GM-CSF receptor has been eliminated or significantly reduced.
  • GMPage-Colony Stimulating Factor refers to a small naturally occurring glycoprotein with internal disulfide bonds having a molecular weight of approximately 23 kDa . In humans, it is encoded by a gene located within the cytokine cluster on human chromosome 5. The sequence of the human gene and protein are known and available under GENBANK Accession Nos. M11220 and AAA52578, respectively. See, also, Lee et al . (1985) Proc. Natl. Acad. Sci . USA 82:4360-4364.
  • the protein has an N- terminal signal sequence, and a C-terminal receptor binding domain (Rasko & Gough (1994) In: The Cytokine Handbook, Thomson, et al . Academic Press, New York, pages 349-369) . Its three-dimensional structure is similar to that of the interleukins , although the amino acid sequences are not similar.
  • GM-CSF is produced in response to a number of inflammatory mediators present in the hemopoietic environment and at peripheral sites of inflammation.
  • GM-CSF is able to stimulate the production of neutrophilic granulocytes, macrophages, and mixed granulocyte -macrophage colonies from bone marrow cells and can stimulate the formation of eosinophil colonies from fetal liver progenitor cells.
  • GM-CSF can also stimulate some functional activities in mature granulocytes and macrophages and inhibits apoptosis of granulocytes and macrophages . 2014/063037
  • GM-CSFR Gramulocyte macrophage-colony stimulating factor receptor
  • the GM-CSF receptor is a member of the haematopoietin receptor superfamily. It is heterodimeric protein composed of an alpha and a beta subunit . The alpha subunit is highly specific for GM-CSF whereas the beta subunit is shared with other cytokine receptors, including IL3 and IL5. This is reflected in a broader tissue distribution of the beta receptor subunit.
  • GM-CSFRa The alpha subunit, GM-CSFRa, is primarily expressed on myeloid cells and non-haematopoetic cells, such as neutrophils, macrophages, eosinophils, dendritic cells, endothelial cells and respiratory epithelial cells.
  • Full length GM- CSFRa is a 400 amino acid type I membrane glycoprotein that belongs to the type I cytokine receptor family, and is composed of a 22 amino acid signal peptide (positions 1- 22), a 298 amino acid extracellular domain (positions 23- 320) , a transmembrane domain from positions 321-345 and a short 55 amino acid intracellular domain.
  • the signal peptide is cleaved to provide the mature form of GM-CSFRa as a 378 amino acid protein.
  • the sequences of nucleic acids encoding the human GM-CSFR alpha and beta proteins are known and available under GENBA K Accession Nos . M73832 and M59941, respectively.
  • the sequences of the human GM-CSFR alpha and beta proteins are known and available under GENBANK Accession Nos. AAA35909 and AAA18171, respectively. See, also, Raines, et al . (1991) Proc . Natl. Acad. Sci. USA 88:8203-8207.
  • GM-CSF is able to bind with relatively low affinity to the a subunit alone (K d 1-5 nM) but not at all to the ⁇ subunit alone.
  • K d 1-5 nM the presence of both a and ⁇ subunits results in a high affinity ligand-receptor complex (Ka »100 pM) .
  • GM-CSF signaling occurs through its initial binding to the GM- CSFRa chain and then cross - 1 inking with a larger subunit the common ⁇ chain to generate the high affinity interaction, which phosphorylates the JAK-STAT pathway.
  • Perforin or "pore-forming protein (pfp) " is a cytolytic protein found in the granules of Cytotoxic T lymphocytes (CTLs) and NK cells. Upon degranulation, perforin inserts itself into the target cell's plasma membrane, forming a pore.
  • CTLs Cytotoxic T lymphocytes
  • pfp pore-forming protein
  • the sequence of the human gene and protein are known and available under GENBANK Accession Wos. NM_001083116 and NP_001076585 , respectively. The perforin gene has been mapped to chromosome 17 in humans (Shinkai ; et al . (1989) Immunogenetics 30:452-457).
  • exon 1 encodes an untranslated sequence, and the entire protein is encoded by a portion of exon 2 and all of exon 3, which also contains a 3' untranslated region.
  • perforin cDNA encoding human (Lichtenheld & Podack (1989) J " . Immunol. 143:4267-4274) perforin indicates that human perforin is 534 amino acids in length and contains 20 cysteine residues, which are believed to form 10 intra-chain disulphide bonds. The amino terminal 100 residues and the carboxy terminal 150 residues are completely unique to perforin.
  • GM-CSF or perforin expression is inhibited or blocked by, e.g., gene deletion, gene disruption, si NA, shRNA or antisense approaches.
  • GM-CSF or perforin activity is inhibited or blocked by, e.g., a GM-CSF or perforin antagonist or antibody; or in the case of GM-CSF, a GM-CSF receptor antagonist or antibody.
  • the expression of endogenous GM-CSF or perforin is blocked by genetically modifying the immune cell.
  • non-homologous end joining is used to edit the genome.
  • Any suitable protocol to modify the genome of a particular immune cell is useful, although in specific embodiments gene modification is achieved using an engineered nuclease such as a zinc finger nuclease (ZFP) , TALE-nuclease (TALEN) , or CRISPR/Cas nuclease.
  • Engineered nuclease technology is based on the engineering of naturally occurring DNA-binding proteins. For example, engineering of homing endonucleases with tailored DNA-binding specificities has been described, (see, Chames, et al . (2005) Nucleic Acids Res.
  • ZFPs and TALEs have been fused to nuclease domains to create ZFNs and TALENs ; functional entities that are able to recognize their intended nucleic acid target through their engineered (ZFP or TALE) DNA binding domains and cause the DNA to be cut near the ZFP or TALE DNA binding site via the nuclease activity.
  • ZFP or TALE engineered DNA binding domains
  • ZFNs have been used for genome modification in a variety of organisms. See, for example, US 2003/0232410; US 2005/0208489; US 2005/0026157; US 2005/0064474; US 2006/0188987; US 2006/0063231; and WO 07/014275.
  • the present invention may involve any nuclease of interest.
  • nucleases include meganucleases, TALENs and zinc finger nucleases.
  • the nuclease may include heterologous DNA-binding and cleavage domains ⁇ e.g., zinc finger nucleases; TALENs ; meganuclease DNA-binding domains with heterologous cleavage domains) or, alternatively, the DNA-binding domain of a naturally- occurring nuclease may be altered to bind to a selected target site (e.g., a meganuclease that has been engineered to bind to site different than the cognate binding site) .
  • a selected target site e.g., a meganuclease that has been engineered to bind to site different than the cognate binding site
  • the nuclease is a meganuclease (homing endonuclease) .
  • Naturally-occurring meganucleases recognize 15-40 base-pair cleavage sites and are commonly grouped into families: the LAGLIDADG family, the GIY-YIG family, the His-Cyst box family, the HNH family, the PD-(D/E)xK family and the Vsr-like family.
  • Exemplary homing endonucleases include I-Scel, I-Ceul, PI- Sce, I-SceW, I-Csml, I-Panl, I-SceII, I-Ppol, I-SceIII, I- Crel, I-Tevl, I-TevII and I-TevIII, the recognition sequences of which are known in the art. See, US 5,420,032; US 6,833,252; Belfort, et al . (1997) Nucleic Acids Res. 25:3379-3388; Dujon, et al . (1989) Gene 82:115-1 18; Perler, et ⁇ al . (1994) Nucleic Acids Res.
  • ZFNs include a zinc finger protein that has been engineered to bind to a target site in a gene of choice and cleavage domain or a cleavage half -domain.
  • zinc finger binding domains can be engineered to bind to a sequence of choice. See, for example, Beerli, et al . (2002) Nature Biotechnol . 20:135-141; Pabo, et al .
  • An engineered zinc finger binding domain can have a novel binding specificity, compared to a naturally-occurring zinc finger protein. Engineering methods include, but are not limited to, rational design and various types of selection.
  • Rational design includes, for example, using databases including triplet (or quadruplet) nucleotide sequences and individual zinc finger amino acid sequences, in which each triplet or quadruplet nucleotide sequence is associated with one or more amino acid sequences of zinc fingers which bind the particular triplet or quadruplet sequence. See, for example, US 6,453,242 and US 6,534,261.
  • Exemplary selection methods including phage display and two-hybrid systems are disclosed in US 5,789,538; US 5,925,523; US 6,007,988; US 6,013,453; US 6,410,248; US 6,140,466; US 6,200,759; and US 6,242,568; as well as WO 98/37186; WO 98/53057; WO 00/27878; WO 01/88197 and GB 2,338,237.
  • enhancement of binding specificity for zinc finger binding domains has been described, for example, in WO 02/077227.
  • zinc finger domains and/or multi- fingered zinc finger proteins may be linked together using any suitable linker sequences. See, e.g., US 6,479,626; US 6,903,185; and US 7,153,949 for exemplary linker sequences 6 or more amino acids in length.
  • the CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) /Cas (CRISPR Associated) nuclease system is an engineered nuclease system based on a bacterial system that can be used for genome engineering. It is based on part of the adaptive immune response of many bacteria and archea . When a virus or plasmid invades a bacterium, segments of the invader's DNA are converted into CRISPR RNAs (crRNA) by the 'immune' response.
  • crRNA CRISPR RNAs
  • This crRNA then associates, through a region of partial complementarity, with another type of RNA called tracrRNA to guide the Cas9 nuclease to a region homologous to the crRNA in the target DNA called a "protospacer . "
  • Cas9 cleaves the DNA to generate blunt ends at the double- stranded break at sites specified by a 20-nucleotide guide sequence contained within the crRNA transcript. Cas9 requires both the crRNA and the tracrRNA for site-specific DNA recognition and cleavage.
  • the crRNA and tracrRNA can be combined into one molecule (the "single guide RNA"), and the crRNA equivalent portion of the single guide RNA can be engineered to guide the Cas9 nuclease to target any desired sequence (see, Jinek et al . (2012) Science 337:816-821; Jinek et al . (2013) eLife 2:e00471; Segal (2013) eLife 2:e00563).
  • the CRISPR/Cas system can be engineered to create a double-stranded break at a desired target in a genome, and repair of the double-stranded break can be influenced by the use of repair inhibitors to cause an increase in error prone repair.
