US20040224402A1 - Generation and isolation of antigen-specific T cells - Google Patents
Generation and isolation of antigen-specific T cells Download PDFInfo
- Publication number
- US20040224402A1 US20040224402A1 US10/742,622 US74262203A US2004224402A1 US 20040224402 A1 US20040224402 A1 US 20040224402A1 US 74262203 A US74262203 A US 74262203A US 2004224402 A1 US2004224402 A1 US 2004224402A1
- Authority
- US
- United States
- Prior art keywords
- cells
- antigen
- antibody
- cancer
- cell
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Abandoned
Links
- 210000001744 T-lymphocyte Anatomy 0.000 title claims abstract description 328
- 239000000427 antigen Substances 0.000 title claims abstract description 264
- 108091007433 antigens Proteins 0.000 title claims abstract description 264
- 102000036639 antigens Human genes 0.000 title claims abstract description 264
- 238000002955 isolation Methods 0.000 title description 10
- 238000000034 method Methods 0.000 claims abstract description 130
- 239000000203 mixture Substances 0.000 claims abstract description 47
- 210000004027 cell Anatomy 0.000 claims description 357
- 239000011324 bead Substances 0.000 claims description 154
- 210000000612 antigen-presenting cell Anatomy 0.000 claims description 88
- 239000003795 chemical substances by application Substances 0.000 claims description 72
- 206010028980 Neoplasm Diseases 0.000 claims description 59
- 239000003446 ligand Substances 0.000 claims description 39
- 241000701022 Cytomegalovirus Species 0.000 claims description 34
- 201000011510 cancer Diseases 0.000 claims description 26
- 241000124008 Mammalia Species 0.000 claims description 24
- 108090000623 proteins and genes Proteins 0.000 claims description 24
- 241000700605 Viruses Species 0.000 claims description 23
- 210000001519 tissue Anatomy 0.000 claims description 21
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 19
- 102000004169 proteins and genes Human genes 0.000 claims description 19
- 230000004936 stimulating effect Effects 0.000 claims description 18
- 210000001616 monocyte Anatomy 0.000 claims description 17
- 241000701044 Human gammaherpesvirus 4 Species 0.000 claims description 16
- 241000725303 Human immunodeficiency virus Species 0.000 claims description 16
- 230000005298 paramagnetic effect Effects 0.000 claims description 16
- 108020004414 DNA Proteins 0.000 claims description 15
- 208000015181 infectious disease Diseases 0.000 claims description 15
- 230000035755 proliferation Effects 0.000 claims description 15
- 241000700584 Simplexvirus Species 0.000 claims description 13
- 108091008048 CMVpp65 Proteins 0.000 claims description 12
- 208000005176 Hepatitis C Diseases 0.000 claims description 12
- 241000701806 Human papillomavirus Species 0.000 claims description 12
- 230000028993 immune response Effects 0.000 claims description 12
- 239000006166 lysate Substances 0.000 claims description 12
- 201000001441 melanoma Diseases 0.000 claims description 12
- 230000003993 interaction Effects 0.000 claims description 11
- 230000005291 magnetic effect Effects 0.000 claims description 11
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 11
- 208000031261 Acute myeloid leukaemia Diseases 0.000 claims description 10
- 208000035473 Communicable disease Diseases 0.000 claims description 10
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 claims description 10
- 230000000735 allogeneic effect Effects 0.000 claims description 10
- 210000004881 tumor cell Anatomy 0.000 claims description 9
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 claims description 8
- 241000894006 Bacteria Species 0.000 claims description 8
- 102000053602 DNA Human genes 0.000 claims description 8
- 206010035226 Plasma cell myeloma Diseases 0.000 claims description 8
- 230000001464 adherent effect Effects 0.000 claims description 8
- 239000002158 endotoxin Substances 0.000 claims description 8
- 230000002401 inhibitory effect Effects 0.000 claims description 8
- 230000001640 apoptogenic effect Effects 0.000 claims description 7
- 229920006008 lipopolysaccharide Polymers 0.000 claims description 7
- 241000228212 Aspergillus Species 0.000 claims description 6
- 208000032791 BCR-ABL1 positive chronic myelogenous leukemia Diseases 0.000 claims description 6
- 206010006187 Breast cancer Diseases 0.000 claims description 6
- 208000026310 Breast neoplasm Diseases 0.000 claims description 6
- 241000222120 Candida <Saccharomycetales> Species 0.000 claims description 6
- 206010009944 Colon cancer Diseases 0.000 claims description 6
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 claims description 6
- 241000233872 Pneumocystis carinii Species 0.000 claims description 6
- 238000011161 development Methods 0.000 claims description 6
- 241001493065 dsRNA viruses Species 0.000 claims description 6
- 208000005252 hepatitis A Diseases 0.000 claims description 6
- 208000002672 hepatitis B Diseases 0.000 claims description 6
- 230000002458 infectious effect Effects 0.000 claims description 6
- 206010022000 influenza Diseases 0.000 claims description 6
- 208000032839 leukemia Diseases 0.000 claims description 6
- 150000002632 lipids Chemical class 0.000 claims description 6
- 230000001338 necrotic effect Effects 0.000 claims description 6
- 244000045947 parasite Species 0.000 claims description 6
- 210000005259 peripheral blood Anatomy 0.000 claims description 6
- 239000011886 peripheral blood Substances 0.000 claims description 6
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 claims description 5
- 208000003174 Brain Neoplasms Diseases 0.000 claims description 5
- 206010008342 Cervix carcinoma Diseases 0.000 claims description 5
- 208000001333 Colorectal Neoplasms Diseases 0.000 claims description 5
- 208000000461 Esophageal Neoplasms Diseases 0.000 claims description 5
- 208000032612 Glial tumor Diseases 0.000 claims description 5
- 206010018338 Glioma Diseases 0.000 claims description 5
- 208000017604 Hodgkin disease Diseases 0.000 claims description 5
- 208000010747 Hodgkins lymphoma Diseases 0.000 claims description 5
- 101000851370 Homo sapiens Tumor necrosis factor receptor superfamily member 9 Proteins 0.000 claims description 5
- 208000008839 Kidney Neoplasms Diseases 0.000 claims description 5
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 5
- 208000034578 Multiple myelomas Diseases 0.000 claims description 5
- 206010030155 Oesophageal carcinoma Diseases 0.000 claims description 5
- 206010033128 Ovarian cancer Diseases 0.000 claims description 5
- 206010061535 Ovarian neoplasm Diseases 0.000 claims description 5
- 206010061902 Pancreatic neoplasm Diseases 0.000 claims description 5
- 208000007452 Plasmacytoma Diseases 0.000 claims description 5
- 206010060862 Prostate cancer Diseases 0.000 claims description 5
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims description 5
- 206010038389 Renal cancer Diseases 0.000 claims description 5
- 208000006265 Renal cell carcinoma Diseases 0.000 claims description 5
- 206010039491 Sarcoma Diseases 0.000 claims description 5
- 102100036856 Tumor necrosis factor receptor superfamily member 9 Human genes 0.000 claims description 5
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 claims description 5
- 238000001574 biopsy Methods 0.000 claims description 5
- 210000001185 bone marrow Anatomy 0.000 claims description 5
- 150000001720 carbohydrates Chemical class 0.000 claims description 5
- 235000014633 carbohydrates Nutrition 0.000 claims description 5
- 201000010881 cervical cancer Diseases 0.000 claims description 5
- 201000004101 esophageal cancer Diseases 0.000 claims description 5
- 206010073071 hepatocellular carcinoma Diseases 0.000 claims description 5
- 201000010982 kidney cancer Diseases 0.000 claims description 5
- 201000005202 lung cancer Diseases 0.000 claims description 5
- 208000020816 lung neoplasm Diseases 0.000 claims description 5
- 210000001165 lymph node Anatomy 0.000 claims description 5
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 claims description 5
- 201000002528 pancreatic cancer Diseases 0.000 claims description 5
- 208000008443 pancreatic carcinoma Diseases 0.000 claims description 5
- 208000008732 thymoma Diseases 0.000 claims description 5
- 201000008827 tuberculosis Diseases 0.000 claims description 5
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 claims description 4
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 claims description 4
- 102000003812 Interleukin-15 Human genes 0.000 claims description 4
- 108090000172 Interleukin-15 Proteins 0.000 claims description 4
- 108010010995 MART-1 Antigen Proteins 0.000 claims description 4
- 102100034256 Mucin-1 Human genes 0.000 claims description 4
- 108010008707 Mucin-1 Proteins 0.000 claims description 4
- 108020004682 Single-Stranded DNA Proteins 0.000 claims description 4
- 210000004700 fetal blood Anatomy 0.000 claims description 4
- 210000000952 spleen Anatomy 0.000 claims description 4
- 210000001541 thymus gland Anatomy 0.000 claims description 4
- NEHKZPHIKKEMAZ-ZFVKSOIMSA-N (2s)-2-[[(2s,3r)-2-[[(2s)-2-[[(2s,3s)-2-[[2-[[(2s,3s)-2-[[2-[[(2s)-2-[[(2s)-2-azaniumylpropanoyl]amino]propanoyl]amino]acetyl]amino]-3-methylpentanoyl]amino]acetyl]amino]-3-methylpentanoyl]amino]-4-methylpentanoyl]amino]-3-hydroxybutanoyl]amino]-3-methylb Chemical compound C[C@H](N)C(=O)N[C@@H](C)C(=O)NCC(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(O)=O NEHKZPHIKKEMAZ-ZFVKSOIMSA-N 0.000 claims description 3
- 101710117545 C protein Proteins 0.000 claims description 3
- 108010003454 EBNA-3B antigen Proteins 0.000 claims description 3
- 102000003886 Glycoproteins Human genes 0.000 claims description 3
- 108090000288 Glycoproteins Proteins 0.000 claims description 3
- 101001005728 Homo sapiens Melanoma-associated antigen 1 Proteins 0.000 claims description 3
- 108090001030 Lipoproteins Proteins 0.000 claims description 3
- 102000004895 Lipoproteins Human genes 0.000 claims description 3
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 claims description 3
- 102000016200 MART-1 Antigen Human genes 0.000 claims description 3
- 102100025050 Melanoma-associated antigen 1 Human genes 0.000 claims description 3
- 102000003425 Tyrosinase Human genes 0.000 claims description 3
- 108060008724 Tyrosinase Proteins 0.000 claims description 3
- 108010026331 alpha-Fetoproteins Proteins 0.000 claims description 3
- 230000036436 anti-hiv Effects 0.000 claims description 3
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 claims description 3
- 230000004064 dysfunction Effects 0.000 claims description 3
- 201000000050 myeloid neoplasm Diseases 0.000 claims description 3
- AEMBWNDIEFEPTH-UHFFFAOYSA-N n-tert-butyl-n-ethylnitrous amide Chemical compound CCN(N=O)C(C)(C)C AEMBWNDIEFEPTH-UHFFFAOYSA-N 0.000 claims description 3
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 3
- 101001109501 Homo sapiens NKG2-D type II integral membrane protein Proteins 0.000 claims description 2
- 102100022680 NKG2-D type II integral membrane protein Human genes 0.000 claims description 2
- 230000001939 inductive effect Effects 0.000 claims description 2
- 210000002741 palatine tonsil Anatomy 0.000 claims description 2
- 102000013529 alpha-Fetoproteins Human genes 0.000 claims 1
- 239000002245 particle Substances 0.000 description 50
- 230000000638 stimulation Effects 0.000 description 44
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 38
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 31
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 31
- 230000008569 process Effects 0.000 description 19
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 18
- 102000004127 Cytokines Human genes 0.000 description 17
- 108090000695 Cytokines Proteins 0.000 description 17
- 230000004913 activation Effects 0.000 description 17
- 102100034922 T-cell surface glycoprotein CD8 alpha chain Human genes 0.000 description 16
- 239000012634 fragment Substances 0.000 description 16
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 description 15
- 238000002617 apheresis Methods 0.000 description 14
- 230000010261 cell growth Effects 0.000 description 14
- 239000000126 substance Substances 0.000 description 14
- 239000000047 product Substances 0.000 description 13
- 238000011282 treatment Methods 0.000 description 13
- 108010002350 Interleukin-2 Proteins 0.000 description 12
- 102000000588 Interleukin-2 Human genes 0.000 description 12
- 241001465754 Metazoa Species 0.000 description 12
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 12
- 239000002243 precursor Substances 0.000 description 12
- 108091008874 T cell receptors Proteins 0.000 description 11
- 230000027455 binding Effects 0.000 description 11
- 210000004369 blood Anatomy 0.000 description 11
- 239000008280 blood Substances 0.000 description 11
- 102000005962 receptors Human genes 0.000 description 11
- 108020003175 receptors Proteins 0.000 description 11
- 201000010099 disease Diseases 0.000 description 10
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 10
- 239000002609 medium Substances 0.000 description 10
- 210000002966 serum Anatomy 0.000 description 10
- 239000000243 solution Substances 0.000 description 10
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 9
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 9
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 9
- 230000006870 function Effects 0.000 description 9
- 239000001963 growth medium Substances 0.000 description 9
- -1 hapten/antibody Substances 0.000 description 9
- 239000008194 pharmaceutical composition Substances 0.000 description 9
- 239000002953 phosphate buffered saline Substances 0.000 description 9
- 230000001177 retroviral effect Effects 0.000 description 9
- 238000001890 transfection Methods 0.000 description 9
- 239000013598 vector Substances 0.000 description 9
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 8
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 8
- 210000003719 b-lymphocyte Anatomy 0.000 description 8
- 238000005516 engineering process Methods 0.000 description 8
- 210000003743 erythrocyte Anatomy 0.000 description 8
- 238000001727 in vivo Methods 0.000 description 8
- 210000003071 memory t lymphocyte Anatomy 0.000 description 8
- 101100347633 Drosophila melanogaster Mhc gene Proteins 0.000 description 7
- 108010047620 Phytohemagglutinins Proteins 0.000 description 7
- 239000007758 minimum essential medium Substances 0.000 description 7
- 230000001885 phytohemagglutinin Effects 0.000 description 7
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 6
- 102100034540 Adenomatous polyposis coli protein Human genes 0.000 description 6
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 6
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 6
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 6
- 102100037877 Intercellular adhesion molecule 1 Human genes 0.000 description 6
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 6
- 108090000978 Interleukin-4 Proteins 0.000 description 6
- 102000004388 Interleukin-4 Human genes 0.000 description 6
- 108091000054 Prion Proteins 0.000 description 6
- 239000012980 RPMI-1640 medium Substances 0.000 description 6
- 210000001772 blood platelet Anatomy 0.000 description 6
- 239000012636 effector Substances 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 239000003102 growth factor Substances 0.000 description 6
- 238000011534 incubation Methods 0.000 description 6
- 238000001802 infusion Methods 0.000 description 6
- 229940028885 interleukin-4 Drugs 0.000 description 6
- 239000003226 mitogen Substances 0.000 description 6
- 229920001184 polypeptide Polymers 0.000 description 6
- 231100000617 superantigen Toxicity 0.000 description 6
- 230000003612 virological effect Effects 0.000 description 6
- 238000005406 washing Methods 0.000 description 6
- 108010090804 Streptavidin Proteins 0.000 description 5
- 230000006044 T cell activation Effects 0.000 description 5
- 239000000872 buffer Substances 0.000 description 5
- 230000002354 daily effect Effects 0.000 description 5
- 210000004443 dendritic cell Anatomy 0.000 description 5
- 230000000694 effects Effects 0.000 description 5
- 238000004520 electroporation Methods 0.000 description 5
- 230000012010 growth Effects 0.000 description 5
- 229940088597 hormone Drugs 0.000 description 5
- 239000005556 hormone Substances 0.000 description 5
- 210000000987 immune system Anatomy 0.000 description 5
- 230000001976 improved effect Effects 0.000 description 5
- 210000000265 leukocyte Anatomy 0.000 description 5
- 230000007774 longterm Effects 0.000 description 5
- 238000007885 magnetic separation Methods 0.000 description 5
- 239000011159 matrix material Substances 0.000 description 5
- 210000003289 regulatory T cell Anatomy 0.000 description 5
- 230000004044 response Effects 0.000 description 5
- 238000000926 separation method Methods 0.000 description 5
- 230000003068 static effect Effects 0.000 description 5
- 238000002560 therapeutic procedure Methods 0.000 description 5
- 238000010361 transduction Methods 0.000 description 5
- 230000026683 transduction Effects 0.000 description 5
- NLXLAEXVIDQMFP-UHFFFAOYSA-N Ammonia chloride Chemical compound [NH4+].[Cl-] NLXLAEXVIDQMFP-UHFFFAOYSA-N 0.000 description 4
- 108090001008 Avidin Proteins 0.000 description 4
- 102100032937 CD40 ligand Human genes 0.000 description 4
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 4
- 102000025850 HLA-A2 Antigen Human genes 0.000 description 4
- 108010074032 HLA-A2 Antigen Proteins 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- 102000043131 MHC class II family Human genes 0.000 description 4
- 108091054438 MHC class II family Proteins 0.000 description 4
- 241001529936 Murinae Species 0.000 description 4
- 239000012124 Opti-MEM Substances 0.000 description 4
- 102000029797 Prion Human genes 0.000 description 4
- 230000006052 T cell proliferation Effects 0.000 description 4
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 4
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 4
- 208000008383 Wilms tumor Diseases 0.000 description 4
- 150000001413 amino acids Chemical class 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 238000013270 controlled release Methods 0.000 description 4
- 238000012258 culturing Methods 0.000 description 4
- 230000018109 developmental process Effects 0.000 description 4
- 238000012239 gene modification Methods 0.000 description 4
- 230000005017 genetic modification Effects 0.000 description 4
- 235000013617 genetically modified food Nutrition 0.000 description 4
- 230000006698 induction Effects 0.000 description 4
- PHEDXBVPIONUQT-RGYGYFBISA-N phorbol 13-acetate 12-myristate Chemical compound C([C@]1(O)C(=O)C(C)=C[C@H]1[C@@]1(O)[C@H](C)[C@H]2OC(=O)CCCCCCCCCCCCC)C(CO)=C[C@H]1[C@H]1[C@]2(OC(C)=O)C1(C)C PHEDXBVPIONUQT-RGYGYFBISA-N 0.000 description 4
- 230000028327 secretion Effects 0.000 description 4
- 238000010561 standard procedure Methods 0.000 description 4
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 4
- 239000011782 vitamin Substances 0.000 description 4
- 235000013343 vitamin Nutrition 0.000 description 4
- 229940088594 vitamin Drugs 0.000 description 4
- 229930003231 vitamin Natural products 0.000 description 4
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 4
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 3
- 241000283690 Bos taurus Species 0.000 description 3
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 3
- 102000019034 Chemokines Human genes 0.000 description 3
- 108010012236 Chemokines Proteins 0.000 description 3
- PMATZTZNYRCHOR-CGLBZJNRSA-N Cyclosporin A Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 3
- 108010036949 Cyclosporine Proteins 0.000 description 3
- 101000599852 Homo sapiens Intercellular adhesion molecule 1 Proteins 0.000 description 3
- 108091006905 Human Serum Albumin Proteins 0.000 description 3
- 102000008100 Human Serum Albumin Human genes 0.000 description 3
- 108010064593 Intercellular Adhesion Molecule-1 Proteins 0.000 description 3
- 102100037850 Interferon gamma Human genes 0.000 description 3
- 108010074328 Interferon-gamma Proteins 0.000 description 3
- 102000003816 Interleukin-13 Human genes 0.000 description 3
- 108090000176 Interleukin-13 Proteins 0.000 description 3
- QJJXYPPXXYFBGM-LFZNUXCKSA-N Tacrolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1\C=C(/C)[C@@H]1[C@H](C)[C@@H](O)CC(=O)[C@H](CC=C)/C=C(C)/C[C@H](C)C[C@H](OC)[C@H]([C@H](C[C@H]2C)OC)O[C@@]2(O)C(=O)C(=O)N2CCCC[C@H]2C(=O)O1 QJJXYPPXXYFBGM-LFZNUXCKSA-N 0.000 description 3
- 101710101607 Toxic shock syndrome toxin-1 Proteins 0.000 description 3
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 3
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 3
- 239000000654 additive Substances 0.000 description 3
- 239000003242 anti bacterial agent Substances 0.000 description 3
- 229940088710 antibiotic agent Drugs 0.000 description 3
- 229960002685 biotin Drugs 0.000 description 3
- 235000020958 biotin Nutrition 0.000 description 3
- 239000011616 biotin Substances 0.000 description 3
- 239000011575 calcium Substances 0.000 description 3
- 229910052791 calcium Inorganic materials 0.000 description 3
- 229940112129 campath Drugs 0.000 description 3
- 238000005119 centrifugation Methods 0.000 description 3
- 229960001265 ciclosporin Drugs 0.000 description 3
- 229930182912 cyclosporin Natural products 0.000 description 3
- 230000016396 cytokine production Effects 0.000 description 3
- 230000009089 cytolysis Effects 0.000 description 3
- 238000012217 deletion Methods 0.000 description 3
- 230000037430 deletion Effects 0.000 description 3
- 239000003937 drug carrier Substances 0.000 description 3
