WO2008039818A2 - Modified t cell receptors and related materials and methods - Google Patents

Modified t cell receptors and related materials and methods Download PDF

Info

Publication number
WO2008039818A2
WO2008039818A2 PCT/US2007/079487 US2007079487W WO2008039818A2 WO 2008039818 A2 WO2008039818 A2 WO 2008039818A2 US 2007079487 W US2007079487 W US 2007079487W WO 2008039818 A2 WO2008039818 A2 WO 2008039818A2
Authority
WO
WIPO (PCT)
Prior art keywords
tcr
amino acid
cells
seq
modified
Prior art date
Application number
PCT/US2007/079487
Other languages
French (fr)
Other versions
WO2008039818A3 (en
Inventor
Paul F. Robbins
Richard A. Morgan
Steven A. Rosenberg
Alan David Bennett
Original Assignee
Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services
Medigene Limited
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 Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services, Medigene Limited filed Critical Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services
Priority to US12/443,111 priority Critical patent/US8088379B2/en
Publication of WO2008039818A2 publication Critical patent/WO2008039818A2/en
Publication of WO2008039818A3 publication Critical patent/WO2008039818A3/en
Priority to US13/304,841 priority patent/US9128080B2/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5091Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing the pathological state of an organism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • PBMC peripheral blood mononuclear cells
  • TCR T cell receptor
  • two out of 17 patients have demonstrated an objective clinical response (Morgan et al., Sciencexpress, e-publication August 31, 2006).
  • the results of this clinical trial demonstrate that normal autologous T lymphocytes, transduced ex vivo with anti-cancer antigen TCR genes and reinfused in cancer patients, can persist and express the transgene long term in vivo and mediate durable regression of large established tumors.
  • approaches to increase the expression and function of the transgene are still needed.
  • the invention provides a modified T cell receptor (TCR) comprising an amino acid sequence of a wild-type (WT) TCR with no more than three amino acid substitutions, wherein the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize target cells when expressed by CD4 + T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD 8 T cells.
  • TCR T cell receptor
  • the invention also provides related polypeptides and proteins, as well as related nucleic acids, recombinant expression vectors, host cells, and populations of cells. Further provided by the invention are antibodies, or an antigen binding portion thereof, and pharmaceutical compositions relating to the modified TCRs of the invention.
  • the invention provides a method of detecting a diseased cell in a host, wherein the diseased cell expresses an antigen characteristic of a disease.
  • the method comprises (a) contacting a sample comprising cells of the host with an inventive modified TCR, polypeptide, protein, nucleic acid, recombinant expression vector, host cell, or population of cells, thereby forming a complex between the modified TCR, polypeptide, protein, nucleic acid, recombinant expression vector, host cell, or population of cells and the antigen, and (b) detecting the complex, wherein detection of the complex is indicative of a diseased cell in the host
  • a method of treating or preventing a disease in a host comprises administering to the host an inventive pharmaceutical composition in an amount effective to treat or prevent the disease in the host.
  • a method of identifying a candidate adoptive immunotherapy TCR is also provided. The method consists of (a) producing a nucleic acid encoding a modified TCR comprising a WT TCR amino acid sequence with no more than three amino acid substitutions, (b) expressing the nucleic acid in CD4 + T cells and CDS + T cells, and (c) assaying the T cells for the ability to recognize target cells and for antigen specificity.
  • a candidate adoptive immunotherapy TCR is identified when the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize target cells when expressed by CD4 + T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD8 + T cells..
  • the invention provides a modified T cell receptor (TCR) comprising an amino acid sequence of a wild-type (WT) TCR with no more than three amino acid substitutions, (e.g., 1, 2, or 3) wherein the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize target cells when expressed by CD4 + T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD8 + T cells.
  • TCR T cell receptor
  • wild-type refers to a TCR which is naturally expressed by a T cell of a host, e.g., a TCR which is endogenous to a T cell of a host.
  • Nucleic acids encoding wild-type TCRs are known in the art and can be obtained from the GenBank database of the National Center for Biotechnology Information (NCBI). For example, wild- type TCR nucleic acid sequences are available as GenBank Accession Nos.
  • nucleic acids encoding wild-type TCRs can be obtained by methods known in the art, such as the PCR-based method described herein (see Example 1).
  • the cells used to obtain the nucleic acids encoding the wild-type TCR are not limited to those used in Example 1. Rather, the cells can be any of the T cells described herein.
  • the wild-type TCR can be entirely synthesized using oligonucleotide primers corresponding to the known sequence.
  • the modified TCR of the invention is marked by one or more enhanced biological properties when expressed in T cells.
  • the modified TCR when compared to the corresponding WT TCR, (i) has an enhanced ability to recognize target cells when expressed by CD4 + T cells and (H) does not exhibit a decrease in antigen specificity when expressed by CDS + T cells.
  • target cells refers to cells which express and present, by way of an MHC molecule, the antigen which is specifically recognized by the modified TCR.
  • recognition target cells refers to the ability of the modified TCR to immunologically recognize (e.g., specifically bind to) an antigen which is expressed and presented by a target cell.
  • the modified TCR of the invention consistently exhibits at least twice the ability to recognize antigen expressed and presented by target cells, as compared to its WT counterpart.
  • the modified TCR of the invention recognizes antigen expressed and presented by target cells at least five times better than its WT counterpart. More preferably, the modified TCRs of the invention recognize antigen at least ten times better than its WT counterpart. Most preferably, the modified TCRs of the invention recognize antigen at least 2 times better than its WT counterpart.
  • the enhanced properties exhibited by the modified TCR consistently exhibits the properties, e.g., ability to recognize antigen expressed and presented by target cells.
  • the modified TCR of the invention exhibits the enhanced properties in at least two assays.
  • the TCR of the invention exhibits the enhanced properties in at least three assays (e.g., in five or more assays, or in ten or more assays).
  • the modified TCR of the invention exhibits an enhanced ability to recognize target cells without exhibiting a decrease in antigen specificity when expressed by CD8 + T cells.
  • the modified TCR is said to retain the antigen specificity of the counterpart WT TCR, e.g., recognizes only the antigen(s) recognized by the WT TCR and does not recognize antigen(s) that are not recognized by the WT TCR.
  • Methods of testing a TCR for the ability to recognize target cells and for antigen specificity are known in the art. For instance, Clay et al., J Immunol, 163: 507-513 (1999), teaches methods of measuring the release of cytokines (e.g., interferon- ⁇ , granulocyte/monocyte colony stimulating factor (GM-CSF) 5 tumor necrosis factor a (TNF- ⁇ ) or interleukin 2 (IL-2)).
  • cytokines e.g., interferon- ⁇ , granulocyte/monocyte colony stimulating factor (GM-CSF) 5 tumor necrosis factor a (TNF- ⁇ ) or interleukin 2 (IL-2)
  • TCR function can be evaluated by measurement of cellular cytoxicity, as described in Zhao et al., J Immunol, 11 A: 4415-4423 (2005). Methods of testing a modified TCR for the ability to recognize target cells and for antigen specificity are described herein as Examples 2 to 4.
  • the modified TCR of the invention can have antigen specificity for any antigen.
  • the phrase "have antigen specificity" as used herein means that the modified TCR can specifically bind to and immunologically recognize an antigen, such that binding of the TCR to the antigen elicits an immune response.
  • the modified TCR of the invention has antigen specificity for an antigen which is characteristic of a disease.
  • the disease can be any disease involving an antigen, as discussed herein, e.g., an infectious disease, an autoimmune disease, or a cancer.
  • the antigen could be, for example, a viral antigen, a bacterial antigen, a cancer antigen, etc.
  • the modified TCR of the invention has antigen specificity for a cancer antigen.
  • cancer antigen refers to any molecule (e.g., protein, peptide, lipid, carbohydrate, etc.) solely or predominantly expressed or over- expressed by a tumor cell or cancer cell, such that the antigen is associated with the tumor or cancer.
  • the cancer antigen additionally can be expressed by normal, non-tumor, or noncancerous cells.
  • normal, non-tumor, or non-cancerous cells are normally expressed by normal, non-tumor, or non-cancerous cells.
  • the tumor or cancer ceils can over-express the antigen or express the antigen at a significantly higher level, as compared to the expression of the antigen by normal, non-tumor, or non-cancerous cells.
  • the cancer antigen additionally can be expressed by cells of a different state of development or maturation.
  • the cancer antigen can be additionally expressed by cells of the embryonic or fetal stage, which cells are not normally found in an adult host.
  • the cancer antigen additionally can be expressed by stem cells or precursor cells, which cells are not normally found in an adult host.
  • the cancer antigen can be an antigen expressed by any cell of any cancer or tumor, including the cancers and tumors described herein.
  • the cancer antigen may be a cancer antigen of only one type of cancer or rumor, such that the cancer antigen is associated with or characteristic of only one type of cancer or tumor.
  • the cancer antigen may be a cancer antigen (e.g., may be characteristic) of more than one type of cancer or tumor.
  • the cancer antigen may be expressed by both breast and prostate cancer cells and not expressed at all by normal, non-tumor, or non-cancer cells.
  • the cancer antigen is a melanoma cancer antigen.
  • the cancer antigen is selected from the group consisting of NY-ESO- 1, MART-I, gplOO, p53, TRP-I, TRP-2, and tyrosinase.
  • the cancer antigen is NY-ESO-I or MART-I .
  • the amino acid substitution(s) can be located in any part of the amino acid sequence of the TCR.
  • the amino acid substitutions are located within the amino acid sequence of the complementary determining region (CDR) of the TCR, which are known in the art. These regions have been defined by elucidation of X-ray crystallographic structures, as well as sequence comparisons which have revealed the presence of regions of high diversity encoded in germline sequences, in the case of CDRl and CDF2 regions, as well as recombinational diversity, in the case of CDR3 region (Lefranc et al, Nucl Acids Res., 27, 209-212 (1999)).
  • the one, two, or three amino acid substitutions are located in the amino acid sequence of a CDR2 or CDR3 of the TCR (e.g., in the CDR2 region of the beta chain of the TCR). More preferably, the amino acid substitutions are located in the amino acid sequence of a CDR2, e.g., CDR2 of an ⁇ chain of a TCR or a ⁇ chain of a TCR. Most preferably, the amino acid substitutions are located in the CDR2 of a ⁇ chain of a TCR.
  • the modified TCR can comprise an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 16 to 121 in which the amino acid substitutions are located at the Xaa's of these sequences.
  • the invention provides a modified TCR comprising two polypeptides (i.e., polypeptide chains), such as an ⁇ chain of a TCR, a ⁇ chain of a TCR 5 a ⁇ chain of a TCR, a ⁇ chain of a TCR, or a combination thereof.
  • the amino acid substitutions of the inventive modified TCRs can be located in the amino acid sequence of either or both polypeptide chains which constitute the TCR.
  • the amino acid substitutions are located in the amino acid sequence of the ⁇ chain of the modified TCR.
  • the amino acid substitutions are located in only the amino acid sequence of the ⁇ chain of the modified TCR.
  • amino acid substitutions of the inventive modified TCR are preferably conservative amino acid substitutions.
  • Conservative amino acid substitutions are known in the art, and include amino acid substitutions in which one amino acid having certain physical and/or chemical properties is exchanged for another amino acid that has the same chemical or physical properties.
  • the conservative amino acid substitution can be an acidic amino acid substituted for another acidic amino acid (e.g., Asp or GIu), an amino acid with a nonpolar side chain substituted for another amino acid with a nonpolar side chain (e.g., Ala, GIy, VaI, De, Leu, Met, Phe, Pro, Tip, VaI, etc.), a basic amino acid substituted for another basic amino acid (Lys, Arg, etc.), an amino acid with a polar side chain substituted for another amino acid with a polar side chain (Asn, Cys, GIn, Ser, Thr, Tyr, etc.), etc.
  • an amino acid with a nonpolar side chain substituted for another amino acid with a nonpolar side chain e.g., Ala, GIy, VaI, De, Leu, Met, Phe, Pro, Tip, VaI, etc.
  • a basic amino acid substituted for another basic amino acid Lys, Arg, etc.
  • the conservative amino acid substitutions are selected from the group consisting of T ⁇ A, G ⁇ A, A ⁇ I, T ⁇ V, A ⁇ M, T ⁇ , A ⁇ V, T ⁇ G, and T- ⁇ S.
  • the polypeptide chains of the inventive modified TCR can comprise any amino acid sequence, provided that the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize antigen when expressed by CD4 + T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD8 + T cells.
  • the modified TCR comprises the amino acid sequence of SEQ ID NO: 8 with no more than three amino acid substitutions.
  • the modified TCR comprises the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 2, or SEQ ID NO: 3, each of which is an amino acid sequence of a variable region of a beta chain of a modified TCR which recognizes the cancer antigen NY-ESO-I.
  • the modified TCR comprises the amino acid sequence of SEQ ID NO: 14, 15, or 16, each of which is the amino acid sequence of a full-length beta chain comprising a constant region and a variable region.
  • the modified TCR can additionally comprise the amino acid sequence of SEQ ID NO: 7, which is the WT alpha chain of the 1G4 NY-ESO-I -specific TCR.
  • the modified TCR comprises the amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 12 with no more than three amino acid substitutions.
  • the modified TCR comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 4 to 6, 13, and 95 to 103, each of which is an amino acid sequence of a variable region of a beta chain of a modified TCR which recognizes the cancer antigen MART-I .
  • the modified TCR comprises the amino acid sequence of any of SEQ ID NOs: 17 to 20, 37 to 39, and 104 to 109, each of which is the amino acid sequence of a full-length beta chain comprising a constant region and a variable region.
  • the modified TCR can additionally comprise the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: I L [0026]
  • the modified TCR comprises the amino acid sequence of any of SEQ ID NOs: 4, 13, and 95 to 103. More preferably, the modified TCR comprises the amino acid sequence of any of SEQ ID NOs: 17, 20, 37 to 39, and 104 to 109. Also, in these instances, it is preferred that the modified TCR additionally comprises the amino acid sequence of SEQ ID NO: 9, which is the WT alpha chain of the F5 MART-I -specific TCR.
  • the modified TCR when the modified TCR comprises the amino acid sequence of SEQ ID NO: 12 with no more than three amino acid substitutions, the modified TCR preferably comprises SEQ ID NO: 5 or SEQ ID NO: 6. More preferably, the modified TCR comprises the amino acid sequence of SEQ ID NO: 18 or 19. Also, in these instances, it is preferred that the modified TCR additionally comprises the amino acid sequence of SEQ ID NO: 11 , which is the WT alpha chain of the F4 MART-I -specific TCR.
  • the modified TCR comprises an amino acid sequence of (i) any one of SEQ ID NOs: 17, 20, 37 to 39, and 104 to 109 and SEQ ID NO: 9, (ii) SEQ ID NO: 18 or 19 and SEQ ID NO: 11, or (iii) SEQ ID NO: 14, 15, or 16 and SEQ ID NO: 7.
  • modified TCRs comprise the amino acid sequences of a mutated ⁇ chain and a WT ⁇ chain of a TCR which recognizes either NY-ESO-I or MART-L
  • the modified TCRs of the invention can comprise one or more immature TCR chains comprising a leader sequence or one or more mature chains in which the leader sequence has been cleaved off.
  • the leader sequence of a TCR chain comprises the amino acids at the N-terminus which together serve as a signal to transport the TCR to the plasma membrane and which amino acids are cleaved off to yield the mature form of the TCR.
  • the modified TCRs described herein can additionally comprise a leader sequence selected from the group consisting of SEQ ID NOs: 1 10-115.
  • the modified TCR comprises the leader sequence of SEQ ID NO: 110; when the modified TCR comprises any of SEQ ID NOs: 1-3, 8, and 14-16, the modified TCR comprises the leader sequence of SEQ ID NO: 111 ; when the modified TCR comprises SEQ ID NO: 11, the modified TCR comprises the leader sequence of SEQ ID NO: 112; when the modified TCR comprises any of SEQ ID NOs: 5, 6, 12, 18, and 19, the modified TCR comprises the leader sequence of SEQ ID NO: 113; when the modified TCR comprises SEQ ID NO: 9, the modified TCR comprises the leader sequence of SEQ ID NO: 114; when the modified TCR comprises any of SEQ ID NOs: 4, 10, 13, 17, 20, 37-39, and 95-109, the modified TCR comprises the leader sequence of SEQ ID NO: 115.
  • polypeptide as used herein includes oligopeptides and refers to a single chain of amino acids connected by one or more peptide bonds.
  • the functional portion can be any portion comprising contiguous amino acids of the modified TCR of which it is a part, provided that the functional portion comprises the amino acid substitutions.
  • the term "functional portion" when used in reference to a modified TCR refers to any part or fragment of the modified TCR of the invention, which part or fragment retains the biological activity of the modified TCR of which it is a part (the parent modified TCR).
  • Functional portions encompass, for example, those parts of a modified TCR that retain the ability to recognize target cells, or detect, treat, or prevent a disease, to a similar extent, the same extent, or to a higher extent, as the parent modified TCR.
  • the functional portion can comprise, for instance, about 10%, 25%, 30%, 50%, 68%, 80%, 90%, 95%, or more, of the parent TCR.
  • the functional portion can comprise additional amino acids at the amino or carboxy terminus of the portion, or at both termini, which additional amino acids are not found in the amino acid sequence of the parent modified TCR.
  • the additional amino acids do not interfere with the biological function of the functional portion, e.g., recognize target cells, detect cancer, treat or prevent cancer, etc. More desirably, the additional amino acids enhance the biological activity, as compared to the biological activity of the parent modified TCR.
  • the polypeptide can comprise a functional portion of either or both of the ⁇ and ⁇ chains of the TCRs of the invention, such as a functional portion comprising one of more of CDRl, CDR2, and CDR3 of the variable region(s) of the ⁇ chain and/or ⁇ chain of a TCR of the invention.
  • the polypeptide can comprise the amino acid sequence of any of SEQ ID NOs: 1 to 6, 13, and 95 to 103.
  • the polypeptides can additionally comprise an amino acid sequence selected from the group consisting of SEQ ID NOs: 7, 9, and 11.
  • the inventive polypeptide can comprise the entire length of an ⁇ or ⁇ chain of one of the modified TCRs described herein.
  • the inventive polypeptide can comprise an amino acid sequence selected from the group consisting of any of SEQ ID NOs: 14 to 20, 37 to 39, and 104 to 109.
  • the polypeptide of the invention can comprise both chains of the TCRs described herein.
  • the inventive polypeptide can comprise: (i) any of SEQ ID NOs: 14 to 16 in combination with SEQ ID NO: 7; (ii) any of SEQ ID NOs: 17, 20, 37 to 39 ; and 104 to 109 in combination with SEQ ID NO: 9; or (iii) SEQ ID NO: 18 or 19 in combination with SEQ ID NO: 11.
  • the invention further provides an isolated or purified protein comprising at least one of the inventive polypeptides described herein.
  • protein is meant a molecule comprising one or more polypeptide chains.
  • the protein of the invention can comprise (i) a first polypeptide chain comprising the amino acid sequence of SEQ ID NO: 1, 2, or 3 and a second polypeptide chain comprising the amino acid sequence of SEQ ID NO: 7, (ii) a first polypeptide chain comprising the amino acid sequence of any of SEQ ID NOs: 4, 13, and 95 to 103 and a second polypeptide chain comprising the amino acid sequence of SEQ ID NO: 9, or (iii) a first polypeptide chain comprising the amino acid sequence of SEQ ID NO: 5 or 6 and a second polypeptide chain comprising the amino acid sequence of SEQ ID NO: 11.
  • the protein of the invention can be a TCR.
  • the protein comprises a single polypeptide chain comprising SEQ ID NO: 1 and SEQ ID NO: 7, or if the first and/or second polypeptide chain(s) of the protein further comprise(s) other amino acid sequences, e.g., an amino acid sequence encoding an immunoglobulin or a portion thereof, then the inventive protein can be a fusion protein.
  • the invention also provides a fusion protein comprising at least one of the inventive polypeptides described herein along with at least one other polypeptide.
  • the other polypeptide can exist as a separate polypeptide of the fusion protein, or can exist as a polypeptide, which is expressed in frame (in tandem) with one of the inventive polypeptides described herein.
  • the other polypeptide can encode any peptidic or proteinaceous molecule, or a portion thereof, including, but not limited to an immunoglobulin, CD3, CD4, CD8, an MHC molecule, etc.
  • the fusion protein can comprise one or more copies of the inventive polypeptide and/or one or more copies of the other polypeptide.
  • the fusion protein can comprise I 5 2, 3, 4, 5, or more, copies of the inventive polypeptide and/or of the other polypeptide.
  • Suitable methods of making fusion proteins are known in the art, and include, for example, recombinant methods. See, for instance, Choi et al. ⁇ MoI Biotechnol. 31: 193- 202 (2005).
  • the protein of the invention can be a recombinant antibody comprising at least one of the inventive polypeptides described herein.
  • "recombinant antibody” refers to a recombinant (e.g., genetically engineered) protein comprising at least one of the polypeptides of the invention and a polypeptide chain of an antibody, or a portion thereof.
  • the polypeptide of an antibody, or portion thereof can be a heavy chain, a light chain, a variable or constant region of a heavy or light chain, a single chain variable fragment (scFv), or an Fc, Fab, or F(ab) 2 ' fragment of an antibody, etc.
  • polypeptide chain of an antibody, or portion thereof can exist as a separate polypeptide of the recombinant antibody.
  • the polypeptide chain of an antibody, or portion thereof can exist as a polypeptide, which is expressed in frame (in tandem) with the polypeptide of the invention.
  • the polypeptide of an antibody, or portion thereof can be a polypeptide of any antibody or any antibody fragment, including any of the antibodies and antibody fragments described herein.
  • the term "functional variant” as used herein refers to a modified TCR, polypeptide, or protein having substantial or significant sequence identity or similarity to a parent modified TCR, polypeptide, or protein, which functional variant retains the biological activity of the modified TCR, polypeptide, or protein of which it is a variant
  • Functional variants encompass, for example, those variants of the TCR, polypeptide, or protein described herein (the parent modified TCR, polypeptide, or protein) that retain the ability to recognize target cells to a similar extent, the same extent, or to a higher extent, as the parent modified TCR, polypeptide, or protein.
  • the functional variant can, for instance, be at least about 30%, 50%, 75%, 80%, 90%, 98% or more identical in amino acid sequence to the parent modified TCR 5 polypeptide, or protein.
  • the functional variant can, for example, comprise the amino acid sequence of the parent TCR, polypeptide, or protein with at least one conservative amino acid substitution.
  • the functional variants can comprise the amino acid sequence of the parent modified TCR, polypeptide, or protein with at least one non-conservative amino acid substitution.
  • the non-conservative amino acid substitution it is preferable for the non-conservative amino acid substitution to not interfere with or inhibit the biological activity of the functional variant.
  • the non-conservative amino acid substitution enhances the biological activity of the functional variant, such that the biological activity of the functional variant is increased as compared to the parent modified TCR, polypeptide, or protein.
  • the modified TCR, polypeptide, or protein can consist essentially of the specified amino acid sequence or sequences described herein, such that other components of the functional variant, e.g., other amino acids, do not materially change the biological activity of the functional variant.
  • the inventive modified TCR, polypeptide, or protein can, for example, consist essentially of an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 to 6, 13, and 95 to 103.
  • the inventive modified TCRs, polypeptides, or proteins can consist essentially of an amino acid sequence selected from the group consisting of SEQ ID NOs: 14 to 20, 37 to 39, and 104 to 109.
  • the modified TCRs, polypeptides, and proteins of the invention can be of any length, i.e., can comprise any number of amino acids, provided that the modified TCRs, polypeptides, or proteins (or functional portions or functional variants thereof) retain their biological activity, e.g., the ability to specifically bind to antigen, detect diseased cells in a host, or treat or prevent disease in a host, etc.
  • the polypeptide can be 50 to 5000 amino acids long, such as 50, 70, 75, 100, 125, 150, 175, 200, 300, 400, 500, 600, 700, 800, 900, 1000 or more amino acids in length.
  • the polypeptides of the invention also include oligopeptides.
  • the modified TCRs, polypeptides, and proteins of the invention (including functional portions and functional variants) of the invention can comprise synthetic amino acids in place of one or more naturally-occurring amino acids.