  • the DNA binding domain is an engineered domain from a TAL effector similar to those derived from the plant pathogens Xanthomonas (see Boch, et al . (2009) Science 326:1509-1512; Moscou & Bogdanove (2009) Science 326:1501) and Ralstonia (see Heuer, et al (2007) Appl. Environ. Microbiol. 73 (13) : 4379-4384 ; WO
  • Nucleases e.g., ZFNs or TALENs
  • Nuclease expression constructs can be readily designed using methods known in the art. See, e.g., US 2003/0232410; US 2005/0208489; US 2005/0026157; US 2005/0064474; US 2006/0188987; US 2006/0063231 and WO 07/014275.
  • Expression of the nuclease may be under the control of a constitutive promoter or an inducible promoter, for example the galactokinase promoter which is activated (de- repressed) in the presence of raffinose and/or galactose and repressed in presence of glucose.
  • a constitutive promoter or an inducible promoter for example the galactokinase promoter which is activated (de- repressed) in the presence of raffinose and/or galactose and repressed in presence of glucose.
  • the engineered nucleases e.g., ZFNs, TALENs , CRISPR/Cas
  • polynucleotides encoding the same may be delivered to a target cell by any suitable means including, for example, by injection of ZFN, TALEN or CRISPR/Cas proteins or by use of ZFN, TALEN or CRISPR/Cas encoding mRNA.
  • ZFNs, TALENs, and CRISPR/Cas may also be delivered using vectors containing sequences encoding one or more of the ZFNs, TALENs, and CRISPR/Cas protein (s) .
  • Any vector systems may be used including, but not limited to, plasmid vectors, retroviral vectors, lentiviral vectors, adenovirus vectors, poxvirus vectors, herpesvirus vectors and adeno-associated virus vectors, etc. See, e.g., US 6,534,261; US 6,607,882; US 6,824,978; US 6,933,113; US 6,979,539; US 7,013,219; and US 7,163,824. Furthermore, it will be apparent that any of these vectors may include one or more zinc finger or TALEN protein- encoding sequences.
  • each vector may include a sequence encoding one or multiple ZFPs, TALENs or CRISPR/Cas systems.
  • Conventional viral and non-viral based gene transfer methods can be used to introduce nucleic acids encoding engineered ZFPs, TALENs or CRISPR/Cas systems into immune cells in vitro.
  • Non-viral vector delivery systems include DNA plasmids, naked nucleic acid, and nucleic acid complexed with a delivery vehicle such as a liposome or poloxamer, lipofection or electroporation .
  • Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell.
  • a lentivirus is used.
  • the expression of GM-CSF or perforin in immune cells is eliminated or reduced post- transcriptionally, e.g., with an antisense, microRNA, siRNA or shRNA molecule.
  • the expression of GM-CSF or. perforin is reduced by at least 70, 80, 90, 95 or 99% as compared to the expression of GM-CSF or perforin in immune cells not contacted with the above-referenced molecule .
  • the term "antisense” refers to a nucleotide sequence that is complementary to a nucleic acid encoding GM-CSF or perforin, e.g., complementary to the coding strand of the double-stranded cDNA molecule or complementary to the mRNA sequence encoding GM-CSF or perforin.
  • the antisense nucleic acid may be complementary to an entire GM-CSF or perforin coding strand, or to only a portion thereof.
  • the antisense molecule is antisense to a "non-coding region" of the coding strand (e.g., the 5' and 3' untranslated regions) .
  • the antisense molecule can be complementary to the region surrounding the translation start site of GM-CSF or perforin mRNA and can be, for example, about 7, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, or more nucleotides in length.
  • An antisense molecule can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art.
  • an antisense molecule can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecule or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used.
  • the molecule also can be produced biologically using an expression vector.
  • the antisense molecule can be produced in vitro and subsequently contacted with an immune cell, or the immune cell can be transformed or transduced with the expression vector such that the antisense molecule is produced in vivo.
  • GM-CSF or perforin short interfering nucleic acid molecules are provided.
  • siRNA that inhibit the expression of the target gene
  • siRNA can also be used to reduce the level of target gene expression.
  • short interfering RNA or “siRNA” as used herein, refers to any nucleic acid molecule capable of inhibiting or down-regulating GM-CSF or perforin gene expression, for example by mediating RNA interference ("RNAi") or gene silencing in a sequence-specific manner.
  • RNAi RNA interference
  • Chemical modifications can also be applied to any siRNA sequence of the present invention.
  • the siRNA can be a double-stranded molecule including self-complementary sense 3037
  • the siRNA can be assembled from two separate oligonucleotides, where one strand is the sense strand and the other is the antisense strand, wherein the antisense and sense strands are self -complementary (i.e. each strand includes a nucleotide sequence that is complementary to nucleotide sequence in the other strand; such as where the antisense strand and sense strand form a duplex or double stranded structure, for example, wherein the double stranded region is about 19 base pairs) .
  • the siRNA is assembled from a single oligonucleotide, where the self -complementary sense and antisense regions of the siRNA are linked by means of a nucleic acid based or non-nucleic acid-based linker.
  • the siRNA can be a circular single- stranded polynucleotide having two or more loop structures and a stem including self-complementary sense and antisense regions, wherein the circular polynucleotide can be processed either in vivo or in vitro to generate an active siRNA molecule capable of mediating RNAi .
  • siRNA molecules need not be limited to those molecules containing only RNA, but further encompasses molecules including chemically-modified nucleotides or those in combination with non-nucleotides .
  • the siRNA molecule of the invention lacks 2'- hydroxy (2 ' -OH) containing nucleotides.
  • Such siRNA molecules can, however, have an attached linker or linkers or other attached or associated groups, moieties, or chains containing one or more nucleotides with 2 1 -OH groups.
  • siRNA molecules of the invention can include ribonucleotides at about 5, 10, 20, 30, 40, or 50% of the nucleotide positions.
  • the modified siRNA molecules of the invention can also be referred to as short interfering modified oligonucleotides "siMON.” As used herein, the term 2014/063037
  • si NA is preferably meant to be equivalent to other terms used to describe nucleic acid molecules that are capable of mediating sequence specific RNAi , for example double- stranded RNA (dsRNA) , micro-RNA (miRNA) , short hairpin RNA (shRNA) , short interfering oligonucleotide, short interfering nucleic acid, short interfering modified oligonucleotide, chemically-modified siRNA, post- transcriptional gene silencing RNA (ptgsRNA) , translational silencing, and others.
  • dsRNA double- stranded RNA
  • miRNA micro-RNA
  • shRNA short hairpin RNA
  • siNA short interfering oligonucleotide
  • ptgsRNA post- transcriptional gene silencing RNA
  • translational silencing and others.
  • RNAi is preferably meant to be equivalent to other terms used to describe sequence specific RNA interference, such as post -transcriptional gene silencing, or epigenetics.
  • siRNA molecules of the invention can be used to epigenetically silence genes at the post- transcriptional level or the pre-transcriptional level.
  • epigenetic regulation of GM-CSF or perforin gene expression by siRNA molecules of the invention can result from siRNA-mediated modification of the chromatin structure to alter GM-CSF or perforin gene expression.
  • siRNA molecules are known in the art and available from commercial sources such as Santa Cruz Biotech.
  • siRNA molecule 5'- AAGCCCACCCAGAGAAGTGTT-3 ' has been shown to decrease the expression of perforin (Zheng, et al . (2007) Blood 109:2049-2057).
  • the antisense molecule of the present invention is an oi-anomeric nucleic acid molecule.
  • An a-anoraeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual a-units, the strands run parallel to each other (Gaultier, et al . (1987) Nucleic Acids, Res. 15:6625-6641).
  • the antisense nucleic acid molecule can also include a 2 ' -o-methylribonucleotide 14 063037
  • an antisense molecule of the invention is a ribozyme .
  • a ribozyme having specificity for GM-CSF- or perforin-encoding nucleic acid molecules can include one or more sequences complementary to the nucleotide sequence of GM-CSF or perforin cDNA disclosed herein, and a sequence having known catalytic sequence responsible for mRNA cleavage (see US 5,093,246; Haselhoff & Gerlach (1988) Nature 334:585-591).
  • a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in a GM-CSF- or perforin-encoding mRNA (see, e.g., US 4,987,071 or US 5,116,742).
  • GM-CSF or perforin mRNA can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules .
  • GM-CSF or perforin expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the GM- CSF or perforin (e.g., a GM-CSF or perforin promoter and/or enhancers) to form triple helical structures that prevent transcription of the GM-CSF or perforin gene in target cells (see generally, Helene (1991) Anticancer Drug Des. 6 (6) .-569-84; Helene, et al . (1992) Ann. N.Y. Acad. Sci . 660:27-36; Maher (1992) Bioassays 14 (12) : 807-15) .
  • nucleotide sequences complementary to the regulatory region of the GM- CSF or perforin e.g., a GM-CSF or perforin promoter and/or enhancers
  • Switchback molecules are synthesized in an alternating 5' -3', 3' -5' manner, such that they base pair with first one strand of a duplex and then the other, eliminating the necessity for a sizeable stretch of either purines or pyrimidines to be present on one strand of a duplex.
  • the antisense molecules may also be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule.
  • the deoxyribose phosphate backbone of the nucleic acid molecule can be modified to generate peptide nucleic acids (see Hyrup, et al . (1996) Bioorg. Med. Chem. 4:5-23) .
  • peptide nucleic acid refers to a nucleic acid mimic, e.g., a DNA mimic, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained.
  • the neutral backbone of a PNA can allow for specific hybridization to DNA and RNA under conditions of low ionic strength.