- 230000001605 fetal effect Effects 0.000 description 3
- 238000000684 flow cytometry Methods 0.000 description 3
- 210000003714 granulocyte Anatomy 0.000 description 3
- 210000002443 helper t lymphocyte Anatomy 0.000 description 3
- 238000009169 immunotherapy Methods 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 238000004519 manufacturing process Methods 0.000 description 3
- 230000001404 mediated effect Effects 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 244000052769 pathogen Species 0.000 description 3
- 210000001539 phagocyte Anatomy 0.000 description 3
- 239000012071 phase Substances 0.000 description 3
- 238000012545 processing Methods 0.000 description 3
- 230000002062 proliferating effect Effects 0.000 description 3
- 230000002829 reductive effect Effects 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 230000019491 signal transduction Effects 0.000 description 3
- 241000894007 species Species 0.000 description 3
- 239000006228 supernatant Substances 0.000 description 3
- 230000004083 survival effect Effects 0.000 description 3
- QJJXYPPXXYFBGM-SHYZHZOCSA-N tacrolimus Natural products CO[C@H]1C[C@H](CC[C@@H]1O)C=C(C)[C@H]2OC(=O)[C@H]3CCCCN3C(=O)C(=O)[C@@]4(O)O[C@@H]([C@H](C[C@H]4C)OC)[C@@H](C[C@H](C)CC(=C[C@@H](CC=C)C(=O)C[C@H](O)[C@H]2C)C)OC QJJXYPPXXYFBGM-SHYZHZOCSA-N 0.000 description 3
- 241001430294 unidentified retrovirus Species 0.000 description 3
- 230000003827 upregulation Effects 0.000 description 3
- DQJCDTNMLBYVAY-ZXXIYAEKSA-N (2S,5R,10R,13R)-16-{[(2R,3S,4R,5R)-3-{[(2S,3R,4R,5S,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy}-5-(ethylamino)-6-hydroxy-2-(hydroxymethyl)oxan-4-yl]oxy}-5-(4-aminobutyl)-10-carbamoyl-2,13-dimethyl-4,7,12,15-tetraoxo-3,6,11,14-tetraazaheptadecan-1-oic acid Chemical compound NCCCC[C@H](C(=O)N[C@@H](C)C(O)=O)NC(=O)CC[C@H](C(N)=O)NC(=O)[C@@H](C)NC(=O)C(C)O[C@@H]1[C@@H](NCC)C(O)O[C@H](CO)[C@H]1O[C@H]1[C@H](NC(C)=O)[C@@H](O)[C@H](O)[C@@H](CO)O1 DQJCDTNMLBYVAY-ZXXIYAEKSA-N 0.000 description 2
- 102100023635 Alpha-fetoprotein Human genes 0.000 description 2
- 102100034452 Alternative prion protein Human genes 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 208000023275 Autoimmune disease Diseases 0.000 description 2
- 102100022005 B-lymphocyte antigen CD20 Human genes 0.000 description 2
- 108010029697 CD40 Ligand Proteins 0.000 description 2
- 108010022366 Carcinoembryonic Antigen Proteins 0.000 description 2
- 102100025475 Carcinoembryonic antigen-related cell adhesion molecule 5 Human genes 0.000 description 2
- 206010057248 Cell death Diseases 0.000 description 2
- 208000020406 Creutzfeldt Jacob disease Diseases 0.000 description 2
- 208000003407 Creutzfeldt-Jakob Syndrome Diseases 0.000 description 2
- 208000010859 Creutzfeldt-Jakob disease Diseases 0.000 description 2
- 229930105110 Cyclosporin A Natural products 0.000 description 2
- 241000450599 DNA viruses Species 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- 241000588724 Escherichia coli Species 0.000 description 2
- 241000233866 Fungi Species 0.000 description 2
- 208000003736 Gerstmann-Straussler-Scheinker Disease Diseases 0.000 description 2
- 206010072075 Gerstmann-Straussler-Scheinker syndrome Diseases 0.000 description 2
- 102000002068 Glycopeptides Human genes 0.000 description 2
- 108010015899 Glycopeptides Proteins 0.000 description 2
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 2
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 2
- 101000897405 Homo sapiens B-lymphocyte antigen CD20 Proteins 0.000 description 2
- 101000868215 Homo sapiens CD40 ligand Proteins 0.000 description 2
- 101000946889 Homo sapiens Monocyte differentiation antigen CD14 Proteins 0.000 description 2
- 101000679851 Homo sapiens Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 2
- 108010008212 Integrin alpha4beta1 Proteins 0.000 description 2
- 102000003814 Interleukin-10 Human genes 0.000 description 2
- 108090000174 Interleukin-10 Proteins 0.000 description 2
- 102000013462 Interleukin-12 Human genes 0.000 description 2
- 108010065805 Interleukin-12 Proteins 0.000 description 2
- 102000000646 Interleukin-3 Human genes 0.000 description 2
- 108010002386 Interleukin-3 Proteins 0.000 description 2
- 102000004889 Interleukin-6 Human genes 0.000 description 2
- 108090001005 Interleukin-6 Proteins 0.000 description 2
- PWKSKIMOESPYIA-BYPYZUCNSA-N L-N-acetyl-Cysteine Chemical compound CC(=O)N[C@@H](CS)C(O)=O PWKSKIMOESPYIA-BYPYZUCNSA-N 0.000 description 2
- 241000222722 Leishmania <genus> Species 0.000 description 2
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 2
- 102100028389 Melanoma antigen recognized by T-cells 1 Human genes 0.000 description 2
- 108010052285 Membrane Proteins Proteins 0.000 description 2
- 102000018697 Membrane Proteins Human genes 0.000 description 2
- 241000713869 Moloney murine leukemia virus Species 0.000 description 2
- 102100035877 Monocyte differentiation antigen CD14 Human genes 0.000 description 2
- 101000686985 Mouse mammary tumor virus (strain C3H) Protein PR73 Proteins 0.000 description 2
- 241000713883 Myeloproliferative sarcoma virus Species 0.000 description 2
- 229930182555 Penicillin Natural products 0.000 description 2
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 2
- 102000007066 Prostate-Specific Antigen Human genes 0.000 description 2
- 108010072866 Prostate-Specific Antigen Proteins 0.000 description 2
- 241000607142 Salmonella Species 0.000 description 2
- 241000242678 Schistosoma Species 0.000 description 2
- 229920002684 Sepharose Polymers 0.000 description 2
- 241000713880 Spleen focus-forming virus Species 0.000 description 2
- 241000295644 Staphylococcaceae Species 0.000 description 2
- 241000191940 Staphylococcus Species 0.000 description 2
- 102000007451 Steroid Receptors Human genes 0.000 description 2
- 108010085012 Steroid Receptors Proteins 0.000 description 2
- 230000024932 T cell mediated immunity Effects 0.000 description 2
- 241000223104 Trypanosoma Species 0.000 description 2
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 2
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 2
- 208000018756 Variant Creutzfeldt-Jakob disease Diseases 0.000 description 2
- 102100023543 Vascular cell adhesion protein 1 Human genes 0.000 description 2
- 238000010317 ablation therapy Methods 0.000 description 2
- 229960004308 acetylcysteine Drugs 0.000 description 2
- 230000003213 activating effect Effects 0.000 description 2
- 235000019270 ammonium chloride Nutrition 0.000 description 2
- 230000003171 anti-complementary effect Effects 0.000 description 2
- 230000000259 anti-tumor effect Effects 0.000 description 2
- 230000000890 antigenic effect Effects 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 208000005881 bovine spongiform encephalopathy Diseases 0.000 description 2
- 239000001506 calcium phosphate Substances 0.000 description 2
- 229910000389 calcium phosphate Inorganic materials 0.000 description 2
- 235000011010 calcium phosphates Nutrition 0.000 description 2
- 125000002091 cationic group Chemical group 0.000 description 2
- 150000001768 cations Chemical class 0.000 description 2
- 230000020411 cell activation Effects 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 238000001311 chemical methods and process Methods 0.000 description 2
- 239000003638 chemical reducing agent Substances 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- 238000005859 coupling reaction Methods 0.000 description 2
- 238000004132 cross linking Methods 0.000 description 2
- 230000007423 decrease Effects 0.000 description 2
- 230000003111 delayed effect Effects 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 230000003828 downregulation Effects 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 210000001671 embryonic stem cell Anatomy 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- 201000006061 fatal familial insomnia Diseases 0.000 description 2
- 239000012847 fine chemical Substances 0.000 description 2
- 230000008014 freezing Effects 0.000 description 2
- 238000007710 freezing Methods 0.000 description 2
- 238000010353 genetic engineering Methods 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- 230000001024 immunotherapeutic effect Effects 0.000 description 2
- 239000007943 implant Substances 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 2
- 229940076264 interleukin-3 Drugs 0.000 description 2
- 229940100601 interleukin-6 Drugs 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- PGHMRUGBZOYCAA-UHFFFAOYSA-N ionomycin Natural products O1C(CC(O)C(C)C(O)C(C)C=CCC(C)CC(C)C(O)=CC(=O)C(C)CC(C)CC(CCC(O)=O)C)CCC1(C)C1OC(C)(C(C)O)CC1 PGHMRUGBZOYCAA-UHFFFAOYSA-N 0.000 description 2
- PGHMRUGBZOYCAA-ADZNBVRBSA-N ionomycin Chemical compound O1[C@H](C[C@H](O)[C@H](C)[C@H](O)[C@H](C)/C=C/C[C@@H](C)C[C@@H](C)C(/O)=C/C(=O)[C@@H](C)C[C@@H](C)C[C@@H](CCC(O)=O)C)CC[C@@]1(C)[C@@H]1O[C@](C)([C@@H](C)O)CC1 PGHMRUGBZOYCAA-ADZNBVRBSA-N 0.000 description 2
- 239000000644 isotonic solution Substances 0.000 description 2
- 210000004698 lymphocyte Anatomy 0.000 description 2
- 210000003563 lymphoid tissue Anatomy 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 239000011777 magnesium Substances 0.000 description 2
- 229910052749 magnesium Inorganic materials 0.000 description 2
- 239000006249 magnetic particle Substances 0.000 description 2
- 230000003211 malignant effect Effects 0.000 description 2
- 210000004962 mammalian cell Anatomy 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 2
- 239000004005 microsphere Substances 0.000 description 2
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 2
- 239000002105 nanoparticle Substances 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 230000007935 neutral effect Effects 0.000 description 2
- 229940049954 penicillin Drugs 0.000 description 2
- 210000005105 peripheral blood lymphocyte Anatomy 0.000 description 2
- 108010058237 plasma protein fraction Proteins 0.000 description 2
- 229940002993 plasmanate Drugs 0.000 description 2
- 239000004033 plastic Substances 0.000 description 2
- 229920003023 plastic Polymers 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- ZAHRKKWIAAJSAO-UHFFFAOYSA-N rapamycin Natural products COCC(O)C(=C/C(C)C(=O)CC(OC(=O)C1CCCCN1C(=O)C(=O)C2(O)OC(CC(OC)C(=CC=CC=CC(C)CC(C)C(=O)C)C)CCC2C)C(C)CC3CCC(O)C(C3)OC)C ZAHRKKWIAAJSAO-UHFFFAOYSA-N 0.000 description 2
- 230000004043 responsiveness Effects 0.000 description 2
- 229960002930 sirolimus Drugs 0.000 description 2
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 description 2
- 230000009870 specific binding Effects 0.000 description 2
- 210000000130 stem cell Anatomy 0.000 description 2
- 238000003860 storage Methods 0.000 description 2
- 229960005322 streptomycin Drugs 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 239000004094 surface-active agent Substances 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 238000010257 thawing Methods 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 125000003396 thiol group Chemical group [H]S* 0.000 description 2
- 230000009466 transformation Effects 0.000 description 2
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 2
- 230000035899 viability Effects 0.000 description 2
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- KZMAWJRXKGLWGS-UHFFFAOYSA-N 2-chloro-n-[4-(4-methoxyphenyl)-1,3-thiazol-2-yl]-n-(3-methoxypropyl)acetamide Chemical compound S1C(N(C(=O)CCl)CCCOC)=NC(C=2C=CC(OC)=CC=2)=C1 KZMAWJRXKGLWGS-UHFFFAOYSA-N 0.000 description 1
- 108010013238 70-kDa Ribosomal Protein S6 Kinases Proteins 0.000 description 1
- 102100031585 ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Human genes 0.000 description 1
- 208000026872 Addison Disease Diseases 0.000 description 1
- 208000032467 Aplastic anaemia Diseases 0.000 description 1
- 241000271566 Aves Species 0.000 description 1
- 208000003950 B-cell lymphoma Diseases 0.000 description 1
- 102100036301 C-C chemokine receptor type 7 Human genes 0.000 description 1
- 108700012434 CCL3 Proteins 0.000 description 1
- 101150013553 CD40 gene Proteins 0.000 description 1
- 101100314454 Caenorhabditis elegans tra-1 gene Proteins 0.000 description 1
- 102000004631 Calcineurin Human genes 0.000 description 1
- 108010042955 Calcineurin Proteins 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 102000000013 Chemokine CCL3 Human genes 0.000 description 1
- 102000001327 Chemokine CCL5 Human genes 0.000 description 1
- 108010055166 Chemokine CCL5 Proteins 0.000 description 1
- 206010008874 Chronic Fatigue Syndrome Diseases 0.000 description 1
- 208000010833 Chronic myeloid leukaemia Diseases 0.000 description 1
- 208000015943 Coeliac disease Diseases 0.000 description 1
- 206010009900 Colitis ulcerative Diseases 0.000 description 1
- 102000008186 Collagen Human genes 0.000 description 1
- 108010035532 Collagen Proteins 0.000 description 1
- 102000007644 Colony-Stimulating Factors Human genes 0.000 description 1
- 108010071942 Colony-Stimulating Factors Proteins 0.000 description 1
- 208000011231 Crohn disease Diseases 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 102100025137 Early activation antigen CD69 Human genes 0.000 description 1
- 201000009273 Endometriosis Diseases 0.000 description 1
- 101710146739 Enterotoxin Proteins 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 239000004593 Epoxy Substances 0.000 description 1
- 101000867232 Escherichia coli Heat-stable enterotoxin II Proteins 0.000 description 1
- 208000004332 Evans syndrome Diseases 0.000 description 1
- 229940124135 Factor VIII inhibitor Drugs 0.000 description 1
- 108010087819 Fc receptors Proteins 0.000 description 1
- 102000009109 Fc receptors Human genes 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 208000001640 Fibromyalgia Diseases 0.000 description 1
- 102100020715 Fms-related tyrosine kinase 3 ligand protein Human genes 0.000 description 1
- 101710162577 Fms-related tyrosine kinase 3 ligand protein Proteins 0.000 description 1
- 229930182566 Gentamicin Natural products 0.000 description 1
- CEAZRRDELHUEMR-URQXQFDESA-N Gentamicin Chemical compound O1[C@H](C(C)NC)CC[C@@H](N)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](NC)[C@@](C)(O)CO2)O)[C@H](N)C[C@@H]1N CEAZRRDELHUEMR-URQXQFDESA-N 0.000 description 1
- 206010018364 Glomerulonephritis Diseases 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 1
- 208000024869 Goodpasture syndrome Diseases 0.000 description 1
- 208000003807 Graves Disease Diseases 0.000 description 1
- 208000015023 Graves' disease Diseases 0.000 description 1
- 102000006354 HLA-DR Antigens Human genes 0.000 description 1
- 108010058597 HLA-DR Antigens Proteins 0.000 description 1
- 239000012981 Hank's balanced salt solution Substances 0.000 description 1
- 208000001204 Hashimoto Disease Diseases 0.000 description 1
- 208000030836 Hashimoto thyroiditis Diseases 0.000 description 1
- 208000035186 Hemolytic Autoimmune Anemia Diseases 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 101000777636 Homo sapiens ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Proteins 0.000 description 1
- 101000716065 Homo sapiens C-C chemokine receptor type 7 Proteins 0.000 description 1
- 101000934374 Homo sapiens Early activation antigen CD69 Proteins 0.000 description 1
- 101000994375 Homo sapiens Integrin alpha-4 Proteins 0.000 description 1
- 101001046686 Homo sapiens Integrin alpha-M Proteins 0.000 description 1
- 101001018097 Homo sapiens L-selectin Proteins 0.000 description 1
- 101000917858 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 1
- 101000917839 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-B Proteins 0.000 description 1
- 101000578784 Homo sapiens Melanoma antigen recognized by T-cells 1 Proteins 0.000 description 1
- 101000946843 Homo sapiens T-cell surface glycoprotein CD8 alpha chain Proteins 0.000 description 1
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 1
- 206010020850 Hyperthyroidism Diseases 0.000 description 1
- 206010021245 Idiopathic thrombocytopenic purpura Diseases 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 208000029462 Immunodeficiency disease Diseases 0.000 description 1
- 206010062016 Immunosuppression Diseases 0.000 description 1
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 1
- 102000004877 Insulin Human genes 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 102100032818 Integrin alpha-4 Human genes 0.000 description 1
- 102100022339 Integrin alpha-L Human genes 0.000 description 1
- 102100022338 Integrin alpha-M Human genes 0.000 description 1
- 102000004125 Interleukin-1alpha Human genes 0.000 description 1
- 108010082786 Interleukin-1alpha Proteins 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- 229930182816 L-glutamine Natural products 0.000 description 1
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 1
- 102100033467 L-selectin Human genes 0.000 description 1
- 108090001090 Lectins Proteins 0.000 description 1
- 102000004856 Lectins Human genes 0.000 description 1
- 102100029185 Low affinity immunoglobulin gamma Fc region receptor III-B Human genes 0.000 description 1
- 108010064548 Lymphocyte Function-Associated Antigen-1 Proteins 0.000 description 1
- 102000008072 Lymphokines Human genes 0.000 description 1
- 108010074338 Lymphokines Proteins 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 108010063954 Mucins Proteins 0.000 description 1
- 201000003793 Myelodysplastic syndrome Diseases 0.000 description 1
- 208000033761 Myelogenous Chronic BCR-ABL Positive Leukemia Diseases 0.000 description 1
- 108010025020 Nerve Growth Factor Proteins 0.000 description 1
- 102000015336 Nerve Growth Factor Human genes 0.000 description 1
- 208000000733 Paroxysmal Hemoglobinuria Diseases 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 208000031845 Pernicious anaemia Diseases 0.000 description 1
- 102100036050 Phosphatidylinositol N-acetylglucosaminyltransferase subunit A Human genes 0.000 description 1
- 208000024777 Prion disease Diseases 0.000 description 1
- 102000003923 Protein Kinase C Human genes 0.000 description 1
- 108090000315 Protein Kinase C Proteins 0.000 description 1
- 201000004681 Psoriasis Diseases 0.000 description 1
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 206010039710 Scleroderma Diseases 0.000 description 1
- 206010040070 Septic Shock Diseases 0.000 description 1
- 208000021386 Sjogren Syndrome Diseases 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- 208000004732 Systemic Vasculitis Diseases 0.000 description 1
- 230000005867 T cell response Effects 0.000 description 1
- 108010092262 T-Cell Antigen Receptors Proteins 0.000 description 1
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 1
- 210000000662 T-lymphocyte subset Anatomy 0.000 description 1
- 208000031981 Thrombocytopenic Idiopathic Purpura Diseases 0.000 description 1
- 102000006601 Thymidine Kinase Human genes 0.000 description 1
- 108020004440 Thymidine kinase Proteins 0.000 description 1
- 206010044248 Toxic shock syndrome Diseases 0.000 description 1
- 231100000650 Toxic shock syndrome Toxicity 0.000 description 1
- 101710120037 Toxin CcdB Proteins 0.000 description 1
- 108700019146 Transgenes Proteins 0.000 description 1
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 1
- 201000006704 Ulcerative Colitis Diseases 0.000 description 1
- 206010046865 Vaccinia virus infection Diseases 0.000 description 1
- 108010000134 Vascular Cell Adhesion Molecule-1 Proteins 0.000 description 1
- 206010047115 Vasculitis Diseases 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 230000032683 aging Effects 0.000 description 1
- VREFGVBLTWBCJP-UHFFFAOYSA-N alprazolam Chemical compound C12=CC(Cl)=CC=C2N2C(C)=NN=C2CN=C1C1=CC=CC=C1 VREFGVBLTWBCJP-UHFFFAOYSA-N 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- 229940024606 amino acid Drugs 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 230000003302 anti-idiotype Effects 0.000 description 1
- 230000000840 anti-viral effect Effects 0.000 description 1
- 239000003146 anticoagulant agent Substances 0.000 description 1
- 229940127219 anticoagulant drug Drugs 0.000 description 1
- 230000030741 antigen processing and presentation Effects 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 239000003443 antiviral agent Substances 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 239000012298 atmosphere Substances 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 201000000448 autoimmune hemolytic anemia Diseases 0.000 description 1
- 201000003710 autoimmune thrombocytopenic purpura Diseases 0.000 description 1
- 230000005784 autoimmunity Effects 0.000 description 1
- LMEKQMALGUDUQG-UHFFFAOYSA-N azathioprine Chemical compound CN1C=NC([N+]([O-])=O)=C1SC1=NC=NC2=C1NC=N2 LMEKQMALGUDUQG-UHFFFAOYSA-N 0.000 description 1
- 229960002170 azathioprine Drugs 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 229960005520 bryostatin Drugs 0.000 description 1
- MJQUEDHRCUIRLF-TVIXENOKSA-N bryostatin 1 Chemical compound C([C@@H]1CC(/[C@@H]([C@@](C(C)(C)/C=C/2)(O)O1)OC(=O)/C=C/C=C/CCC)=C\C(=O)OC)[C@H]([C@@H](C)O)OC(=O)C[C@H](O)C[C@@H](O1)C[C@H](OC(C)=O)C(C)(C)[C@]1(O)C[C@@H]1C\C(=C\C(=O)OC)C[C@H]\2O1 MJQUEDHRCUIRLF-TVIXENOKSA-N 0.000 description 1
- MUIWQCKLQMOUAT-AKUNNTHJSA-N bryostatin 20 Natural products COC(=O)C=C1C[C@@]2(C)C[C@]3(O)O[C@](C)(C[C@@H](O)CC(=O)O[C@](C)(C[C@@]4(C)O[C@](O)(CC5=CC(=O)O[C@]45C)C(C)(C)C=C[C@@](C)(C1)O2)[C@@H](C)O)C[C@H](OC(=O)C(C)(C)C)C3(C)C MUIWQCKLQMOUAT-AKUNNTHJSA-N 0.000 description 1
- 239000007975 buffered saline Substances 0.000 description 1
- 239000003710 calcium ionophore Substances 0.000 description 1
- 244000309466 calf Species 0.000 description 1