  • Such synthetic amino acids are known in the art, and include, for example, aminocyclohexane carboxylic acid, norleucine, ⁇ -amino n-decanoic acid, homoserine, S-acetylaminomethyl-cysteine, trans-3- and trans-4-hydroxyproline, 4-aminophenylalanine, 4- nitrophenylalanine, 4- chlorophenylalanine, 4-carboxyphenylalanine, ⁇ -phenylserine ⁇ -hydroxyphenylalanine, phenylglycine, ⁇ -naphthylalanine, cyclohexylalanine, cyclohexylglycine, indoline-2- carboxylic acid, 1,2 ,3 ,4-tetrahydrois
  • the modified TCRs, polypeptides, and proteins of the invention can be glycosylated, amidated, carboxylated, phosphorylated, esterif ⁇ ed, N-acylated, cyclized via, e.g., a disulfide bridge, or converted into an acid addition salt and/or optionally dimerized or polymerized, or conjugated.
  • the modified TCRs, polypeptides, and proteins of the invention are in the form of a salt, preferably, the polypeptides are in the form of a pharmaceutically acceptable salt.
  • Suitable pharmaceutically acceptable acid addition salts include those derived from mineral acids, such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric, and sulphuric acids, and organic acids, such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, and arylsulphonic acids, for example, p-toluenesulphonic acid.
  • mineral acids such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric, and sulphuric acids
  • organic acids such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, and arylsulphonic acids, for example, p-toluenesulphonic acid.
  • modified TCR, polypeptide, and/or protein of the invention can be obtained by methods known in the art. Suitable methods of de novo synthesizing polypeptides and proteins are described in references, such as Chan et al., Fmoc Solid Phase Peptide Synthesis, Oxford University Press, Oxford, United Kingdom, 2005; Peptide and Protein Drug Analysis, ed. Reid, R,, Marcel Dekker, Inc., 2000; Epitope Mapping, ed. Westwoood et at, Oxford University Press, Oxford, United Kingdom, 2000; and U.S. Patent No. 5,449,752.
  • polypeptides and proteins can be recombinantly produced using the nucleic acids described herein using standard recombinant methods. See, for instance, Sambrook et al., Molecular Cloning: A Laboratory Manual, 3 rd ed., Cold Spring Harbor Press, Cold Spring Harbor, NY 2001; and Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates and John Wiley & Sons, NY, 1994.
  • TCRs, polypeptides, and proteins of the invention can be isolated and/or purified from a source, such as a plant, a bacterium, an insect, a mammal, e.g., a rat, a human, etc. Methods of isolation and purification are well-known in the art.
  • a source such as a plant, a bacterium, an insect, a mammal, e.g., a rat, a human, etc. Methods of isolation and purification are well-known in the art.
  • the TCRs, polypeptides, and/or proteins described herein can be commercially synthesized by companies, such as Synpep (Dublin, CA), Peptide Technologies Corp. (Gaithersburg, MD), and Multiple Peptide Systems (San Diego, CA).
  • the inventive TCRs, polypeptides, and proteins can be synthetic, recombinant, isolated, and/or purified.
  • conjugates e.g., bioconjugates, comprising any of the inventive modified TCRs, polypeptides, or proteins (including any of the functional portions or variants thereof), nucleic acids, recombinant expression vectors, host cells, populations of host cells, or antibodies, or antigen binding portions thereof.
  • Conjugates, as well as methods of synthesizing conjugates in general, are known in the art (See, for instance, Hudecz, F., Methods MoL Biol. 298: 209-223 (2005) and Kirin et al., Inorg Chem. 44(15): 5405-5415 (2005)).
  • nucleic acid comprising a nucleotide sequence encoding any of the modified TCRs, polypeptides, or proteins described herein (including functional portions and functional variants thereof).
  • nucleic acid includes “polynucleotide,” “oligonucleotide,” and “nucleic acid molecule,” and generally means a polymer of DNA or RNA 9 which can be single- stranded or double-stranded, synthesized or obtained (e.g., isolated and/or purified) from natural sources, which can contain natural, non-natural or altered nucleotides, and which can contain a natural, non-natural or altered internucleotide linkage, such as a phosphoroamidate linkage or a phosphorothioate linkage, instead of the phosphodiester found between the nucleotides of an unmodified oligonucleotide.
  • the nucleic acid does not comprise any insertions, deletions, inversions, and/or substitutions. However, it may be suitable in some instances, as discussed herein, for the nucleic acid to comprise one or more insertions, deletions, inversions, and/or substitutions.
  • the nucleic acids of the invention are recombinant.
  • the term "recombinant" refers to (i) molecules that are constructed outside living cells by joining natural or synthetic nucleic acid segments to nucleic acid molecules that can replicate in a living cell, or (ii) molecules that result from the replication of those described in (i) above.
  • the replication can be in vitro replication or in vivo replication.
  • the nucleic acids can be constructed based on chemical synthesis and/or enzymatic ligation reactions using procedures known in the art. See, for example, Sambrook et al., supra, and Ausubel et al., supra.
  • a nucleic acid can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed upon hybridization (e.g., phosphorothioate derivatives and acridme substituted nucleotides).
  • modified nucleotides that can be used to generate the nucleic acids include, but are not limited to, 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxymethyl) uracil, 5- carboxymethylammomethyI-2-thiouridine, 5 -carboxymethylaminomethyluracil , dihydrouracil, beta-D-galactosylqueosine, inosine, N 6 -isopentenyladenine, 1 -methyl guanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N -substituted adenine, 7-methylguanine, 5-methylaminomethyluracil, 5- methoxyaminomethyl-2-thiouracil, beta-D
  • the nucleic acid can comprise any nucleotide sequence which encodes any of the modified TCRs, polypeptides, or proteins, or functional portions or functional variants thereof.
  • the nucleic acid can comprise a nucleotide sequence comprising SEQ ID NO: 22 to 24, 28, 29, 32, or 33.
  • the nucleotide sequence alternatively can comprise a nucleotide sequence which is degenerate to any of these sequences or a combination of degenerate sequences.
  • the nucleotide sequence can comprise a nucleotide sequence comprising a nucleotide sequence degenerate to SEQ ID NO: 22 and a nucleotide sequence degenerate to SEQ ID NO: 23 or comprising a nucleotide sequence degenerate to SEQ ID NO: 23 and a nucleotide sequence degenerate to SEQ ID NO: 25.
  • the invention also provides an isolated or purified nucleic acid comprising a nucleotide sequence which is complementary to the nucleotide sequence of any of the nucleic acids described herein or a nucleotide sequence which hybridizes under stringent conditions to the nucleotide sequence of any of the nucleic acids described herein.
  • the nucleotide sequence which hybridizes under stringent conditions preferably hybridizes under high stringency conditions.
  • high stringency conditions is meant that the nucleotide sequence specifically hybridizes to a target sequence (the nucleotide sequence of any of the nucleic acids described herein) in an amount that is detectably stronger than non-specific hybridization.
  • High stringency conditions include conditions which would distinguish a polynucleotide with an exact complementary sequence, or one containing only a few scattered mismatches from a random sequence that happened to have a few small regions (e.g., 3-10 bases) that matched the nucleotide sequence.
  • Such small regions of complementarity are more easily melted than a full-length complement of 14-17 or more bases, and high stringency hybridization makes them easily distinguishable.
  • Relatively high stringency conditions would include, for example, low salt and/or high temperature conditions, such as provided by about 0.02-0.1 M NaCl or the equivalent, at temperatures of about 50-70 0 C.
  • Such high stringency conditions tolerate little, if any, mismatch between the nucleotide sequence and the template or target strand, and are particularly suitable for detecting expression of any of the inventive TCRs. It is generally appreciated that conditions can be rendered more stringent by the addition of increasing amounts of formamide.
  • the nucleic acids of the invention can be incorporated into a recombinant expression vector.
  • the invention provides recombinant expression vectors comprising any of the nucleic acids of the invention.
  • the term "recombinant expression vector” means a genetically-modified oligonucleotide or polynucleotide construct that permits the expression of an mRNA, protein, polypeptide, or peptide by a host cell, when the construct comprises a nucleotide sequence encoding the mRNA, protein, polypeptide, or peptide, and the vector is contacted with the cell under conditions sufficient to have the mRNA, protein, polypeptide, or peptide expressed within the cell.
  • the vectors of the invention are not naturally-occurring as a whole. However, parts of the vectors can be naturally-occurring.
  • the inventive recombinant expression vectors can comprise any type of nucleotides, including, but not limited to DNA and RNA, which can be single- stranded or double-stranded, synthesized or obtained in part from natural sources, and which can contain natural, non-natural or altered nucleotides.
  • the recombinant expression vectors can comprise naturally-occurring, non-naturally-occuring internucleotide linkages, or both types of linkages.
  • the non-naturally occurring or altered nucleotides or internucleotide linkages does not hinder the transcription or replication of the vector.
  • the recombinant expression vector of the invention can be any suitable recombinant expression vector, and can be used to transform or transfect any suitable host. Suitable vectors include those designed for propagation and expansion or for expression or both, such as plasmids and viruses.
  • the vector can be selected from the group consisting of the pUC series (Fermentas Life Sciences), the pBluescript series (Stratagene, LaJolla, CA), the pET series (Novagen, Madison, WI), the pGEX series (Pharmacia Biotech, Uppsala, Sweden), and the pEX series (Clontech, Palo Alto, CA).
  • Bacteriophage vectors such as ⁇ GTIO, ⁇ GTl 1, ⁇ ZapII (Stratagene), ⁇ EMBL4, and ⁇ NMl 149, also can be used.
  • plant expression vectors include pBIOl, pBI101.2, pBI101.3, pBI121 and pBIN19 (Clontech).
  • animal expression vectors include pEUK-Cl f pMAM and pMAMneo (Clontech).
  • the recombinant expression vector is a viral vector, e.g., a retroviral vector.
  • the recombinant expression vectors of the invention can be prepared using standard recombinant DNA techniques described in, for example, Sambrook et al., supra, and Ausubel et al., supra.
  • Constructs of expression vectors, which are circular or linear, can be prepared to contain a replication system functional in a prokaryotic or eukaryotic host cell.
  • Replication systems can be derived, e.g., from CoIEl, 2 ⁇ plasmid, ⁇ , SV40, bovine papilloma virus, and the like.
  • the recombinant expression vector comprises regulatory sequences, such as transcription and translation initiation and termination codons, which are specific to the type of host (e.g., bacterium, fungus, plant, or animal) into which the vector is to be introduced, as appropriate and taking into consideration whether the vector is DNA- or RNA- based.
  • regulatory sequences such as transcription and translation initiation and termination codons, which are specific to the type of host (e.g., bacterium, fungus, plant, or animal) into which the vector is to be introduced, as appropriate and taking into consideration whether the vector is DNA- or RNA- based.
  • the recombinant expression vector can include one or more marker genes, which allow for selection of transformed or transfected hosts.
  • Marker genes include biocide resistance, e.g., resistance to antibiotics, heavy metals, etc., complementation in an auxotrophic host to provide prototrophy, and the like.
  • Suitable marker genes for the inventive expression vectors include, for instance, neomycin/G418 resistance genes, hygromycin resistance genes, histidinol resistance genes, tetracycline resistance genes, and arnpicillin resistance genes.
  • the recombinant expression vector can comprise a native or normative promoter operably linked to the nucleotide sequence encoding the modified TCR, polypeptide, or protein (including functional portions and functional variants thereof), or to the nucleotide sequence which is complementary to or which hybridizes to the nucleotide sequence encoding the modified TCR, polypeptide, or protein.
  • promoters e.g., strong, weak, inducible, tissue-specific and developmental-specific, is within the ordinary skill of the artisan.
  • the combining of a nucleotide sequence with a promoter is also within the skill of the artisan.
  • the promoter can be a non-viral promoter or a viral promoter, e.g., a cytomegalovirus (CMV) promoter, an SV40 promoter, an RSV promoter, and a promoter found in the long-terminal repeat of the murine stem cell virus.
  • CMV cytomegalovirus
  • the inventive recombinant expression vectors can be designed for either transient expression, for stable expression, or for both. Also, the recombinant expression vectors can be made for constitutive expression or for inducible expression. [0063] Further, the recombinant expression vectors can be made to include a suicide gene.
  • suicide gene refers to a gene that causes the cell expressing the suicide gene to die.
  • the suicide gene can be a gene that confers sensitivity to an agent, e.g., a drug, upon the cell in which the gene is expressed, and causes the cell to die when the cell is contacted with or exposed to the agent.
  • Suicide genes are known in the art (see, for example, Suicide Gene Therapy: Methods and Reviews, Springer, Caroline J. (Cancer Research UK Centre for Cancer Therapeutics at the Institute of Cancer Research, Sutton, Surrey, UK), Humana Press, 2004) and include, for example, the Herpes Simplex Virus (HSV) thymidine kinase (TK) gene, cytosine daminase, purine nucleoside phosphorylase, and nitroreductase.
  • HSV Herpes Simplex Virus
  • TK thymidine kinase
  • the invention further provides a host cell comprising any of the recombinant expression vectors described herein.
  • the term "host cell” refers to any type of cell that can contain the inventive recombinant expression vector.
  • the host cell can be a eukaryotic cell, e.g., plant, animal, fungi, or algae, or can be a prokaryotic cell, e.g., bacteria or protozoa.
  • the host cell can be a cultured cell or a primary cell, i.e., isolated directly from an organism, e.g., a human.
  • the host cell can be an adherent cell or a suspended cell, i.e., a cell that grows in suspension.
  • Suitable host cells are known in the art and include, for instance, DH5 ⁇ E. coli cells, Chinese hamster ovarian cells, monkey VERO cells, COS cells, HEK293 cells, and the like.
  • the host cell is preferably a prokaryotic cell, e.g., a DH5 ⁇ cell
  • the host cell is preferably a mammalian cell. Most preferably, the host cell is a human cell. While the host cell can be of any cell type, can originate from any type of tissue, and can be of any developmental stage, the host cell preferably is a peripheral blood lymphocyte (PBL). More preferably, the host cell is a T cell.
  • PBL peripheral blood lymphocyte
  • the T cell can be any T cell, such as a cultured T cell, e.g., a primary T cell, or a T cell from a cultured T cell line, e.g., Jurkat, SupTl, etc., or a T cell obtained from a mammal. If obtained from a mammal, the T cell can be obtained from numerous sources, including but not limited to blood, bone marrow, lymph node, the thymus, or other tissues or fluids. T cells can also be enriched for or purified.
  • the T cell is a human T cell. More preferably, the T cell is a T cell isolated from a human.
  • the T cell can be any type of T cell and can be of any developmental stage, including but not limited to, CD4 + /CD8 + double positive T cells, CD4 + helper T cells, e.g., Th 1 and Th 2 cells, CD8 + T cells (e.g., cytotoxic T cells), peripheral blood mononuclear cells (PBMCs), peripheral blood leukocytes (PBLs), tumor infiltrating cells (TILs), memory T cells, naive T cells, and the like.
  • the T cell is a CD8 + T cell or a CD4 + T cell.
  • the population of cells can be a heterogeneous population comprising the host cell comprising any of the recombinant expression vectors described, in addition to at least one other cell, e.g., a host cell (e.g., a T cell), which does not comprise any of the recombinant expression vectors, or a cell other than a T cell, e.g., a B cell, a macrophage, a neutrophil, an erythrocyte, a hepatocyte, an endothelial cell, an epithelial cells, a muscle cell, a brain cell, etc.
  • a host cell e.g., a T cell
  • a cell other than a T cell e.g., a B cell, a macrophage, a neutrophil, an erythrocyte, a hepatocyte, an endothelial cell, an epithelial cells, a muscle cell, a brain cell, etc.
  • the population of cells can be a substantially homogeneous population, in which the population comprises mainly of host cells (e.g., consisting essentially of) comprising the recombinant expression vector.
  • the population also can be a clonal population of cells, in which all cells of the population are clones of a single host cell comprising a recombinant expression vector, such that all cells of the population comprise the recombinant expression vector.
  • the population of cells is a clonal population comprising host cells comprising a recombinant expression vector as described herein.
  • the invention further provides an antibody, or antigen binding portion thereof, which specifically binds to an epitope of the modified TCR of the invention, wherein the epitope comprises the amino acid substitutions.
  • the antibody can be any type of immunoglobulin that is known in the art.
  • the antibody can be of any isotype, e.g., IgA 5 IgD 5 IgE, IgG, IgM, etc.
  • the antibody can be monoclonal or polyclonal.
  • the antibody can be a naturally-occurring antibody, e.g., an antibody isolated and/or purified from a mammal, e.g., mouse, rabbit, goat, horse, chicken, hamster, human, etc.
  • the antibody can be a genetically-engineered antibody, e.g., a humanized antibody or a chimeric antibody.
  • the antibody can be in monomeric or polymeric form.
  • the antibody can have any level of affinity or avidity for the functional portion of the inventive modified TCR.
  • the antibody is specific for the epitope of the inventive modified TCR comprising the amino acid substitutions, such that there is minimal cross-reaction with other peptides or proteins.
  • Methods of testing antibodies for the ability to bind to any functional portion of the inventive modified TCR include any antibody-antigen binding assay, such as, for example, radioimmunoassay (RIA), ELISA, Western blot, immunoprecipitation, and competitive inhibition assays (see, e.g., Jane way et al. s infra, and U.S. Patent Application Publication No. 2002/0197266 Al).
  • RIA radioimmunoassay
  • ELISA ELISA
  • Western blot Western blot
  • immunoprecipitation immunoprecipitation
  • competitive inhibition assays see, e.g., Jane way et al. s infra, and U.S. Patent Application Publication No. 2002/0197266 Al.
  • Suitable methods of making antibodies are known in the art. For instance, standard hybridoma methods are described in, e.g., K ⁇ hler and Milstein, Eur. J. Immunol., 5, 51 1-519 (1976), Harlow and Lane (eds.), Antibodies: A Laboratory Manual, CSH Press (1988), and CA. Janeway et al. (eds.), Immunobiology, 5 th Ed., Garland Publishing, New York, NY (2001)). Alternatively, other methods, such as EBV-hybridoma methods (Haskard and Archer, J. Immunol.
  • Phage display furthermore can be used to generate the antibody of the invention.
  • phage libraries encoding antigen-binding variable (V) domains of antibodies can be generated using standard molecular biology and recombinant DNA techniques (see, e.g., Sambrook et al. (eds.), Molecular Cloning, A Laboratory Manual, 3 rd Edition, Cold Spring Harbor Laboratory Press, New York (2001)). Phage encoding a variable region with the desired specificity are selected for specific binding to the desired antigen, and a complete or partial antibody is reconstituted comprising the selected variable domain.
  • Nucleic acid sequences encoding the reconstituted antibody are introduced into a suitable cell line, such as a myeloma cell used for hybridoma production, such that antibodies having the characteristics of monoclonal antibodies are secreted by the cell (see, e.g., Janeway et al., supra, Huse et al., supra, and U.S. Patent 6,265,150).
  • a suitable cell line such as a myeloma cell used for hybridoma production, such that antibodies having the characteristics of monoclonal antibodies are secreted by the cell (see, e.g., Janeway et al., supra, Huse et al., supra, and U.S. Patent 6,265,150).
  • Antibodies can be produced by transgenic mice that are transgenic for specific heavy and light chain immunoglobulin genes. Such methods are known in the art and described in, for example U.S. Patents 5,545 P 806 and 5,569,825, and Janeway et al., supra. [0072] Methods for generating humanized antibodies are well known in the art and are described in detail in, for example, Janeway et al, supra, U.S. Patents 5,225,539, 5,585,089 and 5,693,761, European Patent No. 0239400 Bl, and United Kingdom Patent No. 2188638. Humanized antibodies can also be generated using the antibody resurfacing technology described in U.S.
  • the invention also provides antigen binding portions of any of the antibodies described herein.
  • the antigen binding portion can be any portion that has at least one antigen binding site, such as Fab, F(ab') 2 , dsFv, sFv, diabodies, and triabodies.
  • a single-chain variable region fragment (sFv) antibody fragment which consists of a truncated Fab fragment comprising the variable (V) domain of an antibody heavy chain linked to a V domain of a light antibody chain via a synthetic peptide, can be generated using routine recombinant DNA technology techniques (see, e.g., Janeway et al., supra).
  • disulfide-stabilized variable region fragments (dsFv) can be prepared by recombinant DNA technology (see, e.g., Reiter et al. s Protein Engineering, 7, 697-704 (1994)).
  • Antibody fragments of the invention are not limited to these exemplary types of antibody fragments.
  • the antibody, or antigen binding portion thereof can be modified to comprise a detectable label, such as, for instance, a radioisotope, a fluorophore (e.g., fluorescein isothiocyanate (FITC), phycoerythrin (PE)), an enzyme (e.g., alkaline phosphatase, horseradish peroxidase), and element particles (e.g., gold particles).
  • FITC fluorescein isothiocyanate
  • PE phycoerythrin
  • an enzyme e.g., alkaline phosphatase, horseradish peroxidase
  • element particles e.g., gold particles.
  • inventive modified TCRs, polypeptides, proteins, (including functional portions and functional variants thereof), nucleic acids, recombinant expression vectors, host cells (including populations thereof), and antibodies (including antigen binding portions thereof) can be isolated and/or purified.
  • isolated means having been removed from its natural environment.
  • purified means having been increased in purity, wherein “purity” is a relative term, and not to be necessarily construed as absolute purity.
  • the purity can be at least about 50%, can be greater than 60%, 70% or 80%, or can be 100%.
  • inventive modified TCRs, polypeptides, proteins (including functional portions and variants thereof);, nucleic acids, recombinant expression vectors, host cells (including populations thereof), and antibodies (including antigen binding portions thereof), all of which are collectively referred to as "inventive TCR materials" hereinafter, can be formulated into a composition, such as a pharmaceutical composition, hi this regard, the invention provides a pharmaceutical composition comprising any of the modified TCRs, polypeptides, proteins, functional portions, functional variants, nucleic acids, expression vectors, host cells (including populations thereof), and antibodies (including antigen binding portions thereof), and a pharmaceutically acceptable carrier.
  • inventive pharmaceutical compositions containing any of the inventive TCR materials can comprise more than one inventive TCR material, e.g., a polypeptide and a nucleic acid, or two or more different modified TCRs.
  • the pharmaceutical composition can comprise an inventive TCR material in combination with another pharmaceutically active agents or drugs, such as a chemotherapeutic agents, e.g., asparaginase, busulfan, carboplatin, cisplatin, daunorubicin, doxorubicin, fluorouracil, gemcitabine, hydroxyurea, methotrexate, paclitaxel, rituximab, vinblastine, vincristine, etc.
  • chemotherapeutic agents e.g., asparaginase, busulfan, carboplatin, cisplatin, daunorubicin, doxorubicin, fluorouracil, gemcitabine, hydroxyurea, methotrexate, paclitaxel, rituxim
  • the pharamaceutically acceptable carrier can be any of those conventionally used and is limited only by chemico-physical considerations, such as solubility and lack of reactivity with the active compound(s), and by the route of administration.
  • the pharmaceutically acceptable carriers described herein, for example, vehicles, adjuvants, excipients, and diluents, are well-known to those skilled in the art and are readily available to the public. It is preferred that the pharmaceutically acceptable carrier be one which is chemically inert to the active agent(s) and one which has no detrimental side effects or toxicity under the conditions of use.
  • compositions of the invention are exemplary and are in no way limiting. More than one route can be used to administer the inventive TCR materials, and in certain instances, a particular route can provide a more immediate and more effective response than another route.
  • Topical formulations are well-known to those of skill in the art. Such formulations are particularly suitable in the context of the invention for application to the skin.
  • Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the inventive TCR material dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions.
  • Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant.
  • Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and corn starch.
  • Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, macrocrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and other pharmacologically compatible excipients.
  • Lozenge forms can comprise the inventive TCR material in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the inventive TCR material in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to, such excipients as are known in the art.
  • an inert base such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to, such excipients as are known in the art.