  • the synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup et al . (1996) supra; Perry-0 ' Keefe , et al . (1996) Proc. Natl. Acad. Sci. USA 93:14670-675.
  • the antisense and short interfering RNA molecules of the invention can be directly introduced into an immune cell or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding GM-CSF or perforin to thereby inhibit expression of said GM-CSF or perforin, e.g., by inhibiting transcription and/or translation.
  • the molecules can be delivered to cells using vectors, or by viral mechanisms (such as retroviral or adenoviral infection delivery) . To achieve sufficient intracellular concentrations of the molecules, vector constructs in which the molecule is placed under the control of an appropriate promoter.
  • the immune cells of the invention can be modified with, e.g., siRNA, miRNA, shRNA, ribozyme or antisense, by ex vivo treatment with the appropriate molecule to effect inhibition, reduction or blockade of GM-CSF or perforin expression.
  • immune cells such as primary human T cells (e.g., from human peripheral blood mononuclear cells (PBMC) , bone marrow, or umbilical cord blood) are obtained and a viral or non-viral based nucleic acid transfer method (e.g., as described herein) is used to introduce the siRNA, miRNA, shRNA, ribozyme or antisense into immune cells in vitro.
  • PBMC peripheral blood mononuclear cells
  • a viral or non-viral based nucleic acid transfer method e.g., as described herein
  • Stably transfected immune cells can be selected by measuring the expression of GM-CSF or perforin. Immune cells with a significantly reduced level of GM-CSF or perforin as compared to a normal immune cell are considered deficient in GM-CSF or perforin.
  • the present invention also includes the use of GM-CSF or perforin antagonists.
  • An antagonist of GM-CSF or perforin can inhibit one or more of the activities of the naturally occurring form of the polypeptide by, for example, competitively inhibiting GM-CSF- or perforin-mediated activity.
  • biological activity with reference to perforin refers to the cytolytic activity of a perforin polypeptide; that is, its ability to bind to a target cell membrane and polymerize into pore-like transmembrane channels leading to cell lysis.
  • the activity of perforin also includes the capacity to synergize with other toxins such as granule toxins and other molecules to induce apoptosis .
  • the target cell can be any cell that is capable of being lysed by native perforin.
  • biological activity with reference to GM-CSF refers to the stimulatory activity of GM-CSF toward bone marrow cells U 2014/063037
  • the biological activity of perforin can be assessed by the skilled artisan by any number of means known in the art including, but not limited to, the measurement of target cell lysis, the delivery of granzyme B molecules into the target cell, the measurement of target cell membrane disruption (such as by changes in ion transport) , the induction of apoptosis in the target cell, the modification of vesicular trafficking and the general assessment of target cell death.
  • the target cell may be a red blood cell (RBC) and hence a common means of measuring perforin activity is by a RBC lysis test. It may also be any nucleated cell.
  • the biological activity of GM-CSF can be assessed using conventional methods such as measuring bone marrow cell production of, e.g., neutrophilic granulocytes or macrophages.
  • Perforin activity can be inhibited by perforin antagonists such as small organic molecules, polypeptides or antagonistic antibodies.
  • perforin antagonists such as small organic molecules, polypeptides or antagonistic antibodies.
  • benzylidene-2- thioxoimidazolidinone compounds have been shown to selectively inhibit the activity of perforin. See EP 2515903.
  • soluble forms of a perforin molecule capable of binding in competition with endogenous perforin may be used.
  • Such competitors include fragments of the perforin polypeptide that are able to bind native perforin to inhibit its biological activity, but have no inherent perforin activity of their own. See, e.g., WO 2005/083098.
  • a perforin antagonist may also include antibodies or antigen-binding fragments thereof (including, for example, polyclonal, monoclonal, humanized, anti- idiotypic , chimeric or single chain antibodies, and Fab, F(ab') 2 and Fab expression library fragments, scFV molecules, and epitope- 4 063037
  • perforin binding fragments thereof) , or perforin fragments or other small molecules that bind to a native perforin polypeptide and inhibit the biological activity of perforin.
  • GM-CSF activity can be inhibited by GM-CSF antagonists such as antibodies specifically binding to GM- CSF, peptides, or small organic molecules specific for GM- CSF. Also within the meaning of the term GM-CSF antagonist are antibodies specifically binding to the GM-CSF receptor or small organic molecules specific for the GM-CSF receptor.
  • GM-CSF antagonists also refers to non- antibody scaffold molecules, such as fibronectin scaffolds, ankyrins, maxybodies/avimers , protein A-derived molecules, anticalins, affilins, protein epitope mimetics (PEMs) or the like.
  • GM-CSF antagonists selectively interfere with the induction of signaling by the GM-CSF receptor by causing a reduction in the binding of GM-CSF to the receptor.
  • Such antagonists may include antibodies that bind the GM-CSF receptor a or ⁇ subunit, antibodies that bind to GM-CSF, GM-CSF analogs such as E21R, and other proteins or small molecules that compete for binding of GM-CSF to its receptor or inhibit signaling that normally results from the binding of the ligand to the receptor.
  • An example of a neutralizing anti -GM-CSF antibody is the E10 antibody described in Li, et al . (2006) Proc. Natl. Acad. Sci. USA 103 (10) : 3557-3562.
  • E10 is an IgG class antibody that has an 870 pM binding affinity for GM-CSF.
  • the antibody is specific for binding to human GM-CSF as shown in an ELISA assay, and shows strong neutralizing activity as assessed with a TF1 cell proliferation assay.
  • An additional exemplary neutralizing anti -GM-CSF antibody is the MT203- antibody described by Krinner, et al . (2007) Mol . Immunol. 44:916-25.
  • MT203 is an IgGl class antibody that binds GM- CSF with picomolar affinity. The antibody shows potent inhibitory activity as assessed by TF-1 cell proliferation assay and its ability to block IL-8 production in U937 cells.
  • GM-CSF antibodies are described, e.g., in WO 2006/122797.
  • MOR04357 (Morphosys, Martinsried/Planegg, Germany; Steidl, et al . (2008) Mol. Immunol. 46 (1) : 135-44) may also be used as the GM-CSF antagonist.
  • GM-CSF receptor antagonist can be prepared by fusing the coding region of the sGM- CSFR with the CH2-CH3 regions of murine IgG2a.
  • An exemplary soluble GM-CSF receptor is described by Raines, et al. (1991) Proc . Natl. Acad. Sci . USA 88:8203.
  • GM-CSFRoi-Fc fusion protein An example of a GM-CSFRoi-Fc fusion protein is provided, e.g., in Brown, et al . (1995) Blood 85:1488.
  • a peptide corresponding to residues 17-31 of GM-CSF (the A helix of GM-CSF) has been shown to inhibit high affinity receptor binding
  • a second peptide corresponding to residues 54-78 of GM-CSF (the B and C helices of GM-CSF) has been shown to inhibit low affinity receptor binding (VonFeldt, et al (1995) Peptide Res . 8:20-27, 30-32).
  • GM-CSF antagonists include GM-CSF mutants.
  • GM-CSF having a mutation of amino acid residue 21 of GM-CSF to Arginine or Lysine (E21R or E21K) described by Hercus, et al . (1994) Proc. Natl. Acad. Sci USA 91:5838 has been shown to have in vivo activity in preventing dissemination of GM-CSF-dependent leukemia cells in mouse xenograft models (Iversen, et al . (1997) Blood 90:4910).
  • the GM-CSF antagonist may be a peptide.
  • a GM-CSF peptide antagonist may be a peptide designed to structurally mimic the positions of specific residues on the B and C helices of human GM-CSF that are implicated in receptor binding and bioactivity
  • GM-CSF antagonists include antibodies to the GM-CSF receptor alpha chain or beta chain.
  • the GM-CSF receptor antibody for use in the invention may be to the alpha chain.
  • An anti-GM-CSF receptor antibody employed in the invention can be in any antibody format as explained herein, e.g., intact, chimeric, monoclonal, polyclonal, antibody fragment, humanized, humaneered, and the like. Examples of anti-GM- CSF receptor antibodies, e.g., neutralizing, high-affinity antibodies, suitable for use in the invention are known
  • Neutralizing antibodies and other GM-CSF antagonists may be identified using any number of assays that assess GM-CSF function.
  • cell -based assays for GM-CSF receptor signaling such as assays which determine the rate of proliferation of a GM-CSF-dependent cell line in response to a limiting amount of GM-CSF, are conveniently used.
  • the human TF-1 cell line is suitable for use in such an assay.
  • the neutralizing antibodies of the invention inhibit GM-CSF-stimulated TF-1 cell proliferation by at least 50% when a GM-CSF concentration is used which stimulates 90% maximal TF-1 cell proliferation.
  • the neutralizing antibodies inhibit GM-CSF stimulated proliferation by at least 90%.
  • a neutralizing antibody, or other GM-CSF antagonist for use in the invention has an EC 50 of less than 10 nM. Additional assays suitable for use in identifying neutralizing antibodies suitable for use with the present invention will be well known to persons of skill in the art.
  • polyclonal antibodies can be raised in a mammal by one or more injections of an immunizing agent and, if desired, an adjuvant.
  • the immunizing agent includes perforin, GM-CSF or GM-CSF receptor protein, e.g., a human perforin, GM-CSF or GM-CSF receptor protein, or fragment thereof.
  • an antibody for use in the invention is purified from human plasma.
  • the antibody is typically a polyclonal antibody that is isolated from other antibodies present in human plasma. Such an isolation procedure can be performed, e.g., using known techniques, such as affinity chromatography .
  • an antibody of the invention is a monoclonal antibody.
  • Monoclonal antibodies may be prepared using hybridoma methods, such as those described by Kohler & Milstein (1975) Nature 256:495.
  • an immunizing agent such as human perforin, GM-CSF or GM-CSF receptor protein
  • the lymphocytes may be immunized in vitro.
  • the immunizing agent preferably includes human perforin, GM-CSF or GM-CSF receptor protein, fragments thereof, or fusion protein thereof.