- 230000005907 cancer growth Effects 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 230000021164 cell adhesion Effects 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 230000006037 cell lysis Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000036755 cellular response Effects 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 238000002512 chemotherapy Methods 0.000 description 1
- 239000012829 chemotherapy agent Substances 0.000 description 1
- 208000025302 chronic primary adrenal insufficiency Diseases 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 229920001436 collagen Polymers 0.000 description 1
- 239000000084 colloidal system Substances 0.000 description 1
- 239000000306 component Substances 0.000 description 1
- 239000000599 controlled substance Substances 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 229920001577 copolymer Polymers 0.000 description 1
- 230000000139 costimulatory effect Effects 0.000 description 1
- 108091008034 costimulatory receptors Proteins 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 239000003431 cross linking reagent Substances 0.000 description 1
- 229960004397 cyclophosphamide Drugs 0.000 description 1
- 230000001461 cytolytic effect Effects 0.000 description 1
- 230000003436 cytoskeletal effect Effects 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 201000001981 dermatomyositis Diseases 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 229940126534 drug product Drugs 0.000 description 1
- 210000003162 effector t lymphocyte Anatomy 0.000 description 1
- 239000000147 enterotoxin Substances 0.000 description 1
- 231100000655 enterotoxin Toxicity 0.000 description 1
- CJAONIOAQZUHPN-KKLWWLSJSA-N ethyl 12-[[2-[(2r,3r)-3-[2-[(12-ethoxy-12-oxododecyl)-methylamino]-2-oxoethoxy]butan-2-yl]oxyacetyl]-methylamino]dodecanoate Chemical compound CCOC(=O)CCCCCCCCCCCN(C)C(=O)CO[C@H](C)[C@@H](C)OCC(=O)N(C)CCCCCCCCCCCC(=O)OCC CJAONIOAQZUHPN-KKLWWLSJSA-N 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 238000002710 external beam radiation therapy Methods 0.000 description 1
- 208000037957 feline spongiform encephalopathy Diseases 0.000 description 1
- 210000003754 fetus Anatomy 0.000 description 1
- 239000000835 fiber Substances 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 229960000390 fludarabine Drugs 0.000 description 1
- GIUYCYHIANZCFB-FJFJXFQQSA-N fludarabine phosphate Chemical compound C1=NC=2C(N)=NC(F)=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@@H]1O GIUYCYHIANZCFB-FJFJXFQQSA-N 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 1
- 230000037406 food intake Effects 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 239000012595 freezing medium Substances 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 238000001415 gene therapy Methods 0.000 description 1
- 102000054766 genetic haplotypes Human genes 0.000 description 1
- 229960002518 gentamicin Drugs 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 1
- 210000004837 gut-associated lymphoid tissue Anatomy 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 239000012510 hollow fiber Substances 0.000 description 1
- 230000028996 humoral immune response Effects 0.000 description 1
- 210000004408 hybridoma Anatomy 0.000 description 1
- 208000003532 hypothyroidism Diseases 0.000 description 1
- 230000002989 hypothyroidism Effects 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 238000003018 immunoassay Methods 0.000 description 1
- 230000007813 immunodeficiency Effects 0.000 description 1
- 230000016784 immunoglobulin production Effects 0.000 description 1
- 230000001506 immunosuppresive effect Effects 0.000 description 1
- 239000003018 immunosuppressive agent Substances 0.000 description 1
- 229940125721 immunosuppressive agent Drugs 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 208000027866 inflammatory disease Diseases 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 230000002147 killing effect Effects 0.000 description 1
- 206010023497 kuru Diseases 0.000 description 1
- 239000002523 lectin Substances 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- XIXADJRWDQXREU-UHFFFAOYSA-M lithium acetate Chemical compound [Li+].CC([O-])=O XIXADJRWDQXREU-UHFFFAOYSA-M 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 210000002751 lymph Anatomy 0.000 description 1
- 210000005210 lymphoid organ Anatomy 0.000 description 1
- 230000002934 lysing effect Effects 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 229920002521 macromolecule Polymers 0.000 description 1
- 239000006148 magnetic separator Substances 0.000 description 1
- 238000002826 magnetic-activated cell sorting Methods 0.000 description 1
- 230000031852 maintenance of location in cell Effects 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 150000002739 metals Chemical class 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- MYWUZJCMWCOHBA-VIFPVBQESA-N methamphetamine Chemical compound CN[C@@H](C)CC1=CC=CC=C1 MYWUZJCMWCOHBA-VIFPVBQESA-N 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 230000004660 morphological change Effects 0.000 description 1
- 210000004877 mucosa Anatomy 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- 208000029766 myalgic encephalomeyelitis/chronic fatigue syndrome Diseases 0.000 description 1
- 206010028417 myasthenia gravis Diseases 0.000 description 1
- 229940014456 mycophenolate Drugs 0.000 description 1
- 229960000951 mycophenolic acid Drugs 0.000 description 1
- 229940053128 nerve growth factor Drugs 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 208000015122 neurodegenerative disease Diseases 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 238000010899 nucleation Methods 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 201000003045 paroxysmal nocturnal hemoglobinuria Diseases 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 229940021222 peritoneal dialysis isotonic solution Drugs 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 239000008363 phosphate buffer Substances 0.000 description 1
- 238000007747 plating Methods 0.000 description 1
- 229920000729 poly(L-lysine) polymer Polymers 0.000 description 1
- 208000005987 polymyositis Diseases 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 210000004986 primary T-cell Anatomy 0.000 description 1
- 150000003141 primary amines Chemical group 0.000 description 1
- 210000001948 pro-b lymphocyte Anatomy 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 230000008521 reorganization Effects 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 201000003068 rheumatic fever Diseases 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 229960004641 rituximab Drugs 0.000 description 1
- 239000011435 rock Substances 0.000 description 1
- 229960003452 romidepsin Drugs 0.000 description 1
- OHRURASPPZQGQM-GCCNXGTGSA-N romidepsin Chemical compound O1C(=O)[C@H](C(C)C)NC(=O)C(=C/C)/NC(=O)[C@H]2CSSCC\C=C\[C@@H]1CC(=O)N[C@H](C(C)C)C(=O)N2 OHRURASPPZQGQM-GCCNXGTGSA-N 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 208000008864 scrapie Diseases 0.000 description 1
- 239000012679 serum free medium Substances 0.000 description 1
- 239000004017 serum-free culture medium Substances 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 238000000527 sonication Methods 0.000 description 1
- 150000003431 steroids Chemical class 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 208000011580 syndromic disease Diseases 0.000 description 1
- 229920002994 synthetic fiber Polymers 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 1
- 238000011287 therapeutic dose Methods 0.000 description 1
- 125000002088 tosyl group Chemical group [H]C1=C([H])C(=C([H])C([H])=C1C([H])([H])[H])S(*)(=O)=O 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- 238000011277 treatment modality Methods 0.000 description 1
- 238000009281 ultraviolet germicidal irradiation Methods 0.000 description 1
- 241000712461 unidentified influenza virus Species 0.000 description 1
- 238000002255 vaccination Methods 0.000 description 1
- 208000007089 vaccinia Diseases 0.000 description 1
- 239000012808 vapor phase Substances 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 150000003722 vitamin derivatives Chemical class 0.000 description 1
- 210000005253 yeast cell Anatomy 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4611—T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/464838—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/16—Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/18—Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/12—Drugs for disorders of the urinary system of the kidneys
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P15/00—Drugs for genital or sexual disorders; Contraceptives
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/04—Antibacterial agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/04—Antibacterial agents
- A61P31/06—Antibacterial agents for tuberculosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/10—Antimycotics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P33/00—Antiparasitic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/04—Immunostimulants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K2035/122—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells for inducing tolerance or supression of immune responses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K2035/124—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/515—Animal cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/50—Cell markers; Cell surface determinants
- C12N2501/515—CD3, T-cell receptor complex
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/50—Cell markers; Cell surface determinants
- C12N2501/58—Adhesion molecules, e.g. ICAM, VCAM, CD18 (ligand), CD11 (ligand), CD49 (ligand)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/50—Cell markers; Cell surface determinants
- C12N2501/599—Cell markers; Cell surface determinants with CD designations not provided for elsewhere
Definitions
- the present invention relates generally to methods for generating, isolating, and expanding antigen-specific T cells.
- the present invention also relates to compositions of antigen-specific T cells.
- APC antigen presenting cells
- IL-2 interleukin-2
- IL-2 has been used together with an anti-CD3 antibody to stimulate T-cell proliferation, predominantly expanding the CD8 + subpopulation of T-cells.
- APC signals are thought to be required for optimal T-cell activation, expansion, and long-term survival of the T-cells upon re-infusion.
- MHC-matched APCs as accessory cells presents a significant problem for long-term culture systems because APCs are relatively short-lived.
- APCs must be continually obtained from a source and replenished.
- the necessity for a renewable supply of accessory cells is problematic for treatment of immunodeficiencies in which accessory cells are affected.
- the cells may contaminate the entire T-cell population during long-term culture.
- antigen e.g. tumor/viral antigen
- pulsing of antigen-presenting cells with antigens followed by infusion of cells.
- Expansion of antigen-specific T cells to generate large numbers of antigen-specific T cells often requires labor intensive and expensive cloning, and/or multiple rounds of activation/expansion to achieve therapeutically relevant T cell numbers.
- the present invention provides methods to generate an increased number of highly responsive antigen-specific T cells that have surface receptor and cytokine production characteristics that are more more desirable than other expansion methods.
- the instant invention does not require knowledge of a particular antigen (although known antigens can be used in the context of this invention) and provides for a single, or double, round of expansion to achieve a therapeutically relevant dose of antigen-specific T cells, both of the CD4 and CD8 lineage (and either may be selected if desired).
- the present invention relates to methods for activating, stimulating and isolating antigen-specific T cells.
- the present invention also relates to compositions of antigen-specific T cells and methods of their use in the treatment and prevention of cancer, infectious diseases, autoimmune diseases, immune disfunction related to aging, or any other disease state where antigen-specific T cells are desired for treatment.
- a method for expanding a population of antigen-specific T cells comprising contacting a population of cells wherein at least a portion thereof comprises antigen-specific T cells, with a surface, wherein said surface has attached thereto a first agent and a second agent, wherein said first agent ligates a CD3/TCR complex on said T cells and said second agent ligates an accessory molecule on said T cells, and wherein said ligation by said first and second agent of said T cells induces proliferation of antigen-specific T cells and wherein said surface is present in a ratio of surface to T cells of 1:2 or less. In certain embodiments the ratio of surface to T cells is between about 1:1 and about 1:50 and any ratio therebetween.
- the ratio of surface to T cells is from about 1:2, 1:2.5, 1:5, 1:10, 1:25, 1:50, 1:75, 1:100, or lower.
- the surface includes but is not limited to paramagnetic beads, lipids, and cell surfaces.
- the surface comprises paramagnetic beads conjugated to one or more antibodies.
- the surface can have 1, 2, 3, 4, or more antibodies or natural ligands conjugated thereto.
- Another aspect of the present invention provides a method for generating antigen-specific T cells comprising exposing a first population of cells wherein at least a portion thereof comprises antigen presenting cells (APC) to a surface wherein said surface has antigen attached thereto, such that said surface with antigen attached thereto is ingested by said APC; exposing a second population of cells wherein at least a portion thereof comprises T cells to the population of cells in part (a); thereby generating antigen-specific T cells.
- APC antigen presenting cells
- Antigen may be attached or coupled to, or integrated into a surface by a variety of methods known and available in the art and described herein.
- the antigen is crosslinked to said surface.
- the attachment to said surface is by covalent or noncovalent, electrostatic, or hydrophobic and may be accomplished by a variety of attachment means, including for example, chemical, mechanical, enzymatic, electrostatic, or other means whereby the antigen(s) is capable of stimulating the cells.
- the antibody to an antigen first may be attached to a surface, or avidin or streptavidin may be attached to the surface for binding to a biotinylated antigen.
- the antibody to the ligand may be attached to the surface via an anti-idiotype antibody.
- Another example includes using protein A or protein G, or other non-specific antibody binding molecules, attached to surfaces to bind an antibody.
- antigen may be attached to the surface by chemical means, such as cross-linking to the surface, using commercially available cross-linking reagents (Pierce, Rockford, Ill.) or other means.
- antigens are covalently bound to the surface.
- commercially available tosyl-activated DYNABEADSTM or DYNABEADSTM with epoxy-surface reactive groups are incubated with the polypeptide antigen of interest according to the manufacturer's instructions. Briefly, such conditions typically involve incubation in a phosphate buffer from pH 4 to pH 9.5 at temperatures ranging from 4 to 37 degrees C.
- the APC are in direct contact with the antigen-specific T cells.
- the APC that are in direct contact with said antigen-specific T cells are isolated by exposing said APC to a magnetic field, wherein said surface comprises a paramagnetic, magnetic, or magnetizable component.
- the antigen-specific T cells are expanded by exposing said T cells to a surface wherein said surface has attached thereto a first agent that ligates a first T cell surface moiety of a T cell, and the same or a second surface has attached thereto a second agent that ligates a second moiety of said T cell, wherein said ligation by the first and second agent induces proliferation (expansion) of said antigen-specific T cells.
- at least one agent is an antibody or an antibody fragment.
- the first agent is an antibody or a fragment thereof
- the second agent is an antibody or a fragment thereof.
- the first and the second agents are different antibodies.
- the first agent is an anti-CD3 antibody, an anti-CD2 antibody, or an antibody fragment of an anti-CD3 or anti-CD2 antibody and the second the second agent is an anti-CD28 antibody or antibody fragment thereof.
- the first agent is an anti-CD3 antibody and the second agent is an anti-CD28 antibody.
- the anti-CD3 antibody and the anti-CD28 antibody are present at a ratio of about 1:1 to about 1:100.
- the antigen-specific T cells are expanded by exposing said antigen-specific T cells to a mitogen, such as phytohemagglutinin (PHA), phorbol myristate acetate (PMA) and ionomycin, lipopolysaccharide (LPS), and superantigen.
- a mitogen such as phytohemagglutinin (PHA), phorbol myristate acetate (PMA) and ionomycin, lipopolysaccharide (LPS), and superantigen.
- the antigen of the present invention includes but is not limited to protein, glycoprotein, peptides, antibody/antigen complexes, whole tumor or virus-infected cells, fixed tumor or virus-infected cells, heat-killed tumor or virus-infected cells, tumor lysate, virus lysate, non-soluble cell debris, apoptotic bodies, necrotic cells, whole tumor cells from a tumor or a cell line that have been treated such that they are unable to continue dividing, allogeneic cells that have been treated such that they are unable to continue dividing, irradiated tumor cells, irradiated allogeneic cells, natural or synthetic complex carbohydrates, lipoproteins, lipopolysaccharides, transformed cells or cell line, transfected cells or cell line, transduced cells or cell line, and virally infected cells or cell line.
- antigen is attached to said surface by an antibody/ligand interaction.
- An antibody/ligand interaction includes but is not limited to an interaction between an antibody/ligand pair selected from the group consisting of anti-MART-1 antibody/MART-1 antigen, anti-WT-1 antibody/WT-1, anti-PR1 antibody /PR1, anti-PR3 antibody /PR3, anti-tyrosinase antibody/tyrosinase antigen, anti-MAGE-1 antibody/MAGE-1 antigen, anti-MUC-1 antibody/MUC-1 antigen, anti- ⁇ -fetoprotein antibody/ ⁇ -fetoprotein antigen, anti-Her2Neu antibody/Her2Neu, anti-HIV gp120 antibody/HIV gp120, anti-influenza HA antibody/influenza HA, anti-CMV pp65/CMV pp65, anti-hepatitis C antibody/hepatitis C proteins, anti-EBV EBNA 3B antibody/EBV EBNA 3B antigen, and anti-
- the antigen is chemically attached to a surface.
- the attachment of said antigen to said surface comprises a biotin-avidin interaction.
- the population of cells wherein at least a portion thereof comprises APC is derived from a source selected from the group consisting of a leukapheresis product, peripheral blood, lymph node, tonsil, thymus, tissue biopsy, tumor, spleen, bone marrow, cord blood, CD34 + cells, monocytes, and adherent cells.
- Another aspect of the present invention provides a method for generating and expanding antigen-specific T cells comprising exposing a first population of cells wherein at least a portion thereof comprises antigen presenting cells to antigen such that said antigen is taken up by said APC; exposing a second population of cells wherein at least a portion thereof comprises T cells to the population of cells in part (a); thereby generating antigen-specific T cells; and exposing said antigen-specific T cells of part (b) to a surface wherein said surface has attached thereto a first agent that ligates a first T cell surface moiety of a T cell, and the same or a second surface has attached thereto a second agent that ligates a second moiety of said T cell, wherein said ligation by the first and second agent induces proliferation (expansion) of said antigen-specific T cells.
- At least one agent is an antibody or an antibody fragment.
- the first agent is an antibody or a fragment thereof
- the second agent is an antibody or a fragment thereof.
- the first and the second agents are different antibodies.
- the first agent is an anti-CD3 antibody, an anti-CD2 antibody, or an antibody fragment of an anti-CD3 or anti-CD2 antibody and the second the second agent is an anti-CD28 antibody or antibody fragment thereof.
- the first agent is an anti-CD3 antibody and the second agent is an anti-CD28 antibody.
- the anti-CD3 antibody and the anti-CD28 antibody are present at a ratio of about 1:1 to about 1:100.
- said antigen-specific T cells are isolated by contacting said T cells with antibodies specific for T cell activation markers.
- said antibodies are selected from the group consisting of anti-CD25, anti-CD54, anti-CD69, anti-CD38, anti-CD45RO, anti-CD49d, anti-CD40L, anti-CD137, anti-CD62L, and anti-CD134.
- a further aspect of the present invention provides a population of antigen-specific T cells generated according to any one of the methods described herein.
- An additional aspect of this invention is a composition comprising the antigen-specific T cells according to any of the methods described herein and a pharmaceutically acceptable excipient.
- a further aspect of the present invention provides methods for stimulating an immune response in a mammal comprising, administering to the mammal compositions comprising the antigen-specific T cells of the present invention.
- An additional aspect of the invention provides for reducing the presence of cancer cells in a mammal comprising, exposing the cancer cells to the compositions comprising antigen-specific T cells.