  • the inventive TCR material can be made into aerosol formulations to be administered via inhalation.
  • aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. Such spray formulations also may be used to spray mucosa.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • the inventive TCR material can be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol or hexadecyl alcohol, a glycol, such as propylene glycol or polyethylene glycol, dimethylsulfoxide, glycerol, ketals such as 2,2- dimethyl-l,3-dioxolane-4-methanol, ethers, poly(ethyleneglycol) 400, oils, fatty acids, fatty acid esters or glycerides, or acetylated fatty acid glycerides with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose.
  • a pharmaceutically acceptable surfactant such as a soap or a detergent, suspending
  • Oils which can be used in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, com, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
  • Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts
  • suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfo succinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl- ⁇ -aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (e) mixtures thereof.
  • the parenteral formulations will typically contain from about 0.5% to about 25% by weight of the inventive TCR material in solution. Preservatives and buffers may be used. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations will typically range from about 5% to about 15% by weight. Suitable surfactants include polyethylene glycol sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • HLB hydrophile-lipophile balance
  • parenteral formulations can be presented in unit-dose or multi-dose sealed containers, such as ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use.
  • sterile liquid excipient for example, water
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • injectable formulations are in accordance with the invention.
  • the requirements for effective pharmaceutical carriers for injectable compositions are well-known to those of ordinary skill in the art (see, e.g., Pharmaceutics and Pharmacy Practice, J.B. Lippincott Company, Philadelphia, PA, Banker and Chalmers, eds., pages 238-250 (1982), m ⁇ ASHP Handbook on Injectable Drugs, Toissel, 4th ed., pages 622-630 (1986)).
  • administering cells e.g., dendritic cells
  • the cells are administered via injection.
  • inventive TCR materials can be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases.
  • bases such as emulsifying bases or water-soluble bases.
  • Formulations suitable for vaginal administration can be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
  • the inventive TCR materials of the invention can be formulated as inclusion complexes, such as cyclodextrin inclusion complexes, or liposomes.
  • the amount or dose of the inventive TCR material administered should be sufficient to effect, e.g., a therapeutic or prophylactic response, in the subject or animal over a reasonable time frame.
  • the dose of the inventive TCR material should be sufficient to bind to antigen, or detect, treat or prevent disease in a period of from about 2 hours or longer, e.g., 12 to 24 or more hours, from the time of administration. In certain embodiments, the time period could be even longer.
  • the dose will be determined by the efficacy of the particular inventive TCR material and the condition of the animal (e.g., human), as well as the body weight of the animal (e.g., human) to be treated.
  • Many assays for determining an administered dose are known in the art.
  • an assay which comprises comparing the extent to which target cells are lysed or IFN- ⁇ is secreted by T cells expressing the inventive modified TCR, polypeptide, or protein upon administration of a given dose of such T cells to a mammal among a set of mammals of which is each given a different dose of the T cells, could be used to determine a starting dose to be administered to a mammal.
  • the extent to which target cells are lysed or IFN- ⁇ is secreted upon administration of a certain dose can be assayed by methods known in the art, including, for instance, the methods described herein as Example 2.
  • the dose of the inventive TCR material also will be determined by the existence, nature and extent of any adverse side effects that might accompany the administration of a particular inventive TCR material. Typically, the attending physician will decide the dosage of the inventive TCR material with which to treat each individual patient, taking into consideration a variety of factors, such as age, body weight, general health, diet, sex, inventive TCR material to be administered, route of administration, and the severity of the condition being treated.
  • the dose of the inventive TCR material can be about 0.001 to about 1000 mg/kg body weight of the subject being treated/day, from about 0.01 to about 10 mg/kg body weight/day, about 0.01 mg to about 1 mg/kg body weight/day.
  • inventive TCR materials of the invention can be modified in any number of ways, such that the therapeutic or prophylactic efficacy of the inventive TCR materials is increased through the modification.
  • inventive TCR materials can be conjugated either directly or indirectly through a linker to a targeting moiety.
  • the practice of conjugating compounds, e.g., inventive TCR materials, to targeting moieties is known in the art. See, for instance, Wadwa et al., J Drug Targeting 3: 111 (1995) and U.S. Patent No. 5,087,616.
  • targeting moiety refers to any molecule or agent that specifically recognizes and binds to a cell-surface receptor, such that the targeting moiety directs the delivery of the inventive TCR materials to a population of cells on which surface the receptor is expressed.
  • Targeting moieties include, but are not limited to, antibodies, or fragments thereof, peptides, hormones, growth factors, cytokines, and any other natural or non-natural ligands, which bind to cell surface receptors (e.g., Epithelial Growth Factor Receptor (EGFR), T-cell receptor (TCR), B- cell receptor (BCR), CD28, Platelet-derived Growth Factor Receptor (PDGF), nicotinic acetylcholine receptor (nAChR), etc.).
  • EGFR Epithelial Growth Factor Receptor
  • TCR T-cell receptor
  • BCR B- cell receptor
  • CD28 CD28
  • PDGF Platelet-derived Growth Factor Receptor
  • nAChR nicotinic acetylcholine receptor
  • inventive TCR materials which are not necessary for the function of the inventive TCR materials, are ideal sites for attaching a linker and/or a targeting moiety, provided that the linker and/or targeting moiety, once attached to the inventive TCR materials, do(es) not interfere with the function of the inventive TCR materials, i.e., the ability to bind to antigen, or to detect, treat, or prevent disease.
  • inventive TCR materials can be modified into a depot form, such that the manner in which the inventive TCR materials is released into the body to which it is administered is controlled with respect to time and location within the body (see, for example, U.S. Patent No. 4,450,150).
  • inventive TCR materials can be, for example, an implantable composition comprising the inventive TCR materials and a porous or non-porous material, such as a polymer, wherein the inventive TCR materials is encapsulated by or diffused throughout the material and/or degradation of the non-porous material.
  • the depot is then implanted into the desired location within the body and the inventive TCR materials are released from the implant at a predetermined rate.
  • inventive pharmaceutical compositions, modified TCRs, polypeptides, proteins, nucleic acids, recombinant expression vectors, host cells, or populations of cells can be used in methods of treating or preventing a disease in a host.
  • the inventive modified TCRs are believed to have enhanced biological activity, e.g., ability to recognize antigen, such that the modified TCR (or related inventive polypeptide or protein) when expressed by a cell is able to mediate a stronger immune response against the cell expressing the antigen for which the modified TCR is specific.
  • the invention provides a method of treating or preventing a disease in a host, comprising administering to the host any of the pharmaceutical compositions in an amount effective to treat or prevent the disease in the host.
  • the disease can be any disease involving an antigen, e.g., an infectious disease, an autoimmune disease, a cancer.
  • infectious disease means a disease that can be transmitted from person to person or from organism to organism, and is caused by a microbial agent (e.g., common cold). Infectious diseases are known in the art and include, for example, hepatitis, sexually transmitted diseases (e.g., Chlamydia, gonorrhea), tuberculosis, HIV/ AIDS, diphtheria, hepatitis B, hepatitis C, cholera, and influenza.
  • a microbial agent e.g., common cold.
  • Infectious diseases include, for example, hepatitis, sexually transmitted diseases (e.g., Chlamydia, gonorrhea), tuberculosis, HIV/ AIDS, diphtheria, hepatitis B, hepatitis C, cholera, and influenza.
  • autoimmune disease refers to a disease in which the body produces an immunogenic (i.e., immune system) response to some constituent of its own tissue. In other words the immune system loses its ability to recognize some tissue or system within the body as "self and targets and attacks it as if it were foreign. Autoimmune diseases can be classified into those in which predominantly one organ is affected (e.g., hemolytic anemia and anti-immune thyroiditis), and those in which the autoimmune disease process is diffused through many tissues (e.g., systemic lupus erythematosus). For example, multiple sclerosis is thought to be caused by T cells attacking the sheaths that surround the nerve fibers of the brain and spinal cord.
  • immunogenic i.e., immune system
  • Autoimmune diseases include, for instance, Hashimoto's thyroiditis, Grave's disease, lupus, multiple sclerosis, rheumatic arthritis, hemolytic anemia, anti-immune thyroiditis, systemic lupus erythematosus, celiac disease, Crohn's disease, colitis, diabetes, scleroderma, psoriasis, and the like.
  • the cancer can be any cancer, including any of acute lymphocytic cancer, acute myeloid leukemia, alveolar rhabdomyosarcoma, bone cancer, brain cancer, breast cancer, cancer of the anus, anal canal, or anorectum, cancer of the eye, cancer of the intrahepatic bile duct, cancer of the joints, cancer of the neck, gallbladder, or pleura, cancer of the nose, nasal cavity, or middle ear, cancer of the oral cavity, cancer of the vulva, chronic lymphocytic leukemia, chronic myeloid cancer, colon cancer, esophageal cancer, cervical cancer, gastrointestinal carcinoid tumor.
  • the cancer is melanoma.
  • inventive methods can provide any amount of any level of treatment or prevention of cancer in a mammal.
  • the treatment or prevention provided by the inventive method can include treatment or prevention of one or more conditions or symptoms of the disease, e.g., cancer, being treated or prevented.
  • prevention can encompass delaying the onset of the disease, or a symptom or condition thereof.
  • the method comprises (i) contacting a sample comprising cells of the host with any of the inventive modified TCRs, polypeptides, proteins, nucleic acids, recombinant expression vectors, host cells, and populations of host cells described herein, thereby forming a complex between the antigen which is characteristic of the disease and the inventive modified TCR, polypeptide, protein, nucleic acid, recombinant expression vector, host cell, or population of cells, and (ii) detecting the complex, wherein detection of the complex is indicative of a diseased cell in the host.
  • the diseased cell can be any cell of any disease, which cell expresses an antigen that is characteristic of the disease.
  • the diseased cell can be a cancer cell or an infected cell, for example.
  • the diseased cell is a melanoma cell
  • the inventive modified TCR has antigenic specificity for an antigen that is characteristic of the disease to be treated, prevented, or detected. For instance, if the disease to be treated, prevented or detected is melanoma, the inventive modified TCR has antigenic specificity for a melanoma antigen, e.g., MART-I, NY-ESO-I, gplOO, etc. If a host cell or a population comprising at least one host cell is used in the method, the host cell desirably expresses a TCR having antigenic specificity for the antigen of the disease.
  • the nucleic acid or recombinant expression vector desirably encodes the modified TCR which has antigenic specificity for an antigen of the disease to be treated, prevented, or detected, such that expression of the nucleic acid or recombinant expression vector is achieved in a cell and the TCR expressed by the cell is capable of binding to the antigen of the disease.
  • the sample comprising cells of the host can be a sample comprising whole cells, lysates thereof, or a fraction of the whole cell lysates, e.g., a nuclear or cytoplasmic fraction, a whole protein fraction, or a nucleic acid fraction.
  • the cells can be any cells of the host, e.g., the cells of any organ or tissue, including blood cells.
  • the contacting step can take place in vitro or in vivo with respect to the host.
  • the contacting is an in vitro step.
  • detection of the complex can occur through any number of ways known in the art.
  • the inventive modified TCRs, polypeptides, proteins, nucleic acids, recombinant expression vectors, host cells, populations of cells, or antibodies, or antigen binding portions thereof, described herein can be labeled with a detectable label such as, for instance, a radioisotope, a fluorophore (e.g., fluorescein isothiocyanate (FITC), phycoerythrin (PE)), an enzyme (e.g., alkaline phosphatase, horseradish peroxidase), and element particles (e.g., gold particles).
  • a detectable label such as, for instance, a radioisotope, a fluorophore (e.g., fluorescein isothiocyanate (FITC), phycoerythrin (PE)), an enzyme (e.g., alkaline phosphatase, horseradish peroxidase), and element
  • the cells can be cells that are allogeneic or autologous to the host.
  • the cells are autologous to the host.
  • the host referred to herein can be any host.
  • the host is a mammal.
  • the term "mammal” refers to any mammal, including, but not limited to, mammals of the order Rodentia, such as mice and hamsters, and mammals of the order Logomorpha, such as rabbits. It is preferred that the mammals are from the order Carnivora, including Felines (cats) and Canines (dogs). It is more preferred that the mammals are from the order Artiodactyla, including Bovines (cows) and S wines (pigs) or of the order Perssodactyla, including Equines (horses). It is most preferred that the mammals are of the order Primates, Ceboids, or Simoids (monkeys) or of the order Anthropoids (humans and apes). An especially preferred mammal is the human.
  • a method of identifying a candidate adoptive immunotherapy TCR is also provided.
  • the method consists of (a) producing a nucleic acid encoding a modified TCR comprising a WT TCR amino acid sequence with no more than three amino acid substitutions, (b) expressing the nucleic acid in CD4+ T cells and CD8+ T cells, and (c) assaying the T cells for the ability to recognize target cells and for antigen specificity.
  • a candidate adoptive immunotherapy TCR is identified when the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize target cells when expressed in CD4+ T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD 8+ T cells.
  • candidate adoptive immunotherapy TCR refers to a modified TCR which comprises an amino acid sequence of a WT TCR with no more than three amino acid substitutions and, as compared to the WT TCR (i) has an enhanced ability recognize target cells when expressed in CD4+ T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD8+ T cells.
  • candidate cells are predicted to work well in the context of adoptive immunotherapy, such that the TCR, when expressed in T cells of a patient, will be able to treat or prevent a disease of the patient.
  • Producing a nucleic acid encoding a modified TCR can be carried out by any means known in the art.
  • Examples 2 to 4 describe methods of making such a nucleic acid in which a WT TCR sequence is used as a template for a PCR reaction which utilizes primers which encode the amino acid substitutions.
  • the resulting PCR product, which encodes the amino acid mutations is then in vitro transcribed, thereby producing an RNA encoding the modified TCR.
  • the nucleic acid can be produced by other means, e.g., site directed mutagenesis.
  • the amino acid substitutions preferably are conservative amino acid substitutions. More preferably, the amino acid substitutions are located within a CDR of the TCR, e.g., CDRl, CDR2, CDR3. Most preferably, the amino acid substitutions are located within the CDR2 of a p chain of the TCR. Also, with regard to this method, the modified TCR has at least twice the ability to recognize target cells as the WT TCR and exhibits this enhanced ability in at least two assays.
  • nucleic acids in cells including CD4+ T cells and CD8+ T cells
  • the nucleic acid is an RNA and the RNA is expressed in T cells by methods described in Example 2.
  • the T cells can be assayed for the ability to recognize target cells and for antigen specificity employing methods known in the art.
  • the T cells are assayed as described herein as Example 2.
  • TCRs with respect to the immature TCR sequence i.e., the TCR sequence with the leader sequence.
  • This example demonstrates a method of making a modified TCR of the invention.
  • the WT 1 G4 alpha and beta chains were isolated using RT-PCR. The RT reaction was carried out as described in Huang et al, J Immunol, 172: 6057-6064 (2004), and the fragments were amplified using a high fidelity DNA polymerase, Fusion (New England Biolabs) according to the manufacturer's instructions.
  • the WT 1G4 ⁇ chain was isolated by RT-PCR from an in vitro cultured tumor infiltrating lymphocytes (TIL) sample isolated from a melanoma patient using two primer pairs: the first pair (Primers A and B) was used to isolate the variable region sequence but also contained sequences corresponding to the appropriate J region, while the second primer pair (Primers C and D) was used to isolate the full length constant region but also encoded the J region to allow an overlap with the sequence amplified using the first primer pair.
  • TIL tumor infiltrating lymphocytes
  • the 1 G4 TR ⁇ chain was constructed by RT-PCR from an in vitro cultured TIL sample of using two primer pairs: the first pair (Primers G and H) was used to isolate the 1G4 ⁇ chain variable region sequence including part of the J region, while the second pair (Primers I and J) was used to isolate the 1G4 ⁇ chain constant region but also encoded the J region.
  • the full length construct encoding the ⁇ chain was generated by isolation of the two PCR products on an agarose gel, which were then mixed and used to carry out a second round of amplification using Primers K and L.
  • the sequences of the primers used to obtain the WT 1G4 nucleic acid are set forth in Table 1.
  • the nucleotide sequence of the 1G4 TCR obtained is set forth as SEQ ID NO: 25 (alpha chain) and SEQ ID NO: 21 (beta chain).
  • SEQ ID NO: 25 alpha chain
  • SEQ ID NO: 21 beta chain
  • Primers M and N were used to produce the 5' fragment for each mutant.
  • Primers O and P were used to produce the 3' fragment of 1G4 mutl
  • Primers Q and R were used to produce the V fragment of 1G4 mut2
  • Primers S and T were used to produce the 3 * fragment of 1G4 mut3.
  • the 1G4 PCR products were isolated on agarose gels and the final PCR product generated by amplification of the 5' fragment with the 3' fragments using the full length primer pair (Primers U and V).
  • the sequences of the primers used to generate the mutant beta chains of are set forth in Table 2.
  • the modified TCRs produced in Example 1 were tested for their ability recognize antigen when separately expressed in CD8 + T cells and CD4 + T cells.
  • PBMC from Patient KS were transfected as described in Zhao et al, (2006), supra with (i) RNA encoding the WT alpha chain of the 1G4 NY-ESO-I -specific TCR and (ii) RNA encoding the WT beta chain of the 1G4 TCR, RNA encoding one of the following mutant beta chains of 1G4: 1G4 mutl (G70A; SEQ ID NO: I) 5 1G4 mut2 (A71I, SEQ ID NO: 2), or 1G4 mut3 (G70A/A71I, SEQ ID NO: 3), or DNA encoding Green Fluorescence Protein (GFP).
  • GFP Green Fluorescence Protein
  • Transfected cells were washed and stimulated with or without (T alone) one of the following cells: T2+ pulsed with either 1 ⁇ M NY-ESO-I peptide SLLMWITQC (ESO; SEQ ID NO: 92) or g9-154 peptide KTWGQYWQV (g9-154; SEQ ID NO: 93) or melanoma cells: A375 (HLA-A2 + /NY-ESO-1 + ), 624.38 (HLA-A27NY-ESO-1 + ), 1363 (HL A- A2 + /N Y-ESO- I + ), 526 (HLA-A2 + /NY-ESO-1 " ), and SK (HLA- A2 + /NY-ESO-1 " ).
  • Responder cells (1x10 5 electroporated PBLs) and 1x10 5 stimulator cells (pulsed T2 cells or melanoma cells) were incubated in a 0.2-ml culture volume in individual wells of 96-well plates. Stimulator cells and responder cells were co-cultured for 16 to 24 h. Cytokine secretion of culture supernatants diluted to the linear range of the assay was measured using commercially available ELISA kits (IFN- ⁇ Endogen, Cambridge, MA).
  • modified TCRs of the invention have enhanced antigen specificity when expressed in CD4 + T cells and CDS + T cells.
  • This example demonstrates a method of making modified TCRs specific for the MART-I cancer antigen.
  • the DMF5 (F5) TCR which is specific for the cancer antigen MART-I was isolated and the nucleotide sequence of the F5 TCR obtained is set forth as SEQ ID NO: 30 (alpha chain) and SEQ ID NO: 31 (beta chain).
  • the F5 ⁇ chain was amplified from the cloned gene product (SEQ ID NO: 30) to generate RNA using Primers W and X.
  • the F5 beta chain was amplified from the cloned gene product (SEQ ID NO: 31) to generate RNA using Primers Y and Z.
  • Primers AA and AB were used to produce the 5' fragment for each mutant.
  • Primers AC and AD were used to produce the 3' fragment of F5 mutl
  • Primers AE and AF were used to produce the 3' fragment of F5mut2
  • Primers AG and AH were used to produce the 3' fragment of F5mut3
  • Primers AI and AJ were used to produce the 3' fragment of F5mut 4
  • Primers AK and AL were used to produce the 3' fragment of F5mut5.
  • the PCR products were isolated on agarose gels and the final PCR product generated by amplification of the 5' fragment with the 3' fragments using the full length primer pair (Primers AM and AN).
  • the sequences of the primers used to generate the mutant beta chains of are set forth in Table 5.
  • PBMC from Patients RS, KS, and HS were transfected and assayed as described in Example 2, except that RNA encoding either the WT or mutant F5 beta chains along with the WT alpha chains were used in transfections. Also, T2+ cells were pulsed with either 1 ⁇ g g9-154 peptide or MART-1 peptide AAGIGILTV (MART; SEQ ID NO: 94).
  • melanoma cells 526 (A2 + /MART-1 + ), 624 (A2 + /MART-1 + ), 1359 (A2 + /MART-1 + ), 1363 (A2 + /MART-1 + ), A375 (A2 + /MART-1 + ), 888, 888A2, 397 A2 (A2 + /MART-1 + ), 624.38 (A2 + /MART-1 + ), 1300 (A2 + /MART-1 + ), 397A24 (A2 + /MART-1 + ), 624.28 (A2 + /MART-1 + ), 526 (A2 + /NY-ESO-1 " ), SK23 (A2 + MART-1 + ), and SK (A2 " 7NY- ESO-1+).
  • the amounts of IFN- ⁇ (pg/ml) produced by transfected CD8 + T cells of Patient RS are shown in Table 6, while the amounts of IFN- ⁇ (pg/ml) produced by transfected CD4 + T cells of Patient RS are shown in Table 7.
  • the amounts of IFN- ⁇ (pg/ml) produced by transfected CD8 ⁇ T cells of Patient KS are shown in Table 8, while the amounts of IFN- ⁇ (pg/ml) produced by transfected CD4 + T cells of Patient KS are shown in Table 9.
  • the amounts of IFN- ⁇ (pg/ml) produced by transfected CD8 + T cells of Patient HS are shown in Table 10, while the amounts of IFN- ⁇ (pg/ml) produced by transfected CD4 + T cells of Patient HS are shown in Table 11.
  • the amounts of IFN- ⁇ (pg/ml) produced by transfected CD8 + T cells of Patient KG and MB expressing the modified TCR containing the triple mutant are shown in Tables 12 and 14, while the amounts of IFN- ⁇ (pg/ml) produced by transfected CD4 + T cells of Patient KG and MB expressing the triple mutant are shown in Tables 13 and 15.
  • This example demonstrates a method of making another set of modified TCRs specific for the MART-I cancer antigen.
  • the DMF4 (F4) TCR which is specific for the cancer antigen MART-I was isolated as described in Hughes et al., Hum. Gene Ther. 16: 457-472 (2005).
  • the nucleotide sequence of the F4 TCR obtained is set forth as SEQ ID NO: 26 (alpha chain) and SEQ ID NO: 27 (beta chain).
  • the DMF4 ⁇ chain was amplified using Primers AO and AP to generate RNA.
  • Primers AO and AP For the generation of each mutant F4 TCR, a 5' fragment and a 3' fragment, which encoded the amino acid substitution, were produce by PCR amplification of SEQ ID NO: 27.
  • Primers AQ and AR were used to generate the 5' fragment for each mutant.
  • Primers AS and AT were used to produce the 3' fragment of F4 mutl and Primers AU and AV were used to produce the 3' fragment of F4 mut2.
  • the 5' and 3' fragments were then gel purified, mixed, and amplified using the Primers AW and AX to generate the full length construct.
  • the ⁇ chain of the F4 TCR was amplified from the cloned gene product (SEQ ID NO: 26) to generate RNA using Primers AY and AZ.
  • the sequences of the primers used to generate mutant F4 TCRs are set forth in Table 16.
  • PBMC from Patient HS were obtained as described in Example 1.
  • Cells were transfected and assayed as described in Example 1 , except that nucleic acids encoding the WT F4 alpha chain and the mutant MART-I beta chains (F4 mut) were used and T2+ cells were pulsed with either 1 ⁇ M gplOO peptide KTWGQYWQV (gplOO; SEQ ID NO: 93) or MART-I peptide AAGIGILTV (MART; SEQ ID NO: 94).
  • melanoma cells with phenotypes in () were used: 397- A2 (A2 + /MART-1 + ), 624.38 (A2 + /MART-1 + ), 1300 (A2 + /MART-1 + ), SK23 (A2 + /MART-1 + ), 397-A24 (A2 + /MART-1 ' ), 624.28 (Al + MART-I + X and A375 (A2 + /MART-1 + ).
  • F4 mutants F4 mut 1 (G70A) and F4 mut 2 (G70A/V71 A) were TCRs that have enhanced ability to recognize tumor target cells without having a decrease in antigen specificity.
  • This example demonstrates a method of treating a disease in a host using the inventive TCRs.
  • Adoptive cell transfer is carried out as described in Morgan et al. (2006), supra. Briefly, PBLs are obtained by leukopheresis from a metastatic melanoma patient who is HLA-A*0201 positive. The PBLs are transduced with nucleic acids encoding a WT alpha chain, and a modified beta chain of a TCR specific for either NY-ESO-I or MART-I as described in Example 1. The patient receives the transduced cells at the time of maximum lymphodepletion. One month post-adoptive cell transfer, quantitative RT-PCR assays are carried out to reveal whether the presence of the modified TCRs are expressed by cells of the patient. Tumor regression also is analyzed by the methods described in Morgan et al. (2006), supra.