  • Human monoclonal antibodies can be produced using various techniques known in the art, including phage display libraries (Hoogenboom & Winter (1991) J. Mol. Biol. 227:381; Marks, et al . (1991) J. Mol. Biol. 222:581). Similarly, human antibodies can be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire. This approach is described, e.g., in US 5,545,807; US 5,545,806; US 5,569,825; US 5,625,126; US 5,633,425; US 5,661,016.
  • the antibodies of the invention are chimeric or humanized monoclonal antibodies.
  • Humanized forms of antibodies are chimeric immunoglobulins in which residues from a complementary determining region (CDR) of human antibody are replaced by residues from a CDR of a non-human species such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • CDR complementary determining region
  • an antibody that is employed in the invention can be in any format.
  • the antibody can be a complete antibody including a constant region, e.g., a human constant region, or can be a fragment or derivative of a complete antibody, e.g. , an Fd, a Fab, Fab', F(ab') 2 , a scFv, an Fv fragment, or a single domain antibody, such as a nanobody or a camel id antibody.
  • Such antibodies may additionally be recombinantly engineered by methods well known to persons of skill in the art. As noted above, such antibodies can be produced using known techniques .
  • the antibody is a humaneered antibody.
  • a humaneered antibody is an engineered human antibody having a binding specificity of a reference antibody, obtained by joining a DNA sequence encoding a binding specificity determinant (BSD) from the CDR3 region of the heavy chain of the reference antibody to human VH segment sequence and a light chain CDR3BSD from the reference antibody to a human VL segment sequence.
  • BSD binding specificity determinant
  • the antibodies suitable for use with the present invention have a high affinity binding for human perforin, GM-CSF or GM-CSF receptor protein.
  • High affinity binding between an antibody and an antigen exists if the dissociation constant (K D ) of the antibody is ⁇ about 10 nM, typically ⁇ 1 nM, and preferably ⁇ 100 pM.
  • the antibody has a dissociation rate of about 10 "4 per second or better.
  • a variety of methods can be used to determine the binding affinity of an antibody for its target antigen such as surface plasmon resonance assays, saturation assays, or immunoassays such as ELISA or RIA, as are well known to persons of skill in the art.
  • An exemplary method for determining binding affinity is by surface plasmon resonance analysis on a BIACORE 2000 instrument (Biacore AB, Freiburg, Germany) using CM5 sensor chips, as described by Krinner, et al . (2007) Mol . Immunol. 44 (5) : 916-25.
  • Immune cells deficient in perforin or GM-CSF expression or activity are of use in reducing the toxicity associated with a number of adoptive immunotherapies including, but not limited to, chimeric antigen receptor- bearing lymphocyte therapy, T cell therapy, natural killer cell therapy, gamma/delta T cell therapy, or natural killer T cell therapy, in particular in the treatment of cancer, an infectious disease or other conditions related to immune-dysfunction, e.g. inflammatory diseases, neuronal disorders, diabetes, and cardiovascular disorders.
  • adoptive immunotherapies including, but not limited to, chimeric antigen receptor- bearing lymphocyte therapy, T cell therapy, natural killer cell therapy, gamma/delta T cell therapy, or natural killer T cell therapy, in particular in the treatment of cancer, an infectious disease or other conditions related to immune-dysfunction, e.g. inflammatory diseases, neuronal disorders, diabetes, and cardiovascular disorders.
  • chimeric antigen receptor (CAR) - bearing lymphocyte therapy includes the use of CAR-modified T cells to recognize tumor cells via binding of the CAR to its tumor-associated antigen, independent of T cell receptor-MHC/peptide interactions. As a result T cells are activated and can efficiently eliminate tumor cells.
  • CAR Chimeric antigen receptor
  • CAR refers to engineered receptors, which graft an antigen specificity onto cells (for example T cells such as naive T cells, central memory T cells, effector memory T cells or combination thereof) .
  • CARs are also known as artificial T- cell receptors, chimeric T-cell receptors or chimeric immunoreceptors .
  • Antigens specific for cancer which may be targeted by the CARs include, but are not limited to, any one or more of 4-IBB, 5T4, adenocarcinoma antigen, alpha- fetoprotein, BAFF, B-lymphoma cell, C242 antigen, CA-125, carbonic anhydrase 9 (CA-IX) , C-MET, CCR4, CD152, CD19, CD20, CD200, CD22, CD221, CD23 (IgE receptor), CD28, CD30 (TNFRSF8) , CD33, CD4 , CD40, CD51, CD52, CD56, CD74, CD80, CEA, CNT0888, CTLA-4, DR5 , EGFR, EpCAM, CD3 , FAP, fibronectin extra domain-B, folate receptor 1, GD2 , GD3 ganglioside, glycoprotein 75, GPNMB, HER2/neu, HGF, human scatter factor receptor kinase,
  • CARs which target the above antigens include but are not limited to bispecific CARs, bispecific CARs co-expressed with EGFRt, bispecific CARs co-expressed with EGFRt and CCR, bispecific CARs co-expressed with EGFRt and DHFR (for example mutant DHFR) or bispecific CARs co-expressed with EGFRt and CDR and DHFR (for example mutant DHFR) .
  • the bispecific chimeric antigen receptors target and bind at least two different antigens.
  • pairings of at least two antigens bound by the bispecific CARs of the invention include but are not limited to CD19 and CD20, CD19 and CD22, CD20 and LI -CAM, LI -CAM and GD2 , EGFR and LI -CAM, EGFR and C-MET, EGFR and HER2, C-MET and HER2 and EGFR and R0R1.
  • Antigens specific for inflammatory diseases which may be targeted by the CARs of the invention include but are not limited to any one or more of AOC3 (VAP-1) , CAM- 3001, CCL11 (eotaxin-1) , CD125, CD147 (basigin) , CD154 (CD40L) , CD2, CD20, CD23 (IgE receptor), CD25 (a chain of IL-2 receptor), CD3 , CD4 , CD5 , IF - ⁇ , IFN- ⁇ , IgE, IgE Fc region, IL-1, IL-12, IL-23, IL-13, IL-17, IL-17A, IL-22, IL-4, IL-5, IL-5, IL-6, IL-6 receptor, integrin a4 , integrin 4 ⁇ 7 LFA-1 (CDlla) , MEDI-528, myostatin, OX-40, rhuMAb ⁇ 7, scleroscin, SOST
  • Antigens specific for neuronal disorders which may be targeted by CARs include but are not limited to any one or more of beta amyloid or MABT5102A.
  • Antigens specific for diabetes which may be targeted by CARs include but are not limited to any one or more of L- ⁇ or CD3.
  • Antigens specific for cardiovascular diseases which may be targeted by CARs include but are not limited to any one or more of 3037
  • cardiac myosin C5
  • cardiac myosin CD41 (integrin alpha-lib)
  • fibrin II beta chain
  • ITGB2 CD18
  • sphingosine-l-phosphate C5
  • cardiac myosin CD41 (integrin alpha-lib)
  • fibrin II beta chain
  • ITGB2 CD18
  • sphingosine-l-phosphate C5
  • cardiac myosin CD41 (integrin alpha-lib)
  • fibrin II fibrin II
  • beta chain beta chain
  • ITGB2 CD18
  • sphingosine-l-phosphate sphingosine-l-phosphate
  • Antigens specific for infectious diseases which may be targeted by CARs include but are not limited to any one or more of anthrax toxin, CCR5 , CD4 , clumping factor A, cytomegalovirus, cytomegalovirus glycoprotein B, endotoxin, Escherichia coli, hepatitis B surface antigen, hepatitis B virus, HIV-1, Hsp90, Influenza A hemagglutinin, lipoteichoic acid, Pseudomonas aeruginosa, rabies virus glycoprotein, respiratory syncytial virus and TNF-a.
  • target antigens include but are not limited to surface proteins found on cancer cells in a specific or amplified fashion (e.g. , the IL- 14 receptor, CD19, CD20 and CD40 for B-cell lymphoma, the Lewis Y and CEA antigens for a variety of carcinomas, the Tag72 antigen for breast and colorectal cancer, EGF-R for lung cancer, folate binding protein and the HER-2 protein which is often amplified in human breast and ovarian carcinomas) , or viral proteins (e.g., gpl20 and gp41 envelope proteins of HIV, envelope proteins from the Hepatitis B and C viruses, the glycoprotein B and other envelope glycoproteins of human cytomegalovirus, the envelope proteins from oncoviruses such as Kaposi's sarcoma-associated Herpes virus).
  • gpl20 and gp41 envelope proteins of HIV envelope proteins from the Hepatitis B and C viruses, the glycoprotein B and other envelope glycoproteins of human cytomegalovirus
  • CARs include CD4 , where the ligand is the HIV gpl20 envelope glycoprotein, and other viral receptors, for example ICAM, which is the receptor for the human rhinovirus, and the related receptor molecule for poliovirus. Additional targets of CARs include antigens involved in B-cell associated diseases.
  • CARs may also include a co-stimulatory domain. This domain may enhance cell proliferation, cell survival and development of memory cells. Co-stimulatory domains include, for example, members of the TNFR super family, CD28, CD137 (4-1BB), CD134 (OX40), DaplO, CD27, CD2 , CD5 , ICAM-1, LFA-1, Lck, TNFR-1, TNFR-II, Fas, CD30, CD40 or combinations thereof. If a CAR includes more than one co- stimulatory domain, these domains may be arranged in tandem, optionally separated by a linker.
  • Nucleic acids encoding CARs can be introduced into immune cells as naked DNA or in a suitable vector.
  • the naked DNA or vector can also include nucleic acids encoding one or more engineered nucleases for genetic modification of the immune cells (as described herein) or nucleic acids encoding, e.g., antisense RNA, siRNA, shRNA, or miRNA to block post- transcriptional expression of GM-CSF, G -CSF receptor, or perforin.
  • Methods of stably transfecting immune cells by electroporation using naked DNA are known in the art. See, e.g., US 6,410,319.
  • Naked DNA generally refers to the DNA encoding a chimeric receptor contained in a plasmid expression vector in proper orientation for expression.
  • the use of naked DNA reduces the time required to produce immune cells expressing the chimeric receptor .
  • a viral vector e.g., a retroviral vector, adenoviral vector, adeno-associated viral vector, or lentiviral vector
  • Suitable vectors include, e.g., non- replicating vectors. A large number of vectors are known which are based on viruses, where the copy number of the virus maintained in the cell is low enough to maintain the viability of the cell.
  • Illustrative vectors include the pFB-neo vectors (STRATAGENE ® ) disclosed herein as well as vectors based on HIV, SV40, EBV, HSV or BPV.