- the cancer cells are from a cancer selected from the group consisting of melanoma, non-Hodgkin's lymphoma, Hodgkin's disease, leukemia, plasmocytoma, sarcoma, glioma, thymoma, breast cancer, prostate cancer, colo-rectal cancer, kidney cancer, renal cell carcinoma, pancreatic cancer, esophageal cancer, brain cancer, lung cancer, ovarian cancer, cervical cancer, multiple myeloma, hepatoma, acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), and chronic lymphocytic leukemia (CLL).
- ALL acute lymphoblastic leukemia
- AML acute myelogenous leukemia
- One aspect of the present invention provides a method for inhibiting the development of a cancer in a mammal, comprising administering to the mammal the composition comprising antigen-specific T cells fo the present invention.
- the cancer cells are from a cancer selected from the group consisting of melanoma, non-Hodgkin's lymphoma, Hodgkin's disease, leukemia, plasmocytoma, sarcoma, glioma, thymoma, breast cancer, prostate cancer, colo-rectal cancer, kidney cancer, renal cell carcinoma, pancreatic cancer, esophageal cancer, brain cancer, lung cancer, ovarian cancer, cervical cancer, multiple myeloma, hepatoma, acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), and chronic lymphocytic leukemia (CLL).
- ALL acute lymphoblastic leukemia
- AML acute my
- a further aspect of the present invention provides a method for ameliorating an immune response dysfunction in a mammal comprising administering to the mammal the compositions comprising antigen-specific T cells generated using any one of the methods described herein.
- an infectious organism can include but is not limited to a virus, a single-stranded RNA virus, a single-stranded DNA virus, a double-stranded DNA virus, Human Immunodeficiency Virus (HIV), Hepatitis A, B, or C virus, Herpes Simplex Virus (HSV), Human Papilloma Virus (HPV), Cytomegalovirus (CMV), Epstein-Barr virus (EBV), a parasite, a bacterium, M. tuberculosis, Pneumocystis carinii, Candida, Aspergillus.
- HIV Human Immunodeficiency Virus
- HSV Herpes Simplex Virus
- HPV Human Papilloma Virus
- CMV Cytomegalovirus
- EBV Epstein-Barr virus
- An additional aspect of the present invention provides a method for inhibiting the development of an infectious disease in a mammal, comprising administering to the mammal the compositions comprising antigen-specific T cells generated using any one of the methods described herein.
- an infectious disease can be caused by an infectious organism including but not limited to a virus, an RNA virus, a DNA virus, Human Immunodeficiency Virus (HIV), Hepatitis A, B, or C virus, Herpes Simplex Virus (HSV), Human Papilloma Virus (HPV), Cytomegalovirus (CMV), Epstein-Barr virus (EBV), a parasite, a bacterium, M. tuberculosis, Pneumocystis carinii, Candida, Aspergillus.
- HIV Human Immunodeficiency Virus
- HPV Human Papilloma Virus
- CMV Cytomegalovirus
- EBV Epstein-Barr virus
- FIG. 1 is a photograph showing the tight association of antigen-specific T cells and bead-loaded antigen presenting cells (APC) post magnetic separation.
- FIG. 2 is a plot showing upregulation of CD25 in re-stimulated memory CD8 CMV tetramer+ T cells expanded ex vivo.
- Panel A is a negative control from an HLA-A2+, CMV ⁇ donor.
- Panel B is a negative control showing uncoated bead stimulation from an HLA-A2+, CMV+ donor.
- Panel C shows CMV antigen-coated bead stimulation of cells from an HLA-A2+, CMV+ donor.
- FIG. 3 is a plot showing the effect of varying bead:cell ratio on expansion or deletion of CMV-specific T cells.
- FIG. 4 panels A and B is a bar graph showing the effect on T cell expansion of sequential bead addition at varying bead:cell ratios at varying times during cutlure.
- Panel A shows a comparison of total T cell expansion over 15 days, comparing standard static cuture (beads at day 0 at either 1:2.5 or 1:5 bead to cell ratio) or additional beads added at day 5, 7, or 9 at 1:10, 1:25, 1:50 or 1:100 bead to cell ratios.
- Panel B shows CMV-specific T cell expansion under the same experimental conditions as Panel A.
- FIG. 5 is a graph showing the effect on T cell expansion of low bead:T cell ratio and sequential addition of beads on Melanoma gp100(M)-specific T cells.
- FIG. 6 is a graph depicting the fold increase of T-cells over time following stimulation with anti-CD3 and anti-CD28 co-immobilized beads with varying ratios of anti-CD3:CD28 antibodies attached thereto.
- FIG. 7 is a graph depicting the fold increase of CMVpp65-specific T-cells over time following stimulation with anti-CD3 and anti-CD28 co-immobilized beads with varying ratios of anti-CD3:CD28 antibodies attached thereto.
- biocompatible refers to the property of being predominantly non-toxic to living cells.
- stimulation refers to a primary response induced by ligation of a cell surface moiety.
- such stimulation entails the ligation of a receptor and a subsequent signal transduction event.
- stimulation of a T-cell refers to the ligation of a T-cell surface moiety that in one embodiment subsequently induces a signal transduction event, such as binding the TCR/CD3 complex.
- the stimulation event may activate a cell and upregulate or downregulate expression or secretion of a molecule, such as downregulation of TGF- ⁇ .
- ligation of cell surface moieties may result in the reorganization of cytoskeletal structures, or in the coalescing of cell surface moieties, each of which could serve to enhance, modify, or alter subsequent cell responses.
- activation refers to the state of a cell following sufficient cell surface moiety ligation to induce a noticeable biochemical or morphological change.
- T-cells such activation, refers to the state of a T-cell that has been sufficiently stimulated to induce cellular proliferation.
- Activation of a T-cell may also induce cytokine production and performance of regulatory or cytolytic effector functions. Within the context of other cells, this term infers either up or down regulation of a particular physico-chemical process.
- target cell refers to any cell that is intended to be stimulated by cell surface moiety ligation.
- an “antibody”, as used herein, includes both polyclonal and monoclonal antibodies; primatized (e.g., humanized); murine; mouse-human; mouse-primate; and chimeric; and may be an intact molecule, a fragment thereof (such as scFv, Fv, Fd, Fab, Fab′ and F(ab)′ 2 fragments), or multimers or aggregates of intact molecules and/or fragments; and may occur in nature or be produced, e.g., by immunization, synthesis or genetic engineering; an “antibody fragment,” as used herein, refers to fragments, derived from or related to an antibody, which bind antigen and which in some embodiments may be derivatized to exhibit structural features that facilitate clearance and uptake, e.g., by the incorporation of galactose residues. This includes, e.g., F(ab), F(ab)′ 2 , scFv, light chain variable region (V L ), heavy chain
- protein includes proteins, polypeptides and peptides; and may be an intact molecule, a fragment thereof, or multimers or aggregates of intact molecules and/or fragments; and may occur in nature or be produced, e.g., by synthesis (including chemical and/or enzymatic) or genetic engineering.
- agent refers to a molecule that binds to a defined population of cells.
- the agent may bind any cell surface moiety, such as a receptor, an antigenic determinant, or other binding site present on the target cell population.
- the agent may be a protein, peptide, antibody and antibody fragments thereof, fusion proteins, synthetic molecule, an organic molecule (e.g., a small molecule), or the like.
- antibodies are used as a prototypical example of such an agent.
- agent that binds a cell surface moiety and “cell surface moiety”, as used herein, are used in the context of a ligand/anti-ligand pair. Accordingly, these molecules should be viewed as a complementary/anti-complementary set of molecules that demonstrate specific binding, generally of relatively high affinity (an affinity constant, K a , of about 10 6 M ⁇ 1 or tighter).
- Antigen-presenting cell refers to those cells that normally initiate the responses of na ⁇ ve and/or memory T cells to antigen.
- APC refers to any cell capable of antigen presentation.
- APCs include, but are not limited to, dendritic cells, monocytes, macrophages, and B cells.
- An APC may express high levels of MHC class II, ICAM-1 and B7-2.
- a “co-stimulatory signal”, as used herein, refers to a signal, which in combination with a primary signal, such as TCR/CD3 ligation, leads to T-cell proliferation.
- a “ligand/anti-ligand pair”, as used herein, refers to a complementary/anti-complementary set of molecules that demonstrate specific binding, generally of relatively high affinity (an affinity constant, K a , of at least about 10 6 M ⁇ 1 ,).
- affinity constants of the ligand/anti-ligand pairs useful in the context of the present invention might be lower or in some cases higher.
- the streptavidin on-rate is comparable to that of monomeric avidin while its off-rate is seven times lower.
- the dissociation constant was determined to be 1.3 ⁇ 10( ⁇ 8)M.
- ligand/anti-ligand pairs enzyme/inhibitor hapten/antibody, lectin/carbohydrate, ligand/receptor, and biotin/avidin or streptavidin.
- receptors and other cell surface moieties are anti-ligands, while agents (e.g., antibodies and antibody fragments) reactive therewith are considered ligands.
- Separatation includes any means of substantially purifying one component from another (e.g., by filtration, magnetic attraction, etc.).
- “Quiescent”, as used herein, refers to a cell state wherein the cell is not actively proliferating.
- a “surface”, as used herein, refers to any surface capable of having an agent attached thereto and includes, without limitation, metals, glass, plastics, co-polymers, colloids, lipids, cell surfaces, and the like. Essentially any surface that is capable of retaining an agent bound or attached thereto.
- a prototypical example of a surface used herein, is a particle such as a bead. As such, the terms “surface” and “particle” are used herein interchangeably.
- Immuno response or responsiveness refers to activation of cells of the immune system, including but not limited to, T-cells, such that a particular effector function(s) of a particular cell is induced. Effector functions may include, but are not limited to, proliferation, secretion of cytokines, secretion of antibodies, expression of regulatory and/or adhesion molecules, and the ability to induce cytolysis.
- Stimulating an immune response refers to any stimulation such that activation and induction of effector functions of cells of the immune system are achieved.
- Immuno response dysfunction refers to the inappropriate activation and/or proliferation, or lack thereof, of cells of the immune system, and/or the inappropriate secretion, or lack thereof, of cytokines, and/or the inappropriate or inadequate induction of other effector functions of cells of the immune system, such as expression of regulatory, adhesion, and/or homing receptors, and the induction of cytolysis.
- preventing or “inhibiting” the development of a cancer or cancer cells” as used herein, refers to the occurrence of the cancer being prevented or the onset of the cancer being delayed.
- treating means that the cancer growth is inhibited, which is reflected by, e.g., tumor volume or numbers of malignant cells.
- Tumor volume may be determined by various known procedures, e.g., obtaining two dimensional measurements with a dial caliper.
- Preventing or inhibiting the development of an infectious disease means the occurrence of the infectious disease is prevented or the onset of the infectious disease is delayed, or the spread of an existing infection is reversed.
- “Ameliorate” as used herein, is defined as: to make better; improve (The American Heritage College Dictionary, 3 rd Edition, Houghton Mifflin Company, 2000).
- Particles may include a colloidal particle, a microsphere, nanoparticle, a bead, or the like.
- commercially available surfaces such as beads or other particles, are useful (e.g., Miltenyi Particles, Miltenyi Biotec, Germany; Sepharose beads, Pharmacia Fine Chemicals, Sweden; DYNABEADSTM, Dynal Inc., Oslo, Norway; PURABEADSTM, Prometic Biosciences, magnetic beads from Immunicon, Huntingdon Valley, Pa., microspheres from Bangs Laboratories, Inc., Fishers, Ind.).
- Paraamagnetic particles refer to particles, as defined above, that localize in response to a magnetic field.
- Antigen refers to any molecule 1) capable of being specifically recognized, either in its entirety or fragments thereof, and bound by the “idotypic” portion (antigen-binding region) of a mAb or its derviative; 2) containing peptide sequences which can be bound by MHC and then, in the context of MHC presentation, can specifically engage its cognate T cell antigen receptor.
- To “load” an APC with antigen refers to exposing an APC to antigen or antigenic peptide for a period of time sufficient for the APC to uptake, process, and present the antigen, bound by MHC molecules, to T cells.
- the antigen, especially peptide can be bound by MHC molecules and presented to T cells without being taken up and processed by the APC.
- animal or “mammal” as used herein, encompasses all mammals, including humans.
- the animal of the present invention is a human subject.
- exposing refers to bringing into the state or condition of immediate proximity or direct contact.
- lysate refers to the supernatant and non-soluble cell debris resulting from lysis of cells.
- lysis buffers known in the art may be used (see for example Current Protocols in Immunology, John Wiley & Sons, New York. N.Y.). Cell lysis may also be carried out by freeze-thaw procedures or other means (e.g. sonication, etc.).
- apoptotic body as used herein, is defined as the smaller, intact, membrane-bound fragments that result from apoptotic cells.
- proliferation means to grow or multiply by producing new cells.
- infectious disease refers to any disease that is caused by an infectious organism.
- Infectious organisms may comprise viruses, (e.g., RNA viruses, DNA viruses, human immunodeficiency virus (HIV), hepatitis A, B, and C virus, herpes simplex virus (HSV), cytomegalovirus (CMV) Epstein-Barr virus (EBV), human papilloma virus (HPV)), parasites (e.g., protozoan and metazoan pathogens such as Plasmodia species, Leishmania species, Schistosoma species, Trypanosoma species), bacteria (e.g., Mycobacteria, in particular, M.
- viruses e.g., RNA viruses, DNA viruses, human immunodeficiency virus (HIV), hepatitis A, B, and C virus, herpes simplex virus (HSV), cytomegalovirus (CMV) Epstein-Barr virus (EBV), human papilloma virus (HPV)
- fungi e.g., Candida species, Aspergillus species
- Pneumocystis carinii fungi
- prions known prions infect animals to cause scrapie, a transmissible, degenerative disease of the nervous system of sheep and goats, as well as bovine spongiform encephalopathy (BSE), or “mad cow disease”, and feline spongiform encephalopathy of cats.
- BSE bovine spongiform encephalopathy
- prion diseases known to affect humans are (1) kuru, (2) Creutzfeldt-Jakob Disease (CJD), (3) Gerstmann-Straussler-Scheinker Disease (GSS), and (4) fatal familial insomnia (FFI)).
- CJD Creutzfeldt-Jakob Disease
- GSS Gerstmann-Straussler-Scheinker Disease
- FFI fatal familial insomnia
- T cells can be obtained from a number of sources, including peripheral blood mononuclear cells, bone marrow, thymus, tissue biopsy, tumor, lymph node tissue, gut associated lymphoid tissue, mucosa associated lymphoid tissue, spleen tissue, or any other lymphoid tissue, and tumors.
- T cells can be obtained from T cell lines and from autologous or allogeneic sources.
- T cells may also be obtained from a xenogeneic source, for example, from mouse, rat, non-human primate, and pig.
- cells from the circulating blood of an individual are obtained by apheresis or leukapheresis.
- the apheresis product typically contains lymphocytes, including T cells, monocytes, granulocytes, B cells, other nucleated white blood cells, red blood cells, and platelets.
- the cells collected by apheresis or leukapheresis may be washed to remove the plasma fraction and to place the cells in an appropriate buffer or media for subsequent processing steps.
- the cells are washed with phosphate buffered saline (PBS).
- the wash solution lacks calcium and may lack magnesium or may lack many if not all divalent cations.
- a washing step may be accomplished by methods known to those in the art, such as by using a semi-automated “flow-through” centrifuge (for example, the Cobe 2991 cell processor, Baxter) according to the manufacturer's instructions.
- a semi-automated “flow-through” centrifuge for example, the Cobe 2991 cell processor, Baxter
- the cells may be resuspended in a variety of biocompatible buffers, such as, for example, Ca ++ /Mg ++ free PBS.
- the undesirable components of the apheresis sample may be removed and the cells directly resuspended in culture media.
- T cells are isolated from peripheral blood lymphocytes by lysing the red blood cells and by centrifugation through a PERCOLLTM gradient.
- a specific subpopulation of T cells such as CD28 + , CD4 + , CD8 + , CD45RA + , and CD45RO + T cells, can be further isolated by positive or negative selection techniques.
- CD3 + , CD28 + T cells can be positively selected using CD3/CD28 conjugated magnetic beads (e.g., DYNABEADS® M-450 CD3/CD28 T Cell Expander).
- enrichment of a T cell population by negative selection can be accomplished with a combination of antibodies directed to surface markers unique to the negatively selected cells.
- a preferred method is cell sorting and/or selection via negative magnetic immunoadherence or flow cytometry that uses a cocktail of monoclonal antibodies directed to cell surface markers present on the cells negatively selected.
- a monoclonal antibody cocktail typically includes antibodies to CD14, CD20, CD11b, CD16, HLA-DR, and CD8.
- Another method for preparing T cells for stimulation is to freeze the cells after the washing step, which does not require the monocyte-removal step.
- the freeze and subsequent thaw step provides a more uniform product by removing granulocytes and, to some extent, monocytes in the cell population.
- the cells may be suspended in a freezing solution. While many freezing solutions and parameters are known in the art and will be useful in this context, one method involves using PBS containing 20% DMSO and 8% human serum albumin (HSA), or other suitable cell freezing media. This is then diluted 1:1 with media so that the final concentration of DMSO and HSA are 10% and 4%, respectively. The cells are then frozen to ⁇ 80° C. at a rate of 1° per minute and stored in the vapor phase of a liquid nitrogen storage tank.
- HSA human serum albumin
- the source of antigen-presenting cell is typically a tissue source comprising APC or APC precursors that are capable of proliferating and maturing in vitro into professional APC (pAPC) when loaded with antigen and/or treated with the necessary cytokines or factors.
- “Professional APC” (pAPC) or “antigen-presenting cell” (APC), as used herein, refers to those cells that normally initiate the responses of na ⁇ ve and/or memory T cells to antigen.
- Professional APCs include, but are not limited to, DC, macrophages, and B cells.
- pAPC may express high levels of MHC class II, ICAM-1 and B7-2.
- APC precursor cells are capable of proliferating and maturing in vitro into dendritic cells (DC). While many tissue sources may be used, typical tissue sources comprise spleen, thymus, tissue biopsy, tumor, afferent lymph, lymph nodes, bone marrow, apheresis or leukapheresis product, and/or peripheral blood. In certain embodiments, apheresis product, bone marrow and peripheral blood are preferred sources. Fetal tissue, fetal or umbilical cord blood, which is also rich in growth factors may also be used as a source of blood for obtaining APC and/or precursor APC. Exemplary precursor cells may be, but are not limited to, embryonic stem cells, CD34 + cells, monocyte progenitors, monocytes, and pre-B cells.
- APC may be derived from precursor cells comprising monocytes or CD34 + cells.
- the source of APC and/or precursor APC is an apheresis or leukapheresis product.
- Cells are collected using apheresis procedures known in the art. See, for example, Bishop et al., Blood, vol. 83, No. 2, pp. 610-616 (1994). Briefly, cells are collected using conventional devices, for example, a Haemonetics Model V50 apheresis device (Haemonetics, Braintree, Mass.).
- Apheresis product typically contains lymphocytes, including T cells, monocytes, granulocytes, B cells, other nucleated white blood cells, red blood cells, and platelets.
- the cells collected by apheresis may be washed to remove the plasma fraction and to place the cells in an appropriate buffer or media for subsequent processing steps.
- the cells are washed with phosphate buffered saline (PBS).
- PBS phosphate buffered saline
- the wash solution lacks calcium and may lack magnesium or may lack many if not all divalent cations.
- a washing step may be accomplished by methods known to those in the art, such as by using a semi-automated “flow-through” centrifuge (for example, the Cobe 2991 cell processor, Gambro BCT, Lakewood, Colo.) according to the manufacturer's instructions.
- the cells may be resuspended in a variety of biocompatible buffers, such as, for example, Ca-free, Mg-free PBS.
- a variety of biocompatible buffers such as, for example, Ca-free, Mg-free PBS.
- the undesirable components of the apheresis sample may be removed and the cells directly resuspended in culture media.
- blood leukocytes may be obtained using conventional methods that maintain their viability.
- blood is diluted into medium (preferably RPMI) that may or may not contain heparin (about 100 U/ml) or other suitable anticoagulant.
- the volume of blood to medium is about 1 to 1.
- Cells are concentrated by centrifugation of the blood in medium at about 1000 rpm (150 g) at 4° C. Platelets and red blood cells are depleted by resuspending the cells in any number of solutions known in the art that will lyse erythrocytes, for example ammonium chloride.
- the mixture may be medium and ammonium chloride (at a final concentration of about 0.839 percent) at about 1:1 by volume.
- Cells may be concentrated by centrifugation and washed in the desired solution until a population of leukocytes, substantially free of platelets and red blood cells, is obtained, typically about two times.
- Any isotonic solution commonly used in tissue culture may be used as the medium for separating blood leukocytes from platelets and red blood cells. Examples of such isotonic solutions are phosphate buffered saline, Hanks balanced salt solution, or complete growth media including for example RPMI 1640, DMEM, MEM, HAMS F-12, X-Vivo 15, or X-Vivo 20.