Abstract

The invention is directed to a modified T cell receptor (TCR) comprising an amino acid sequence of a wild-type (WT) TCR with no more than three amino acid substitutions, wherein the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize target cells when expressed by CD4+ T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD8+ T cells. Polypeptides, proteins, nucleic acids, recombinant expression vectors, host cells, populations of cells, antibodies, and pharmaceutical compositions related to the modified TCR also are part of the invention. Further, the invention is directed to methods of detecting a diseased cell in a host, methods of treating or preventing a disease in a host, and methods of identifying a candidate adoptive immunotherapy TCR.

Description

MODIFIED T CELL RECEPTORS AND RELATED MATERIALS AND METHODS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This patent application claims the benefit of U.S. Provisional Patent Application No. 60/847,447 filed September 26, 2006, which is incorporated by reference.
INCORPORATION-BY-REFERENCE OF MATERIAL SUBMITTED
ELECTRONICALLY
[0002] Incorporated by reference in its entirety herein is a computer-readable nucleotide/amino acid sequence listing submitted concurrently herewith and identified as follows: One 122,662 Byte ASCII (Text) file named "701866ST25.TXT," created on September 25, 2007.
BACKGROUND OF THE INVENTION
[0003] In an ongoing adoptive transfer clinical trial, cancer patients received autologous peripheral blood mononuclear cells (PBMC) that were transduced with nucleic acids encoding a T cell receptor (TCR) specific for the melanoma antigen MART-I. Thus far, two out of 17 patients have demonstrated an objective clinical response (Morgan et al., Sciencexpress, e-publication August 31, 2006). The results of this clinical trial demonstrate that normal autologous T lymphocytes, transduced ex vivo with anti-cancer antigen TCR genes and reinfused in cancer patients, can persist and express the transgene long term in vivo and mediate durable regression of large established tumors. However, approaches to increase the expression and function of the transgene are still needed.
[0004] There remains a need in. the art for modified TCRs for use in treating patients with disease. The invention provides such T cell receptors and methods of treating cancer.
BRIEF SUMMARY OF THE INVENTION
[0005] The invention provides a modified T cell receptor (TCR) comprising an amino acid sequence of a wild-type (WT) TCR with no more than three amino acid substitutions, wherein the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize target cells when expressed by CD4+ T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD 8 T cells. [0006] The invention also provides related polypeptides and proteins, as well as related nucleic acids, recombinant expression vectors, host cells, and populations of cells. Further provided by the invention are antibodies, or an antigen binding portion thereof, and pharmaceutical compositions relating to the modified TCRs of the invention. [0007] Further, the invention provides a method of detecting a diseased cell in a host, wherein the diseased cell expresses an antigen characteristic of a disease. The method comprises (a) contacting a sample comprising cells of the host with an inventive modified TCR, polypeptide, protein, nucleic acid, recombinant expression vector, host cell, or population of cells, thereby forming a complex between the modified TCR, polypeptide, protein, nucleic acid, recombinant expression vector, host cell, or population of cells and the antigen, and (b) detecting the complex, wherein detection of the complex is indicative of a diseased cell in the host
[0008] Also provided by the invention is a method of treating or preventing a disease in a host. The method comprises administering to the host an inventive pharmaceutical composition in an amount effective to treat or prevent the disease in the host. [0009] A method of identifying a candidate adoptive immunotherapy TCR is also provided. The method consists of (a) producing a nucleic acid encoding a modified TCR comprising a WT TCR amino acid sequence with no more than three amino acid substitutions, (b) expressing the nucleic acid in CD4+ T cells and CDS+ T cells, and (c) assaying the T cells for the ability to recognize target cells and for antigen specificity. A candidate adoptive immunotherapy TCR is identified when the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize target cells when expressed by CD4+ T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD8+ T cells..
DETAILED DESCRIPTION OF THE INVENTION
[0010] The invention provides a modified T cell receptor (TCR) comprising an amino acid sequence of a wild-type (WT) TCR with no more than three amino acid substitutions, (e.g., 1, 2, or 3) wherein the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize target cells when expressed by CD4+ T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD8+ T cells.
[0011] The term "wild-type" as used herein refers to a TCR which is naturally expressed by a T cell of a host, e.g., a TCR which is endogenous to a T cell of a host. Nucleic acids encoding wild-type TCRs are known in the art and can be obtained from the GenBank database of the National Center for Biotechnology Information (NCBI). For example, wild- type TCR nucleic acid sequences are available as GenBank Accession Nos. NGJ)01333, NGJ)OOOIo, NGJ)Ol 337, NGJ)01332, NGJ)Ol 336, AF043179, HSJ004872, Ml 3863, Z81026, AF397440, AY124793, and the like. Also, nucleic acids encoding wild-type TCRs can be obtained by methods known in the art, such as the PCR-based method described herein (see Example 1). The cells used to obtain the nucleic acids encoding the wild-type TCR are not limited to those used in Example 1. Rather, the cells can be any of the T cells described herein. In addition, the wild-type TCR can be entirely synthesized using oligonucleotide primers corresponding to the known sequence.
[0012] The modified TCR of the invention is marked by one or more enhanced biological properties when expressed in T cells. Specifically, the modified TCR, when compared to the corresponding WT TCR, (i) has an enhanced ability to recognize target cells when expressed by CD4+ T cells and (H) does not exhibit a decrease in antigen specificity when expressed by CDS+ T cells. The term "target cells" as used herein refers to cells which express and present, by way of an MHC molecule, the antigen which is specifically recognized by the modified TCR. The phrase "recognize target cells" as used herein refers to the ability of the modified TCR to immunologically recognize (e.g., specifically bind to) an antigen which is expressed and presented by a target cell. The term "enhanced" as used herein means that the modified TCR of the invention consistently exhibits at least twice the ability to recognize antigen expressed and presented by target cells, as compared to its WT counterpart. Preferably, the modified TCR of the invention recognizes antigen expressed and presented by target cells at least five times better than its WT counterpart. More preferably, the modified TCRs of the invention recognize antigen at least ten times better than its WT counterpart. Most preferably, the modified TCRs of the invention recognize antigen at least 2 times better than its WT counterpart.
[0013] The enhanced properties exhibited by the modified TCR consistently exhibits the properties, e.g., ability to recognize antigen expressed and presented by target cells. By "consistently" is meant that the modified TCR of the invention exhibits the enhanced properties in at least two assays. Preferably, the TCR of the invention exhibits the enhanced properties in at least three assays (e.g., in five or more assays, or in ten or more assays). [0014] The modified TCR of the invention exhibits an enhanced ability to recognize target cells without exhibiting a decrease in antigen specificity when expressed by CD8+ T cells. In this respect, the modified TCR is said to retain the antigen specificity of the counterpart WT TCR, e.g., recognizes only the antigen(s) recognized by the WT TCR and does not recognize antigen(s) that are not recognized by the WT TCR. [0015] Methods of testing a TCR for the ability to recognize target cells and for antigen specificity are known in the art. For instance, Clay et al., J Immunol, 163: 507-513 (1999), teaches methods of measuring the release of cytokines (e.g., interferon-γ, granulocyte/monocyte colony stimulating factor (GM-CSF)5 tumor necrosis factor a (TNF-α) or interleukin 2 (IL-2)). In addition, TCR function can be evaluated by measurement of cellular cytoxicity, as described in Zhao et al., J Immunol, 11 A: 4415-4423 (2005). Methods of testing a modified TCR for the ability to recognize target cells and for antigen specificity are described herein as Examples 2 to 4.
[0016] The modified TCR of the invention can have antigen specificity for any antigen. The phrase "have antigen specificity" as used herein means that the modified TCR can specifically bind to and immunologically recognize an antigen, such that binding of the TCR to the antigen elicits an immune response.
[0017] Preferably, the modified TCR of the invention has antigen specificity for an antigen which is characteristic of a disease. The disease can be any disease involving an antigen, as discussed herein, e.g., an infectious disease, an autoimmune disease, or a cancer. The antigen could be, for example, a viral antigen, a bacterial antigen, a cancer antigen, etc. [0018] More preferably, the modified TCR of the invention has antigen specificity for a cancer antigen. The term "cancer antigen" as used herein refers to any molecule (e.g., protein, peptide, lipid, carbohydrate, etc.) solely or predominantly expressed or over- expressed by a tumor cell or cancer cell, such that the antigen is associated with the tumor or cancer. The cancer antigen additionally can be expressed by normal, non-tumor, or noncancerous cells. However, in such a situation, the expression of the cancer antigen by normal, non-tumor, or non-cancerous cells is not as robust as the expression by tumor or cancer cells. In this regard, the tumor or cancer ceils can over-express the antigen or express the antigen at a significantly higher level, as compared to the expression of the antigen by normal, non-tumor, or non-cancerous cells. Also, the cancer antigen additionally can be expressed by cells of a different state of development or maturation. For instance, the cancer antigen can be additionally expressed by cells of the embryonic or fetal stage, which cells are not normally found in an adult host. Alternatively, the cancer antigen additionally can be expressed by stem cells or precursor cells, which cells are not normally found in an adult host. Another group of cancer antigens are represented by the differentiation antigens that are expressed in only a limited set of tissues in the adult, such as the melanocytes differentiation antigens, whose expression is limited to normal melanocytes. Although it is not known why these molecules elicit immune responses, the limited expression pattern of these proteins may allow these molecules to be recognized by the immune system. [0019] The cancer antigen can be an antigen expressed by any cell of any cancer or tumor, including the cancers and tumors described herein. The cancer antigen may be a cancer antigen of only one type of cancer or rumor, such that the cancer antigen is associated with or characteristic of only one type of cancer or tumor. Alternatively, the cancer antigen may be a cancer antigen (e.g., may be characteristic) of more than one type of cancer or tumor. For example, the cancer antigen may be expressed by both breast and prostate cancer cells and not expressed at all by normal, non-tumor, or non-cancer cells. In a preferred embodiment of the invention, the cancer antigen is a melanoma cancer antigen. In a more preferred embodiment, the cancer antigen is selected from the group consisting of NY-ESO- 1, MART-I, gplOO, p53, TRP-I, TRP-2, and tyrosinase. In a most preferred embodiment, the cancer antigen is NY-ESO-I or MART-I .
[0020] With respect to the inventive modified TCR, the amino acid substitution(s) can be located in any part of the amino acid sequence of the TCR. Preferably, the amino acid substitutions are located within the amino acid sequence of the complementary determining region (CDR) of the TCR, which are known in the art. These regions have been defined by elucidation of X-ray crystallographic structures, as well as sequence comparisons which have revealed the presence of regions of high diversity encoded in germline sequences, in the case of CDRl and CDF2 regions, as well as recombinational diversity, in the case of CDR3 region (Lefranc et al, Nucl Acids Res., 27, 209-212 (1999)). Preferably, the one, two, or three amino acid substitutions are located in the amino acid sequence of a CDR2 or CDR3 of the TCR (e.g., in the CDR2 region of the beta chain of the TCR). More preferably, the amino acid substitutions are located in the amino acid sequence of a CDR2, e.g., CDR2 of an α chain of a TCR or a β chain of a TCR. Most preferably, the amino acid substitutions are located in the CDR2 of a β chain of a TCR. For example, the modified TCR can comprise an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 16 to 121 in which the amino acid substitutions are located at the Xaa's of these sequences. [0021] The invention provides a modified TCR comprising two polypeptides (i.e., polypeptide chains), such as an α chain of a TCR, a β chain of a TCR5 a γ chain of a TCR, a δ chain of a TCR, or a combination thereof. The amino acid substitutions of the inventive modified TCRs can be located in the amino acid sequence of either or both polypeptide chains which constitute the TCR. In a preferred embodiment of the invention, the amino acid substitutions are located in the amino acid sequence of the β chain of the modified TCR. In a more preferred embodiment, the amino acid substitutions are located in only the amino acid sequence of the β chain of the modified TCR.
[0022] The amino acid substitutions of the inventive modified TCR are preferably conservative amino acid substitutions. Conservative amino acid substitutions are known in the art, and include amino acid substitutions in which one amino acid having certain physical and/or chemical properties is exchanged for another amino acid that has the same chemical or physical properties. For instance, the conservative amino acid substitution can be an acidic amino acid substituted for another acidic amino acid (e.g., Asp or GIu), an amino acid with a nonpolar side chain substituted for another amino acid with a nonpolar side chain (e.g., Ala, GIy, VaI, De, Leu, Met, Phe, Pro, Tip, VaI, etc.), a basic amino acid substituted for another basic amino acid (Lys, Arg, etc.), an amino acid with a polar side chain substituted for another amino acid with a polar side chain (Asn, Cys, GIn, Ser, Thr, Tyr, etc.), etc. Preferably, the conservative amino acid substitutions are selected from the group consisting of T→A, G^A, A→I, T→V, A→M, T→ϊ, A→V, T→G, and T-→S. [0023] The polypeptide chains of the inventive modified TCR can comprise any amino acid sequence, provided that the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize antigen when expressed by CD4+ T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD8+ T cells. [0024] In a preferred embodiment of the invention, the modified TCR comprises the amino acid sequence of SEQ ID NO: 8 with no more than three amino acid substitutions. In a more preferred embodiment of the invention, the modified TCR comprises the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 2, or SEQ ID NO: 3, each of which is an amino acid sequence of a variable region of a beta chain of a modified TCR which recognizes the cancer antigen NY-ESO-I. In a most preferred embodiment of the invention, the modified TCR comprises the amino acid sequence of SEQ ID NO: 14, 15, or 16, each of which is the amino acid sequence of a full-length beta chain comprising a constant region and a variable region. The modified TCR can additionally comprise the amino acid sequence of SEQ ID NO: 7, which is the WT alpha chain of the 1G4 NY-ESO-I -specific TCR. [0025] In another preferred embodiment, the modified TCR comprises the amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 12 with no more than three amino acid substitutions. In a more preferred embodiment, the modified TCR comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 4 to 6, 13, and 95 to 103, each of which is an amino acid sequence of a variable region of a beta chain of a modified TCR which recognizes the cancer antigen MART-I . In a most preferred embodiment of the invention, the modified TCR comprises the amino acid sequence of any of SEQ ID NOs: 17 to 20, 37 to 39, and 104 to 109, each of which is the amino acid sequence of a full-length beta chain comprising a constant region and a variable region. The modified TCR can additionally comprise the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: I L [0026] Preferably, when the modified TCR comprises the amino acid sequence of SEQ ID NO: 10 with no more than three amino acid substitutions, the modified TCR comprises the amino acid sequence of any of SEQ ID NOs: 4, 13, and 95 to 103. More preferably, the modified TCR comprises the amino acid sequence of any of SEQ ID NOs: 17, 20, 37 to 39, and 104 to 109. Also, in these instances, it is preferred that the modified TCR additionally comprises the amino acid sequence of SEQ ID NO: 9, which is the WT alpha chain of the F5 MART-I -specific TCR.
[0027] Likewise, when the modified TCR comprises the amino acid sequence of SEQ ID NO: 12 with no more than three amino acid substitutions, the modified TCR preferably comprises SEQ ID NO: 5 or SEQ ID NO: 6. More preferably, the modified TCR comprises the amino acid sequence of SEQ ID NO: 18 or 19. Also, in these instances, it is preferred that the modified TCR additionally comprises the amino acid sequence of SEQ ID NO: 11 , which is the WT alpha chain of the F4 MART-I -specific TCR.
[0028] In a preferred embodiment, the modified TCR comprises an amino acid sequence of (i) any one of SEQ ID NOs: 17, 20, 37 to 39, and 104 to 109 and SEQ ID NO: 9, (ii) SEQ ID NO: 18 or 19 and SEQ ID NO: 11, or (iii) SEQ ID NO: 14, 15, or 16 and SEQ ID NO: 7. Such modified TCRs comprise the amino acid sequences of a mutated β chain and a WT α chain of a TCR which recognizes either NY-ESO-I or MART-L
[0029] The modified TCRs of the invention can comprise one or more immature TCR chains comprising a leader sequence or one or more mature chains in which the leader sequence has been cleaved off. As one of ordinary skill in the art appreciates, the leader sequence of a TCR chain comprises the amino acids at the N-terminus which together serve as a signal to transport the TCR to the plasma membrane and which amino acids are cleaved off to yield the mature form of the TCR. In this regard, the modified TCRs described herein can additionally comprise a leader sequence selected from the group consisting of SEQ ID NOs: 1 10-115. Preferably, when the modified TCR comprises SEQ ID NO: 7, the modified TCR comprises the leader sequence of SEQ ID NO: 110; when the modified TCR comprises any of SEQ ID NOs: 1-3, 8, and 14-16, the modified TCR comprises the leader sequence of SEQ ID NO: 111 ; when the modified TCR comprises SEQ ID NO: 11, the modified TCR comprises the leader sequence of SEQ ID NO: 112; when the modified TCR comprises any of SEQ ID NOs: 5, 6, 12, 18, and 19, the modified TCR comprises the leader sequence of SEQ ID NO: 113; when the modified TCR comprises SEQ ID NO: 9, the modified TCR comprises the leader sequence of SEQ ID NO: 114; when the modified TCR comprises any of SEQ ID NOs: 4, 10, 13, 17, 20, 37-39, and 95-109, the modified TCR comprises the leader sequence of SEQ ID NO: 115.
[0030] Also provided by the invention is an isolated or purified polypeptide comprising a functional portion of any of the modified TCRs described herein, wherein the functional portion comprises the amino acid substitutions. The term "polypeptide" as used herein includes oligopeptides and refers to a single chain of amino acids connected by one or more peptide bonds.
[0031] With respect to the inventive polypeptides, the functional portion can be any portion comprising contiguous amino acids of the modified TCR of which it is a part, provided that the functional portion comprises the amino acid substitutions. The term "functional portion" when used in reference to a modified TCR refers to any part or fragment of the modified TCR of the invention, which part or fragment retains the biological activity of the modified TCR of which it is a part (the parent modified TCR). Functional portions encompass, for example, those parts of a modified TCR that retain the ability to recognize target cells, or detect, treat, or prevent a disease, to a similar extent, the same extent, or to a higher extent, as the parent modified TCR. In reference to the parent modified TCR, the functional portion can comprise, for instance, about 10%, 25%, 30%, 50%, 68%, 80%, 90%, 95%, or more, of the parent TCR.
[0032] The functional portion can comprise additional amino acids at the amino or carboxy terminus of the portion, or at both termini, which additional amino acids are not found in the amino acid sequence of the parent modified TCR. Desirably, the additional amino acids do not interfere with the biological function of the functional portion, e.g., recognize target cells, detect cancer, treat or prevent cancer, etc. More desirably, the additional amino acids enhance the biological activity, as compared to the biological activity of the parent modified TCR.
[0033] The polypeptide can comprise a functional portion of either or both of the α and β chains of the TCRs of the invention, such as a functional portion comprising one of more of CDRl, CDR2, and CDR3 of the variable region(s) of the α chain and/or β chain of a TCR of the invention. In this regard, the polypeptide can comprise the amino acid sequence of any of SEQ ID NOs: 1 to 6, 13, and 95 to 103. The polypeptides can additionally comprise an amino acid sequence selected from the group consisting of SEQ ID NOs: 7, 9, and 11. [0034] Alternatively or additionally, the inventive polypeptide can comprise the entire length of an α or β chain of one of the modified TCRs described herein. In this regard, the inventive polypeptide can comprise an amino acid sequence selected from the group consisting of any of SEQ ID NOs: 14 to 20, 37 to 39, and 104 to 109. Alternatively, the polypeptide of the invention can comprise both chains of the TCRs described herein. For example, the inventive polypeptide can comprise: (i) any of SEQ ID NOs: 14 to 16 in combination with SEQ ID NO: 7; (ii) any of SEQ ID NOs: 17, 20, 37 to 39; and 104 to 109 in combination with SEQ ID NO: 9; or (iii) SEQ ID NO: 18 or 19 in combination with SEQ ID NO: 11.
[0035] The invention further provides an isolated or purified protein comprising at least one of the inventive polypeptides described herein. By "protein" is meant a molecule comprising one or more polypeptide chains.
[0036] The protein of the invention can comprise (i) a first polypeptide chain comprising the amino acid sequence of SEQ ID NO: 1, 2, or 3 and a second polypeptide chain comprising the amino acid sequence of SEQ ID NO: 7, (ii) a first polypeptide chain comprising the amino acid sequence of any of SEQ ID NOs: 4, 13, and 95 to 103 and a second polypeptide chain comprising the amino acid sequence of SEQ ID NO: 9, or (iii) a first polypeptide chain comprising the amino acid sequence of SEQ ID NO: 5 or 6 and a second polypeptide chain comprising the amino acid sequence of SEQ ID NO: 11. In this instance, the protein of the invention can be a TCR.
[0037] Alternatively, if, for example, the protein comprises a single polypeptide chain comprising SEQ ID NO: 1 and SEQ ID NO: 7, or if the first and/or second polypeptide chain(s) of the protein further comprise(s) other amino acid sequences, e.g., an amino acid sequence encoding an immunoglobulin or a portion thereof, then the inventive protein can be a fusion protein. In this regard, the invention also provides a fusion protein comprising at least one of the inventive polypeptides described herein along with at least one other polypeptide. The other polypeptide can exist as a separate polypeptide of the fusion protein, or can exist as a polypeptide, which is expressed in frame (in tandem) with one of the inventive polypeptides described herein. The other polypeptide can encode any peptidic or proteinaceous molecule, or a portion thereof, including, but not limited to an immunoglobulin, CD3, CD4, CD8, an MHC molecule, etc.
[0038] The fusion protein can comprise one or more copies of the inventive polypeptide and/or one or more copies of the other polypeptide. For instance, the fusion protein can comprise I5 2, 3, 4, 5, or more, copies of the inventive polypeptide and/or of the other polypeptide. Suitable methods of making fusion proteins are known in the art, and include, for example, recombinant methods. See, for instance, Choi et al.} MoI Biotechnol. 31: 193- 202 (2005).
[0039] The protein of the invention can be a recombinant antibody comprising at least one of the inventive polypeptides described herein. As used herein, "recombinant antibody" refers to a recombinant (e.g., genetically engineered) protein comprising at least one of the polypeptides of the invention and a polypeptide chain of an antibody, or a portion thereof. The polypeptide of an antibody, or portion thereof, can be a heavy chain, a light chain, a variable or constant region of a heavy or light chain, a single chain variable fragment (scFv), or an Fc, Fab, or F(ab)2' fragment of an antibody, etc. The polypeptide chain of an antibody, or portion thereof, can exist as a separate polypeptide of the recombinant antibody. Alternatively, the polypeptide chain of an antibody, or portion thereof, can exist as a polypeptide, which is expressed in frame (in tandem) with the polypeptide of the invention. The polypeptide of an antibody, or portion thereof, can be a polypeptide of any antibody or any antibody fragment, including any of the antibodies and antibody fragments described herein.
[0040] Included in the scope of the invention are functional variants of the inventive modified TCRs, polypeptides, and proteins described herein. The term "functional variant" as used herein refers to a modified TCR, polypeptide, or protein having substantial or significant sequence identity or similarity to a parent modified TCR, polypeptide, or protein, which functional variant retains the biological activity of the modified TCR, polypeptide, or protein of which it is a variant Functional variants encompass, for example, those variants of the TCR, polypeptide, or protein described herein (the parent modified TCR, polypeptide, or protein) that retain the ability to recognize target cells to a similar extent, the same extent, or to a higher extent, as the parent modified TCR, polypeptide, or protein. In reference to the parent modified TCR, polypeptide, or protein, the functional variant can, for instance, be at least about 30%, 50%, 75%, 80%, 90%, 98% or more identical in amino acid sequence to the parent modified TCR5 polypeptide, or protein.
[0041] The functional variant can, for example, comprise the amino acid sequence of the parent TCR, polypeptide, or protein with at least one conservative amino acid substitution. Alternatively or additionally, the functional variants can comprise the amino acid sequence of the parent modified TCR, polypeptide, or protein with at least one non-conservative amino acid substitution. In this case, it is preferable for the non-conservative amino acid substitution to not interfere with or inhibit the biological activity of the functional variant. Preferably, the non-conservative amino acid substitution enhances the biological activity of the functional variant, such that the biological activity of the functional variant is increased as compared to the parent modified TCR, polypeptide, or protein.
[0042] The modified TCR, polypeptide, or protein can consist essentially of the specified amino acid sequence or sequences described herein, such that other components of the functional variant, e.g., other amino acids, do not materially change the biological activity of the functional variant. In this regard, the inventive modified TCR, polypeptide, or protein can, for example, consist essentially of an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 to 6, 13, and 95 to 103. Also, for instance, the inventive modified TCRs, polypeptides, or proteins can consist essentially of an amino acid sequence selected from the group consisting of SEQ ID NOs: 14 to 20, 37 to 39, and 104 to 109. [0043] The modified TCRs, polypeptides, and proteins of the invention (including functional portions and functional variants) can be of any length, i.e., can comprise any number of amino acids, provided that the modified TCRs, polypeptides, or proteins (or functional portions or functional variants thereof) retain their biological activity, e.g., the ability to specifically bind to antigen, detect diseased cells in a host, or treat or prevent disease in a host, etc. For example, the polypeptide can be 50 to 5000 amino acids long, such as 50, 70, 75, 100, 125, 150, 175, 200, 300, 400, 500, 600, 700, 800, 900, 1000 or more amino acids in length. In this regard, the polypeptides of the invention also include oligopeptides.