  • the immune cells are introduced or administered to the subject to treat cancer, an infectious disease or other conditions related to immune-dysfunction, e.g., inflammatory diseases, neuronal disorders, diabetes, and cardiovascular disorders.
  • an infectious disease or other conditions related to immune-dysfunction e.g., inflammatory diseases, neuronal disorders, diabetes, and cardiovascular disorders.
  • the immune cells according to the invention can be made into a pharmaceutical composition or made implant -appropriate for administration in vivo, with appropriate carriers or diluents, which further can be pharmaceutically acceptable.
  • suitable carriers or diluents which further can be pharmaceutically acceptable.
  • the means of making such a composition or an implant have been described in the art (see, for instance, Remington's Pharmaceutical Sciences, 16th Ed., Mack, ed.
  • the immune cells can be formulated into a preparation in semisolid or liquid form, such as a capsule, solution, injection, inhalant, or aerosol, in the usual ways for their respective route of administration. Means known in the art can be utilized to prevent or minimize release and absorption of the composition until it reaches the target tissue or organ, or to ensure timed-orelease of the composition. Desirably, however, a pharmaceutically acceptable form is employed which does not ineffectuate the immune cells.
  • the immune cells can be made into a pharmaceutical composition containing a balanced salt solution, preferably Hanks' balanced salt solution, or normal saline.
  • a pharmaceutical composition of the present invention can be used alone or in combination with other well-established agents useful for treating, e.g., cancer or an infectious disease. Whether delivered alone or in combination with other agents, the pharmaceutical composition of the present invention can be delivered via various routes and to various sites in a mammalian, particularly human, body to achieve a particular effect.
  • a particular route can provide a more immediate and more effective reaction than another route.
  • intradermal delivery may be advantageously used over inhalation for the treatment of melanoma.
  • Local- or systemic delivery can be accomplished by administration comprising application or instillation of the formulation into body cavities, inhalation or insufflation of an aerosol, or by parenteral introduction, comprising intramuscular, intravenous, intraportal, intrahepatic, peritoneal, subcutaneous, or intradermal administration.
  • a composition of the present invention can be provided in unit dosage form wherein each dosage unit, e.g., an injection, contains a predetermined amount of the composition, alone or in appropriate combination with other active agents.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of the composition of the present invention, alone or in combination with other active agents, calculated in an amount sufficient to produce the desired effect, in association with a pharmaceutically acceptable diluent, carrier, or vehicle, where appropriate.
  • the specifications for the novel unit dosage forms of the present invention depend on the particular pharmacodynamics associated with the pharmaceutical composition in the particular subject.
  • an effective amount or sufficient number of immune cells is present in the composition and introduced into a subject such that long-term, specific response is established to, e.g., reduce the size of a tumor, eliminate tumor growth or regrowth, or eliminate an infection than would otherwise result in the absence of such treatment.
  • the amount of immune cells introduced into the subject causes a measurable decrease in the signs or symptoms of the disease being treated when compared to otherwise same conditions wherein the immune cells are not present.
  • the amount of immune cells administered should take into account the route of administration and should be such that a sufficient number of the immune cells will be introduced so as to achieve the desired therapeutic response.
  • the amounts of each active agent included in the compositions described herein e.g., the amount per each cell to be contacted or the amount per certain body weight
  • the concentration of immune cells desirably should be sufficient to provide in the subject being treated at least from about 1x10 s to about lxlO 9 immune cells, even more desirably, from about 1x10 7 to about 5x10 8 immune cells, although any suitable amount can be utilized either above, e.g., greater than 5xl0 8 cells, or below, e.g., less than lxlO 7 cells.
  • the dosing schedule can be based on well- established cell-based therapies (see, e.g., Topalian and Rosenberg (1987) Acta Haematol. 78 Suppl 1:75-6; US 4,690,915) or an alternate continuous infusion strategy can be employed.
  • schedule can vary depending on whether the compositions are administered in combination with other pharmaceutical compositions, or depending on interindividual differences in pharmacokinetics, drug disposition, and metabolism.
  • One skilled in the art readily can make any necessary adjustments in accordance with the exigencies of the particular situation.
  • the immune cells of the invention can be used in adoptive cell therapies for the treatment of a variety of cancers .
  • Forms of cancer that may be treated in accordance with this invention include squamous cell carcinoma, leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, Burketts lymphoma, acute or chronic myelogenous leukemias, promyelocytic leukemia, fibrosarcoma, rhabdomyoscarcoma; melanoma, seminoma, teratocarcinoma, neuroblastoma, glioma, astrocytoma, neuroblastoma, glioma, schwannomas; fibrosarcoma, rhabdomyoscaroma , osteosarcoma, melanoma,
  • Immune cells also can be useful in the treatment of other carcinomas of the bladder, breast, colon, kidney, liver, lung, ovary, prostate, pancreas, stomach, cervix, thyroid or skin. Immune cells also may be useful in treatment of other hematopoietic tumors of lymphoid lineage, other hematopoietic tumors of myeloid lineage, other tumors of mesenchymal origin, other tumors of the central or peripheral nervous system, and/or other tumors of mesenchymal origin.
  • the immune cells of the invention also may be useful in reducing cancer progression in prostate cancer cells, melanoma cells (e.g., cutaneous melanoma cells, ocular melanoma cells, and/or lymph node- associated melanoma cells) , breast cancer cells, colon cancer cells, and lung cancer cells.
  • melanoma cells e.g., cutaneous melanoma cells, ocular melanoma cells, and/or lymph node- associated melanoma cells
  • breast cancer cells e.g., colon cancer cells, and lung cancer cells.
  • the immune cells of the invention can be used to treat both tumorigenic and non- tumorigenic cancers ⁇ e.g., non-tumor- forming hematopoietic cancers) .
  • the immune cells of the invention are particularly useful in the treatment of epithelial cancers (e.g., carcinomas) and/or colorectal cancers, breast cancers, lung cancers, vaginal cancers, cervical cancers, and/or squamous cell carcinomas (e.g., of the head and neck) .
  • Additional potential targets include sarcomas and lymphomas.
  • Additional advantageous targets include solid tumors and/or disseminated tumors (e.g., myeloid and lymphoid tumors, which can be acute or chronic) .
  • the immune cells of the invention are also of use in adoptive cell therapies for the treatment of an infectious disease caused by bacteria, protozoa, fungi or viruses.
  • a viral infection is treated.
  • Viral infections that can be treated include, but are not limited to, hepatitis type A, hepatitis type B, hepatitis type C, influenza, varicella, adenovirus, herpes simplex type I (HSV-1) , herpes simplex type 2 (HSV-2), rinderpest, rhinovirus, echovirus, rotavirus, respiratory syncytial virus, papilloma virus, papilloma virus, cytomegalovirus (CMV, e.g., HCMV) , echinovirus, arbovirus, huntavirus, coxsackie virus, mumps virus, measles virus, rubella virus, polio virus, and/or human immunodeficiency virus type I or type
  • CMV e.g.,
  • kits for adoptive cell therapy can include an engineered nuclease for GM-CSF or perforin gene deletion or gene disruption; one or more microRNAs, siRNAs, shRNAs , or antisense molecule that inhibit the expression of GM-CSF or perforin; or a GM-CSF, GM-CSF receptor, or perforin antagonist.
  • the kit includes isolated immune cells including, but are not limited to T- lymphocytes, naive T cells, memory T cells, effector memory cells, natural killer cells, hematopoietic stem cells or pluripotent embryonic/ induced stem cells capable of giving rise to therapeutically relevant progeny.
  • the kit further includes nucleic acids encoding a CAR.
  • the above-referenced reagents are typically provided, either individually or together, in a container and can be in liquid (ready-to-use) or lyophilized forms.
  • the kit can include a compatible pharmaceutical carrier as well as instructions for preparing immune cells with reduced toxicity and administering the same to a subject in need of treatment.
  • Example 1 LentiCRISPR Vectors to Eliminate Human GM-CSF
  • Target guide sequences (20 nt) with 3' protospacer adjacent motif (PAM) sequence (NGG) were selected according to established methods (Shalem, et al . (2014) Science 343:83-7) .
  • sgRNA vectors were sequence verified, then used to co- transfect 293T cells with packaging plasmids pCMV-VSVg and pCMV-dR8.91 to make lentivirus stocks.
  • U937 and THP-1 cells were transduced with lentiviruses expressing the huGM-CSF target sgRNAs LGH-1, LGH-2, LGH-3 and LGH-4.
  • Day 1 post-transduction cells were transferred to RPMI media containing puromycin (2.0 g/mL) , and maintained in puromycin media for 10 days.
  • THP-1 cells expressing LGH- 1 and LGH-3, and U937 cells expressing LGH-2 and LGH-4 were subjected to single-cell dilution to isolate single clones.
  • Four clones for each construct were expanded in puromycin media before harvesting and isolating genomic DNA.
  • Genomic regions containing the LGH-1 and LGH-2 recognition site in GM-CSF Exon 1 and the LGM-3 and LGH-4 region in Exon 3 were PCR-amplified and the -500 bp amplicons were cloned into vector pGE T. Sequencing of amplicons (from two different single cell colonies per group) revealed the insertions or deletions (indels) introduced by the CRISPR machinery.
  • sgRNA target sites are underlined.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Hematology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