- APC and/or APC precursor cells may also purified by elutriation, using, for example, a Beckman J6ME centrifuge equipped with a J5.0 rotor and a 40 ml elutriation chamber.
- isolation of APC and/or precursor APC is performed by preincubating ficolled whole blood or apheresed peripheral blood with one or more varieties of irrelevant or non-antibody coupled paramagnetic particles (approx. 1 vial of beads or 4 ⁇ 10 9 beads to one batch of cells (typically from about 5 ⁇ 10 8 to about 2 ⁇ 10 10 cells) for about 30 minutes to 2 hours at 22 to 37 degrees C., followed by magnetic removal of cells which have attached to or engulfed the paramagnetic particles.
- Such separation can be performed using standard methods available in the art.
- any magnetic separation methodology may be used including a variety of which are commercially available, (e.g., DYNAL® Magnetic Particle Concentrator (DYNAL MPC®)).
- DYNAL® Magnetic Particle Concentrator DYNAL MPC®
- Assurance of isolation can be monitored by a variety of methodologies known to those of ordinary skill in the art, including flow cytometric analysis of cells before and after said isolation.
- APC obtained from treatment of the tissue source may be cultured to form a primary culture in an appropriate culture container or vessel in an appropriate culture medium.
- the culture medium is supplemented with one or more cytokines.
- the appropriate culture container or vessel may be any container with tissue culture compatible surface. Examples include various bags (e.g., Lifecell culture bags), flasks, roller bottles, petri dishes and multi-well containing plates made for use in tissue culture. Surfaces treated with a substance, for example collagen or poly-L-lysine, or antibodies specific for a particular cell type to promote cell adhesion may also be used provided they allow for the differential attachment of cells as described below. Surfaces may be also be chemically treated, for example by ionization. Cells are plated at an initial cell density from about 10 5 to 10 7 cells/cm 2 . In one aspect, cells are plated at 10 6 cell s/cm 2 .
- the primary cultures from the selected tissue source are allowed to incubate at about 37° C. under standard tissue culture conditions of humidity, CO 2 , and pH until a population of cells has adhered to the substrate sufficiently to allow for the separation of nonadherent cells.
- Some immature APC in blood initially are nonadherent to plastic, particularly immature DC, in contrast to monocytes, so that the precursors can be separated after overnight culture.
- Monocytes and fibroblasts are believed to comprise the majority of adherent cells and usually adhere to the substrate within about 30 minutes to about 24 hours.
- nonadherent cells are separated from adherent cells between about 1 to 16 hours. Nonadherent cells may be separated at about 1 to 2 hours. Any method which does not dislodge significant quantities of adherent cells may be used to separate the adherent from nonadherent cells.
- the cells are dislodged by simple shaking or pipetting. Pipetting is most preferred.
- Adherent cells comprising precursor APC (e.g., monocytes) isolated according to the methods of the invention are allowed to incubate at about 37° C. under standard tissue culture conditions of humidity, CO 2 , and pH until a population of cells has reached an immature APC stage.
- adherent cells are allowed to incubate for a period of between 4 hours and 7 days.
- incubation times and conditions may vary.
- Immature APC refers to an intermediate differentiation state of an APC wherein the APC has the capacity to endocytose or phagocytose antigen, foreign bodies, necrotic and/or apoptosing tissue and/or cells.
- Immature APC may be CD14 ⁇ or CD14 + depending on the origin of the precursor cells.
- Immature APC may also express CD1a, CD40, CD86, CD54, and intermediate levels of MHC class II (levels of marker expression on sample cells can be compared by flow cytometric analysis to levels of expression on MHC class II-negative cells and cells known to express high levels of MHC class II).
- Immature APC typically do not express CCR7.
- PBMC comprising APC and T cells can be exposed to antigen as described herein and the resulting antigen-specific T cells further expanded as described herein.
- the APCs or the T cells described herein be derived from an autologous source.
- the APC and T cells can be obtained from a matched or unmatched donor, or from a cell line, a T cell line, or other cells grown in vitro. Methods for matching haplotypes are known in the art.
- the APC and T cells or supernatant therefrom may be obtained from a xenogeneic source, for example, mouse, rat, non-human primate, and porcine cells may be used.
- the source of antigen may be, but is not limited to, protein, including glycoprotein, peptides (including pools of overlapping peptides), superantigens (e.g., SEA, SEB, TSST-1) antibody/antigen complexes, tumor lysate, viral lysate (e.g., CMV lysate and the like), non-soluble cell debris, apoptotic bodies, necrotic cells, whole cells which are live, fixed, irradiated, heat-killed or otherwise manipulated, whole tumor cells from a tumor or a cell line that have been treated such that they are unable to continue dividing, allogeneic cells that have been treated such that they are unable to continue dividing, irradiated tumor cells, irradiated allogeneic cells, natural or synthetic complex carbohydrates, lipoproteins, lipopolysaccharides, RNA or a translation product of said RNA, and DNA or a polypeptide encoded by said DNA.
- protein including glycoprotein, peptid
- Non-transformed cells are typically irradiated with gamma rays in the range of about 3000 to 3600 rads, more preferably at about 3300 rads.
- Lymphoblastoid or tumor cell lines are typically irradiated with gamma rays in the range of about 6000 to 10,000 rads, more preferably at about 8000 rads.
- Necrotic and apoptotic cells may be generated by physical, chemical, or biological means. Necrotic cells are typically generated by freeze-thawing, while apoptotic cells are generated using UV irradiation. UV and gamma irradiation, and freeze-thawing procedures are well known in the art and are described, for example, in Current Protocols in Molecular Biology or Current Protocols in Immunology, John Wiley & Sons, New York. N.Y.
- Antigen source may also comprise non-transformed, transformed, transfected, or transduced cells or cell lines.
- Cells may be transformed, transfected, or transduced using any of a variety of expression or retroviral vectors known to those of ordinary skill in the art that may be employed to express recombinant antigens. Expression may also be achieved in any appropriate host cell that has been transformed, transfected, or transduced with an expression or retroviral vector containing a DNA molecule encoding recombinant antigen(s). Any number of transfection, transformation, and transduction protocols known to those in the art may be used, for example those outlined in Current Protocols in Molecular Biology, John Wiley & Sons, New York.
- recombinant vaccinia vectors and cells infected with said vaccina vectors may be used as a source of antigen.
- Recombinant antigen may include any number of defined tumor antigens described below.
- antigen may comprise viral antigens such as CMV pp65, HIV pg120, and the like.
- antigen may comprise defined tumor antigens such as the melanoma antigen Melan-A (also referred to as melanoma antigen recognized by T cells or MART-1), melanoma antigen-encoding genes 1, 2, and 3 (MAGE-1, -2, -3), melanoma GP100, carcinoembryonic antigen (CEA), the breast cancer angtigen, Her-2/Neu, serum prostate specific antigen (PSA), Wilm's Tumor (WT-1), PR1, PR3 (antigens implicated in the graft-versus-leukemia (GVL) effect in chronic myeloid leukemia), mucin antigens, MUC-1, -2, -3, -4, B cell lymphoma idotypes, and the like.
- melanoma antigen also referred to as melanoma antigen recognized by T cells or MART-1
- bead:cell ratios can lead to different outcomes with respect to expansion of antigen-specific T cells.
- bead:cell ratios can be varied to selectively expand or delete antigen-specific (memory) T cells.
- the particular bead:cell ratio used selectively expands antigen-specific T cells.
- antigen-specific T cells are activated by direct contact of a population of cells wherein at least a portion thereof comprises T cells (e.g., a leukaphersis product from an individual, blood sample, tumor biopsy, etc.), with a surface, wherein said surface has attached thereto a first agent that ligates a first T cell surface moiety of a T cell, and the same or a second surface has attached thereto a second agent that ligates a second moiety of said T cell, wherein said ligation by the first and second agent induces proliferation (expansion) of antigen-specific T cells present within the population of cells.
- T cells e.g., a leukaphersis product from an individual, blood sample, tumor biopsy, etc.
- the antigen-specific T cells are sensitized to further stimulation.
- the quantity of the CD3/TCR (and CD28) receptors that are bound by ligands determines the signal strength.
- stimulation with high bead:cell ratios provides a high concentration of stimulating antibody (i.e., “strong signal”), leading to over-stimulation of antigen-specific T cells, causing them to die, either by apoptosis or other mechanisms.
- Using lower bead:cell ratios provides a stimulation signal to antigen-specific T cells that does not over-stimulate, but rather induces rapid proliferation of these cells.
- antigen-specific T cells are activated by culturing T cells isolated as described herein above, with APC that have been loaded with antigen.
- suitable APC are plated in culture dishes and exposed to a source of antigen as described herein, in a sufficient amount and for a sufficient period of time to allow the antigen to bind and/or be taken up by the APC.
- antigen is exposed to the APC for a period of time between 24 hours and 4 days.
- the antigen is exposed to the APC for 36, 48, or 72 hours.
- the antigen is exposed to the APC for 2.5, 3, 3.5, or 4 days.
- antigen may be exposed to the APC for periods longer than 4 days, for example 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, or 10 days.
- the amount and time necessary to achieve binding and uptake of the antigen by the APC may differ depending on the source and type of antigen and may be determined by those of ordinary skill in the art by immunoassay or binding assay. Other methods known to those of skill in the art may be used to detect the presence of antigen in the context of MHC on the APC following their exposure to antigen.
- PBMC e.g., from blood, a leukapheris product, etc.
- PBMC from a subject are cultured directly in the presence of antigen, as described herein, to load APC with the antigen and to activate/stimulate antigen-specific T cells present in the PBMC.
- PBMC may be collected from an individual, contacted with an antigen of interest, such as a tumor antigen, or a viral lysate, etc.
- an antigen of interest such as a tumor antigen, or a viral lysate, etc.
- the APC present in the PBMC are loaded with the antigen, which is then presented to the T cells present in the sample.
- the antigen-specific T cells of the present invention may be stimulated with peptide-MHC tetramers, see for example Altman, et al., Science 1998 Jun. 19; 280(5371):1821.
- the APC of the present invention may be loaded with antigen through genetic modification.
- Genetic modification may comprise RNA or DNA transfection using any number of techniques known in the art, for example electroporation (using e.g., the Gene Pulser II, BioRad, Richmond, Calif.), various cationic lipids, (LIPOFECTAMINETM, Life Technologies, Carlsbad, Calif.), or other techniques such as calcium phosphate transfection as described in Current Protocols in Molecular Biology, John Wiley & Sons, New York. N.Y.
- RNA or DNA in 500 ⁇ l of Opti-MEM can be mixed with a cationic lipid at a concentration of 10 to 100 ⁇ g, and incubated at room temperature for 20 to 30 minutes.
- suitable lipids include LIPOFECTINTM, LIPOFECTAMINETM.
- the resulting nucleic acid-lipid complex is then added to 1-3 ⁇ 10 6 cells, preferably 2 ⁇ 10 6 , antigen-presenting cells in a total volume of approximately 2 ml (e.g., in Opti-MEM), and incubated at 37° C. for 2 to 4 hours.
- the APC may also be transduced using viral transduction methodologies as described below.
- APC are loaded with antigen attached to, coated on, or otherwise immobilized on particles, such as beads.
- particles such as beads.
- commercially available beads or other particles are useful, e.g., Miltenyi Particles, Miltenyi Biotec, Germany; Sepharose beads, Pharmacia Fine Chemicals, Sweden; DYNABEADSTM, Dynal Inc., New York.
- paramagnetic particles or beads are particularly suitable. Such paramagnetic beads or particles are commercially available, for example, those produced by Dynal AS under the trade name DynabeadsTM. Exemplary DynabeadsTM in this regard are M-280, M-450, and M-500.
- whole cells which are live, fixed, irradiated, heat-killed or ohterwise manipulated, are immobilized to ingestable beads, via for example antibody/ligand specific means or chemical means.
- tumor cell or virus-infected cell lysates, or antigen-preparations can be attached or otherwise immobilized to the beads (which may be paramagnetic or otherwise selectable).
- coated or antigen/cell/lysate-attached beads can be mixed with human or other animal peripheral blood preparations (or other compositions containing some percentage of antigen-presenting cells (particularly those capable of ingesting particles and then processing and presenting antigens associated with the particles).
- Phagocytic cells will ingest the beads/particles, process antigens associated with the particles, and present them to T cells in the cell mix. As noted elsewhere herein, only T cells with specificity for the variety of presented antigens will interact in a positive manner with the APC. APC containing paramagnetic or otherwise selectable beads can then be isolated carrying with them antigen-specific T cells.
- the particles of the present invention comprise a cell surface, such as described in U.S. patent application Ser. No. 10/336,224, PCT/US03/00339.
- antigen can be attached to the cells via antibody/ligand specific means as described herein or through genetic modification.
- Any number of transfection, transformation, and transduction protocols known to those in the art may be used, for example those outlined in Current Protocols in Molecular Biology, John Wiley & Sons, New York. N.Y., or in numerous kits available commercially (e.g., Invitrogen Life Technologies, Carlsbad, Calif.). Such techniques may result in stable transformants or may be transient.
- electroporation which may be performed on a variety of cell types, including mammalian cells, yeast cells and bacteria, using commercially available equipment.
- Optimal conditions for electroporation are experimentally determined for the particular host cell type, and general guidelines for optimizing electroporation may be obtained from manufacturers.
- Other suitable methods for transfection will depend upon the type of cell used (e.g., the lithium acetate method for yeast), and will be apparent to those of ordinary skill in the art.
- cells may be maintained in conditions that promote expression of the polynucleotide within the cell. Appropriate conditions depend upon the expression system and cell type, and will be apparent to those skilled in the art.
- Antigen may be attached to the particles, such as beads, by antibody/ligand specific means, e.g. through particles, such as beads, conjugated to an antibody or antibodies.
- Suitable antibody/ligand pairs may include, but are not limited to anti-MART-1 antibody/MART-1 antigen, anti-WT-1 antibody/WT-1, anti-PR1 antibody /PR1, anti-PR3 antibody/PR3, anti-tyrosinase antibody/tyrosinase antigen, anti-MAGE-1 antibody/MAGE-1 antigen, anti-MUC-1 antibody/MUC-1 antigen, anti- ⁇ -fetoprotein antibody/ ⁇ -fetoprotein antigen, anti-Her2Neu antibody/Her2Neu, anti-HIV gp120 antibody/HIV gp120, anti-influenza HA antibody/influenza HA, anti-CMV pp65/CMV pp65, anti-hepatitis C antibody/hepatitis C proteins, anti-EBV EBNA 3B antibody/EBV
- antigen binding interactions may be suitable for attaching antigen to particles, such as beads, for example, receptor/ligand interactions may be utilized.
- the antigen/protein is attached to the particles, such as beads by chemical means, e.g. antigen/protein can be bound through non-covalent association of the antigen and bead, simply by incubating/contacting the two together for a time and under conditions sufficient for association to occur.
- antigen may be attached to the particles, such as beads by a biotin/avidin or streptavidin interaction.
- hydrophobic “naked” beads with p-toluenesulphonyl (tosyl) reactive groups are used.
- Proteins are adsorbed hydrophobically on initial coupling with covalent binding of primary amine groups (NH 2 ) and sulphydryl groups (SH) occurring overnight. Coupling reactions can be performed at neutral pH however high pH and incubation at 37° C. can promote covalent binding.
- primary amine groups NH 2
- SH sulphydryl groups
- T cells isolated from a tissue source are exposed to antigen-loaded APC described herein for a time sufficient for T cells specific for a given antigen to be activated, for example as described in U.S. Pat. No. 5,827,642, or as described in Riddell, et al., 1990, J. Immunol. Methods, 128:189-201.
- T cells are exposed to antigen-loaded APC for a period of between about several hours to about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or about 20 days.
- the T cells are exposed to antigen, or antigen-loaded APC as described herein in vivo.
- antigen or antigen-loaded APC may be administered to an individual in order to stimulate and activate the T cells in vivo.
- the T cells may then be expanded either in vivo or ex vivo using the methods as described herein, such as with anti-CD3/anti-CD28 beads.
- the quantity and frequency of administration will be determined by such factors as the condition of the individual, and the type and severity of disease, although appropriate dosages may be determined by clinical trials.
- the T cells are exposed to antigen in vivo in an individual prior to onset of a disease or prior to treatment with other known therapies.
- the antigen-specific T cells are generated and then isolated and expanded and preserved for later use.
- isolation of antigen-specific T cells in direct contact with APC loaded with antigen immobilized on particles, such as beads is performed by magnetic isolation of cells which have attached to or engulfed paramagnetic particles.
- Such separation can be performed using standard methods available in the art.
- any magnetic separation methodology may be used including a variety of which are commercially available, (e.g., DYNAL® Magnetic Particle Concentrator (DYNAL MPC®), MACS, Miltenyi Biotec, Germany).
- DYNAL® Magnetic Particle Concentrator DYNAL MPC®
- MACS Miltenyi Biotec
- APC containing paramagnetic (or otherwise selectable) beads can then be isolated (via magnet or otherwise) carrying with them antigen-specific T cells.
- These antigen-specific T cells can then be activated/expanded by a variety of means, such as via XCELLERATETM technologies as described herein and U.S. patent application Ser. Nos. 10/350,305; 10/187,467; 10/133,236; 09/960,264; 09/794,230; PCT/US01/06139; and PCT/US02/28161.
- antigen-specific T cells are isolated by positive selection. Such isolation can be carried out on T cells freshly isolated from a subject or on T cells that have been exposed to antigen or antigen-loaded APC as described herein. Numerous immunoselection methods known to skilled artisans may be used. Such techniques are described, for example, in Current Protocols in Immunology, John Wiley & Sons, New York. N.Y. Markers that may be useful for the positive selection of antigen-specific cells include, but are not limited to, CD25, CD54, CD69, CD38, CD45RO, CD49d, CD40L, CD137, CD62L, and CD134. In one embodiment, fluorescence activated cell sorting may also be used to isolate desired antigen-specific T cells. In an additional embodiment, antigen-specific T cells may be isolated using peptide-MHC tetramers, see for example Altman, et al., Science 1998 Jun. 19; 280(5371):1821.
- antigen-specific T cells may be genetically modified. Genetic modification may comprise RNA or DNA transfection using any number of techniques known in the art, for example electroporation (using e.g., the Gene Pulser II, BioRad, Richmond, Calif.), various cationic lipids, (LIPOFECTAMINETM, Life Technologies, Carlsbad, Calif.), or other techniques such as calcium phosphate transfection as described in Current Protocols in Molecular Biology, John Wiley & Sons, New York. N.Y.
- electroporation using e.g., the Gene Pulser II, BioRad, Richmond, Calif.
- various cationic lipids (LIPOFECTAMINETM, Life Technologies, Carlsbad, Calif.)
- calcium phosphate transfection as described in Current Protocols in Molecular Biology, John Wiley & Sons, New York. N.Y.
- RNA or DNA in 500 ⁇ l of Opti-MEM can be mixed with a cationic lipid at a concentration of 10 to 100 ⁇ g, and incubated at room temperature for 20 to 30 minutes.
- suitable lipids include LIPOFECTINTM, LIPOFECTAMINETM.
- the resulting nucleic acid-lipid complex is then added to 1-3 ⁇ 10 6 cells, preferably 2 ⁇ 10 6 , antigen-presenting cells in a total volume of approximately 2 ml (e.g., in Opti-MEM), and incubated at 37° C. for 2 to 4 hours.
- the APC may also be transduced using viral transduction methodologies as described below.
- the retroviral vector may be an amphotropic retroviral vector, preferably a vector characterized in that it has a long terminal repeat sequence (LTR), e.g., a retroviral vector derived from the Moloney murine leukemia virus (MoMLV), myeloproliferative sarcoma virus (MPSV), murine embryonic stem cell virus (MESV), murine stem cell virus (MSCV), spleen focus forming virus (SFFV), or adeno-associated virus (AAV).
- LTR long terminal repeat sequence
- MoMLV Moloney murine leukemia virus
- MPSV myeloproliferative sarcoma virus
- MMV murine embryonic stem cell virus
- MSCV murine stem cell virus
- SFFV spleen focus forming virus
- AAV adeno-associated virus
- retroviral vectors are derived from murine retroviruses.
- Retroviruses adaptable for use in accordance with the present invention can, however, be derived from any avian or mammalian cell source. These retroviruses are preferably amphotropic, meaning that they are capable of infecting host cells of several species, including humans.
- the gene to be expressed replaces the retroviral gag, pol and/or env sequences.
- a number of illustrative retroviral systems have been described (e.g., U.S. Pat. Nos. 5,219,740; 6,207,453; 5,219,740; Miller and Rosman (1989) BioTechniques 7:980-990; Miller, A. D. (1990) Human Gene Therapy 1:5-14; Scarpa et al.