[0044] The modified TCRs, polypeptides, and proteins of the invention (including functional portions and functional variants) of the invention can comprise synthetic amino acids in place of one or more naturally-occurring amino acids. Such synthetic amino acids are known in the art, and include, for example, aminocyclohexane carboxylic acid, norleucine, α-amino n-decanoic acid, homoserine, S-acetylaminomethyl-cysteine, trans-3- and trans-4-hydroxyproline, 4-aminophenylalanine, 4- nitrophenylalanine, 4- chlorophenylalanine, 4-carboxyphenylalanine, β-phenylserine β-hydroxyphenylalanine, phenylglycine, α-naphthylalanine, cyclohexylalanine, cyclohexylglycine, indoline-2- carboxylic acid, 1,2 ,3 ,4-tetrahydroisoquinoIine-3 -carboxylic acid, aminomalonic acid, aminomalonic acid monoamide, N'-benzyl-N'-methyl-lysine, N',N'-dibenzyl-lysine, 6- hydroxylysine, ornithine, α-aminocyclopentane carboxylic acid, α-aminocyclohexane carboxylic acid, α-aminocycloheptane carboxylic acid, α-(2-amino-2-norbornane)-carboxylic acid, α,γ-diaminobutyric acid, α,β-diaminopropionic acid, homophenylalanine, and α-tert- butylglycine.
[0045] The modified TCRs, polypeptides, and proteins of the invention (including functional portions and functional variants) can be glycosylated, amidated, carboxylated, phosphorylated, esterifϊed, N-acylated, cyclized via, e.g., a disulfide bridge, or converted into an acid addition salt and/or optionally dimerized or polymerized, or conjugated. [0046] When the modified TCRs, polypeptides, and proteins of the invention (including functional portions and functional variants) are in the form of a salt, preferably, the polypeptides are in the form of a pharmaceutically acceptable salt. Suitable pharmaceutically acceptable acid addition salts include those derived from mineral acids, such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric, and sulphuric acids, and organic acids, such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, and arylsulphonic acids, for example, p-toluenesulphonic acid.
[0047] The modified TCR, polypeptide, and/or protein of the invention (including functional portions and functional variants thereof) can be obtained by methods known in the art. Suitable methods of de novo synthesizing polypeptides and proteins are described in references, such as Chan et al., Fmoc Solid Phase Peptide Synthesis, Oxford University Press, Oxford, United Kingdom, 2005; Peptide and Protein Drug Analysis, ed. Reid, R,, Marcel Dekker, Inc., 2000; Epitope Mapping, ed. Westwoood et at, Oxford University Press, Oxford, United Kingdom, 2000; and U.S. Patent No. 5,449,752. Also, polypeptides and proteins can be recombinantly produced using the nucleic acids described herein using standard recombinant methods. See, for instance, Sambrook et al., Molecular Cloning: A Laboratory Manual, 3rd ed., Cold Spring Harbor Press, Cold Spring Harbor, NY 2001; and Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates and John Wiley & Sons, NY, 1994. Further, some of the TCRs, polypeptides, and proteins of the invention (including functional portions and functional variants thereof) can be isolated and/or purified from a source, such as a plant, a bacterium, an insect, a mammal, e.g., a rat, a human, etc. Methods of isolation and purification are well-known in the art. Alternatively, the TCRs, polypeptides, and/or proteins described herein (including functional portions and functional variants thereof) can be commercially synthesized by companies, such as Synpep (Dublin, CA), Peptide Technologies Corp. (Gaithersburg, MD), and Multiple Peptide Systems (San Diego, CA). In this respect, the inventive TCRs, polypeptides, and proteins can be synthetic, recombinant, isolated, and/or purified.
[0048] Included in the scope of the invention are conjugates, e.g., bioconjugates, comprising any of the inventive modified TCRs, polypeptides, or proteins (including any of the functional portions or variants thereof), nucleic acids, recombinant expression vectors, host cells, populations of host cells, or antibodies, or antigen binding portions thereof. Conjugates, as well as methods of synthesizing conjugates in general, are known in the art (See, for instance, Hudecz, F., Methods MoL Biol. 298: 209-223 (2005) and Kirin et al., Inorg Chem. 44(15): 5405-5415 (2005)).
[0049] Further provided by the invention is a nucleic acid comprising a nucleotide sequence encoding any of the modified TCRs, polypeptides, or proteins described herein (including functional portions and functional variants thereof).
[0050] By "nucleic acid" as used herein includes "polynucleotide," "oligonucleotide," and "nucleic acid molecule," and generally means a polymer of DNA or RNA9 which can be single- stranded or double-stranded, synthesized or obtained (e.g., isolated and/or purified) from natural sources, which can contain natural, non-natural or altered nucleotides, and which can contain a natural, non-natural or altered internucleotide linkage, such as a phosphoroamidate linkage or a phosphorothioate linkage, instead of the phosphodiester found between the nucleotides of an unmodified oligonucleotide. It is generally preferred that the nucleic acid does not comprise any insertions, deletions, inversions, and/or substitutions. However, it may be suitable in some instances, as discussed herein, for the nucleic acid to comprise one or more insertions, deletions, inversions, and/or substitutions. [0051] Preferably, the nucleic acids of the invention are recombinant. As used herein, the term "recombinant" refers to (i) molecules that are constructed outside living cells by joining natural or synthetic nucleic acid segments to nucleic acid molecules that can replicate in a living cell, or (ii) molecules that result from the replication of those described in (i) above. For purposes herein, the replication can be in vitro replication or in vivo replication. [0052] The nucleic acids can be constructed based on chemical synthesis and/or enzymatic ligation reactions using procedures known in the art. See, for example, Sambrook et al., supra, and Ausubel et al., supra. For example, a nucleic acid can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed upon hybridization (e.g., phosphorothioate derivatives and acridme substituted nucleotides). Examples of modified nucleotides that can be used to generate the nucleic acids include, but are not limited to, 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxymethyl) uracil, 5- carboxymethylammomethyI-2-thiouridine, 5 -carboxymethylaminomethyluracil , dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1 -methyl guanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N -substituted adenine, 7-methylguanine, 5-methylaminomethyluracil, 5- methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'- methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil- 5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2- thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, 3- (3-amino-3-N-2~carboxypropyl) uracil, and 2,6-diaminopurine. Alternatively, one or more of the nucleic acids of the invention can be purchased from companies, such as Macromolecular Resources (Fort Collins, CO) and Synthegen (Houston, TX).
[0053] The nucleic acid can comprise any nucleotide sequence which encodes any of the modified TCRs, polypeptides, or proteins, or functional portions or functional variants thereof. For example, the nucleic acid can comprise a nucleotide sequence comprising SEQ ID NO: 22 to 24, 28, 29, 32, or 33. The nucleotide sequence alternatively can comprise a nucleotide sequence which is degenerate to any of these sequences or a combination of degenerate sequences. For example, the nucleotide sequence can comprise a nucleotide sequence comprising a nucleotide sequence degenerate to SEQ ID NO: 22 and a nucleotide sequence degenerate to SEQ ID NO: 23 or comprising a nucleotide sequence degenerate to SEQ ID NO: 23 and a nucleotide sequence degenerate to SEQ ID NO: 25. [0054] The invention also provides an isolated or purified nucleic acid comprising a nucleotide sequence which is complementary to the nucleotide sequence of any of the nucleic acids described herein or a nucleotide sequence which hybridizes under stringent conditions to the nucleotide sequence of any of the nucleic acids described herein. [0055] The nucleotide sequence which hybridizes under stringent conditions preferably hybridizes under high stringency conditions. By "high stringency conditions" is meant that the nucleotide sequence specifically hybridizes to a target sequence (the nucleotide sequence of any of the nucleic acids described herein) in an amount that is detectably stronger than non-specific hybridization. High stringency conditions include conditions which would distinguish a polynucleotide with an exact complementary sequence, or one containing only a few scattered mismatches from a random sequence that happened to have a few small regions (e.g., 3-10 bases) that matched the nucleotide sequence. Such small regions of complementarity are more easily melted than a full-length complement of 14-17 or more bases, and high stringency hybridization makes them easily distinguishable. Relatively high stringency conditions would include, for example, low salt and/or high temperature conditions, such as provided by about 0.02-0.1 M NaCl or the equivalent, at temperatures of about 50-70 0C. Such high stringency conditions tolerate little, if any, mismatch between the nucleotide sequence and the template or target strand, and are particularly suitable for detecting expression of any of the inventive TCRs. It is generally appreciated that conditions can be rendered more stringent by the addition of increasing amounts of formamide. [0056] The nucleic acids of the invention can be incorporated into a recombinant expression vector. In this regard, the invention provides recombinant expression vectors comprising any of the nucleic acids of the invention. For purposes herein, the term "recombinant expression vector" means a genetically-modified oligonucleotide or polynucleotide construct that permits the expression of an mRNA, protein, polypeptide, or peptide by a host cell, when the construct comprises a nucleotide sequence encoding the mRNA, protein, polypeptide, or peptide, and the vector is contacted with the cell under conditions sufficient to have the mRNA, protein, polypeptide, or peptide expressed within the cell. The vectors of the invention are not naturally-occurring as a whole. However, parts of the vectors can be naturally-occurring. The inventive recombinant expression vectors can comprise any type of nucleotides, including, but not limited to DNA and RNA, which can be single- stranded or double-stranded, synthesized or obtained in part from natural sources, and which can contain natural, non-natural or altered nucleotides. The recombinant expression vectors can comprise naturally-occurring, non-naturally-occuring internucleotide linkages, or both types of linkages. Preferably, the non-naturally occurring or altered nucleotides or internucleotide linkages does not hinder the transcription or replication of the vector. [0057] The recombinant expression vector of the invention can be any suitable recombinant expression vector, and can be used to transform or transfect any suitable host. Suitable vectors include those designed for propagation and expansion or for expression or both, such as plasmids and viruses. The vector can be selected from the group consisting of the pUC series (Fermentas Life Sciences), the pBluescript series (Stratagene, LaJolla, CA), the pET series (Novagen, Madison, WI), the pGEX series (Pharmacia Biotech, Uppsala, Sweden), and the pEX series (Clontech, Palo Alto, CA). Bacteriophage vectors, such as λGTIO, λGTl 1, λZapII (Stratagene), λEMBL4, and λNMl 149, also can be used. Examples of plant expression vectors include pBIOl, pBI101.2, pBI101.3, pBI121 and pBIN19 (Clontech). Examples of animal expression vectors include pEUK-Clf pMAM and pMAMneo (Clontech). Preferably, the recombinant expression vector is a viral vector, e.g., a retroviral vector.
[0058] The recombinant expression vectors of the invention can be prepared using standard recombinant DNA techniques described in, for example, Sambrook et al., supra, and Ausubel et al., supra. Constructs of expression vectors, which are circular or linear, can be prepared to contain a replication system functional in a prokaryotic or eukaryotic host cell. Replication systems can be derived, e.g., from CoIEl, 2 μ plasmid, λ, SV40, bovine papilloma virus, and the like.
[0059] Desirably, the recombinant expression vector comprises regulatory sequences, such as transcription and translation initiation and termination codons, which are specific to the type of host (e.g., bacterium, fungus, plant, or animal) into which the vector is to be introduced, as appropriate and taking into consideration whether the vector is DNA- or RNA- based.
[0060] The recombinant expression vector can include one or more marker genes, which allow for selection of transformed or transfected hosts. Marker genes include biocide resistance, e.g., resistance to antibiotics, heavy metals, etc., complementation in an auxotrophic host to provide prototrophy, and the like. Suitable marker genes for the inventive expression vectors include, for instance, neomycin/G418 resistance genes, hygromycin resistance genes, histidinol resistance genes, tetracycline resistance genes, and arnpicillin resistance genes. [0061] The recombinant expression vector can comprise a native or normative promoter operably linked to the nucleotide sequence encoding the modified TCR, polypeptide, or protein (including functional portions and functional variants thereof), or to the nucleotide sequence which is complementary to or which hybridizes to the nucleotide sequence encoding the modified TCR, polypeptide, or protein. The selection of promoters, e.g., strong, weak, inducible, tissue-specific and developmental-specific, is within the ordinary skill of the artisan. Similarly,, the combining of a nucleotide sequence with a promoter is also within the skill of the artisan. The promoter can be a non-viral promoter or a viral promoter, e.g., a cytomegalovirus (CMV) promoter, an SV40 promoter, an RSV promoter, and a promoter found in the long-terminal repeat of the murine stem cell virus.
[0062] The inventive recombinant expression vectors can be designed for either transient expression, for stable expression, or for both. Also, the recombinant expression vectors can be made for constitutive expression or for inducible expression. [0063] Further, the recombinant expression vectors can be made to include a suicide gene. As used herein, the term "suicide gene" refers to a gene that causes the cell expressing the suicide gene to die. The suicide gene can be a gene that confers sensitivity to an agent, e.g., a drug, upon the cell in which the gene is expressed, and causes the cell to die when the cell is contacted with or exposed to the agent. Suicide genes are known in the art (see, for example, Suicide Gene Therapy: Methods and Reviews, Springer, Caroline J. (Cancer Research UK Centre for Cancer Therapeutics at the Institute of Cancer Research, Sutton, Surrey, UK), Humana Press, 2004) and include, for example, the Herpes Simplex Virus (HSV) thymidine kinase (TK) gene, cytosine daminase, purine nucleoside phosphorylase, and nitroreductase.
[0064] The invention further provides a host cell comprising any of the recombinant expression vectors described herein. As used herein, the term "host cell" refers to any type of cell that can contain the inventive recombinant expression vector. The host cell can be a eukaryotic cell, e.g., plant, animal, fungi, or algae, or can be a prokaryotic cell, e.g., bacteria or protozoa. The host cell can be a cultured cell or a primary cell, i.e., isolated directly from an organism, e.g., a human. The host cell can be an adherent cell or a suspended cell, i.e., a cell that grows in suspension. Suitable host cells are known in the art and include, for instance, DH5α E. coli cells, Chinese hamster ovarian cells, monkey VERO cells, COS cells, HEK293 cells, and the like. For purposes of amplifying or replicating the recombinant expression vector, the host cell is preferably a prokaryotic cell, e.g., a DH5α cell For purposes of producing a recombinant modified TCR, polypeptide, or protein, the host cell is preferably a mammalian cell. Most preferably, the host cell is a human cell. While the host cell can be of any cell type, can originate from any type of tissue, and can be of any developmental stage, the host cell preferably is a peripheral blood lymphocyte (PBL). More preferably, the host cell is a T cell.
[0065] For purposes herein, the T cell can be any T cell, such as a cultured T cell, e.g., a primary T cell, or a T cell from a cultured T cell line, e.g., Jurkat, SupTl, etc., or a T cell obtained from a mammal. If obtained from a mammal, the T cell can be obtained from numerous sources, including but not limited to blood, bone marrow, lymph node, the thymus, or other tissues or fluids. T cells can also be enriched for or purified. Preferably, the T cell is a human T cell. More preferably, the T cell is a T cell isolated from a human. The T cell can be any type of T cell and can be of any developmental stage, including but not limited to, CD4+/CD8+ double positive T cells, CD4+ helper T cells, e.g., Th1 and Th2 cells, CD8+ T cells (e.g., cytotoxic T cells), peripheral blood mononuclear cells (PBMCs), peripheral blood leukocytes (PBLs), tumor infiltrating cells (TILs), memory T cells, naive T cells, and the like. Preferably, the T cell is a CD8+ T cell or a CD4+ T cell.
[0066] Also provided by the invention is a population of cells comprising at least one host cell described herein. The population of cells can be a heterogeneous population comprising the host cell comprising any of the recombinant expression vectors described, in addition to at least one other cell, e.g., a host cell (e.g., a T cell), which does not comprise any of the recombinant expression vectors, or a cell other than a T cell, e.g., a B cell, a macrophage, a neutrophil, an erythrocyte, a hepatocyte, an endothelial cell, an epithelial cells, a muscle cell, a brain cell, etc. Alternatively, the population of cells can be a substantially homogeneous population, in which the population comprises mainly of host cells (e.g., consisting essentially of) comprising the recombinant expression vector. The population also can be a clonal population of cells, in which all cells of the population are clones of a single host cell comprising a recombinant expression vector, such that all cells of the population comprise the recombinant expression vector. In one embodiment of the invention, the population of cells is a clonal population comprising host cells comprising a recombinant expression vector as described herein.
[0067] The invention further provides an antibody, or antigen binding portion thereof, which specifically binds to an epitope of the modified TCR of the invention, wherein the epitope comprises the amino acid substitutions. The antibody can be any type of immunoglobulin that is known in the art. For instance, the antibody can be of any isotype, e.g., IgA5 IgD5 IgE, IgG, IgM, etc. The antibody can be monoclonal or polyclonal. The antibody can be a naturally-occurring antibody, e.g., an antibody isolated and/or purified from a mammal, e.g., mouse, rabbit, goat, horse, chicken, hamster, human, etc. Alternatively, the antibody can be a genetically-engineered antibody, e.g., a humanized antibody or a chimeric antibody. The antibody can be in monomeric or polymeric form. Also, the antibody can have any level of affinity or avidity for the functional portion of the inventive modified TCR. Desirably, the antibody is specific for the epitope of the inventive modified TCR comprising the amino acid substitutions, such that there is minimal cross-reaction with other peptides or proteins.
[0068] Methods of testing antibodies for the ability to bind to any functional portion of the inventive modified TCR are known in the art and include any antibody-antigen binding assay, such as, for example, radioimmunoassay (RIA), ELISA, Western blot, immunoprecipitation, and competitive inhibition assays (see, e.g., Jane way et al.s infra, and U.S. Patent Application Publication No. 2002/0197266 Al).
[0069] Suitable methods of making antibodies are known in the art. For instance, standard hybridoma methods are described in, e.g., Kδhler and Milstein, Eur. J. Immunol., 5, 51 1-519 (1976), Harlow and Lane (eds.), Antibodies: A Laboratory Manual, CSH Press (1988), and CA. Janeway et al. (eds.), Immunobiology, 5th Ed., Garland Publishing, New York, NY (2001)). Alternatively, other methods, such as EBV-hybridoma methods (Haskard and Archer, J. Immunol. Methods, 74(2), 361-67 (1984), and Roder et al., Methods Enzymol, 121, 140-67 (1986)), and bacteriophage vector expression systems (see, e.g., Huse et al, Science, 246, 1275-81 (1989)) are known in the art. Further, methods of producing antibodies in non-human animals are described in, e.g., U.S. Patents 5,545,806, 5,569,825, and 5,714,352, and U.S. Patent Application Publication No. 2002/0197266 Al). [0070] Phage display furthermore can be used to generate the antibody of the invention. In this regard, phage libraries encoding antigen-binding variable (V) domains of antibodies can be generated using standard molecular biology and recombinant DNA techniques (see, e.g., Sambrook et al. (eds.), Molecular Cloning, A Laboratory Manual, 3rd Edition, Cold Spring Harbor Laboratory Press, New York (2001)). Phage encoding a variable region with the desired specificity are selected for specific binding to the desired antigen, and a complete or partial antibody is reconstituted comprising the selected variable domain. Nucleic acid sequences encoding the reconstituted antibody are introduced into a suitable cell line, such as a myeloma cell used for hybridoma production, such that antibodies having the characteristics of monoclonal antibodies are secreted by the cell (see, e.g., Janeway et al., supra, Huse et al., supra, and U.S. Patent 6,265,150).
[0071] Antibodies can be produced by transgenic mice that are transgenic for specific heavy and light chain immunoglobulin genes. Such methods are known in the art and described in, for example U.S. Patents 5,545P806 and 5,569,825, and Janeway et al., supra. [0072] Methods for generating humanized antibodies are well known in the art and are described in detail in, for example, Janeway et al, supra, U.S. Patents 5,225,539, 5,585,089 and 5,693,761, European Patent No. 0239400 Bl, and United Kingdom Patent No. 2188638. Humanized antibodies can also be generated using the antibody resurfacing technology described in U.S. Patent 5,639,641 and Pedersen et al., J. MoI. Biol, 235, 959-973 (1994). [0073] The invention also provides antigen binding portions of any of the antibodies described herein. The antigen binding portion can be any portion that has at least one antigen binding site, such as Fab, F(ab')2, dsFv, sFv, diabodies, and triabodies. [0074J A single-chain variable region fragment (sFv) antibody fragment, which consists of a truncated Fab fragment comprising the variable (V) domain of an antibody heavy chain linked to a V domain of a light antibody chain via a synthetic peptide, can be generated using routine recombinant DNA technology techniques (see, e.g., Janeway et al., supra). Similarly, disulfide-stabilized variable region fragments (dsFv) can be prepared by recombinant DNA technology (see, e.g., Reiter et al.s Protein Engineering, 7, 697-704 (1994)). Antibody fragments of the invention, however, are not limited to these exemplary types of antibody fragments.
[0075] Also, the antibody, or antigen binding portion thereof, can be modified to comprise a detectable label, such as, for instance, a radioisotope, a fluorophore (e.g., fluorescein isothiocyanate (FITC), phycoerythrin (PE)), an enzyme (e.g., alkaline phosphatase, horseradish peroxidase), and element particles (e.g., gold particles). [0076] The inventive modified TCRs, polypeptides, proteins, (including functional portions and functional variants thereof), nucleic acids, recombinant expression vectors, host cells (including populations thereof), and antibodies (including antigen binding portions thereof), can be isolated and/or purified. The term "isolated" as used herein means having been removed from its natural environment. The term "purified" as used herein means having been increased in purity, wherein "purity" is a relative term, and not to be necessarily construed as absolute purity. For example, the purity can be at least about 50%, can be greater than 60%, 70% or 80%, or can be 100%.
[0077J The inventive modified TCRs, polypeptides, proteins (including functional portions and variants thereof);, nucleic acids, recombinant expression vectors, host cells (including populations thereof), and antibodies (including antigen binding portions thereof), all of which are collectively referred to as "inventive TCR materials" hereinafter, can be formulated into a composition, such as a pharmaceutical composition, hi this regard, the invention provides a pharmaceutical composition comprising any of the modified TCRs, polypeptides, proteins, functional portions, functional variants, nucleic acids, expression vectors, host cells (including populations thereof), and antibodies (including antigen binding portions thereof), and a pharmaceutically acceptable carrier. The inventive pharmaceutical compositions containing any of the inventive TCR materials can comprise more than one inventive TCR material, e.g., a polypeptide and a nucleic acid, or two or more different modified TCRs. Alternatively, the pharmaceutical composition can comprise an inventive TCR material in combination with another pharmaceutically active agents or drugs, such as a chemotherapeutic agents, e.g., asparaginase, busulfan, carboplatin, cisplatin, daunorubicin, doxorubicin, fluorouracil, gemcitabine, hydroxyurea, methotrexate, paclitaxel, rituximab, vinblastine, vincristine, etc.
[0078J With respect to pharmaceutical compositions, the pharamaceutically acceptable carrier can be any of those conventionally used and is limited only by chemico-physical considerations, such as solubility and lack of reactivity with the active compound(s), and by the route of administration. The pharmaceutically acceptable carriers described herein, for example, vehicles, adjuvants, excipients, and diluents, are well-known to those skilled in the art and are readily available to the public. It is preferred that the pharmaceutically acceptable carrier be one which is chemically inert to the active agent(s) and one which has no detrimental side effects or toxicity under the conditions of use.
[0079] The choice of carrier will be determined in part by the particular inventive TCR material, as well as by the particular method used to administer the inventive TCR material. Accordingly, there are a variety of suitable formulations of the pharmaceutical composition of the invention. The following formulations for oral, aerosol, parenteral, subcutaneous, intravenous, intramuscular, intraarterial, intrathecal, interperitoneal, rectal, and vaginal administration are exemplary and are in no way limiting. More than one route can be used to administer the inventive TCR materials, and in certain instances, a particular route can provide a more immediate and more effective response than another route. [0080] Topical formulations are well-known to those of skill in the art. Such formulations are particularly suitable in the context of the invention for application to the skin.
[0081] Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the inventive TCR material dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions. Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant. Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and corn starch. Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, macrocrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and other pharmacologically compatible excipients. Lozenge forms can comprise the inventive TCR material in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the inventive TCR material in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to, such excipients as are known in the art.
[0082] The inventive TCR material, alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation. These aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. Such spray formulations also may be used to spray mucosa.
[0083] Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The inventive TCR material can be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol or hexadecyl alcohol, a glycol, such as propylene glycol or polyethylene glycol, dimethylsulfoxide, glycerol, ketals such as 2,2- dimethyl-l,3-dioxolane-4-methanol, ethers, poly(ethyleneglycol) 400, oils, fatty acids, fatty acid esters or glycerides, or acetylated fatty acid glycerides with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose. or carboxymethylcellulose, or emulsifying agents and other pharmaceutical adjuvants. [0084] Oils, which can be used in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, com, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
[0085] Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts, and suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfo succinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl- β-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (e) mixtures thereof. [0086] The parenteral formulations will typically contain from about 0.5% to about 25% by weight of the inventive TCR material in solution. Preservatives and buffers may be used. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations will typically range from about 5% to about 15% by weight. Suitable surfactants include polyethylene glycol sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol. The parenteral formulations can be presented in unit-dose or multi-dose sealed containers, such as ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