L'invention concerne des méthodes de prévention ou réduction de toxicité de la thérapie cellulaire adoptive à l'aide d'une population de cellules dont l'expression ou l'activité de GM-CSF, du récepteur de GM-SCF ou de perforine est déficiente, ainsi que des nécessaires pour la préparation de cellules déficientes dont l'expression ou l'activité de GM-CSF, du récepteur de GM-CSF ou de perforine est déficiente.
PCT/US2014/063037 2013-11-04 2014-10-30 Méthodes de prévention de toxicité de la thérapie cellulaire adoptive WO2015066262A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361899431P 2013-11-04 2013-11-04
US61/899,431 2013-11-04

Publications (1)

Publication Number Publication Date
WO2015066262A1 true WO2015066262A1 (fr) 2015-05-07

Family

ID=53005096

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/063037 WO2015066262A1 (fr) 2013-11-04 2014-10-30 Méthodes de prévention de toxicité de la thérapie cellulaire adoptive

Country Status (1)

Country Link
WO (1) WO2015066262A1 (fr)

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016174461A1 (fr) * 2015-04-30 2016-11-03 Ucl Business Plc Cellules t exprimant un récepteur des cellules t (tcr) gamma-delta et un récepteur antigénique chimérique (car)
WO2018052142A1 (fr) * 2016-09-16 2018-03-22 キッセイ薬品工業株式会社 Cellules modifiées génétiquement et leur procédé de production
WO2018058002A1 (fr) 2016-09-23 2018-03-29 Fred Hutchinson Cancer Research Center Tcr spécifiques de l'antigène ha-1 d'histocompatibilité (h) mineure et ses utilisations
WO2018064076A1 (fr) 2016-09-27 2018-04-05 Cero Therapeutics, Inc. Molécules de récepteurs d'engloutissement chimériques
WO2018184074A1 (fr) * 2017-04-07 2018-10-11 Cynata Therapeutics Limited Procédé de traitement d'un effet secondaire d'une thérapie cellulaire par des lymphocytes t récepteurs d'antigènes chimériques (car)
WO2018218038A1 (fr) 2017-05-24 2018-11-29 Effector Therapeutics, Inc. Méthodes et compositions d'immunothérapie cellulaire
WO2019067328A1 (fr) 2017-09-26 2019-04-04 Cero Therapeutics, Inc. Molécules de récepteur d'engloutissement chimérique et méthodes d'utilisation
WO2019070680A3 (fr) * 2017-10-02 2019-05-23 Humanigen, Inc. Méthodes de traitement de la toxicité associée aux immunothérapies utilisant un antagoniste du gm-csf
WO2019177151A1 (fr) * 2018-03-16 2019-09-19 キッセイ薬品工業株式会社 Cellule génétiquement modifiée et son procédé de production
US20190284271A1 (en) * 2017-10-02 2019-09-19 Humanigen, Inc. Methods of treating immunotherapy-related toxicity using a gm-csf antagonist
WO2019191339A1 (fr) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Vecteurs d'expression pour récepteurs d'envahissement chimériques, cellules hôtes génétiquement modifiées, et leurs utilisations
WO2019191340A1 (fr) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Compositions d'immunothérapie cellulaire et utilisations associées
WO2019191334A1 (fr) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Récepteurs tim4 chimériques et leurs utilisations
WO2019191332A1 (fr) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Récepteurs d'engloutissement chimériques et leurs utilisations pour des maladies neurodégénératives
US20190338023A1 (en) * 2017-10-02 2019-11-07 Humanigen, Inc. Methods of treating immunotherapy-related toxicity using a gm-csf antagonist
WO2019232425A1 (fr) * 2018-06-01 2019-12-05 Washington University Suppression du syndrome de libération des cytokines lors d'une thérapie cellulaire par récepteur antigénique chimérique
WO2019232409A1 (fr) * 2018-05-31 2019-12-05 Washington University Procédés d'édition génomique et d'activation de cellules
WO2019232370A1 (fr) 2018-06-01 2019-12-05 Mayo Foundation For Medical Education And Research Matériels et procédés de traitement du cancer
CN110760001A (zh) * 2019-07-29 2020-02-07 浙江启新生物技术有限公司 Gm-csf敲低及其中和单链抗体分泌的嵌合抗原受体t细胞构建与应用
WO2020055932A3 (fr) * 2018-09-10 2020-05-22 Humanigen, Inc. Méthodes de traitement de toxicité liée à une immunothérapie au moyen d'un antagoniste du gm-csf
US10881688B2 (en) 2015-04-15 2021-01-05 Tc Biopharm Ltd Modified gamma delta T cells and uses thereof
EP3771744A1 (fr) * 2019-07-29 2021-02-03 Thyas Co. Ltd. Procédé de production de lymphocytes t spécifiques d'un antigène
WO2021067875A1 (fr) 2019-10-03 2021-04-08 Cero Therapeutics, Inc. Récepteurs tim4 chimériques et leurs utilisations
US11034748B2 (en) 2017-03-15 2021-06-15 Fred Hutchinson Cancer Research Center High affinity MAGE-A1-specific TCRs and uses thereof
WO2021127261A1 (fr) * 2019-12-17 2021-06-24 The General Hospital Corporation Cellules immunitaires modifiées à toxicité réduite et leurs utilisations
WO2021139734A1 (fr) * 2020-01-08 2021-07-15 上海交通大学医学院附属瑞金医院 Lymphocyte t inactivant le gm-csf humain, et son utilisation
CN113194715A (zh) * 2018-10-31 2021-07-30 赫曼尼根公司 用于治疗癌症的材料和方法
US20210290677A1 (en) * 2017-10-02 2021-09-23 Humanigen, Inc. Methods of treating immunotherapy-related toxicity using a gm-csf antagonist
US11130805B2 (en) 2017-10-02 2021-09-28 Humanigen, Inc. Methods of treating CART-T cell therapy-induced neuroinflammation using a GM-CSF antagonist
WO2021222092A3 (fr) * 2020-04-27 2022-01-20 Vanderbilt University Lymphocytes dépourvus de fonction perforine
WO2022036285A1 (fr) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Compositions et méthodes de traitement du cancer à l'aide de récepteurs tim chimériques en association avec des inhibiteurs de la poly(adp-ribose)polymérase
WO2022036287A1 (fr) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Récepteurs chimériques anti-cd72 et utilisations de ceux-ci
WO2022036265A1 (fr) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Récepteurs chimériques tim et leurs utilisations
RU2787828C2 (ru) * 2018-06-01 2023-01-12 Мэйо Фаундейшн Фор Медикал Эдьюкейшн Энд Рисерч Материалы и способы лечения рака
WO2022266538A3 (fr) * 2021-06-18 2023-01-19 Artisan Development Labs, Inc. Compositions et procédés de ciblage, d'édition ou de modification de gènes humains
WO2023010097A1 (fr) 2021-07-28 2023-02-02 Cero Therapeutics, Inc. Récepteurs tim4 chimériques et leurs utilisations