- genetically modified antigen-specific T cells can be isolated by any one of numerous immunoselection methods known to skilled artisans using antibodies or other receptors/ligands specific for the protein or proteins expressed from the transgene. Such techniques are known in the art, for example, in Current Protocols in Immunology, John Wiley & Sons, New York. N.Y.
- the antigen-specific T cells may be genetically modified to express a suicide gene, e.g. the herpes simplex virus thymidine kinase (HSV-TK) as described in Bonini, et al., 1997 Science, 276(5319):1719-24, and/or other surface markers (e.g., truncated nerve growth factor (dNGFR)) for in vivo tracking and/or control of infused antigen-specific T cells.
- HSV-TK herpes simplex virus thymidine kinase
- dNGFR truncated nerve growth factor
- the antigen-specific T cells may be genetically modified to express a protein for targeting the T cells to a particular tissue of interest.
- CMOS complementary metal-oxide-semiconductor
- a bioreactor e.g., CellCube (Corning Science Products) or CELL-PHARM, (CD-Medical, Inc. of Hialeah, Fla.)
- petri dishes e.g., petri dishes and multi-well containing plates made for use in tissue culture, or any container capable of holding cells, preferably in a sterile environment.
- a bioreactor is also useful.
- Suitable complete growth media for the culture of the APC and antigen-specific T cells of the present invention include for example RPMI 1640, DMEM, MEM, ⁇ -MEM, AIM-V, HAMS F-12, X-Vivo 15, or X-Vivo 20.
- the media can comprise a cytokine, such as IL-2, IFN- ⁇ , IL-4, GM-CSF, IL-10, IL-12, TGF ⁇ , and TNF- ⁇ , or a vitamin.
- the medium comprises surfactant, an antibody, plasmanate or a reducing agent (e.g. N-acetyl-cysteine, 2-mercaptoethanol).
- the growth medium for the cells at each step of the method of the invention should allow for the survival of the APC and/or the antigen-specific T cells.
- Any growth medium typically used to culture cells may be used according to the method of the invention provided the medium is supplemented with the appropriate cytokines, serum, antibiotics, vitamins, amino acids or other necessary additives.
- the cytokines may be, but are not limited to, granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin 4 (IL-4), or IL-13.
- cytokines and growth factors that may be added to the growth medium include but are not limited to interleukin 1 ⁇ (IL-1 ⁇ ) and ⁇ (IL-1 ⁇ ), IL-2, tumor necrosis factor alpha (TNF- ⁇ ), interleukin 3 (IL-3), monocyte colony stimulating factor (M-CSF), granulocyte colony-stimulating factor (G-CSF), stem cell factor (SCF), interleukin 6 (IL-6), interleukin 15 (IL-15), and Flt3-ligand.
- IL-1 ⁇ interleukin 1 ⁇
- IL-1 ⁇ tumor necrosis factor alpha
- IL-3 interleukin 3
- M-CSF monocyte colony stimulating factor
- G-CSF granulocyte colony-stimulating factor
- SCF stem cell factor
- IL-6 interleukin 6
- IL-15 interleukin 15
- Flt3-ligand Flt3-ligand.
- Preferred media include RPMI 1640, AIM-V, DMEM, MEM, ⁇ -MEM, F-12, X-Vivo 15, and X-Vivo 20, with added amino acids and vitamins, either serum-free or supplemented with an appropriate amount of serum (or plasma) or a defined set of hormones, and an amount of cytokine(s) sufficient to support the expansion of the antigen-specific T cells.
- the preferred media comprises 1 liter of X-Vivo 15, BioWhittaker; with 50 ml heat inactivated pooled human serum, 20 ml 1 M Hepes, 10 ml 200 mM L-glutamine with or without about 100,000 I.U. IL-2.
- media may include lipids and/or sources of protein.
- RPMI 1640 supplemented with 1-5% human AB serum preferred.
- Mixtures of cytokines may also be used.
- Cells may also be adapted to grow in other sera, such as fetal calf (bovine) serum (FCS/FBS), at other concentrations of serum, or in serum-free media.
- FCS/FBS fetal calf serum
- serum-free medium supplemented with hormones is also suitable for culturing the APC precursors.
- Media may, but does not necessarily, contain antibiotics to minimize growth of bacteria in the cultures. Penicillin, streptomycin or gentamicin or combinations containing them are preferred.
- the medium, or a portion of the medium, in which the cells are cultured should be periodically replenished to provide fresh nutrients including GM-CSF, IL-4, IL-13, IL-15 and/or other cytokines.
- Expansion of the antigen-specific T cells of the present invention is carried out by cell surface moiety ligation that re-stimulates the antigen-specific T cells to proliferate.
- the antigen-specific T cells are first isolated by methods described herein following exposure to antigen loaded APC.
- the antigen-specific T cells are expanded directly from the culture with antigen-loaded APC present without an isolation step.
- antigen-specific T cells are activated and expanded using XCELLERATETM processes as described herein and in U.S. patent application Ser. Nos. 10/350,305; 10/187,467; 10/133,236; 09/960,264; 09/794,230, with no addition of antigen or antigen-coated particles.
- antigen-specific T cells that have been previously stimulated or activated in vivo (e.g.
- memory T cells are expanded by an agent providing a primary activation signal such as an anti-CD3 antibody and an agent providing a co-stimulatory signal, such as an anti-CD28 antibody, with both agents co-immobilized to the same surface, such as a paramagnetic bead.
- an agent providing a primary activation signal such as an anti-CD3 antibody
- an agent providing a co-stimulatory signal such as an anti-CD28 antibody
- both agents co-immobilized to the same surface such as a paramagnetic bead.
- varying the bead:cell ratios during this expansion phase in particular using low bead:cell ratios, favors expansion of antigen-specific T cells.
- bead to cell ratios of 1:200, 1:150, 1:125, 1:110, 1:100, 1:75, 1:50, 1:25, 1:20, 1:15, 1:10, 1:5 or 1:2.5 are used to expand antigen-specific T cells.
- a particular advantage of this aspect of the present invention is that it is not
- expansion is carried out by re-stimulating a population of antigen-specific T cells and simultaneously stimulating an accessory molecule on the surface of the antigen-specific T cells with a ligand which binds the accessory molecule, as described for example, in U.S. patent application Ser. Nos. 10/350,305, 10/187,467, 10/133,236, 09/960,264, 09/794,230, 08/253,694, 08/403,253, 08/435,816, 08/592,711, 09/183,055, 09/350,202, and 09/252,150, and U.S. Pat. Nos. 5,858,358; 6,352,694; and 5,883,223.
- re-stimulation may be accomplished by cell surface moiety ligation, such as through the T cell receptor (TCR)/CD3 complex or the CD2 surface protein.
- TCR T cell receptor
- a number of anti-human CD3 monoclonal antibodies are commercially available, exemplary are, clone BC3 (XR-CD3; Fred Hutchinson Cancer Research Center, Seattle, Wash.), OKT3, prepared from hybridoma cells obtained from the American Type Culture Collection, and monoclonal antibody G19-4.
- stimulatory forms of anti-CD2 antibodies are known and available. Stimulation through CD2 with anti-CD2 antibodies is typically accomplished using a combination of at least two different anti-CD2 antibodies.
- Stimulatory combinations of anti-CD2 antibodies that have been described include the following: the T11.3 antibody in combination with the T11.1 or T11.2 antibody (Meuer et al., Cell 36:897-906, 1984), and the 9.6 antibody (which recognizes the same epitope as T11.1) in combination with the 9-1 antibody (Yang et al., J. Immunol. 137:1097-1100, 1986).
- Other antibodies that bind to the same epitopes as any of the above-described antibodies can also be used.
- Additional antibodies, or combinations of antibodies can be prepared and identified by standard techniques. Re-stimulation may also be achieved through contact with antigen, peptide, protein, peptide-MHC tetramers (see Altman, et al Science 1996 Oct.
- superantigens e.g., Staphylococcus enterotoxin A (SEA), Staphylococcus enterotoxin B (SEB), Toxic Shock Syndrome Toxin 1 (TSST-1)
- endotoxin or through a variety of mitogens, including but not limited to, phytohemagglutinin (PHA), phorbol myristate acetate (PMA) and ionomycin, lipopolysaccharide (LPS), T cell mitogen, and IL-2.
- PHA phytohemagglutinin
- PMA phorbol myristate acetate
- LPS lipopolysaccharide
- T cell mitogen and IL-2.
- the antigen-specific cell population may be stimulated or restimulated as described herein, such as by contact with an anti-CD3 antibody or an anti-CD2 antibody immobilized on a surface, or by contact with a protein kinase C activator (e.g., bryostatin) in conjunction with a calcium ionophore.
- a protein kinase C activator e.g., bryostatin
- a ligand that binds the accessory molecule is used.
- a population of CD4 + cells can be contacted with an anti-CD3 antibody and an anti-CD28 antibody, under conditions appropriate for stimulating proliferation of the T-cells.
- an anti-CD3 antibody and the anti-CD28 antibody B-T3, XR-CD28 (Diaclone, Besançon, France) can be used as can other methods commonly known in the art (Berg et al., Transplant Proc. 30(8):3975-3977, 1998; Haanen et al., J. Exp. Med. 190(9):1319-1328, 1999; Garland et al., J. Immunol Meth. 227(1-2):53-63, 1999).
- a co-stimulatory or accessory molecule on the surface of the T cells such as CD28
- a ligand that binds the accessory molecule can be used to stimulate T cells.
- Exemplary anti-CD28 antibodies or fragments thereof useful in the context of the present invention include monoclonal antibody 9.3 (IgG2 a ) (Bristol-Myers Squibb, Princeton, N.J.), monoclonal antibody KOLT-2 (IgG1), 15E8 (IgG1), 248.23.2 (IgM), clone B-T3 (XR-CD28; Diaclone, Besan wonder, France) and EX5.3D10 (IgG2 a ) (ATCC HB11373).
- Exemplary natural ligands include the B7 family of proteins, such as B7-1 (CD80) and B7-2 (CD86) (Freedman et al., J. Immunol.
- activation of a T-cell population may be enhanced by co-stimulation of other T-cell integral membrane proteins.
- binding of the T-cell integrin LFA-1 to its natural ligand, ICAM-1 may enhance activation of cells.
- Another cell surface molecule that may act as a co-stimulator for T-cells is VCAM-1 (CD106) that binds very-late-antigen-4 (VLA-4) on T-cells.
- VCAM-1 CD106
- VLA-4 very-late-antigen-4
- Ligation of 4-1BB (CD137), a co-stimulatory receptor expressed on activated T cells, and/or NKG2D may also be useful in the context of the present invention to amplify T-cell mediated immunity. It should be noted that more than one costimulatory molecule as described herein may be stimulated at a time, and in any combination, such that desired expansion of the T cells occurs.
- binding homologues of a natural ligand can also be used in accordance with the present invention.
- Other agents may include natural and synthetic ligands. Agents may include, but are not limited to, other antibodies or fragments thereof, a peptide, polypeptide, growth factor, cytokine, chemokine, glycopeptide, soluble receptor, steroid, hormone, mitogen, such as PHA, or other superantigens.
- the primary stimulatory signal and the co-stimulatory signal for the T-cell may be provided by different protocols.
- the agents providing each signal may be in solution or coupled to a surface. When coupled to a surface, the agents may be coupled to the same surface (i.e., in “cis” formation) or to separate surfaces (i.e., in “trans” formation).
- one agent may be coupled to a surface and the other agent in solution.
- the agent providing the co-stimulatory signal is bound to a cell surface and the agent providing the primary activation signal is in solution or coupled to a surface. In certain embodiments, both agents can be in solution.
- the agents may be in soluble form, and then cross-linked to a surface, such as a cell expressing Fc receptors or an antibody or other binding agent which will bind to the agents.
- the two agents are immobilized on beads, either on the same bead, i.e., “cis,” or to separate beads, i.e., “trans.”
- the agent providing the primary activation signal is an anti-CD3 antibody and the agent providing the co-stimulatory signal is an anti-CD28 antibody; and both agents are co-immobilized to the same surface, such as a bead, in equivalent molecular amounts.
- a 1:1 ratio of each antibody bound to the beads for CD4 + T-cell expansion and T-cell growth is used.
- One aspect of the present invention stems from the surprising finding that using lower ratios of anti-CD3:anti-CD28 antibodies bound to the beads results in improved expansion of T cells, including antigen-specific T cells.
- a ratio of anti CD3:CD28 antibodies bound to the beads is used such that an increase in T cell expansion is observed as compared to the expansion observed using a ratio of 1:1. In one particular embodiment an increase of from about 0.5 to about 3 fold is observed as compared to the expansion observed using a ratio of 1:1. In one embodiment, the ratio of CD3:CD28 antibody bound to the beads ranges from 100:1 to 1:100 and all integer values there between.
- more anti-CD28 antibody is bound to the particles than anti-CD3 antibody, i.e. the ratio of CD3:CD28 is less than one.
- the ratio of anti CD28 antibody to anti CD3 antibody bound to the beads is greater than 2:1.
- a 1:100 CD3:CD28 ratio of antibody bound to beads is used.
- a 1:75 CD3:CD28 ratio of antibody bound to beads is used.
- a 1:50 CD3:CD28 ratio of antibody bound to beads is used.
- a 1:30 CD3:CD28 ratio of antibody bound to beads is used.
- a 1:10 CD3:CD28 ratio of antibody bound to beads is used.
- a 1:3 CD3:CD28 ratio of antibody bound to the beads is used.
- a 3:1 CD3:CD28 ratio of antibody bound to the beads is used.
- Ratios of particles to cells from 1:500 to 500:1 and any integer values in between may be used to stimulate T-cells.
- the ratio of particle to cells may dependant on particle size relative to the target cell. For example, small sized beads could only bind a few cells, while larger beads could bind many.
- the ratio of particles to cells ranges from 1:100 to 100:1 and any integer values in-between and in further embodiments the ratio comprises 1:9 to 9:1 and any integer values in between, can also be used to stimulate T-cells.
- the ratio of anti-CD3- and anti-CD28-coupled particles to T-cells that result in T-cell stimulation and expansion can vary as noted above, however in certain embodiments the ratio may be 1:150 or lower. Certain preferred ratios include 1:150, 1:100, 1:75, 1:50, 1:40, 1:30, 1:25, 1:20, 1:15, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2.5, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 15:1, and 20:1 with one preferred ratio being 1:1 particles per T-cell. In one embodiment, a ratio of particles to cells of 1:1 or less is used.
- a preferred particle:cell ratio is 1:2.5 or 1:5.
- the ratio of particles to cells can be varied depending on the day of stimulation.
- the ratio of particles to cells is from 1:5, 1:2.5, 1:1 to 10:1 on the first day and additional particles are added to the cells every day or every other day thereafter for up to 10 days, at final ratios of from 1:1, 1:5, 1:20, 1:25, 1:50, or 1:100 (based on cell counts on the day of addition).
- the ratio of particles to cells is 1:2.5, 1:5, or 1:1 on the first day of stimulation and adjusted to 1:5 on the third and fifth days of stimulation.
- the ratio of particles to cells is 1:2.5, 1:5, or 1:1 on the first day of stimulation and adjusted to 1:10, 1:20, 1:25, 1:50, or 1:100 at day 5, 7, or 9.
- particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:5 on the third and fifth days of stimulation.
- the ratio of particles to cells is 2:1 on the first day of stimulation and adjusted to 1:10 on the third and fifth days of stimulation.
- particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:10 on the third and fifth days of stimulation.
- ratios may be suitable for use in the present invention.
- bead:cell ratios can be varied to selectively expand or delete antigen-specific (memory) T cells.
- the particular bead:cell ratio used selectively deletes antigen-specific T cells.
- the particular bead:cell ratio used selectively expands antigen-specific T cells.
- bead to cell ratios of 1:100, 1:50, 1:25, 1:5 or 1:2.5 and the like are used to expand antigen-specific T cells.
- Low bead:cell ratio can help preserve and promote expansion of memory (antigen-specific) T cells.
- compositions and methods described herein can be used to expand specific populations of T cells, or to delete specific populations of T cells, for use in any variety of immunotherapeutic settings described herein.
- the particle:cell ratios described herein can be used in any combination with the various ratios of antibodies bound on the beads.
- beads containing about 1:5 to 1:10 ratio of anti-CD3/anti-CD28 antibodies bound thereto can be used at a ratio of about 1:5 to 1:10 particles:cell.
- beads containing a 1:1 ratio of anti-CD3/anti-CD28 antibodies bound thereto can be used at a ratio of about 1:5 particles:cell, etc.
- the ratio of anti-CD3:anti-CD28 antibody bound to the beads ranges from 100:1 to 1:100 and all integer values there between and such beads can be used at a ratio of particle:cell of anywhere from about 1:500 to 500:1 and any integer values in between, in any combination.
- T-cell proliferation is monitored periodically (e.g., daily) by, for example, examining the size or measuring the volume of the T-cells, such as with a Coulter Counter.
- a resting T-cell has a mean diameter of about 6.8 microns, and upon initial activation and stimulation, in the presence of the stimulating ligand, the T-cell mean diameter will increase to over 12 microns by day 4 and begin to decrease by about day 6.
- the T-cells When the mean T-cell diameter decreases to approximately 8 microns, the T-cells may be reactivated and re-stimulated to induce further proliferation of the T-cells.
- the rate of T-cell proliferation and time for T-cell re-stimulation can be monitored by assaying for the presence of cell surface molecules, such as, CD154, CD54, CD25, CD137, CD134, which are induced on activated T-cells.
- T-cell stimulation is performed with anti-CD3 and anti-CD28 antibodies co-immobilized on beads (3 ⁇ 28 beads), for a period of time sufficient for the cells to return to a quiescent state (low or no proliferation) (approximately 8-14 days after initial stimulation).
- the stimulation signal is then removed from the cells and the cells are washed and infused back into the patient.
- the cells at the end of the stimulation phase are rendered “super-inducible” by the methods of the present invention, as demonstrated by their ability to respond to antigens and the ability of these cells to demonstrate a memory-like phenotype, as is evidence by the examples.
- the activated T-cells demonstrate a robust response characterized by unique phenotypic properties, such as sustained CD154 expression and increased cytokine production.
- the cells such as T-cells
- the beads and the cells are subsequently separated, and then the cells are cultured.
- the agent-coated beads and cells prior to culture, are not separated but are cultured together.
- the beads and cells are first concentrated by application of a force, resulting in cell surface moiety ligation, thereby inducing cell stimulation.
- the time of exposure to stimulatory agents such as anti-CD3/anti-CD28 (i.e., CD3xCD28)-coated particles, such as beads, may be modified or tailored to obtain a desired T-cell phenotype.
- stimulatory agents such as anti-CD3/anti-CD28 (i.e., CD3xCD28)-coated particles, such as beads
- T H helper T-cells
- CD4 + T-cells express important immune-regulatory molecules, such as GM-CSF, CD40L, and IL-2, for example.
- CD4-mediated help a method, such as that described herein, which preserves or enhances the CD4:CD8 ratio could be of significant benefit.
- the T cell activation approaches described herein can also be utilized, by for example, pre-selecting for CD8 + cells prior to stimulation and/or culture. Such situations may exist where increased levels of IFN- ⁇ is preferred.
- T H 1-type cells versus T H 2-type cells (or vice versa), or supernatants therefrom.
- regulatory T cells e.g., Autoimmun Rev. 2002 August;1(4):190-7; Curr Opin Immunol. 2002 December; 14(6):771-8.
- times of cell surface moiety ligation that induces re-stimulation may be varied or pulsed. For example expansion times may be varied to obtain the specific phenotype of interest and/or different types of stimulatory agents may be used (e.g., antibodies or fragments thereof, a peptide, polypeptide, MHC/peptide tetramer, growth factor, cytokine, chemokine, glycopeptide, soluble receptor, steroid, hormone, mitogen, such as PHA, or other superantigens).
- stimulatory agents e.g., antibodies or fragments thereof, a peptide, polypeptide, MHC/peptide tetramer, growth factor, cytokine, chemokine, glycopeptide, soluble receptor, steroid, hormone, mitogen, such as PHA, or other superantigens.
- the stimulation and/or expansion time may be four weeks or less, 2 weeks or less, 10 days or less, or 8 days or less (four weeks or less includes all time ranges from 4 weeks down to 1 day (24 hours)). In some embodiments, stimulation and expansion may be carried out for 6 days or less, 4 days or less, 2 days or less, and in other embodiments for as little as 24 or less hours, and preferably 4-6 hours or less (these ranges include any integer values in between).
- the population of T-cells may not increase in number as dramatically, but the population will provide robust and healthy activated antigen-specific T-cells that can continue to proliferate in vivo and more closely resemble the natural effector T-cell pool.