[0087] Injectable formulations are in accordance with the invention. The requirements for effective pharmaceutical carriers for injectable compositions are well-known to those of ordinary skill in the art (see, e.g., Pharmaceutics and Pharmacy Practice, J.B. Lippincott Company, Philadelphia, PA, Banker and Chalmers, eds., pages 238-250 (1982), mάASHP Handbook on Injectable Drugs, Toissel, 4th ed., pages 622-630 (1986)). Preferably, when administering cells, e.g., dendritic cells, the cells are administered via injection. [0088] Additionally, the inventive TCR materials, or compositions comprising such inventive TCR materials, can be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases. Formulations suitable for vaginal administration can be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
[0089] It will be appreciated by one of skill in the art that, in addition to the above- described pharmaceutical compositions, the inventive TCR materials of the invention can be formulated as inclusion complexes, such as cyclodextrin inclusion complexes, or liposomes. [0090] For purposes of the invention, the amount or dose of the inventive TCR material administered should be sufficient to effect, e.g., a therapeutic or prophylactic response, in the subject or animal over a reasonable time frame. For example, the dose of the inventive TCR material should be sufficient to bind to antigen, or detect, treat or prevent disease in a period of from about 2 hours or longer, e.g., 12 to 24 or more hours, from the time of administration. In certain embodiments, the time period could be even longer. The dose will be determined by the efficacy of the particular inventive TCR material and the condition of the animal (e.g., human), as well as the body weight of the animal (e.g., human) to be treated. [0091] Many assays for determining an administered dose are known in the art. For purposes of the invention, an assay, which comprises comparing the extent to which target cells are lysed or IFN-γ is secreted by T cells expressing the inventive modified TCR, polypeptide, or protein upon administration of a given dose of such T cells to a mammal among a set of mammals of which is each given a different dose of the T cells, could be used to determine a starting dose to be administered to a mammal. The extent to which target cells are lysed or IFN-γ is secreted upon administration of a certain dose can be assayed by methods known in the art, including, for instance, the methods described herein as Example 2.
[0092] The dose of the inventive TCR material also will be determined by the existence, nature and extent of any adverse side effects that might accompany the administration of a particular inventive TCR material. Typically, the attending physician will decide the dosage of the inventive TCR material with which to treat each individual patient, taking into consideration a variety of factors, such as age, body weight, general health, diet, sex, inventive TCR material to be administered, route of administration, and the severity of the condition being treated. By way of example and not intending to limit the invention, the dose of the inventive TCR material can be about 0.001 to about 1000 mg/kg body weight of the subject being treated/day, from about 0.01 to about 10 mg/kg body weight/day, about 0.01 mg to about 1 mg/kg body weight/day.
[Θ093] One of ordinary skill in the art will readily appreciate that the inventive TCR materials of the invention can be modified in any number of ways, such that the therapeutic or prophylactic efficacy of the inventive TCR materials is increased through the modification. For instance, the inventive TCR materials can be conjugated either directly or indirectly through a linker to a targeting moiety. The practice of conjugating compounds, e.g., inventive TCR materials, to targeting moieties is known in the art. See, for instance, Wadwa et al., J Drug Targeting 3: 111 (1995) and U.S. Patent No. 5,087,616. The term "targeting moiety" as used herein, refers to any molecule or agent that specifically recognizes and binds to a cell-surface receptor, such that the targeting moiety directs the delivery of the inventive TCR materials to a population of cells on which surface the receptor is expressed. Targeting moieties include, but are not limited to, antibodies, or fragments thereof, peptides, hormones, growth factors, cytokines, and any other natural or non-natural ligands, which bind to cell surface receptors (e.g., Epithelial Growth Factor Receptor (EGFR), T-cell receptor (TCR), B- cell receptor (BCR), CD28, Platelet-derived Growth Factor Receptor (PDGF), nicotinic acetylcholine receptor (nAChR), etc.). The term "linker" as used herein, refers to any agent or molecule that bridges the inventive TCR materials Io the targeting moiety. One of ordinary skill in the art recognizes that sites on the inventive TCR materials, which are not necessary for the function of the inventive TCR materials, are ideal sites for attaching a linker and/or a targeting moiety, provided that the linker and/or targeting moiety, once attached to the inventive TCR materials, do(es) not interfere with the function of the inventive TCR materials, i.e., the ability to bind to antigen, or to detect, treat, or prevent disease. [0094] Alternatively, the inventive TCR materials can be modified into a depot form, such that the manner in which the inventive TCR materials is released into the body to which it is administered is controlled with respect to time and location within the body (see, for example, U.S. Patent No. 4,450,150). Depot forms of inventive TCR materials can be, for example, an implantable composition comprising the inventive TCR materials and a porous or non-porous material, such as a polymer, wherein the inventive TCR materials is encapsulated by or diffused throughout the material and/or degradation of the non-porous material. The depot is then implanted into the desired location within the body and the inventive TCR materials are released from the implant at a predetermined rate. [0095] It is contemplated that the inventive pharmaceutical compositions, modified TCRs, polypeptides, proteins, nucleic acids, recombinant expression vectors, host cells, or populations of cells can be used in methods of treating or preventing a disease in a host. Without being bound to a particular theory, the inventive modified TCRs are believed to have enhanced biological activity, e.g., ability to recognize antigen, such that the modified TCR (or related inventive polypeptide or protein) when expressed by a cell is able to mediate a stronger immune response against the cell expressing the antigen for which the modified TCR is specific. In this regard, the invention provides a method of treating or preventing a disease in a host, comprising administering to the host any of the pharmaceutical compositions in an amount effective to treat or prevent the disease in the host. [0096] The disease can be any disease involving an antigen, e.g., an infectious disease, an autoimmune disease, a cancer.
[0097] For purposes herein, "infectious disease" means a disease that can be transmitted from person to person or from organism to organism, and is caused by a microbial agent (e.g., common cold). Infectious diseases are known in the art and include, for example, hepatitis, sexually transmitted diseases (e.g., Chlamydia, gonorrhea), tuberculosis, HIV/ AIDS, diphtheria, hepatitis B, hepatitis C, cholera, and influenza.
[0098J For purposes herein, "autoimmune disease" refers to a disease in which the body produces an immunogenic (i.e., immune system) response to some constituent of its own tissue. In other words the immune system loses its ability to recognize some tissue or system within the body as "self and targets and attacks it as if it were foreign. Autoimmune diseases can be classified into those in which predominantly one organ is affected (e.g., hemolytic anemia and anti-immune thyroiditis), and those in which the autoimmune disease process is diffused through many tissues (e.g., systemic lupus erythematosus). For example, multiple sclerosis is thought to be caused by T cells attacking the sheaths that surround the nerve fibers of the brain and spinal cord. This results in loss of coordination, weakness, and blurred vision. Autoimmune diseases are known in the art and include, for instance, Hashimoto's thyroiditis, Grave's disease, lupus, multiple sclerosis, rheumatic arthritis, hemolytic anemia, anti-immune thyroiditis, systemic lupus erythematosus, celiac disease, Crohn's disease, colitis, diabetes, scleroderma, psoriasis, and the like. [0099] With respect to the inventive methods, the cancer can be any cancer, including any of acute lymphocytic cancer, acute myeloid leukemia, alveolar rhabdomyosarcoma, bone cancer, brain cancer, breast cancer, cancer of the anus, anal canal, or anorectum, cancer of the eye, cancer of the intrahepatic bile duct, cancer of the joints, cancer of the neck, gallbladder, or pleura, cancer of the nose, nasal cavity, or middle ear, cancer of the oral cavity, cancer of the vulva, chronic lymphocytic leukemia, chronic myeloid cancer, colon cancer, esophageal cancer, cervical cancer, gastrointestinal carcinoid tumor. Hodgkin lymphoma, hypopharynx cancer, kidney cancer, larynx cancer, liver cancer, lung cancer, malignant mesothelioma, melanoma, multiple myeloma, nasopharynx cancer, non-Hodgkin lymphoma, ovarian cancer, pancreatic cancer, peritoneum, omentum, and mesentery cancer, pharynx cancer, prostate cancer, rectal cancer, renal cancer (e.g., renal cell carcinoma (RCC)), small intestine cancer, soft tissue cancer, stomach cancer, testicular cancer, thyroid cancer, ureter cancer, and urinary bladder cancer. Preferably, the cancer is melanoma.
[0100] The terms "treat," and "prevent" as well as words stemming therefrom, as used herein, do not necessarily imply 100% or complete treatment or prevention. Rather, there are varying degrees of treatment or prevention of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect. In this respect, the inventive methods can provide any amount of any level of treatment or prevention of cancer in a mammal. Furthermore, the treatment or prevention provided by the inventive method can include treatment or prevention of one or more conditions or symptoms of the disease, e.g., cancer, being treated or prevented. Also, for purposes herein, "prevention" can encompass delaying the onset of the disease, or a symptom or condition thereof.
[0101] Also provided is a method of detecting a diseased cell in a host, wherein the diseased cell expresses an antigen characteristic of a disease. The method comprises (i) contacting a sample comprising cells of the host with any of the inventive modified TCRs, polypeptides, proteins, nucleic acids, recombinant expression vectors, host cells, and populations of host cells described herein, thereby forming a complex between the antigen which is characteristic of the disease and the inventive modified TCR, polypeptide, protein, nucleic acid, recombinant expression vector, host cell, or population of cells, and (ii) detecting the complex, wherein detection of the complex is indicative of a diseased cell in the host.
[0102] The diseased cell can be any cell of any disease, which cell expresses an antigen that is characteristic of the disease. The diseased cell can be a cancer cell or an infected cell, for example. Preferably, the diseased cell is a melanoma cell
[0103] In the method of treating or preventing a disease or of detecting a diseased cell, the inventive modified TCR has antigenic specificity for an antigen that is characteristic of the disease to be treated, prevented, or detected. For instance, if the disease to be treated, prevented or detected is melanoma, the inventive modified TCR has antigenic specificity for a melanoma antigen, e.g., MART-I, NY-ESO-I, gplOO, etc. If a host cell or a population comprising at least one host cell is used in the method, the host cell desirably expresses a TCR having antigenic specificity for the antigen of the disease. If an inventive nucleic acid or recombinant expression vector is used in the method, the nucleic acid or recombinant expression vector desirably encodes the modified TCR which has antigenic specificity for an antigen of the disease to be treated, prevented, or detected, such that expression of the nucleic acid or recombinant expression vector is achieved in a cell and the TCR expressed by the cell is capable of binding to the antigen of the disease.
[0104] With respect to the inventive method of detecting a diseased cell in a host, the sample comprising cells of the host can be a sample comprising whole cells, lysates thereof, or a fraction of the whole cell lysates, e.g., a nuclear or cytoplasmic fraction, a whole protein fraction, or a nucleic acid fraction. If the sample comprises whole cells, the cells can be any cells of the host, e.g., the cells of any organ or tissue, including blood cells. [0105] For purposes of the inventive detecting method, the contacting step can take place in vitro or in vivo with respect to the host. Preferably, the contacting is an in vitro step. [0106] Also, detection of the complex can occur through any number of ways known in the art. For instance, the inventive modified TCRs, polypeptides, proteins, nucleic acids, recombinant expression vectors, host cells, populations of cells, or antibodies, or antigen binding portions thereof, described herein, can be labeled with a detectable label such as, for instance, a radioisotope, a fluorophore (e.g., fluorescein isothiocyanate (FITC), phycoerythrin (PE)), an enzyme (e.g., alkaline phosphatase, horseradish peroxidase), and element particles (e.g., gold particles).
[0107 J For purposes of the inventive methods, wherein host cells or populations of cells are administered to the host, the cells can be cells that are allogeneic or autologous to the host. Preferably, the cells are autologous to the host.
[0108] The host referred to herein can be any host. Preferably, the host is a mammal. As used herein, the term "mammal" refers to any mammal, including, but not limited to, mammals of the order Rodentia, such as mice and hamsters, and mammals of the order Logomorpha, such as rabbits. It is preferred that the mammals are from the order Carnivora, including Felines (cats) and Canines (dogs). It is more preferred that the mammals are from the order Artiodactyla, including Bovines (cows) and S wines (pigs) or of the order Perssodactyla, including Equines (horses). It is most preferred that the mammals are of the order Primates, Ceboids, or Simoids (monkeys) or of the order Anthropoids (humans and apes). An especially preferred mammal is the human.
[0109] A method of identifying a candidate adoptive immunotherapy TCR is also provided. The method consists of (a) producing a nucleic acid encoding a modified TCR comprising a WT TCR amino acid sequence with no more than three amino acid substitutions, (b) expressing the nucleic acid in CD4+ T cells and CD8+ T cells, and (c) assaying the T cells for the ability to recognize target cells and for antigen specificity. A candidate adoptive immunotherapy TCR is identified when the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize target cells when expressed in CD4+ T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD 8+ T cells.
[0110] The term "candidate adoptive immunotherapy TCR" as used herein refers to a modified TCR which comprises an amino acid sequence of a WT TCR with no more than three amino acid substitutions and, as compared to the WT TCR (i) has an enhanced ability recognize target cells when expressed in CD4+ T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD8+ T cells. Such candidate cells are predicted to work well in the context of adoptive immunotherapy, such that the TCR, when expressed in T cells of a patient, will be able to treat or prevent a disease of the patient. [0111] Producing a nucleic acid encoding a modified TCR can be carried out by any means known in the art. For instance, Examples 2 to 4 describe methods of making such a nucleic acid in which a WT TCR sequence is used as a template for a PCR reaction which utilizes primers which encode the amino acid substitutions. The resulting PCR product, which encodes the amino acid mutations is then in vitro transcribed, thereby producing an RNA encoding the modified TCR. Alternatively, the nucleic acid can be produced by other means, e.g., site directed mutagenesis.
[0112] With respect to the method of identifying a candidate adoptive immunotherapy TCR, the amino acid substitutions preferably are conservative amino acid substitutions. More preferably, the amino acid substitutions are located within a CDR of the TCR, e.g., CDRl, CDR2, CDR3. Most preferably, the amino acid substitutions are located within the CDR2 of a p chain of the TCR. Also, with regard to this method, the modified TCR has at least twice the ability to recognize target cells as the WT TCR and exhibits this enhanced ability in at least two assays.
[0113] Methods of expressing nucleic acids in cells (including CD4+ T cells and CD8+ T cells) are known in the art, as discussed herein. Preferably, the nucleic acid is an RNA and the RNA is expressed in T cells by methods described in Example 2. [0114] The T cells can be assayed for the ability to recognize target cells and for antigen specificity employing methods known in the art. Preferably, the T cells are assayed as described herein as Example 2.
EXAMPLES
[0115] The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope.
[0116] The following examples describe the amino acid substitutions of the modified
TCRs with respect to the immature TCR sequence (i.e., the TCR sequence with the leader sequence).
EXAMPLE 1
[0117] This example demonstrates a method of making a modified TCR of the invention. [0118] The WT 1 G4 alpha and beta chains were isolated using RT-PCR. The RT reaction was carried out as described in Huang et al, J Immunol, 172: 6057-6064 (2004), and the fragments were amplified using a high fidelity DNA polymerase, Fusion (New England Biolabs) according to the manufacturer's instructions. Specifically, the WT 1G4 β chain was isolated by RT-PCR from an in vitro cultured tumor infiltrating lymphocytes (TIL) sample isolated from a melanoma patient using two primer pairs: the first pair (Primers A and B) was used to isolate the variable region sequence but also contained sequences corresponding to the appropriate J region, while the second primer pair (Primers C and D) was used to isolate the full length constant region but also encoded the J region to allow an overlap with the sequence amplified using the first primer pair. The full length construct encoding the β chain was generated by isolation of the two PCR products on an agarose gel, which products were then mixed and used to carry out a second round of amplification using Primers E and F.
[0119] The 1 G4 TR α chain was constructed by RT-PCR from an in vitro cultured TIL sample of using two primer pairs: the first pair (Primers G and H) was used to isolate the 1G4 α chain variable region sequence including part of the J region, while the second pair (Primers I and J) was used to isolate the 1G4 α chain constant region but also encoded the J region. The full length construct encoding the α chain was generated by isolation of the two PCR products on an agarose gel, which were then mixed and used to carry out a second round of amplification using Primers K and L. The sequences of the primers used to obtain the WT 1G4 nucleic acid are set forth in Table 1.
K*
Figure imgf000033_0001
[0120] The nucleotide sequence of the 1G4 TCR obtained is set forth as SEQ ID NO: 25 (alpha chain) and SEQ ID NO: 21 (beta chain). For generation of each mutant 1G4 TCR, a 5" fragment and a 3' fragment, which encoded the amino acid substitution, were produced by PCR amplification of SEQ ID NO: 21. Primers M and N were used to produce the 5' fragment for each mutant. Primers O and P were used to produce the 3' fragment of 1G4 mutl, Primers Q and R were used to produce the V fragment of 1G4 mut2, and Primers S and T were used to produce the 3* fragment of 1G4 mut3. The 1G4 PCR products were isolated on agarose gels and the final PCR product generated by amplification of the 5' fragment with the 3' fragments using the full length primer pair (Primers U and V). The sequences of the primers used to generate the mutant beta chains of are set forth in Table 2.
TABLE 2
Figure imgf000035_0001
mutated nucleotides noted by shading
[0121] The resulting PCR products were in vitro transcribed into RNA as described in
Zhao et al., MoI. Ther. 13: 151-159 (2006).
[0122] This example demonstrates that the modified TCRs of the invention can be made.
EXAMPLE 2
[0123] This example demonstrates the biological activity of the modified TCRs of the invention as compared to their wild-type counterparts.
[0124] The modified TCRs produced in Example 1 were tested for their ability recognize antigen when separately expressed in CD8+ T cells and CD4+ T cells. PBMC from Patient KS were transfected as described in Zhao et al, (2006), supra with (i) RNA encoding the WT alpha chain of the 1G4 NY-ESO-I -specific TCR and (ii) RNA encoding the WT beta chain of the 1G4 TCR, RNA encoding one of the following mutant beta chains of 1G4: 1G4 mutl (G70A; SEQ ID NO: I)5 1G4 mut2 (A71I, SEQ ID NO: 2), or 1G4 mut3 (G70A/A71I, SEQ ID NO: 3), or DNA encoding Green Fluorescence Protein (GFP).
[0125] Transfected cells were washed and stimulated with or without (T alone) one of the following cells: T2+ pulsed with either 1 μM NY-ESO-I peptide SLLMWITQC (ESO; SEQ ID NO: 92) or g9-154 peptide KTWGQYWQV (g9-154; SEQ ID NO: 93) or melanoma cells: A375 (HLA-A2+/NY-ESO-1+), 624.38 (HLA-A27NY-ESO-1+), 1363 (HL A- A2+/N Y-ESO- I+), 526 (HLA-A2+/NY-ESO-1"), and SK (HLA- A2+/NY-ESO-1"). [0126) Responder cells (1x105 electroporated PBLs) and 1x105 stimulator cells (pulsed T2 cells or melanoma cells) were incubated in a 0.2-ml culture volume in individual wells of 96-well plates. Stimulator cells and responder cells were co-cultured for 16 to 24 h. Cytokine secretion of culture supernatants diluted to the linear range of the assay was measured using commercially available ELISA kits (IFN-γ Endogen, Cambridge, MA). [0127] The amount of IFN-γ (pg/ml) produced by transfected CD8+ T cells are shown in Table 3, while the amount of IFN-γ (pg/ml) produced by transfected CD4+ T cells are shown in Table 4. TABLE 3
Figure imgf000037_0001
TABLE 4
Figure imgf000038_0001
[0128] This example demonstrated that the modified TCRs of the invention have enhanced antigen specificity when expressed in CD4+ T cells and CDS+ T cells.
EXAMPLE 3
[0129] This example demonstrates a method of making modified TCRs specific for the MART-I cancer antigen.
[0130] The DMF5 (F5) TCR which is specific for the cancer antigen MART-I was isolated and the nucleotide sequence of the F5 TCR obtained is set forth as SEQ ID NO: 30 (alpha chain) and SEQ ID NO: 31 (beta chain).
[0131] The F5 α chain was amplified from the cloned gene product (SEQ ID NO: 30) to generate RNA using Primers W and X. The F5 beta chain was amplified from the cloned gene product (SEQ ID NO: 31) to generate RNA using Primers Y and Z. [0132] For generation of each mutant F5 TCR, a 5' fragment and a 3' fragment, which encoded the amino acid substitution, were produced by PCR amplification of SEQ ID NO: 31. Primers AA and AB were used to produce the 5' fragment for each mutant. Primers AC and AD were used to produce the 3' fragment of F5 mutl, Primers AE and AF were used to produce the 3' fragment of F5mut2, Primers AG and AH were used to produce the 3' fragment of F5mut3, Primers AI and AJ were used to produce the 3' fragment of F5mut 4, and Primers AK and AL were used to produce the 3' fragment of F5mut5. The PCR products were isolated on agarose gels and the final PCR product generated by amplification of the 5' fragment with the 3' fragments using the full length primer pair (Primers AM and AN). The sequences of the primers used to generate the mutant beta chains of are set forth in Table 5.
TABLE 5
Figure imgf000040_0001
Figure imgf000041_0001
[0133] PBMC from Patients RS, KS, and HS were transfected and assayed as described in Example 2, except that RNA encoding either the WT or mutant F5 beta chains along with the WT alpha chains were used in transfections. Also, T2+ cells were pulsed with either 1 μg g9-154 peptide or MART-1 peptide AAGIGILTV (MART; SEQ ID NO: 94). Also, the following melanoma cells were used: 526 (A2+/MART-1+), 624 (A2+/MART-1+), 1359 (A2+/MART-1+), 1363 (A2+/MART-1+), A375 (A2+/MART-1+), 888, 888A2, 397 A2 (A2+/MART-1+), 624.38 (A2+/MART-1+), 1300 (A2+/MART-1+), 397A24 (A2+/MART-1+), 624.28 (A2+/MART-1+), 526 (A2+/NY-ESO-1"), SK23 (A2+ MART-1+), and SK (A2"7NY- ESO-1+).
[0134] The amounts of IFN-γ (pg/ml) produced by transfected CD8+ T cells of Patient RS are shown in Table 6, while the amounts of IFN-γ (pg/ml) produced by transfected CD4+ T cells of Patient RS are shown in Table 7. The amounts of IFN-γ (pg/ml) produced by transfected CD8÷ T cells of Patient KS are shown in Table 8, while the amounts of IFN-γ (pg/ml) produced by transfected CD4+ T cells of Patient KS are shown in Table 9. The amounts of IFN-γ (pg/ml) produced by transfected CD8+ T cells of Patient HS are shown in Table 10, while the amounts of IFN-γ (pg/ml) produced by transfected CD4+ T cells of Patient HS are shown in Table 11. The amounts of IFN-γ (pg/ml) produced by transfected CD8+ T cells of Patient KG and MB expressing the modified TCR containing the triple mutant are shown in Tables 12 and 14, while the amounts of IFN-γ (pg/ml) produced by transfected CD4+ T cells of Patient KG and MB expressing the triple mutant are shown in Tables 13 and 15.
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000044_0002
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
TABLE 14
O
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
[0135] This example demonstrated that the modified F5 receptors F5 mutl (T52A) and F5 mut5 (T49V/A50M/T52A) have enhanced capability to recognize target cells in CD4+ T cells without having a decreased antigen specificity in CD8+ T cells.
EXAMPLE 4
[0136] This example demonstrates a method of making another set of modified TCRs specific for the MART-I cancer antigen.
[0137] The DMF4 (F4) TCR which is specific for the cancer antigen MART-I was isolated as described in Hughes et al., Hum. Gene Ther. 16: 457-472 (2005). The nucleotide sequence of the F4 TCR obtained is set forth as SEQ ID NO: 26 (alpha chain) and SEQ ID NO: 27 (beta chain).
[0138] The DMF4 β chain was amplified using Primers AO and AP to generate RNA. For the generation of each mutant F4 TCR, a 5' fragment and a 3' fragment, which encoded the amino acid substitution, were produce by PCR amplification of SEQ ID NO: 27. Primers AQ and AR were used to generate the 5' fragment for each mutant. Primers AS and AT were used to produce the 3' fragment of F4 mutl and Primers AU and AV were used to produce the 3' fragment of F4 mut2. The 5' and 3' fragments were then gel purified, mixed, and amplified using the Primers AW and AX to generate the full length construct. The α chain of the F4 TCR was amplified from the cloned gene product (SEQ ID NO: 26) to generate RNA using Primers AY and AZ. The sequences of the primers used to generate mutant F4 TCRs are set forth in Table 16.
TABLE 16
Figure imgf000055_0001
Figure imgf000056_0001
[0139] PBMC from Patient HS were obtained as described in Example 1. Cells were transfected and assayed as described in Example 1 , except that nucleic acids encoding the WT F4 alpha chain and the mutant MART-I beta chains (F4 mut) were used and T2+ cells were pulsed with either 1 μM gplOO peptide KTWGQYWQV (gplOO; SEQ ID NO: 93) or MART-I peptide AAGIGILTV (MART; SEQ ID NO: 94). Also, the following melanoma cells (with phenotypes in ()) were used: 397- A2 (A2+/MART-1+), 624.38 (A2+/MART-1+), 1300 (A2+/MART-1+), SK23 (A2+/MART-1+), 397-A24 (A2+/MART-1'), 624.28 (Al+MART-I+X and A375 (A2+/MART-1+).
[0140] The amounts of IFN-γ (pg/ml) produced by transfected CD8+ T cells of Patient HS are shown in Table 17, while the amounts of IFN-γ (pg/ml) produced by transfected CD4+ T cells of Patient HS are shown in Table 18.
TABLE 17
Figure imgf000058_0001
TABLE 18
Figure imgf000059_0001
[0141] This example demonstrated that the F4 mutants F4 mut 1 (G70A) and F4 mut 2 (G70A/V71 A) were TCRs that have enhanced ability to recognize tumor target cells without having a decrease in antigen specificity.
EXAMPLE 5
[0142] This example demonstrates a method of treating a disease in a host using the inventive TCRs.
[0143] Adoptive cell transfer is carried out as described in Morgan et al. (2006), supra. Briefly, PBLs are obtained by leukopheresis from a metastatic melanoma patient who is HLA-A*0201 positive. The PBLs are transduced with nucleic acids encoding a WT alpha chain, and a modified beta chain of a TCR specific for either NY-ESO-I or MART-I as described in Example 1. The patient receives the transduced cells at the time of maximum lymphodepletion. One month post-adoptive cell transfer, quantitative RT-PCR assays are carried out to reveal whether the presence of the modified TCRs are expressed by cells of the patient. Tumor regression also is analyzed by the methods described in Morgan et al. (2006), supra.
[0144] All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
[0145] The use of the terms "a" and "an" and "the" and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms "comprising," "having," "including," and "containing" are to be construed as open-ended terms (i.e., meaning "including, but not limited to,") unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
[0146] Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