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080206280A1 (en) * 2007-02-26 2008-08-28 University Of Pittsburgh- Of The Commonwealth System Of Higher Education Vaccine for activating helper function of cd8+ tcells
US20120065249A1 (en) * 2004-10-29 2012-03-15 Topigen Pharmaceuticals Inc. Antisense oligonucleotides for treating allergy and neoplastic cell proliferation
US20130259799A1 (en) * 2007-09-18 2013-10-03 Amgen Inc. Human gm-csf antigen binding proteins

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120065249A1 (en) * 2004-10-29 2012-03-15 Topigen Pharmaceuticals Inc. Antisense oligonucleotides for treating allergy and neoplastic cell proliferation
US20080206280A1 (en) * 2007-02-26 2008-08-28 University Of Pittsburgh- Of The Commonwealth System Of Higher Education Vaccine for activating helper function of cd8+ tcells
US20130259799A1 (en) * 2007-09-18 2013-10-03 Amgen Inc. Human gm-csf antigen binding proteins

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BARBER ET AL.: "Immunotherapy with chimeric NKG2D receptors leads to long-term tumor-free survival and development of host antitumor immunity in murine ovarian cancer.", THE JOUMAL OF IMMUNOLOGY, vol. 180.1, 2008, pages 72 - 78 *