- the mixture may be cultured for several hours (about 3 hours) to about 14 days or any hourly integer value in between. In another embodiment, the mixture may be cultured for 21 days. In one embodiment of the invention the beads and the T-cells are cultured together for about eight days. In another embodiment, the beads and T-cells are cultured together for 2-3 days. Several cycles of stimulation may also be desired such that culture time of T cells can be 60 days or more.
- Conditions appropriate for T-cell culture include an appropriate media (e.g., Minimal Essential Media or RPMI Media 1640 or, X-vivo 15, (BioWhittaker)) that may contain factors necessary for proliferation and viability, including serum (e.g., fetal bovine or human serum), interleukin-2 (IL-2), insulin, IFN- ⁇ , IL-4, GM-CSF, IL-10, IL-12, TGF ⁇ , and TNF- ⁇ . or any other additives for the growth of cells known to the skilled artisan.
- Other additives for the growth of cells include, but are not limited to, surfactant, plasmanate, and reducing agents such as N-acetyl-cysteine and 2-mercaptoethanol.
- Media can include RPMI 1640, AIM-V, DMEM, MEM, ⁇ -MEM, F-12, X-Vivo 15, and X-Vivo 20, with added amino acids and vitamins, either serum-free or supplemented with an appropriate amount of serum (or plasma) or a defined set of hormones, and/or an amount of cytokine(s) sufficient for the growth and expansion of T-cells.
- Antibiotics e.g., penicillin and streptomycin, are included only in experimental cultures, not in cultures of cells that are to be infused into a subject.
- the target cells are maintained under conditions necessary to support growth, for example, an appropriate temperature (e.g., 37° C.) and atmosphere (e.g., air plus 5% CO 2 ).
- Feeder cells can encompass a variety of cell types, including, irradiated peripheral blood lymphocytes (autologous or allogeneic) alone or in combination with EBV-transformed B cell lines (autologous or allogeneic), immortalized or non-immortalized cell lines of the myelomoncytic lineage, such as macrophges, dentritic cells, red blood cells, B-cells, tumor cell lines such as U937, Jurkat, Daudi, MOLT-4, HUT, CEM, Colo 205, HTB-13, and HTB-70.
- Feeder cells need not be of human origin as long as they provide feeder function, e.g. the ability to facilitate the survival and growth of primary T cells and ther derived antigen-specific clones.
- compositions of the present invention may be administered either alone, or as a pharmaceutical composition in combination with diluents and/or with other components such as IL-2 or other cytokines or cell populations.
- pharmaceutical compositions of the present invention may comprise a target cell population as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients.
- compositions may comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as ethylenediaminetetraacetic acid (EDTA) or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives.
- buffers such as neutral buffered saline, phosphate buffered saline and the like
- carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol
- proteins polypeptides or amino acids such as glycine
- antioxidants such as ethylenediaminetetraacetic acid (EDTA) or glutathione
- adjuvants e.g., aluminum hydroxide
- preservatives e.g., aluminum hydroxide
- a related embodiment of the present invention further provides a pharmaceutical composition comprising the antigen-specific T cells, and a pharmaceutically acceptable carrier.
- the pharmaceutically acceptable carrier should be sterilized by techniques known to those skilled in the art.
- compositions of the present invention may be administered in a manner appropriate to the disease to be treated (or prevented).
- the quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease, although appropriate dosages may be determined by clinical trials.
- the present invention also provides methods for preventing, inhibiting, or reducing the presence of a cancer or malignant cells in an animal, which comprise administering to an animal an anti-cancer effective amount of the subject antigen-specific T cells.
- the cancers contemplated by the present invention against which the immune response is induced, or which is to be prevented, inhibited, or reduced in presence, may include but are not limited to melanoma, non-Hodgkin's lymphoma, Hodgkin's disease, leukemia, plasmocytoma, sarcoma, glioma, thymoma, breast cancer, prostate cancer, colo-rectal cancer, kidney cancer, renal cell carcinoma, pancreatic cancer, esophageal cancer, brain cancer, lung cancer, ovarian cancer, cervical cancer, multiple myeloma, hepatoma, acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), low-grade lymphoma, and other neoplasms known in the art.
- ALL acute lymphoblastic leukemia
- AML acute myelogenous le
- compositions as described herein can be used to induce or enhance responsiveness to pathogenic organisms, such as viruses, (e.g., single stranded RNA viruses, single stranded DNA viruses, human immunodeficiency virus (HIV), hepatitis A, B, and C virus, herpes simplex virus (HSV), cytomegalovirus (CMV) Epstein-Barr virus (EBV), Human Papilloma Virus (HPV)), parasites (e.g., protozoan and metazoan pathogens such as Plasmodia species, Leishmania species, Schistosoma species, Trypanosoma species), bacteria (e.g., Mycobacteria, Salmonella, Streptococci, E. coli, Staphylococci ), fungi (e.g., Candida species, Aspergillus species) and Pneumocystis carinii.
- viruses e.g., single stranded RNA viruses, single stranded
- the methods of the present invention can be used in conjunction with the generation of T regulatory cells for specific immunosuppression in the case of inflammatory disease, autoimmunity, and foreign graft acceptance.
- Regulatory T cells can be generated and expanded using the methods of the present invention.
- the regulatory T cells can be antigen-specific and/or polyclonal.
- Regulatory T cells can be generated using art-recognized techniques as described for example, in Woo, et al., J. Immunol. 2002 May 1;168(9):4272-6; Shevach, E. M., Annu. Rev. Immunol. 2000, 18:423; Stephens, et al., Eur. J. Immunol.
- T cells of the present invention can be used for the treatment of autoimmune diseases such as, but not limited to, rheumatoid arthritis, multiple sclerosis, insulin dependent diabetes, Addison's disease, celiac disease, chronic fatigue syndrome, inflammatory bowel disease, ulcerativecolitis, Crohn's disease, Fibromyalgia, systemic lupus erythematosus, psoriasis, Sjogren's syndrome, hyperthyroidism/Graves disease, hypothyroidism/Hashimoto's disease, Insulin-dependent diabetes (type 1), Myasthenia Gravis, endometriosis, scleroderma, pernicious anemia, Goodpasture syndrome, Wegener's disease, glomerulonephritis, aplastic anemia, par
- the immune response induced in the animal by administering the subject compositions of the present invention may include cellular immune responses mediated by cytotoxic T cells, capable of killing tumor and infected cells, and helper T cell responses.
- Humoral immune responses mediated primarily by helper T cells capable of activating B cells thus leading to antibody production, may also be induced.
- a variety of techniques may be used for analyzing the type of immune responses induced by the compositions of the present invention, which are well described in the art; e.g., Coligan et al. Current Protocols in Immunology, John Wiley & Sons Inc. (1994).
- an immunologically effective amount When “an immunologically effective amount”, “an anti-tumor effective amount”, “an tumor-inhibiting effective amount”, or “therapeutic amount” is indicated, the precise amount of the compositions of the present invention to be administered can be determined by a physician with consideration of individual differences in age, weight, tumor size, extent of infection or metastasis, and condition of the patient. It can generally be stated that a pharmaceutical composition comprising the subject antigen-specific T cells, may be administered at a dosage of 10 4 to 10 7 APC/kg body weight, preferably 10 5 to 10 6 APC/kg body weight, including all integer values within those ranges. Antigen-specific T cells compositions may also be administered multiple times at these dosages.
- the cells can be administered by using infusion techniques that are commonly known in immunotherapy (see, e.g., Rosenberg et al., New Eng. J. of Med. 319:1676, 1988).
- the optimal dosage and treatment regime for a particular patient can readily be determined by one skilled in the art of medicine by monitoring the patient for signs of disease and adjusting the treatment accordingly.
- antigen-specific T cells are administered approximately at 2 ⁇ 10 9 to 2 ⁇ 10 11 cells to the patient.
- lower numbers of cells in the range of 10 6 /kilogram (10 6 -10 11 per patient) may be administered.
- T cells are administered at 1 ⁇ 10 5 , 1 ⁇ 10 6 , 1 ⁇ 10 7 , 1 ⁇ 10 8 , 2 ⁇ 10 8 , 2 ⁇ 10 9 , 1 ⁇ 10 10 , 2 ⁇ 10 10 , 1 ⁇ 10 11 , 5 ⁇ 10 11 , or 1 ⁇ 10 12 cells to the subject.
- T cell compositions may be administered multiple times at dosages within these ranges.
- the antigen-specific T cells may be autologous or heterologous to the patient undergoing therapy.
- the treatment may also include administration of mitogens (e.g., PHA) or lymphokines, cytokines, and/or chemokines (e.g., GM-CSF, IL-4, IL-13, Flt3-L, RANTES, MIP1 ⁇ , etc.) as described herein to enhance induction of the immune response.
- mitogens e.g., PHA
- lymphokines e.g., IL-4, IL-13, Flt3-L, RANTES, MIP1 ⁇ , etc.
- chemokines e.g., GM-CSF, IL-4, IL-13, Flt3-L, RANTES, MIP1 ⁇ , etc.
- compositions of the subject pharmaceutical compositions may be carried out in any convenient manner, including by aerosol inhalation, injection, ingestion, transfusion, implantation or transplantation.
- the compositions of the present invention may be administered to a patient subcutaneously, intradermally, intramuscularly, by intravenous (i.v.) injection, or intraperitoneally.
- the antigen-specific T cell compositions of the present invention are administered to a patient by intradermal or subcutaneous injection.
- the antigen-specific T cell compositions of the present invention are preferably administered by i.v. injection.
- the compositions of antigen-specific T cells may be injected directly into a tumor or lymph node.
- the pharmaceutical composition can be delivered in a controlled release system.
- a pump may be used (see Langer, 1990, Science 249:1527-1533; Sefton 1987, CRC Crit. Ref. Biomed. Eng. 14:201; Buchwald et al., 1980; Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321:574).
- polymeric materials can be used (see Medical Applications of Controlled Release, 1974, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla.; Controlled Drug Bioavailability, Drug Product Design and Performance, 1984, Smolen and Ball (eds.), Wiley, New York; Ranger and Peppas, 1983; J. Macromol. Sci. Rev. Macromol. Chem. 23:61; see also Levy et al., 1985, Science 228:190; During et al., 1989, Ann. Neurol. 25:351; Howard et al., 1989, J. Neurosurg. 71:105).
- a controlled release system can be placed in proximity of the therapeutic target, thus requiring only a fraction of the systemic dose (see, e.g., Medical Applications of Controlled Release, 1984, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla., vol. 2, pp. 115-138).
- the antigen-specific T cell compositions of the present invention may also be administered using any number of matrices.
- Matrices have been utilized for a number of years within the context of tissue engineering (see, e.g., Principles of Tissue Engineering (Lanza, Langer, and Chick (eds.)), 1997.
- the present invention utilizes such matrices within the novel context of acting as an artificial lymphoid organ to support, maintain, or modulate the immune system, typically through modulation of T cells. Accordingly, the present invention can utilize those matrix compositions and formulations which have demonstrated utility in tissue engineering. Accordingly, the type of matrix that may be used in the compositions, devices and methods of the invention is virtually limitless and may include both biological and synthetic matrices.
- compositions and devices set forth by U.S. Pat. Nos. 5,980,889; 5,913,998; 5,902,745; 5,843,069; 5,787,900; or 5,626,561 are utilized.
- Matrices comprise features commonly associated with being biocompatible when administered to a mammalian host. Matrices may be formed from both natural or synthetic materials. The matrices may be non-biodegradable in instances where it is desirable to leave permanent structures or removable structures in the body of an animal, such as an implant; or biodegradable.
- the matrices may take the form of sponges, implants, tubes, telfa pads, fibers, hollow fibers, lyophilized components, gels, powders, porous compositions, or nanoparticles.
- matrices can be designed to allow for sustained release seeded cells or produced cytokine or other active agent.
- the matrix of the present invention is flexible and elastic, and may be described as a semisolid scaffold that is permeable to substances such as inorganic salts, aqueous fluids and dissolved gaseous agents including oxygen.
- a matrix is used herein as an example of a biocompatible substance.
- the current invention is not limited to matrices and thus, wherever the term matrix or matrices appears these terms should be read to include devices and other substances which allow for cellular retention or cellular traversal, are biocompatible, and are capable of allowing traversal of macromolecules either directly through the substance such that the substance itself is a semi-permeable membrane or used in conjunction with a particular semi-permeable substance.
- the cells of the present invention are administered to a patient in conjunction with (e.g. before, simulataneously or following) any number of relevant treatment modalities, including but not limited to treatment with agents such as antiviral agents, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies, cytoxin, fludaribine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, and irradiation.
- agents such as antiviral agents, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies, cytoxin, fludaribine, cyclosporin, FK506,
- the cell compositions of the present invention are administered to a patient in conjunction with (e.g.
- T-cell ablative therapy using either chemotherapy agents such as, fludarabine, external-beam radiation therapy (XRT), cyclophosphamide, or antibodies such as OKT3 or CAMPATH.
- chemotherapy agents such as, fludarabine, external-beam radiation therapy (XRT), cyclophosphamide, or antibodies such as OKT3 or CAMPATH.
- the cell compositions of the present invention are administered following B-cell ablative therapy such as agents that react with CD20, e.g. Rituxan.
- the dosage of the above treatments to be administered to a patient will vary with the precise nature of the condition being treated and the recipient of the treatment. The scaling of dosages for human administration can be performed according to art-accepted practices.
- the dose for CAMPATH for example, will generally be in the range 1 to about 100 mg for an adult patient, usually administered daily for a period between 1 and 30 days.
- the preferred daily dose is 1 to 10 mg per day although in some instances larger doses of up to 40 mg per day may be used
- CMV cytomegalovirus
- CMV lysate prepared using standard techniques was mixed at room temperature for 1-2 hours with Dynabead M-450 while rotating. Beads were then washed once, and added to PBMC. Within hours, the beads were phagocytosed in the APC. Within 72 hours, CMVpp65-HLA-A2 tetramers detected CD25-high (activated) T cell specific for CMV pp65. Magnetic selection of the bead-loaded APC with the associated antigen-specific T cells was carried out at day 5, thereby enriching for CMV-specific T cells. As shown in FIG. 1, following magnetic separation, CMV-specific T cells were still tightly associated with bead-loaded APC. It should be noted that magnetic separation can be carried out anywhere from about day 1 to about day 10.
- antigen-coated beads were used to activate CMV-specific CD8 + T cells ex vivo.
- PBMC from CMV pp65 tetramer-positive and tetramer-negative donors were stimulated with paramagnetic Dynal M-450 beads coated with CMV lysate.
- CMV pp65 tetramer-negative PBMC were cultured with CMV-lysate coated beads (FIG. 2, panel A)
- CMV pp65 tetramer-positive PBMC were cultured with “naked” beads (no CMV antigen) (FIG. 2, panel B).
- CMV pp65 tetramer-positive PBMC were cultured with CMV-lysate coated beads (FIG. 2, panel C).
- CMV-specific T cells Following stimulation, activation of CMV-specific T cells was measured on Day 10 by CMV pp65 HLA-A2 tetramer stain and CD25 expression as an indicator of activation. As shown in FIG. 2, up-regulation of CD25 was observed in memory CD8 CMV tetramer+T cells expanded ex vivo using antigen-coated beads.
- Antigen-coated beads can be used to activate and stimulate antigen-specific T cells. These antigen-specific T cells can then be enriched as described in Example 1 and elsewhere herein. These antigen-specific T cells can be further expanded as described herein and in U.S. patent application Ser. Nos. 10/350,305, 10/187,467, 10/133,236, 09/960,264, and 09/794,230.
- the antigen-specific T cells of the present invention can be used in any number of immunotherapeutic settings as described herein.
- This example shows that the bead:cell ratio can have a profound effect on expansion of different populations of T cells.
- a high bead:cell ratio (3:1-10:1, 20:1 and higher) tends to induce death in antigen-specific T cells while a lower bead:cell ratio (1:1-1:10, 1:20, 1:30, 1:40, 1:50 or lower) leads to expansion of antigen-specific T cells.
- the data described below show that lower bead:cell ratios lead to improved cell expansion in polyclonal cell populations as well.
- this example shows that lower bead:cell ratios improve overall cell expansion.
- the cells and beads are processed over a MaxSep Magnetic Separator in order to remove the beads and any monocytic/phagocytic cells that are attached to the beads.
- a volume containing a total of 5 ⁇ 10 8 CD3 + T-cells is taken and set-up with 1.5 ⁇ 10 9 DYNABEADS® M-450 CD3/CD28 T to initiate the XCELLERATETM process (approx. 3:1 beads to T-cells).
- the mixture of cells and DYNABEADS® M-450 CD3/CD28 T are then incubated at 37° C., 5% CO 2 for approximately 8 days to generate XCELLERATED T-cells for a first infusion.
- the remaining monocyte-depleted PBMC are cryopreserved until a second or further cell product expansion (approximately 21 days later) at which time they are thawed, washed and then a volume containing a total of 5 ⁇ 10 8 CD3 + T-cells is taken and set-up with 1.5 ⁇ 10 9 DYNABEADS® M-450 CD3/CD28 T to initiate the XCELLERATE Process for a second infusion.
- the CD3 + T-cells activate and expand.
- the anti-CD3 mAb used is BC3 (XR-CD3; Fred Hutchinson Cancer Research Center, Seattle, Wash.), and the anti-CD28 mAb (B-T3, XR-CD28) is obtained from Diaclone, Besançon, France.
- the monocyte depleted cells were mixed by rotation for 30 minutes with varying amounts of beads as summarized below in Table 1.
- the beads used in this Example comprised the DYNABEADS® M-450 CD3CD28 T with a 1:1 CD3:CD28 antibody ratio bound on the beads.
- Varying Bead:Cell Ratios can Selectively Expand or Delete Memory CD8 T cells Fold Increase Bead:Cell Ratio Polyclonal T cells
- CMV Antigen-Specific T cells 10:1 149 0 5:1 294 0 3:1 346 1.4 1:1 562 20.6 1:5 113 53 1:10 79 45.8
- stimulation with high bead:cell ratios provides a high concentration of stimulating antibody (i.e., “strong signal”), leading to over-stimulation of antigen-specific T cells, causing them to die, either by apoptosis or other mechanisms.
- strong signal i.e., “strong signal”
- Using lower bead:cell ratios provides a stimulation signal to antigen-specific T cells that does not over-stimulate, but rather induces rapid proliferation of these cells.
- Cells were prepared and stimulated essentially as described in Example 3 with the following modifications: as shown in FIG. 4, panels A and B, cells were cultured either at a starting static culture with a bead:cell ratio of 1:2.5 or 1:5 OR at 1:2.5 or 1:5 starting ratio with additional beads added at day 5, 7, or 9 at 1:10, 1:25, 1:50 or 1:100 ratios as noted.
- a comparison of total T cell expansion over 15 days shows an increase in expansion of cells when beads are added sequentially over culturing time, in cultures with both starting bead:cell ratios of 1:2.5 and 1:5.
- Comparison of CMV-specific T cell expansion over 15 days also shows an increase in expansion of antigen-specific cells when beads are added sequentially during culture (see FIG. 4 panel A and FIG.
- This example describes a model system for assessing CD4 T cell subsets in the Xcellerate® expansion process.
- Toxic Shock Syndrome Toxin is a superantigen that specifically stimulates CD4+ T cells expressing TCR V ⁇ 2.
- PBMC are composed of between 1-25% V ⁇ 2 TCR T cells.
- a CD4 + V ⁇ 2 specific cell line is generated by stimulating PBMC with TSST for 9-14 days until T cells proliferate out of log phase.
- These “antigen experienced” V ⁇ 2 T cells are then mixed back at varying percentages of the total culture (e.g., 1%, 2%) with a V ⁇ 2 depleted na ⁇ ve PBMC culture and stimulated with CD3/CD28 beads at varying bead:cell ratios as described herein in the Xcellerate® process.
- T cell expansion was evaluated using varying concentrations of anti-CD3:anti-CD28 antibody ratios on the 3 ⁇ 28 DYNABEADS® M-450.
- the process referred to as XCELLERATE IITM was used, as described in U.S. patent application Ser. No. 10/187,467. Briefly, this process is similar to XCELLERATE ITM as described in Example 3 with some modifications in which no separate monocyte depletion step was utilized and in certain processes the cells were frozen prior to initial contact with beads and further concentration and stimulation were performed.
- FIG. 6 surprisingly, about a 68-fold expansion after 8 days of culture was observed with an anti-CD3:CD28 ratio of 1:10 antibodies on the beads.
- This example describes the T cells expansion using essentially the Xcellerate II process as described in U.S. Patent Application Nos. 10/350,305; 10/187,467; 10/133,236; 09/960,264; 09/794,230; PCT/US01/06139; and PCT/US02/28161, followed by seeding cells into the Wave Bioreactor.