Claims

CLAIM:
1. A modified T cell receptor (TCR) comprising an amino acid sequence of a wild-type (WT) TCR with no more than three amino acid substitutions, wherein the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize target cells when expressed by CD4+ T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD8÷ T cells.
2. The modified TCR of claim 1, wherein the amino acid substitutions are located in a complementary determining region (CDR) of the TCR.
3. The modified TCR of claim 2, wherein the CDR is a CDR2.
4. The modified TCR of any of claims 1 to 3, wherein the amino acid substitutions are located on a beta chain of the TCR.
5. The modified TCR of any of claims 1 to 4, wherein the amino acid substitutions are conservative amino acid substitutions.
6. The modified TCR of claim 5, wherein the conservative amino acid substitutions are selected from the group consisting of T→A, G-→A, A→I, T-→ V, A→M, T→I, A→V, T→G, and T→S.
7. The modified TCR of any of claims 1 to 6, wherein the modified TCR has antigen specificity for a cancer antigen.
8. The modified TCR claim 7, wherein the cancer antigen is selected from a group consisting of NY-ESO-I, MART-I5 gplOO, p53, TRP-I, TRP-2, and tyrosinase.
9. The modified TCR of claim 8, wherein the WT TCR comprises the amino acid sequence of SEQ ID NO: 8.
10. The modified TCR of claim 9, wherein the modified TCR comprises the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 2, or SEQ ID NO: 3.
11. The modified TCR of claim 10, wherein the modified TCR comprises the amino acid sequence of SEQ ID NO. 14, SEQ ID NO. 15, or SEQ ID NO. 16.
12. The modified TCR of claim 10 or 11 , wherein the modified TCR additionally comprises the amino acid sequence of SEQ ID NO: 7.
13. The modified TCR of claim 8, wherein the cancer antigen is MART- 1.
14. The modified TCR of claim 13, wherein the WT TCR comprises the amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 12.
15. The modified TCR of claim 14, wherein the modified TCR comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 4 to 6, 13, and 95 to 103.
16. The modified TCR of claim 15, wherein the modified TCR comprises an amino acid sequence selected from the group consisting of SEQ ID NOs. 17 to 20, 37 to 39, and 104 to 109.
17. The modified TCR of claim 15 or 16, wherein the modified TCR additionally comprises an amino acid sequence of SEQ ID NO. 9 or SEQ ID NO. 11.
18. The modified TCR of claim 17, wherein the modified TCR comprises the amino acid sequence of:
(i) any of SEQ ID NOs: 17, 20, 37 to 30, and 104 to 109 in combination with SEQ ID NO. 9; or
(ii) SEQ ID NO: 18 or 19 in combination with SEQ ID NO: 1 1.
19. The modified TCR of any of claims 1 to 18 comprising a leader sequence selected from the group consisting of SEQ ID NOs: 110 to 115.
20. An isolated polypeptide comprising a functional portion of the modified TCR of any of claims 1 to 19, wherein the functional portion comprises the amino acid substitutions.
21. The isolated polypeptide of claim 20 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs. 1 to 6, 13, and 95 to 103.
22. The isolated polypeptide of claim 21 comprising an amino acid sequence selected from the group consisting of SEQ NOs. 14 to 20, 37 to 39, and 104 to 109.
23. The isolated polypeptide of any of claims 20 to 22, wherein the polypeptide additionally comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 7, 9, and 11.
24. An isolated protein comprising the polypeptide of any of claims 20 to 23.
25. The isolated protein of claim 24, comprising (i) a first polypeptide chain comprising the amino acid sequence of SEQ ID NO. 1, 2, or 3 and a second polypeptide chain comprising the amino acid sequence of SEQ ID NO: 7, (ii) a first polypeptide chain comprising the amino acid sequence of any of SEQ ID NOs: 4, 13, and 95 to 103 and a second polypeptide chain comprising the amino acid sequence of SEQ ID NO: 9, or (iii) a first polypeptide chain comprising the amino acid sequence of SEQ ID NO: 5 or 6 and a second polypeptide chain comprising the amino acid sequence of SEQ ID NO: 11.
26. An isolated nucleic acid comprising a nucleotide sequence encoding the modified TCR of any of claims 1 to 19, the polypeptide of any of claims 20 to 23, or the protein of claim 24 or 25.
27. The isolated nucleic acid of claim 26 comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs.: 22 to 24, 28, 29, 32 and 33.
28. A recombinant expression vector comprising the nucleic acid of claim 26 or 27.
29. An isolated host cell comprising the recombinant expression vector of claim 28.
30. The isolated host cell of claim 29, wherein the cell is a peripheral blood lymphocyte (PBL).
31. The isolated host cell of claim 30, wherein the PBL is a CD8+ T cell or a CD4+ T cell.
32. A population of cells comprising at least one host cell of any of claims 29 to 31.
33. An antibody, or antigen binding portion thereof, which specifically binds an epitope of the modified TCR of any of claims 1 to 19, wherein the epitope comprises the amino acid substitutions.
34. A pharmaceutical composition comprising the modified TCR of any of claims 1 to 19, the polypeptide of any of claims 20 to 23, the protein of claim 24 or 25, the nucleic acid of claim 26 or 27, the recombinant expression vector of claim 28, the host cell of any of claims 29 to 31, or the population of cells of claim 32.
35. A method of detecting a diseased cell in a host, wherein the diseased cell expresses an antigen characteristic of a disease, comprising (a) contacting a sample comprising cells of the host with the modified TCR of any of claims 1 to 19, the polypeptide of any of claims 20 to 23, the protein of claim 24 or 25, the nucleic acid of claim 26 or 27, the recombinant expression vector of claim 28, the host cell of any of claims 29 to 31, or the population of cells of claim 32, thereby forming a complex between the modified TCR, polypeptide, protein, nucleic acid, recombinant expression vector, host cell, or cells and the antigen, and
(b) detecting the complex, wherein detection of the complex is indicative of a diseased cell in the host.
36. The method of claim 35, wherein the diseased cell is a cancer cell or an infected cell.
37. The method of claim 36, wherein the cancer cell is a melanoma cell.
38. The method of any of claims 35 to 37, wherein the host is a human.
39. The method of any of claims 35 to 38, wherein the method is an in vitro method.
40. A method of treating or preventing a disease in a host, comprising administering to the host the pharmaceutical composition of claim 34 in an amount effective to treat or prevent the disease in the host.
41. The method of claim 40, wherein the disease is a cancer or an infectious disease.
42. The method of claim 41, wherein the cancer is melanoma.
43. The method of any of claims 40 to 42, wherein the host is a human.
44. The method of any of claims 40 to 43, wherein the host cells are cells that are autologous to the host.
45. The method of any of claims 40 to 44, wherein the cells of the population are cells that are autologous to the host.
46. A method of identifying a candidate adoptive immunotherapy T cell receptor (TCR), consisting of:
(a) producing a nucleic acid encoding a modified TCR comprising a WT TCR amino acid sequence with no more than three amino acid substitutions,
(b) expressing the nucleic acid in CD4+ T cells and CD8÷ T cells, and (c) assaying the T cells for the ability to recognize target cells and for antigen specificity, wherein a candidate adoptive immunotherapy TCR is identified when the modified TCR, as compared to the WT TCR, (i) has an enhanced ability to recognize target cells when expressed by CD4+ T cells and (ii) does not exhibit a decrease in antigen specificity when expressed by CD8+ T cells.
47. The method of claim 46, wherein the amino acid substitutions are conservative amino acid substitutions.
48. The method of claim 46 or 47, wherein the amino acid substitutions are located within a CDR of the WT TCR.
49. The method of claim 48, wherein the CDR is a CDR2 of the beta chain of the modified TCR.
50. The method of any of claims 46 to 49, wherein the modified TCR has at least twice the ability to recognize target cells as the WT TCR.
PCT/US2007/079487 2006-09-26 2007-09-26 Modified t cell receptors and related materials and methods WO2008039818A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/443,111 US8088379B2 (en) 2006-09-26 2007-09-26 Modified T cell receptors and related materials and methods
US13/304,841 US9128080B2 (en) 2006-09-26 2011-11-28 Modified T cell receptors and related materials and methods

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US84744706P 2006-09-26 2006-09-26
US60/847,447 2006-09-26

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/443,111 A-371-Of-International US8088379B2 (en) 2006-09-26 2007-09-26 Modified T cell receptors and related materials and methods
US13/304,841 Division US9128080B2 (en) 2006-09-26 2011-11-28 Modified T cell receptors and related materials and methods

Publications (2)

Publication Number Publication Date
WO2008039818A2 true WO2008039818A2 (en) 2008-04-03
WO2008039818A3 WO2008039818A3 (en) 2008-06-05

Family

ID=38926230

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/079487 WO2008039818A2 (en) 2006-09-26 2007-09-26 Modified t cell receptors and related materials and methods

Country Status (2)

Country Link
US (2) US8088379B2 (en)
WO (1) WO2008039818A2 (en)

Cited By (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009042570A2 (en) * 2007-09-25 2009-04-02 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Modified t cell receptors and related materials and methods
WO2010089412A1 (en) 2009-02-09 2010-08-12 Helmholtz Zentrum Muenchen Deutsches Forschungszentrum Fuer Gesundheit Und Umwelt (Gmbh) Repertoire of allo-restricted peptide-specific t cell receptor sequences and use thereof
US8524234B2 (en) 2011-11-03 2013-09-03 Tolera Therapeutics, Inc Antibody for selective inhibition of T-cell responses
US8556882B2 (en) 2009-04-30 2013-10-15 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Inducible interleukin-12
US8697854B2 (en) 2008-11-24 2014-04-15 Helmholtz Zentrum München Deutsches Forschungszentrum Für Gesundheit Und Umwelt Gmbh High affinity T cell receptor and use thereof
US8722049B2 (en) 2011-11-03 2014-05-13 Tolera Therapeutics, Inc. Antibody and methods for selective inhibition of T-cell responses
US8785601B2 (en) 2009-01-28 2014-07-22 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services T cell receptors and related materials and methods of use
WO2015091823A1 (en) * 2013-12-19 2015-06-25 Friedrich-Alexander Universität Erlangen-Nürnberg Influenza-specific t-cell receptor and its uses in the detection, prevention and/or treatment of influenza
US20150313978A1 (en) * 2012-12-14 2015-11-05 Universitätsmedizin Der Johannes Gutenberg- Universität Mainz Novel MHC-Independent Tumor-Associated Antigens
US9238063B2 (en) 2001-03-16 2016-01-19 Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundlheit und Umwelt (GmbH) Semi-allogenic anti-tumour vaccine with HLA haplo-identical antigen-presenting cells
EP2877489A4 (en) * 2012-07-27 2016-04-13 Univ Illinois Engineering t-cell receptors
WO2016100977A1 (en) 2014-12-19 2016-06-23 The Broad Institute Inc. Methods for profiling the t-cel- receptor repertoire
WO2017069958A2 (en) 2015-10-09 2017-04-27 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
WO2017075465A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting gata3
WO2017075451A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting pou2af1
WO2017075478A2 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by use of immune cell gene signatures
WO2017087708A1 (en) 2015-11-19 2017-05-26 The Brigham And Women's Hospital, Inc. Lymphocyte antigen cd5-like (cd5l)-interleukin 12b (p40) heterodimers in immunity
WO2017184590A1 (en) 2016-04-18 2017-10-26 The Broad Institute Inc. Improved hla epitope prediction
WO2018035364A1 (en) 2016-08-17 2018-02-22 The Broad Institute Inc. Product and methods useful for modulating and evaluating immune responses
WO2018049025A2 (en) 2016-09-07 2018-03-15 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses
WO2018067991A1 (en) 2016-10-07 2018-04-12 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
WO2018148671A1 (en) 2017-02-12 2018-08-16 Neon Therapeutics, Inc. Hla-based methods and compositions and uses thereof
WO2018191553A1 (en) 2017-04-12 2018-10-18 Massachusetts Eye And Ear Infirmary Tumor signature for metastasis, compositions of matter methods of use thereof
WO2018195019A1 (en) 2017-04-18 2018-10-25 The Broad Institute Inc. Compositions for detecting secretion and methods of use
US10149898B2 (en) 2017-08-03 2018-12-11 Taiga Biotechnologies, Inc. Methods and compositions for the treatment of melanoma
WO2019027465A1 (en) 2017-08-03 2019-02-07 Taiga Biotechnologies, Inc. Methods and compositions for the treatment of melanoma
WO2019060746A1 (en) 2017-09-21 2019-03-28 The Broad Institute, Inc. Systems, methods, and compositions for targeted nucleic acid editing
WO2019094983A1 (en) 2017-11-13 2019-05-16 The Broad Institute, Inc. Methods and compositions for treating cancer by targeting the clec2d-klrb1 pathway
WO2019232542A2 (en) 2018-06-01 2019-12-05 Massachusetts Institute Of Technology Methods and compositions for detecting and modulating microenvironment gene signatures from the csf of metastasis patients
WO2020010565A1 (en) * 2018-07-12 2020-01-16 深圳华大生命科学研究院 Mart-1(27-35) epitope-specific t cell receptor
WO2020041387A1 (en) 2018-08-20 2020-02-27 The Brigham And Women's Hospital, Inc. Degradation domain modifications for spatio-temporal control of rna-guided nucleases
WO2020041384A1 (en) 2018-08-20 2020-02-27 The Broad Institute, Inc. 3-phenyl-2-cyano-azetidine derivatives, inhibitors of rna-guided nuclease activity
WO2020068304A2 (en) 2018-08-20 2020-04-02 The Broad Institute, Inc. Inhibitors of rna-guided nuclease target binding and uses thereof
WO2020072700A1 (en) 2018-10-02 2020-04-09 Dana-Farber Cancer Institute, Inc. Hla single allele lines
WO2020081730A2 (en) 2018-10-16 2020-04-23 Massachusetts Institute Of Technology Methods and compositions for modulating microenvironment
WO2020092455A2 (en) 2018-10-29 2020-05-07 The Broad Institute, Inc. Car t cell transcriptional atlas
WO2020131586A2 (en) 2018-12-17 2020-06-25 The Broad Institute, Inc. Methods for identifying neoantigens
US10760055B2 (en) 2005-10-18 2020-09-01 National Jewish Health Conditionally immortalized long-term stem cells and methods of making and using such cells
WO2020186101A1 (en) 2019-03-12 2020-09-17 The Broad Institute, Inc. Detection means, compositions and methods for modulating synovial sarcoma cells
WO2020191079A1 (en) 2019-03-18 2020-09-24 The Broad Institute, Inc. Compositions and methods for modulating metabolic regulators of t cell pathogenicity
US10786534B2 (en) 2013-03-11 2020-09-29 Taiga Biotechnologies, Inc. Production and use of red blood cells
US10801070B2 (en) 2013-11-25 2020-10-13 The Broad Institute, Inc. Compositions and methods for diagnosing, evaluating and treating cancer
US10835585B2 (en) 2015-05-20 2020-11-17 The Broad Institute, Inc. Shared neoantigens
WO2020236967A1 (en) 2019-05-20 2020-11-26 The Broad Institute, Inc. Random crispr-cas deletion mutant
WO2020243371A1 (en) 2019-05-28 2020-12-03 Massachusetts Institute Of Technology Methods and compositions for modulating immune responses
WO2021030627A1 (en) 2019-08-13 2021-02-18 The General Hospital Corporation Methods for predicting outcomes of checkpoint inhibition and treatment thereof
WO2021041922A1 (en) 2019-08-30 2021-03-04 The Broad Institute, Inc. Crispr-associated mu transposase systems
US10953048B2 (en) 2012-07-20 2021-03-23 Taiga Biotechnologies, Inc. Enhanced reconstitution and autoreconstitution of the hematopoietic compartment
US10975442B2 (en) 2014-12-19 2021-04-13 Massachusetts Institute Of Technology Molecular biomarkers for cancer immunotherapy
US11116796B2 (en) 2016-12-02 2021-09-14 Taiga Biotechnologies, Inc. Nanoparticle formulations
US11183272B2 (en) 2018-12-21 2021-11-23 Biontech Us Inc. Method and systems for prediction of HLA class II-specific epitopes and characterization of CD4+ T cells
US11180751B2 (en) 2015-06-18 2021-11-23 The Broad Institute, Inc. CRISPR enzymes and systems
US11203758B2 (en) 2014-10-31 2021-12-21 The Trustees Of The University Of Pennsylvania Altering gene expression in modified T cells and uses thereof
US11369678B2 (en) 2008-08-28 2022-06-28 Taiga Biotechnologies, Inc. Compositions and methods for modulating immune cells
WO2022187280A1 (en) 2021-03-01 2022-09-09 Dana-Farber Cancer Institute, Inc. Personalized redirection and reprogramming of t cells for precise targeting of tumors
US11452768B2 (en) 2013-12-20 2022-09-27 The Broad Institute, Inc. Combination therapy with neoantigen vaccine
WO2022261018A1 (en) 2021-06-07 2022-12-15 Providence Health & Services - Oregon Cxcr5, pd-1, and icos expressing tumor reactive cd4 t cells and their use
US11549149B2 (en) 2017-01-24 2023-01-10 The Broad Institute, Inc. Compositions and methods for detecting a mutant variant of a polynucleotide
US11639374B2 (en) 2015-12-22 2023-05-02 Immunocore Limited T cell receptors specific for the NY-ESO-1 tumor antigen-HLA-A*02 complex
US11667695B2 (en) 2008-05-16 2023-06-06 Taiga Biotechnologies, Inc. Antibodies and processes for preparing the same
US11725237B2 (en) 2013-12-05 2023-08-15 The Broad Institute Inc. Polymorphic gene typing and somatic change detection using sequencing data
US11732257B2 (en) 2017-10-23 2023-08-22 Massachusetts Institute Of Technology Single cell sequencing libraries of genomic transcript regions of interest in proximity to barcodes, and genotyping of said libraries
US11739156B2 (en) 2019-01-06 2023-08-29 The Broad Institute, Inc. Massachusetts Institute of Technology Methods and compositions for overcoming immunosuppression
US11793787B2 (en) 2019-10-07 2023-10-24 The Broad Institute, Inc. Methods and compositions for enhancing anti-tumor immunity by targeting steroidogenesis
US11844800B2 (en) 2019-10-30 2023-12-19 Massachusetts Institute Of Technology Methods and compositions for predicting and preventing relapse of acute lymphoblastic leukemia
US11865168B2 (en) 2019-12-30 2024-01-09 Massachusetts Institute Of Technology Compositions and methods for treating bacterial infections
US11897953B2 (en) 2017-06-14 2024-02-13 The Broad Institute, Inc. Compositions and methods targeting complement component 3 for inhibiting tumor growth
US11913075B2 (en) 2017-04-01 2024-02-27 The Broad Institute, Inc. Methods and compositions for detecting and modulating an immunotherapy resistance gene signature in cancer
WO2024077256A1 (en) 2022-10-07 2024-04-11 The General Hospital Corporation Methods and compositions for high-throughput discovery ofpeptide-mhc targeting binding proteins
US11957695B2 (en) 2018-04-26 2024-04-16 The Broad Institute, Inc. Methods and compositions targeting glucocorticoid signaling for modulating immune responses
US11963966B2 (en) 2017-03-31 2024-04-23 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating ovarian tumors
US11981922B2 (en) 2020-10-05 2024-05-14 Dana-Farber Cancer Institute, Inc. Methods and compositions for the modulation of cell interactions and signaling in the tumor microenvironment

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8562988B2 (en) * 2005-10-19 2013-10-22 Ibc Pharmaceuticals, Inc. Strategies for improved cancer vaccines
WO2008039818A2 (en) 2006-09-26 2008-04-03 Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Modified t cell receptors and related materials and methods
EP2719774B8 (en) 2008-11-07 2020-04-22 Adaptive Biotechnologies Corporation Methods of monitoring conditions by sequence analysis
US8748103B2 (en) 2008-11-07 2014-06-10 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
US8628927B2 (en) 2008-11-07 2014-01-14 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
US9365901B2 (en) 2008-11-07 2016-06-14 Adaptive Biotechnologies Corp. Monitoring immunoglobulin heavy chain evolution in B-cell acute lymphoblastic leukemia
US9528160B2 (en) 2008-11-07 2016-12-27 Adaptive Biotechnolgies Corp. Rare clonotypes and uses thereof
US9506119B2 (en) 2008-11-07 2016-11-29 Adaptive Biotechnologies Corp. Method of sequence determination using sequence tags
DK3059337T3 (en) 2009-01-15 2019-07-22 Adaptive Biotechnologies Corp Adaptive immunity profiling and methods for producing monoclonal antibodies
KR20120044941A (en) 2009-06-25 2012-05-08 프레드 헛친슨 켄서 리서치 센터 Method of measuring adaptive immunity
US10385475B2 (en) 2011-09-12 2019-08-20 Adaptive Biotechnologies Corp. Random array sequencing of low-complexity libraries
US9279159B2 (en) 2011-10-21 2016-03-08 Adaptive Biotechnologies Corporation Quantification of adaptive immune cell genomes in a complex mixture of cells
EP3904536A1 (en) 2011-12-09 2021-11-03 Adaptive Biotechnologies Corporation Diagnosis of lymphoid malignancies and minimal residual disease detection
US9499865B2 (en) 2011-12-13 2016-11-22 Adaptive Biotechnologies Corp. Detection and measurement of tissue-infiltrating lymphocytes
EP2823060B1 (en) 2012-03-05 2018-02-14 Adaptive Biotechnologies Corporation Determining paired immune receptor chains from frequency matched subunits
JP5756247B1 (en) 2012-05-08 2015-07-29 アダプティブ バイオテクノロジーズ コーポレイション Composition and method for measuring and calibrating amplification bias in multiplex PCR reactions
PT2852613T (en) 2012-05-22 2019-05-20 Us Health Murine anti-ny-eso-1 t cell receptors
WO2014039044A1 (en) 2012-09-06 2014-03-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of producing t memory stem cell populations
EP2904111B1 (en) 2012-10-01 2017-12-06 Adaptive Biotechnologies Corporation Immunocompetence assessment by adaptive immune receptor diversity and clonality characterization
US10150996B2 (en) 2012-10-19 2018-12-11 Adaptive Biotechnologies Corp. Quantification of adaptive immune cell genomes in a complex mixture of cells
US9717758B2 (en) 2013-05-17 2017-08-01 University Of Massachusetts High-affinity DMF5 T cell receptor (TCR) variants
US9708657B2 (en) 2013-07-01 2017-07-18 Adaptive Biotechnologies Corp. Method for generating clonotype profiles using sequence tags
WO2015077607A1 (en) 2013-11-22 2015-05-28 The Board Of Trustees Of The University Of Illinois Engineered high-affinity human t cell receptors
WO2015134787A2 (en) 2014-03-05 2015-09-11 Adaptive Biotechnologies Corporation Methods using randomer-containing synthetic molecules
US11390921B2 (en) 2014-04-01 2022-07-19 Adaptive Biotechnologies Corporation Determining WT-1 specific T cells and WT-1 specific T cell receptors (TCRs)
US10066265B2 (en) 2014-04-01 2018-09-04 Adaptive Biotechnologies Corp. Determining antigen-specific t-cells
US10526391B2 (en) 2014-07-22 2020-01-07 The University Of Notre Dame Du Lac Molecular constructs and uses thereof
WO2016069886A1 (en) 2014-10-29 2016-05-06 Adaptive Biotechnologies Corporation Highly-multiplexed simultaneous detection of nucleic acids encoding paired adaptive immune receptor heterodimers from many samples
US10246701B2 (en) 2014-11-14 2019-04-02 Adaptive Biotechnologies Corp. Multiplexed digital quantitation of rearranged lymphoid receptors in a complex mixture
EP3498866A1 (en) 2014-11-25 2019-06-19 Adaptive Biotechnologies Corp. Characterization of adaptive immune response to vaccination or infection using immune repertoire sequencing
WO2016138122A1 (en) 2015-02-24 2016-09-01 Adaptive Biotechnologies Corp. Methods for diagnosing infectious disease and determining hla status using immune repertoire sequencing
AU2016242967B2 (en) 2015-04-01 2021-07-01 Adaptive Biotechnologies Corp. Method of identifying human compatible T cell receptors specific for an antigenic target
WO2017070042A1 (en) 2015-10-20 2017-04-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of producing t cell populations using akt inhibitors
EP3410849B1 (en) 2016-02-05 2023-07-05 Institut Pasteur Use of inhibitors of adam12 as adjuvants in tumor therapies
WO2017139199A1 (en) 2016-02-10 2017-08-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Inducible arginase
US10428325B1 (en) 2016-09-21 2019-10-01 Adaptive Biotechnologies Corporation Identification of antigen-specific B cell receptors
WO2019103857A1 (en) 2017-11-22 2019-05-31 Iovance Biotherapeutics, Inc. Expansion of peripheral blood lymphocytes (pbls) from peripheral blood
KR20200063215A (en) 2017-10-06 2020-06-04 오슬로 유니버시테시케후스 에이치에프 Chimeric antigen receptor
WO2019075055A1 (en) 2017-10-11 2019-04-18 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of producing t cell populations using p38 mapk inhibitors
US11254980B1 (en) 2017-11-29 2022-02-22 Adaptive Biotechnologies Corporation Methods of profiling targeted polynucleotides while mitigating sequencing depth requirements
KR20190076297A (en) 2017-12-22 2019-07-02 (주)메디톡스 MART-1 Specific T Cell Receptor and Use Thereof
AU2019218785A1 (en) 2018-02-09 2020-09-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Tethered interleukin-15 and interleukin-21
AU2019260656A1 (en) 2018-04-24 2020-12-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of producing T cell populations using hydroxycitric acid and/or a salt thereof
KR20200068263A (en) 2018-12-05 2020-06-15 (주)메디톡스 NY-ESO-1 Specific T Cell Receptor and Use Thereof
KR20210136050A (en) 2019-03-01 2021-11-16 이오반스 바이오테라퓨틱스, 인크. Expansion of tumor-infiltrating lymphocytes from liquid tumors and their therapeutic use
CN113906140A (en) * 2019-03-18 2022-01-07 路德维格癌症研究院 A2/NY-ESO-1 specific T cell receptor and application thereof
WO2021221783A1 (en) 2020-05-01 2021-11-04 Massachusetts Institute Of Technology Methods for identifying chimeric antigen receptor-targeting ligands and uses thereof
EP4211228A1 (en) 2020-09-08 2023-07-19 The United States of America, as represented by the Secretary, Department of Health and Human Services T cell phenotypes associated with response to adoptive cell therapy
EP4243937A2 (en) 2020-11-13 2023-09-20 The United States of America, as represented by the Secretary, Department of Health and Human Services Enhanced antigen reactivity of immune cells expressing a mutant non-signaling cd3 zeta chain
CA3217738A1 (en) 2021-05-05 2022-05-04 Immatics Biotechnologies Gmbh Antigen binding proteins specifically binding prame
WO2023130040A2 (en) 2021-12-31 2023-07-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services T cell therapy with vaccination as a combination immunotherapy against cancer