Cited By (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10881688B2 (en) 2015-04-15 2021-01-05 Tc Biopharm Ltd Modified gamma delta T cells and uses thereof
WO2016174461A1 (fr) * 2015-04-30 2016-11-03 Ucl Business Plc Cellules t exprimant un récepteur des cellules t (tcr) gamma-delta et un récepteur antigénique chimérique (car)
JPWO2018052142A1 (ja) * 2016-09-16 2019-06-24 キッセイ薬品工業株式会社 遺伝子改変細胞及びその作製方法
WO2018052142A1 (fr) * 2016-09-16 2018-03-22 キッセイ薬品工業株式会社 Cellules modifiées génétiquement et leur procédé de production
US10683355B2 (en) 2016-09-16 2020-06-16 Kissei Pharmaceutical Co., Ltd. Genetically-modified cells and method for producing same
WO2018058002A1 (fr) 2016-09-23 2018-03-29 Fred Hutchinson Cancer Research Center Tcr spécifiques de l'antigène ha-1 d'histocompatibilité (h) mineure et ses utilisations
US10538574B2 (en) 2016-09-23 2020-01-21 Fred Hutchinson Cancer Research Center TCRS specific for minor histocompatibility (H) antigen HA-1 and uses thereof
US11655282B2 (en) 2016-09-27 2023-05-23 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules
EP4089116A1 (fr) 2016-09-27 2022-11-16 Cero Therapeutics, Inc. Molécules de récepteurs d'engloutissement chimériques
WO2018064076A1 (fr) 2016-09-27 2018-04-05 Cero Therapeutics, Inc. Molécules de récepteurs d'engloutissement chimériques
US11034748B2 (en) 2017-03-15 2021-06-15 Fred Hutchinson Cancer Research Center High affinity MAGE-A1-specific TCRs and uses thereof
WO2018184074A1 (fr) * 2017-04-07 2018-10-11 Cynata Therapeutics Limited Procédé de traitement d'un effet secondaire d'une thérapie cellulaire par des lymphocytes t récepteurs d'antigènes chimériques (car)
US11564947B2 (en) 2017-05-24 2023-01-31 Effector Therapeutics Inc. Methods and compositions for cellular immunotherapy
US10780119B2 (en) 2017-05-24 2020-09-22 Effector Therapeutics Inc. Methods and compositions for cellular immunotherapy
WO2018218038A1 (fr) 2017-05-24 2018-11-29 Effector Therapeutics, Inc. Méthodes et compositions d'immunothérapie cellulaire
US11708423B2 (en) 2017-09-26 2023-07-25 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules and methods of use
WO2019067328A1 (fr) 2017-09-26 2019-04-04 Cero Therapeutics, Inc. Molécules de récepteur d'engloutissement chimérique et méthodes d'utilisation
US20190338023A1 (en) * 2017-10-02 2019-11-07 Humanigen, Inc. Methods of treating immunotherapy-related toxicity using a gm-csf antagonist
JP2020536118A (ja) * 2017-10-02 2020-12-10 ヒューマニジェン,インコーポレイテッド Gm−csfアンタゴニストを使用した免疫療法関連毒性の治療方法
JP7320498B2 (ja) 2017-10-02 2023-08-03 ヒューマニジェン,インコーポレイテッド Gm-csfアンタゴニストを使用した免疫療法関連毒性の治療方法
WO2019070680A3 (fr) * 2017-10-02 2019-05-23 Humanigen, Inc. Méthodes de traitement de la toxicité associée aux immunothérapies utilisant un antagoniste du gm-csf
US10927168B2 (en) 2017-10-02 2021-02-23 Humanicen, Inc. Method of reducing tumor relapse rate in immunotherapy by administration of lenzilumab
US11673962B2 (en) 2017-10-02 2023-06-13 Humanigen, Inc. Method of increasing the efficacy of CAR-T immunotherapy using lenzilumab
US20210290677A1 (en) * 2017-10-02 2021-09-23 Humanigen, Inc. Methods of treating immunotherapy-related toxicity using a gm-csf antagonist
US11130805B2 (en) 2017-10-02 2021-09-28 Humanigen, Inc. Methods of treating CART-T cell therapy-induced neuroinflammation using a GM-CSF antagonist
CN111757744A (zh) * 2017-10-02 2020-10-09 人源股份有限公司 使用gm-csf拮抗剂治疗免疫疗法相关毒性的方法
US20190284271A1 (en) * 2017-10-02 2019-09-19 Humanigen, Inc. Methods of treating immunotherapy-related toxicity using a gm-csf antagonist
US10870703B2 (en) 2017-10-02 2020-12-22 Humanigen, Inc. Methods of treating immunotherapy-related toxicity using a GM-CSF antagonist
US20190194343A1 (en) * 2017-10-02 2019-06-27 Humanigen, Inc. Methods of treating immunotherapy-related toxicity using a gm-csf antagonist
US10899831B2 (en) 2017-10-02 2021-01-26 Humanigen, Inc. Method of reducing the level of non-GM-CSF cytokines/chemokines in immunotherapy-related toxicity
EP3691663A4 (fr) * 2017-10-02 2021-08-18 Humanigen, Inc. Méthodes de traitement de la toxicité associée aux immunothérapies utilisant un antagoniste du gm-csf
JP7296133B2 (ja) 2018-03-16 2023-06-22 国立大学法人信州大学 遺伝子改変細胞及びその作製方法
JPWO2019177151A1 (ja) * 2018-03-16 2021-03-11 国立大学法人信州大学 遺伝子改変細胞及びその作製方法
WO2019177151A1 (fr) * 2018-03-16 2019-09-19 キッセイ薬品工業株式会社 Cellule génétiquement modifiée et son procédé de production
WO2019191340A1 (fr) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Compositions d'immunothérapie cellulaire et utilisations associées
WO2019191339A1 (fr) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Vecteurs d'expression pour récepteurs d'envahissement chimériques, cellules hôtes génétiquement modifiées, et leurs utilisations
WO2019191334A1 (fr) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Récepteurs tim4 chimériques et leurs utilisations
WO2019191332A1 (fr) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Récepteurs d'engloutissement chimériques et leurs utilisations pour des maladies neurodégénératives
CN112867498A (zh) * 2018-05-31 2021-05-28 华盛顿大学 使用嵌合抗原受体基因组编辑和转导t细胞以用于治疗t和b细胞恶性肿瘤的方法
WO2019232409A1 (fr) * 2018-05-31 2019-12-05 Washington University Procédés d'édition génomique et d'activation de cellules
CN112533942A (zh) * 2018-06-01 2021-03-19 华盛顿大学 嵌合抗原受体细胞疗法中细胞因子释放综合征的抑制
RU2787828C2 (ru) * 2018-06-01 2023-01-12 Мэйо Фаундейшн Фор Медикал Эдьюкейшн Энд Рисерч Материалы и способы лечения рака
US20230074186A1 (en) * 2018-06-01 2023-03-09 Washington University Suppression of cytokine release syndrome in chimeric antigen receptor cell therapy
CN112512533A (zh) * 2018-06-01 2021-03-16 妙佑医学教育研究基金会 用于治疗癌症的材料与方法
JP2021525530A (ja) * 2018-06-01 2021-09-27 ワシントン・ユニバーシティWashington University キメラ抗原受容体細胞治療薬を用いたサイトカイン放出症候群の抑制
JP2021525519A (ja) * 2018-06-01 2021-09-27 マヨ ファウンデーション フォー メディカル エデュケーション アンド リサーチMayo Foundation For Medical Education And Research 癌を治療するための物質及び方法
WO2019232370A1 (fr) 2018-06-01 2019-12-05 Mayo Foundation For Medical Education And Research Matériels et procédés de traitement du cancer
WO2019232425A1 (fr) * 2018-06-01 2019-12-05 Washington University Suppression du syndrome de libération des cytokines lors d'une thérapie cellulaire par récepteur antigénique chimérique
JP7411578B2 (ja) 2018-06-01 2024-01-11 マヨ ファウンデーション フォー メディカル エデュケーション アンド リサーチ 癌を治療するための物質及び方法
JP2022517461A (ja) * 2018-09-10 2022-03-09 ヒューマニゲン インコーポレイティッド Gm-csfアンタゴニストを使用した免疫療法関連毒性の治療方法
EP3849572A4 (fr) * 2018-09-10 2022-07-13 Humanigen, Inc. Méthodes de traitement de toxicité liée à une immunothérapie au moyen d'un antagoniste du gm-csf
WO2020055932A3 (fr) * 2018-09-10 2020-05-22 Humanigen, Inc. Méthodes de traitement de toxicité liée à une immunothérapie au moyen d'un antagoniste du gm-csf
CN113164520A (zh) * 2018-09-10 2021-07-23 赫曼尼根公司 使用gm-csf拮抗剂治疗免疫疗法相关毒性的方法
JP2022513412A (ja) * 2018-10-31 2022-02-07 ヒューマニゲン インコーポレイティッド 癌を治療するための物質及び方法
CN113194715A (zh) * 2018-10-31 2021-07-30 赫曼尼根公司 用于治疗癌症的材料和方法
EP3873205A4 (fr) * 2018-10-31 2022-05-11 Humanigen, Inc. Substances et méthodes pour le traitement du cancer
CN113194715B (zh) * 2018-10-31 2024-03-19 赫曼尼根公司 用于治疗癌症的材料和方法
CN110760001A (zh) * 2019-07-29 2020-02-07 浙江启新生物技术有限公司 Gm-csf敲低及其中和单链抗体分泌的嵌合抗原受体t细胞构建与应用
EP3771744A1 (fr) * 2019-07-29 2021-02-03 Thyas Co. Ltd. Procédé de production de lymphocytes t spécifiques d'un antigène
WO2021067875A1 (fr) 2019-10-03 2021-04-08 Cero Therapeutics, Inc. Récepteurs tim4 chimériques et leurs utilisations
WO2021127261A1 (fr) * 2019-12-17 2021-06-24 The General Hospital Corporation Cellules immunitaires modifiées à toxicité réduite et leurs utilisations
WO2021139734A1 (fr) * 2020-01-08 2021-07-15 上海交通大学医学院附属瑞金医院 Lymphocyte t inactivant le gm-csf humain, et son utilisation
WO2021222092A3 (fr) * 2020-04-27 2022-01-20 Vanderbilt University Lymphocytes dépourvus de fonction perforine
WO2022036265A1 (fr) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Récepteurs chimériques tim et leurs utilisations
WO2022036287A1 (fr) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Récepteurs chimériques anti-cd72 et utilisations de ceux-ci
WO2022036285A1 (fr) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Compositions et méthodes de traitement du cancer à l'aide de récepteurs tim chimériques en association avec des inhibiteurs de la poly(adp-ribose)polymérase
WO2022266538A3 (fr) * 2021-06-18 2023-01-19 Artisan Development Labs, Inc. Compositions et procédés de ciblage, d'édition ou de modification de gènes humains
WO2023010097A1 (fr) 2021-07-28 2023-02-02 Cero Therapeutics, Inc. Récepteurs tim4 chimériques et leurs utilisations

Similar Documents

Publication Publication Date Title
WO2015066262A1 (fr) Méthodes de prévention de toxicité de la thérapie cellulaire adoptive
JP7037577B2 (ja) 高親和性mage-a1特異的tcr及びその使用
US20190175651A1 (en) Immortalized car-t cells genetically modified to elminate t-cell receptor and beta 2-microglobulin expression
KR20210020932A (ko) Bcma 키메라 항원 수용체 및 이의 용도
JP2020195393A (ja) 養子細胞療法用の操作された細胞
KR20210008502A (ko) 암을 치료하기 위한 방법 및 조성물
JP2021516996A (ja) 生物学的に関連する直交サイトカイン/受容体対
KR20220104217A (ko) Cd19 및 cd22 키메라 항원 수용체 및 이의 용도
BR112020009336A2 (pt) receptor quimérico de antígenos de alvejamento de bcma, receptor quimérico de antígenos de alvejamento de cd19 e terapias de combinação
CN114026116A (zh) Ras新抗原特异性结合蛋白及其用途
JP2018519841A (ja) 養子細胞療法用の操作された細胞
JP2021502330A (ja) Tigitおよび/またはcd112rを標的とするか、またはcd226過剰発現を含む、組成物および免疫治療の方法
KR102617818B1 (ko) 인간 t 세포 수용체 알파 불변 영역 유전자에 대한 특이성을 갖는 최적화된 조작된 뉴클레아제
KR20170002381A (ko) 동종이형 이식에 양립성인 t-세포들을 만들어내는 방법
CN113227141A (zh) 抗cd33免疫细胞癌症疗法
JP7471289B2 (ja) 抗liv1免疫細胞癌療法
WO2018218231A1 (fr) Antagonistes de monomères, d'homodimères cd5-like (cd5l) d'antigènes lymphocytaires, et d'hétérodimères de l'interleukine 12b (p40) et leurs procédés d'utilisation
JP2020535796A (ja) strepタグ特異的キメラ受容体およびその使用
KR20200079312A (ko) 면역요법을 위한 t 세포 내 cblb의 crispr-cas9 편집 방법, 조성물 및 성분
CN112955472A (zh) 抗ptk7免疫细胞癌症疗法
JP2024519515A (ja) 人工多能性幹細胞からガンマ-デルタt細胞を生成するための組成物および方法
JP2024513454A (ja) キメラ抗原受容体細胞のための人工細胞死/リポーター系ポリペプチドの組合せおよびその使用
US20230233677A1 (en) Compositions and methods for enhancing immune response
WO2018218223A1 (fr) Agonistes de monomères, d'homodimères cd5-like (cd5l) d'antigènes lymphocytaires, et d'hétérodimères de l'interleukine 12b (p40) et leurs procédés d'utilisation
WO2024030970A2 (fr) Édition génique des gènes cibles pour améliorer les fonctions des cellules tueuses naturelles

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14858768

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14858768

Country of ref document: EP

Kind code of ref document: A1