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005114215A2 (en) * 2004-05-19 2005-12-01 Avidex Ltd Method of improving t cell receptors
WO2005113595A2 (en) * 2004-05-19 2005-12-01 Avidex Ltd High affinity ny-eso t cell receptor

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4450150A (en) * 1973-05-17 1984-05-22 Arthur D. Little, Inc. Biodegradable, implantable drug delivery depots, and method for preparing and using the same
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
IN165717B (en) * 1986-08-07 1989-12-23 Battelle Memorial Institute
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5545806A (en) * 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
ES2108048T3 (en) * 1990-08-29 1997-12-16 Genpharm Int PRODUCTION AND USE OF LOWER TRANSGENIC ANIMALS CAPABLE OF PRODUCING HETEROLOGICAL ANTIBODIES.
JP3266311B2 (en) * 1991-05-02 2002-03-18 生化学工業株式会社 Novel polypeptide and anti-HIV agent using the same
US5639641A (en) * 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
US6001975A (en) * 1993-03-18 1999-12-14 Ludwig Institute For Cancer Research Precursor processed to at least one tumor rejection antigen presented by HLA-A2
US5620886A (en) 1993-03-18 1997-04-15 Ludwig Institute For Cancer Research Isolated nucleic acid sequence coding for a tumor rejection antigen precursor processed to at least one tumor rejection antigen presented by HLA-A2
US5856091A (en) * 1993-03-18 1999-01-05 Ludwig Institute For Cancer Research Isolated nucleic acid sequence coding for a tumor rejection antigen precursor processed to at least one tumor rejection antigen presented by HLA-A2
US5837476A (en) * 1995-03-03 1998-11-17 Ludwig Institute Methods for determining disorders by assaying for a non-tyrosinase, tumor rejection antigen precursor
US6265150B1 (en) * 1995-06-07 2001-07-24 Becton Dickinson & Company Phage antibodies
US5714352A (en) * 1996-03-20 1998-02-03 Xenotech Incorporated Directed switch-mediated DNA recombination
DK1066380T3 (en) 1998-05-19 2002-01-28 Avidex Ltd Soluble T cell receptor
WO2001058479A1 (en) * 2000-02-08 2001-08-16 The Penn State Research Foundation Immunotherapy using interleukin 13 receptor subunit alpha 2
PT1421115E (en) 2001-08-31 2005-07-29 Avidex Ltd T SOLUVEL CELL RECEIVER
ATE432290T1 (en) 2002-11-09 2009-06-15 Immunocore Ltd T CELL RECEPTOR ßDISPLAYß
US20060135418A1 (en) 2002-12-03 2006-06-22 Avidex Ltd. Receptors
JP5563194B2 (en) * 2004-06-29 2014-07-30 イムノコア リミテッド Cells expressing modified T cell receptors
GB0511124D0 (en) * 2005-06-01 2005-07-06 Avidex Ltd High affinity melan-a t cell receptors
ATE550356T1 (en) * 2006-05-03 2012-04-15 Us Gov Health & Human Serv CHIMERIC T-CELL RECEPTORS AND CORRESPONDING MATERIALS AND METHODS OF USE
WO2008039818A2 (en) 2006-09-26 2008-04-03 Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Modified t cell receptors and related materials and methods

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005114215A2 (en) * 2004-05-19 2005-12-01 Avidex Ltd Method of improving t cell receptors
WO2005113595A2 (en) * 2004-05-19 2005-12-01 Avidex Ltd High affinity ny-eso t cell receptor

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
DUNN STEVEN M ET AL: "Directed evolution of human T cell receptor CDR2 residues by phage display dramatically enhances affinity for cognate peptide-MHC without increasing apparent cross-reactivity" PROTEIN SCIENCE, vol. 15, no. 4, April 2006 (2006-04), pages 710-721, XP002465974 ISSN: 0961-8368 *
LI YI ET AL: "Directed evolution of human T-cell receptors with picomolar affinities by phage display" NATURE BIOTECHNOLOGY, NATURE PUBLISHING GROUP, NEW YORK, NY, US, vol. 23, no. 3, March 2005 (2005-03), pages 349-354, XP002336795 ISSN: 1087-0156 *
MORRIS ET AL: "Generation of tumor-specific T-cell therapies" BLOOD REVIEWS, CHURCHILL LIVINGSTONE, vol. 20, no. 2, March 2006 (2006-03), pages 61-69, XP005302162 ISSN: 0268-960X *
PURBHOO MARCO A ET AL: "Quantifying and imaging NY-ESO-1/LAGE-1-derived epitopes on tumor cells using high affinity T cell receptors" JOURNAL OF IMMUNOLOGY, vol. 176, no. 12, June 2006 (2006-06), pages 7308-7316, XP002465973 ISSN: 0022-1767 *
ZHAO YANGBING ET AL: "High-affinity TCRs generated by phage display provide CD4(+) T cells with the ability to recognize and kill tumor cell lines" JOURNAL OF IMMUNOLOGY, vol. 179, no. 9, November 2007 (2007-11), pages 5845-5854, XP002465972 ISSN: 0022-1767 *
ZHAO YANGBING ET AL: "Primary human lymphocytes transduced with NY-ESO-1 antigen-specific TCR genes recognize and kill diverse human tumor cell lines" JOURNAL OF IMMUNOLOGY, WILLIAMS & WILKINS CO, US, vol. 174, no. 7, April 2005 (2005-04), pages 4415-4423, XP002343301 ISSN: 0022-1767 *

Cited By (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9597384B2 (en) 2001-03-16 2017-03-21 Helmholtz Zentrum München Deutsches Forschungszentrum Für Gesundheit Und Umwelt (Gmbh) Semi-allogenic anti-tumour vaccine with HLA haplo-identical antigen-presenting cells
US9238063B2 (en) 2001-03-16 2016-01-19 Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundlheit und Umwelt (GmbH) Semi-allogenic anti-tumour vaccine with HLA haplo-identical antigen-presenting cells
US10760055B2 (en) 2005-10-18 2020-09-01 National Jewish Health Conditionally immortalized long-term stem cells and methods of making and using such cells
WO2009042570A3 (en) * 2007-09-25 2009-06-04 Us Health Modified t cell receptors and related materials and methods
WO2009042570A2 (en) * 2007-09-25 2009-04-02 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Modified t cell receptors and related materials and methods
US11667695B2 (en) 2008-05-16 2023-06-06 Taiga Biotechnologies, Inc. Antibodies and processes for preparing the same
US11369678B2 (en) 2008-08-28 2022-06-28 Taiga Biotechnologies, Inc. Compositions and methods for modulating immune cells
US8697854B2 (en) 2008-11-24 2014-04-15 Helmholtz Zentrum München Deutsches Forschungszentrum Für Gesundheit Und Umwelt Gmbh High affinity T cell receptor and use thereof
US9862755B2 (en) 2008-11-24 2018-01-09 Max-Delbrueck-Centrum Fuer Molekulare Medizin High affinity T cell receptor and use thereof
US10626159B2 (en) 2008-11-24 2020-04-21 Max-Delbrueck-Centrum Fuer Molekulare Medizin High affinity T cell receptor and use thereof
US8785601B2 (en) 2009-01-28 2014-07-22 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services T cell receptors and related materials and methods of use
US9688739B2 (en) 2009-01-28 2017-06-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services T cell receptors and related materials and methods of use
WO2010089412A1 (en) 2009-02-09 2010-08-12 Helmholtz Zentrum Muenchen Deutsches Forschungszentrum Fuer Gesundheit Und Umwelt (Gmbh) Repertoire of allo-restricted peptide-specific t cell receptor sequences and use thereof
US9409969B2 (en) 2009-02-09 2016-08-09 Helmholtz Zentrum München Deutsches Forschungszentrum Für Gesundheit Und Umwelt (Gmbh) Repertoire of allo-restricted peptide-specific T cell receptor sequences and use thereof
US8556882B2 (en) 2009-04-30 2013-10-15 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Inducible interleukin-12
US8722049B2 (en) 2011-11-03 2014-05-13 Tolera Therapeutics, Inc. Antibody and methods for selective inhibition of T-cell responses
US8524234B2 (en) 2011-11-03 2013-09-03 Tolera Therapeutics, Inc Antibody for selective inhibition of T-cell responses
US8968739B2 (en) 2011-11-03 2015-03-03 Tolera Therapeutics, Inc. Antibody and methods for selective inhibition of T-cell responses
US10953048B2 (en) 2012-07-20 2021-03-23 Taiga Biotechnologies, Inc. Enhanced reconstitution and autoreconstitution of the hematopoietic compartment
EP2877489A4 (en) * 2012-07-27 2016-04-13 Univ Illinois Engineering t-cell receptors
US11384133B2 (en) 2012-07-27 2022-07-12 The Board Of Trustees Of The University Of Illinois Engineering t cell receptors
US9861688B2 (en) 2012-12-14 2018-01-09 Universitaetsmedizin Der Johannes Gutenberg-Universitaet Mainz T cell receptors with MHC independent binding to GM-CSF receptor alpha chain
US10987413B2 (en) 2012-12-14 2021-04-27 Biontech Rna Pharmaceuticals Gmbh MHC-independent tumor-associated antigens
US20150313978A1 (en) * 2012-12-14 2015-11-05 Universitätsmedizin Der Johannes Gutenberg- Universität Mainz Novel MHC-Independent Tumor-Associated Antigens
US10786534B2 (en) 2013-03-11 2020-09-29 Taiga Biotechnologies, Inc. Production and use of red blood cells
US10801070B2 (en) 2013-11-25 2020-10-13 The Broad Institute, Inc. Compositions and methods for diagnosing, evaluating and treating cancer
US11834718B2 (en) 2013-11-25 2023-12-05 The Broad Institute, Inc. Compositions and methods for diagnosing, evaluating and treating cancer by means of the DNA methylation status
US11725237B2 (en) 2013-12-05 2023-08-15 The Broad Institute Inc. Polymorphic gene typing and somatic change detection using sequencing data
US10370427B2 (en) 2013-12-19 2019-08-06 Friedrich-Alexander Universität Erlangen-Nürnberg Influenza-specific T-cell receptor and its uses in the detection, prevention and/or treatment of influenza
WO2015091823A1 (en) * 2013-12-19 2015-06-25 Friedrich-Alexander Universität Erlangen-Nürnberg Influenza-specific t-cell receptor and its uses in the detection, prevention and/or treatment of influenza
US11452768B2 (en) 2013-12-20 2022-09-27 The Broad Institute, Inc. Combination therapy with neoantigen vaccine
US11208661B2 (en) 2014-10-31 2021-12-28 The Trustees Of The University Of Pennsylvania Altering gene expression in modified T cells and uses thereof
US11203758B2 (en) 2014-10-31 2021-12-21 The Trustees Of The University Of Pennsylvania Altering gene expression in modified T cells and uses thereof
US10993997B2 (en) 2014-12-19 2021-05-04 The Broad Institute, Inc. Methods for profiling the t cell repertoire
EP3757211A1 (en) 2014-12-19 2020-12-30 The Broad Institute, Inc. Methods for profiling the t-cell-receptor repertoire
WO2016100977A1 (en) 2014-12-19 2016-06-23 The Broad Institute Inc. Methods for profiling the t-cel- receptor repertoire
US10975442B2 (en) 2014-12-19 2021-04-13 Massachusetts Institute Of Technology Molecular biomarkers for cancer immunotherapy
US11939637B2 (en) 2014-12-19 2024-03-26 Massachusetts Institute Of Technology Molecular biomarkers for cancer immunotherapy
US10835585B2 (en) 2015-05-20 2020-11-17 The Broad Institute, Inc. Shared neoantigens
US11180751B2 (en) 2015-06-18 2021-11-23 The Broad Institute, Inc. CRISPR enzymes and systems
WO2017069958A2 (en) 2015-10-09 2017-04-27 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
US11180730B2 (en) 2015-10-28 2021-11-23 The Broad Institute, Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting GATA3
WO2017075451A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting pou2af1
US11186825B2 (en) 2015-10-28 2021-11-30 The Broad Institute, Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting POU2AF1
WO2017075465A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting gata3
WO2017075478A2 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by use of immune cell gene signatures
US11001622B2 (en) 2015-11-19 2021-05-11 The Brigham And Women's Hospital, Inc. Method of treating autoimmune disease with lymphocyte antigen CD5-like (CD5L) protein
US11884717B2 (en) 2015-11-19 2024-01-30 The Brigham And Women's Hospital, Inc. Method of treating autoimmune disease with lymphocyte antigen CD5-like (CD5L) protein
WO2017087708A1 (en) 2015-11-19 2017-05-26 The Brigham And Women's Hospital, Inc. Lymphocyte antigen cd5-like (cd5l)-interleukin 12b (p40) heterodimers in immunity
US11639374B2 (en) 2015-12-22 2023-05-02 Immunocore Limited T cell receptors specific for the NY-ESO-1 tumor antigen-HLA-A*02 complex
WO2017184590A1 (en) 2016-04-18 2017-10-26 The Broad Institute Inc. Improved hla epitope prediction
US11630103B2 (en) 2016-08-17 2023-04-18 The Broad Institute, Inc. Product and methods useful for modulating and evaluating immune responses
WO2018035364A1 (en) 2016-08-17 2018-02-22 The Broad Institute Inc. Product and methods useful for modulating and evaluating immune responses
WO2018049025A2 (en) 2016-09-07 2018-03-15 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses
WO2018067991A1 (en) 2016-10-07 2018-04-12 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
US11116796B2 (en) 2016-12-02 2021-09-14 Taiga Biotechnologies, Inc. Nanoparticle formulations
US11549149B2 (en) 2017-01-24 2023-01-10 The Broad Institute, Inc. Compositions and methods for detecting a mutant variant of a polynucleotide
US11650211B2 (en) 2017-02-12 2023-05-16 Biontech Us Inc. HLA-based methods and compositions and uses thereof
EP4287191A2 (en) 2017-02-12 2023-12-06 BioNTech US Inc. Hla-based methods and compositions and uses thereof
WO2018148671A1 (en) 2017-02-12 2018-08-16 Neon Therapeutics, Inc. Hla-based methods and compositions and uses thereof
US11963966B2 (en) 2017-03-31 2024-04-23 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating ovarian tumors
US11913075B2 (en) 2017-04-01 2024-02-27 The Broad Institute, Inc. Methods and compositions for detecting and modulating an immunotherapy resistance gene signature in cancer
WO2018191553A1 (en) 2017-04-12 2018-10-18 Massachusetts Eye And Ear Infirmary Tumor signature for metastasis, compositions of matter methods of use thereof
WO2018195019A1 (en) 2017-04-18 2018-10-25 The Broad Institute Inc. Compositions for detecting secretion and methods of use
US11897953B2 (en) 2017-06-14 2024-02-13 The Broad Institute, Inc. Compositions and methods targeting complement component 3 for inhibiting tumor growth
US10864259B2 (en) 2017-08-03 2020-12-15 Taiga Biotechnologies, Inc. Methods and compositions for the treatment of melanoma
US10149898B2 (en) 2017-08-03 2018-12-11 Taiga Biotechnologies, Inc. Methods and compositions for the treatment of melanoma
WO2019027465A1 (en) 2017-08-03 2019-02-07 Taiga Biotechnologies, Inc. Methods and compositions for the treatment of melanoma
EP4026554A1 (en) 2017-08-03 2022-07-13 Taiga Biotechnologies, Inc. Methods and compositions for the treatment of melanoma
WO2019060746A1 (en) 2017-09-21 2019-03-28 The Broad Institute, Inc. Systems, methods, and compositions for targeted nucleic acid editing
US11732257B2 (en) 2017-10-23 2023-08-22 Massachusetts Institute Of Technology Single cell sequencing libraries of genomic transcript regions of interest in proximity to barcodes, and genotyping of said libraries
WO2019094983A1 (en) 2017-11-13 2019-05-16 The Broad Institute, Inc. Methods and compositions for treating cancer by targeting the clec2d-klrb1 pathway
US11957695B2 (en) 2018-04-26 2024-04-16 The Broad Institute, Inc. Methods and compositions targeting glucocorticoid signaling for modulating immune responses
WO2019232542A2 (en) 2018-06-01 2019-12-05 Massachusetts Institute Of Technology Methods and compositions for detecting and modulating microenvironment gene signatures from the csf of metastasis patients
CN112469733B (en) * 2018-07-12 2022-04-19 深圳华大生命科学研究院 MART-1(27-35) epitope-specific T cell receptor
WO2020010565A1 (en) * 2018-07-12 2020-01-16 深圳华大生命科学研究院 Mart-1(27-35) epitope-specific t cell receptor
CN112469733A (en) * 2018-07-12 2021-03-09 深圳华大生命科学研究院 MART-1(27-35) epitope-specific T cell receptor
WO2020041387A1 (en) 2018-08-20 2020-02-27 The Brigham And Women's Hospital, Inc. Degradation domain modifications for spatio-temporal control of rna-guided nucleases
WO2020041384A1 (en) 2018-08-20 2020-02-27 The Broad Institute, Inc. 3-phenyl-2-cyano-azetidine derivatives, inhibitors of rna-guided nuclease activity
WO2020068304A2 (en) 2018-08-20 2020-04-02 The Broad Institute, Inc. Inhibitors of rna-guided nuclease target binding and uses thereof
WO2020072700A1 (en) 2018-10-02 2020-04-09 Dana-Farber Cancer Institute, Inc. Hla single allele lines
WO2020081730A2 (en) 2018-10-16 2020-04-23 Massachusetts Institute Of Technology Methods and compositions for modulating microenvironment
WO2020092455A2 (en) 2018-10-29 2020-05-07 The Broad Institute, Inc. Car t cell transcriptional atlas
WO2020131586A2 (en) 2018-12-17 2020-06-25 The Broad Institute, Inc. Methods for identifying neoantigens
US11183272B2 (en) 2018-12-21 2021-11-23 Biontech Us Inc. Method and systems for prediction of HLA class II-specific epitopes and characterization of CD4+ T cells
US11739156B2 (en) 2019-01-06 2023-08-29 The Broad Institute, Inc. Massachusetts Institute of Technology Methods and compositions for overcoming immunosuppression
WO2020186101A1 (en) 2019-03-12 2020-09-17 The Broad Institute, Inc. Detection means, compositions and methods for modulating synovial sarcoma cells
WO2020191079A1 (en) 2019-03-18 2020-09-24 The Broad Institute, Inc. Compositions and methods for modulating metabolic regulators of t cell pathogenicity
WO2020236967A1 (en) 2019-05-20 2020-11-26 The Broad Institute, Inc. Random crispr-cas deletion mutant
WO2020243371A1 (en) 2019-05-28 2020-12-03 Massachusetts Institute Of Technology Methods and compositions for modulating immune responses
WO2021030627A1 (en) 2019-08-13 2021-02-18 The General Hospital Corporation Methods for predicting outcomes of checkpoint inhibition and treatment thereof
WO2021041922A1 (en) 2019-08-30 2021-03-04 The Broad Institute, Inc. Crispr-associated mu transposase systems
US11793787B2 (en) 2019-10-07 2023-10-24 The Broad Institute, Inc. Methods and compositions for enhancing anti-tumor immunity by targeting steroidogenesis
US11844800B2 (en) 2019-10-30 2023-12-19 Massachusetts Institute Of Technology Methods and compositions for predicting and preventing relapse of acute lymphoblastic leukemia
US11865168B2 (en) 2019-12-30 2024-01-09 Massachusetts Institute Of Technology Compositions and methods for treating bacterial infections
US11981922B2 (en) 2020-10-05 2024-05-14 Dana-Farber Cancer Institute, Inc. Methods and compositions for the modulation of cell interactions and signaling in the tumor microenvironment
WO2022187280A1 (en) 2021-03-01 2022-09-09 Dana-Farber Cancer Institute, Inc. Personalized redirection and reprogramming of t cells for precise targeting of tumors
WO2022261018A1 (en) 2021-06-07 2022-12-15 Providence Health & Services - Oregon Cxcr5, pd-1, and icos expressing tumor reactive cd4 t cells and their use
WO2024077256A1 (en) 2022-10-07 2024-04-11 The General Hospital Corporation Methods and compositions for high-throughput discovery ofpeptide-mhc targeting binding proteins

Also Published As

Publication number Publication date
US8088379B2 (en) 2012-01-03
WO2008039818A3 (en) 2008-06-05
US20120071420A1 (en) 2012-03-22
US20100034834A1 (en) 2010-02-11
US9128080B2 (en) 2015-09-08

Similar Documents

Publication Publication Date Title
US9128080B2 (en) Modified T cell receptors and related materials and methods
US10407485B2 (en) Murine anti-NY-ESO-1 T cell receptors
US20240084007A1 (en) M971 chimeric antigen receptors
AU2017225049B2 (en) Anti-cd22 chimeric antigen receptors
EP2016102B1 (en) Chimeric t cell receptors and related materials and methods of use
US9567387B2 (en) T cell receptors and related materials and methods of use
EP2828290B1 (en) Anti-mesothelin chimeric antigen receptors
EP3533802B1 (en) Anti-ssx-2 t cell receptors and related materials and methods of use
US20100297093A1 (en) Modified t cell receptors and related materials and methods
EP3077416B1 (en) Thymic stromal lymphopoietin receptor-specific chimeric antigen receptors and methods using same
WO2010075417A1 (en) Survivin specific t cell receptor for treating cancer

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: 07843200

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12443111

Country of ref document: US

122 Ep: pct application non-entry in european phase

Ref document number: 07843200

Country of ref document: EP

Kind code of ref document: A2