- CD3 + cells were culture-expanded for ⁇ 2 additional days at which point the contents of the culture bags were then seeded into a 20 L Wave Bioreactor containing a 10 L volume of media. The cells were then cultured at 37° C., 5% CO 2 with the wave motion at 15 rocks/minute and with perfusion at 1 ml/minute.
- the Wave Bioreactor provides an unexpected and dramatic improvement to the expansion process. Furthermore, hitherto unobserved cell densities and final absolute cell yields were achieved using The Wave Bioreactor.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- Medicinal Chemistry (AREA)
- Organic Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Immunology (AREA)
- Biomedical Technology (AREA)
- Oncology (AREA)
- Cell Biology (AREA)
- Microbiology (AREA)
- Communicable Diseases (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Hematology (AREA)
- Biotechnology (AREA)
- Epidemiology (AREA)
- Mycology (AREA)
- Virology (AREA)
- Pulmonology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Gastroenterology & Hepatology (AREA)
- Dermatology (AREA)
- Urology & Nephrology (AREA)
- Tropical Medicine & Parasitology (AREA)
- Diabetes (AREA)
- Neurosurgery (AREA)
- Neurology (AREA)
- Endocrinology (AREA)
Priority Applications (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US10/742,622 US20040224402A1 (en) | 2003-05-08 | 2003-12-19 | Generation and isolation of antigen-specific T cells |
US11/674,304 US20070212767A1 (en) | 2003-05-08 | 2007-02-13 | Generation and isolation of antigen-specific t cells |
US12/251,224 US7977095B2 (en) | 2003-05-08 | 2008-10-14 | Generation and isolation of antigen-specific T cells |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US46912203P | 2003-05-08 | 2003-05-08 | |
US10/742,622 US20040224402A1 (en) | 2003-05-08 | 2003-12-19 | Generation and isolation of antigen-specific T cells |
Related Child Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US11/674,304 Division US20070212767A1 (en) | 2003-05-08 | 2007-02-13 | Generation and isolation of antigen-specific t cells |
Publications (1)
Publication Number | Publication Date |
---|---|
US20040224402A1 true US20040224402A1 (en) | 2004-11-11 |
Family
ID=33476667
Family Applications (3)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US10/742,622 Abandoned US20040224402A1 (en) | 2003-05-08 | 2003-12-19 | Generation and isolation of antigen-specific T cells |
US11/674,304 Abandoned US20070212767A1 (en) | 2003-05-08 | 2007-02-13 | Generation and isolation of antigen-specific t cells |
US12/251,224 Expired - Lifetime US7977095B2 (en) | 2003-05-08 | 2008-10-14 | Generation and isolation of antigen-specific T cells |
Family Applications After (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US11/674,304 Abandoned US20070212767A1 (en) | 2003-05-08 | 2007-02-13 | Generation and isolation of antigen-specific t cells |
US12/251,224 Expired - Lifetime US7977095B2 (en) | 2003-05-08 | 2008-10-14 | Generation and isolation of antigen-specific T cells |
Country Status (11)
Country | Link |
---|---|
US (3) | US20040224402A1 (ja) |
EP (1) | EP1623017B1 (ja) |
JP (2) | JP2006524991A (ja) |
AT (1) | ATE481476T1 (ja) |
AU (1) | AU2003300359A1 (ja) |
CA (1) | CA2525519A1 (ja) |
DE (1) | DE60334250D1 (ja) |
DK (1) | DK1623017T3 (ja) |
MX (1) | MXPA05012080A (ja) |
TW (1) | TW200502391A (ja) |
WO (1) | WO2004104185A1 (ja) |
Cited By (72)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP1879615A2 (en) * | 2005-05-11 | 2008-01-23 | The Trustees Of The University Of Pennsylvania | Methods for the rapid expansion of antigen specific t-cells |
US20080261307A1 (en) * | 2007-03-16 | 2008-10-23 | University Of Ulsan Foundation For Industry Cooperation | Method of isolating and proliferating autologous antigen-specific CD8+ T Cell using anti-4-1BB antibodies |
US20100254958A1 (en) * | 2007-10-24 | 2010-10-07 | Anne Letsch | Antigen-Specific T-Cell Preparations from Bone Marrow |
US20110229448A1 (en) * | 2007-09-20 | 2011-09-22 | St Vincent's Hospital Sydney Limited | method for identifying antigen-specific regulatory t cells |
US20130181333A1 (en) * | 2012-01-17 | 2013-07-18 | Chipmos Technologies Inc. | Semiconductor package structure and manufacturing method thereof |
EP2635305A1 (en) * | 2010-11-02 | 2013-09-11 | Baylor Research Institute | Enhancement of pathogen-specific memory th17 cell responses |
KR101503341B1 (ko) | 2014-03-12 | 2015-03-18 | 국립암센터 | 자가암항원 특이적 cd8+ t 세포의 분리 및 증식방법 |
KR20150141962A (ko) * | 2013-03-14 | 2015-12-21 | 더 존스 홉킨스 유니버시티 | 나노스케일 인공 항원 제시 세포 |
WO2016100977A1 (en) | 2014-12-19 | 2016-06-23 | The Broad Institute Inc. | Methods for profiling the t-cel- receptor repertoire |
WO2016189159A1 (en) * | 2015-05-28 | 2016-12-01 | Ge Healthcare Bio-Sciences Ab | Semi-static cell culture |
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 |
CN107148285A (zh) * | 2014-11-25 | 2017-09-08 | Adc治疗股份有限公司 | 吡咯并苯并二氮杂*‑抗体缀合物 |
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 |
WO2018080997A1 (en) * | 2016-10-24 | 2018-05-03 | Gpb Scientific, Llc | Deterministic lateral displacement in the preparation of cells and compositions for therapeutic uses |
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. | PHARMACEUTICAL METHODS AND COMPOSITIONS FOR THE TREATMENT OF MELANOMA |
WO2019046052A1 (en) * | 2017-09-01 | 2019-03-07 | Gpb Scientific, Llc | METHODS FOR PREPARING THERAPEUTICALLY ACTIVE CELLS USING MICROFLUIDIC |
WO2019094983A1 (en) | 2017-11-13 | 2019-05-16 | The Broad Institute, Inc. | Methods and compositions for treating cancer by targeting the clec2d-klrb1 pathway |
US10294454B2 (en) | 2016-08-24 | 2019-05-21 | General Electric Company | Methods and kits for cell activation |
EP3495382A1 (en) * | 2012-10-11 | 2019-06-12 | UTI Limited Partnership | Methods and compositions for treating multiple sclerosis and related disorders |
US10324011B2 (en) | 2013-03-15 | 2019-06-18 | The Trustees Of Princeton University | Methods and devices for high throughput purification |
CN110241086A (zh) * | 2012-06-11 | 2019-09-17 | 威尔逊沃夫制造公司 | 用于过继细胞疗法的改进的细胞培养方法 |
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 |
CN110785487A (zh) * | 2017-06-22 | 2020-02-11 | Tcer公司 | T细胞扩增方法及用途 |
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 |
CN111107871A (zh) * | 2017-07-21 | 2020-05-05 | 伯克利之光生命科技公司 | 抗原呈递合成表面、共价官能化表面、活化t细胞及其用途 |
CN111182918A (zh) * | 2017-08-04 | 2020-05-19 | 特鲁提诺生物科学公司 | 用于激活免疫细胞的方法 |
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 |
US10774151B2 (en) | 2017-01-06 | 2020-09-15 | Eutilex Co., Ltd. | Anti-human 4-1BB antibodies and uses thereof |
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 |
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 |
US10905773B2 (en) | 2012-10-11 | 2021-02-02 | Uti Limited Partnership | Methods and compositions for treating multiple sclerosis and related disorders |
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 |
US10976232B2 (en) | 2015-08-24 | 2021-04-13 | Gpb Scientific, Inc. | Methods and devices for multi-step cell purification and concentration |
US10988516B2 (en) | 2012-03-26 | 2021-04-27 | Uti Limited Partnership | Methods and compositions for treating inflammation |
US11000596B2 (en) | 2010-11-12 | 2021-05-11 | UTI Limited Parttiership | Compositions and methods for the prevention and treatment of cancer |
US11116796B2 (en) | 2016-12-02 | 2021-09-14 | Taiga Biotechnologies, Inc. | Nanoparticle formulations |
US11142746B2 (en) | 2013-03-15 | 2021-10-12 | University Of Maryland, Baltimore | High efficiency microfluidic purification of stem cells to improve transplants |
US11338024B2 (en) | 2013-11-04 | 2022-05-24 | Uti Limited Partnership | Methods and compositions for sustained immunotherapy |
US11369678B2 (en) | 2008-08-28 | 2022-06-28 | Taiga Biotechnologies, Inc. | Compositions and methods for modulating immune cells |
US11493428B2 (en) | 2013-03-15 | 2022-11-08 | Gpb Scientific, Inc. | On-chip microfluidic processing of particles |
US11667695B2 (en) | 2008-05-16 | 2023-06-06 | Taiga Biotechnologies, Inc. | Antibodies and processes for preparing the same |
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 |
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 |
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) | 2019-10-03 | 2024-05-14 | Dana-Farber Cancer Institute, Inc. | Methods and compositions for the modulation of cell interactions and signaling in the tumor microenvironment |
US11994512B2 (en) | 2018-01-04 | 2024-05-28 | Massachusetts Institute Of Technology | Single-cell genomic methods to generate ex vivo cell systems that recapitulate in vivo biology with improved fidelity |
WO2024124044A1 (en) | 2022-12-07 | 2024-06-13 | The Brigham And Women’S Hospital, Inc. | Compositions and methods targeting sat1 for enhancing anti¬ tumor immunity during tumor progression |
US12011480B2 (en) | 2015-05-06 | 2024-06-18 | Uti Limited Partnership | Nanoparticle compositions for sustained therapy |
US12036240B2 (en) | 2018-06-14 | 2024-07-16 | The Broad Institute, Inc. | Compositions and methods targeting complement component 3 for inhibiting tumor growth |
US12043870B2 (en) | 2017-10-02 | 2024-07-23 | The Broad Institute, Inc. | Methods and compositions for detecting and modulating an immunotherapy resistance gene signature in cancer |
US12049643B2 (en) | 2017-07-14 | 2024-07-30 | The Broad Institute, Inc. | Methods and compositions for modulating cytotoxic lymphocyte activity |
KR102713342B1 (ko) * | 2017-06-22 | 2024-10-02 | 네오갭 테라퓨틱스 아베 | T-세포 증식 방법 및 용도 |
Families Citing this family (41)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CA2423806C (en) * | 2000-09-29 | 2009-12-22 | Molecular Probes, Inc. | Modified carbocyanine dyes and their conjugates |
CA2629306A1 (en) * | 2005-11-23 | 2007-05-31 | Genentech, Inc. | Methods and compositions related to b cell assays |
GB0603081D0 (en) | 2006-02-15 | 2006-03-29 | Dynal Biotech Asa Oslo | Method |
EP2010906B1 (de) | 2006-03-24 | 2014-06-11 | Miltenyi Biotec GmbH | Verwendung des 4-1bb rezeptors zur identifizierung und/oder separation aktivierter regulatorischer th-zellen (treg) |
EP1840569A1 (de) * | 2006-03-28 | 2007-10-03 | Deutsches Rheuma-Forschungszentrum Berlin | Verwendung des 4-1BB Rezeptors zur Identifizierung und/oder Separation aktivierter regulatorischer Th-Zellen (Treg) |
CA2828401C (en) * | 2011-02-28 | 2023-01-10 | Cadila Pharmaceuticals Limited | Therapeutic cancer vaccine |
JP5840857B2 (ja) * | 2011-04-08 | 2016-01-06 | 国立大学法人 東京大学 | 細胞傷害性t細胞誘導用組成物 |
EP2817625A2 (en) | 2012-02-23 | 2014-12-31 | Stage Cell Therapeutics GmbH | Chromatographic isolation of cells and other complex biological materials |
CN102816734A (zh) * | 2012-05-09 | 2012-12-12 | 阮润生 | 一种肿瘤抗原特异性t细胞的获取方法 |
US8921055B2 (en) | 2012-10-30 | 2014-12-30 | Berkeley Lights, Inc. | Detecting cells secreting a protein of interest |
CA2902448C (en) | 2013-03-01 | 2023-04-18 | The United States Of America, As Represented By The Secretary, Department Of Health And Human Services | Methods of producing enriched populations of tumor reactive t cells from peripheral blood |
CN103740643B (zh) * | 2014-01-21 | 2015-09-30 | 山东省齐鲁干细胞工程有限公司 | 一种mhc限制性杀伤t细胞的体外诱导培养方法 |
EP3988920A1 (en) | 2014-04-16 | 2022-04-27 | Juno Therapeutics GmbH | Methods, kits and apparatus for expanding a population of cells |
CN110819595A (zh) * | 2014-04-25 | 2020-02-21 | 蓝鸟生物公司 | 制备过继性细胞疗法的改善方法 |
SG10201809379UA (en) | 2014-04-25 | 2018-11-29 | Bluebird Bio Inc | Mnd promoter chimeric antigen receptors |
US10479975B2 (en) | 2014-06-06 | 2019-11-19 | Bluebird Bio, Inc. | Methods of making T cell compositions |
WO2015191874A1 (en) * | 2014-06-12 | 2015-12-17 | Children's National Medical Center | Generation of broadly-specific, virus-immune cells targeting multiple hiv antigens for preventive and therapeutic use |
AU2015283704A1 (en) | 2014-07-01 | 2016-12-15 | Pfizer Inc. | Bispecific heterodimeric diabodies and uses thereof |
CN106795217B (zh) | 2014-07-24 | 2021-08-06 | 蓝鸟生物公司 | Bcma嵌合抗原受体 |
SG11201704727WA (en) | 2014-12-12 | 2017-07-28 | Bluebird Bio Inc | Bcma chimeric antigen receptors |
EP4248991A3 (en) * | 2015-08-21 | 2024-01-10 | Mayo Foundation for Medical Education and Research | Methods and materials for expanding antigen-specific t cells in culture |
MA44909A (fr) | 2015-09-15 | 2018-07-25 | Acerta Pharma Bv | Association thérapeutique d'un inhibiteur du cd19 et d'un inhibiteur de la btk |
MA45488A (fr) | 2015-10-22 | 2018-08-29 | Juno Therapeutics Gmbh | Procédés, kits et appareil de culture de cellules |
RU2761555C2 (ru) | 2015-10-22 | 2021-12-09 | Джуно Терапьютикс Гмбх | Способы, наборы, средства и устройства для трансдукции |
MA45489A (fr) * | 2015-10-22 | 2018-08-29 | Juno Therapeutics Gmbh | Procédés de culture de cellules, kits et appareil associés |
AU2016344745A1 (en) * | 2015-10-28 | 2018-05-17 | Life Technologies As | Selective expansion of different subpopulations of T cells by the alteration of cell surfacing signals and signal ratio |
US10934525B2 (en) | 2015-10-30 | 2021-03-02 | Children's National Medical Center | Generating HPV antigen-specific cells from a naive T cell population |
US11479755B2 (en) | 2015-12-07 | 2022-10-25 | 2Seventy Bio, Inc. | T cell compositions |
WO2017139065A1 (en) | 2016-02-08 | 2017-08-17 | American Gene Technologies International Inc. | Hiv vaccination and immunotherapy |
WO2017161092A1 (en) * | 2016-03-16 | 2017-09-21 | Neximmune, Inc. | Production of antigen-specific t-cells |
CA3028982A1 (en) * | 2016-07-08 | 2018-01-11 | American Gene Technologies International Inc. | Hiv pre-immunization and immunotherapy |
MX2019005277A (es) | 2016-11-04 | 2019-09-27 | Bluebird Bio Inc | Composiciones de celulas t con car anti-bcma. |
KR101862502B1 (ko) * | 2017-01-02 | 2018-05-29 | 성균관대학교산학협력단 | 재구성 인공 암세포, 이의 제조 방법, 및 이를 포함하는 항암 조성물 |
TW201842335A (zh) | 2017-04-27 | 2018-12-01 | 德商朱諾醫療公司 | 寡聚粒子試劑及其使用方法 |
MA49981A (fr) | 2017-08-09 | 2020-06-17 | Juno Therapeutics Inc | Procédés et compositions de préparation de cellules génétiquement modifiées |
CA3185343A1 (en) * | 2017-09-01 | 2019-03-07 | Lonza Walkersville, Inc. | End-to-end cell therapy automation |
US20210030802A1 (en) * | 2018-04-07 | 2021-02-04 | Constant Biotechnology, Llc | Glucocorticoid-resistant leukocytes and their use in the treatment of cancers and viruses |
EP3876979A4 (en) | 2018-11-08 | 2022-08-24 | NexImmune, Inc. | COMPOSITIONS OF T LYMPHOCYTES WITH ENHANCED PHENOTYPIC PROPERTIES |
EP4034641A4 (en) | 2019-09-26 | 2024-01-03 | NantBio, Inc. | PRIMARY MULTIPLICATION OF T CELLS |
WO2022187319A2 (en) * | 2021-03-02 | 2022-09-09 | University Of Louisville Research Foundation, Inc. | Acoustofluidic separation of cells and particles via acoustic radiation force dynamics |
NL2028681B1 (en) * | 2021-04-02 | 2022-10-17 | Nutcracker Therapeutics Inc | Materials and methods for generating antigen-specific t cells and treating diseases |
Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5595881A (en) * | 1994-08-09 | 1997-01-21 | Anergen, Inc. | Method for the detection of antigen presenting cells |
US5827642A (en) * | 1994-08-31 | 1998-10-27 | Fred Hutchinson Cancer Research Center | Rapid expansion method ("REM") for in vitro propagation of T lymphocytes |
US5858358A (en) * | 1992-04-07 | 1999-01-12 | The United States Of America As Represented By The Secretary Of The Navy | Methods for selectively stimulating proliferation of T cells |
US5874307A (en) * | 1994-06-28 | 1999-02-23 | The Institute Of Physical And Chemical Research | Process for induction culture of cytotoxic T lymphocytes having killing activity against tumor cells |
US6534055B1 (en) * | 1988-11-23 | 2003-03-18 | Genetics Institute, Inc. | Methods for selectively stimulating proliferation of T cells |
US20030124122A1 (en) * | 2000-02-24 | 2003-07-03 | Xcyte Therapies, Inc. | Simultaneous stimulation and concentration of cells |
US20030235908A1 (en) * | 2000-02-24 | 2003-12-25 | Xcyte Therapies, Inc. | Activation and expansion of cells |
US6905874B2 (en) * | 2000-02-24 | 2005-06-14 | Xcyte Therapies, Inc. | Simultaneous stimulation and concentration of cells |
Family Cites Families (79)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5081029A (en) * | 1985-09-25 | 1992-01-14 | Oncogen | Methods of adoptive immunotherapy for treatment of aids |
AU7873187A (en) * | 1986-08-08 | 1988-02-24 | University Of Minnesota | Method of culturing leukocytes |
US5219740A (en) * | 1987-02-13 | 1993-06-15 | Fred Hutchinson Cancer Research Center | Retroviral gene transfer into diploid fibroblasts for gene therapy |
US5057423A (en) * | 1987-12-18 | 1991-10-15 | University Of Pittsburgh | Method for the preparation of pure LAK-active lymphocytes |
US6010902A (en) * | 1988-04-04 | 2000-01-04 | Bristol-Meyers Squibb Company | Antibody heteroconjugates and bispecific antibodies for use in regulation of lymphocyte activity |
US6352694B1 (en) * | 1994-06-03 | 2002-03-05 | Genetics Institute, Inc. | Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells |
US20020076407A1 (en) * | 1988-11-23 | 2002-06-20 | Carl H. June | Method for selectively stimulating proliferation of t cells |
US6905680B2 (en) * | 1988-11-23 | 2005-06-14 | Genetics Institute, Inc. | Methods of treating HIV infected subjects |
US5635697A (en) * | 1989-03-01 | 1997-06-03 | Symbol Technologies, Inc. | Method and apparatus for decoding two-dimensional bar code |
US5763266A (en) * | 1989-06-15 | 1998-06-09 | The Regents Of The University Of Michigan | Methods, compositions and devices for maintaining and growing human stem and/or hematopoietics cells |
DE3923279A1 (de) * | 1989-07-14 | 1990-01-18 | Will W Prof Dr Minuth | Minusheets ist ein neues produkt, um zellen in beliebigen behaeltnissen in hochdifferenzierter form auf einer moeglichst natuerlichen unterlage zu kultivieren |
AU6642390A (en) * | 1989-10-27 | 1991-05-31 | Arch Development Corporation | Methods and compositions for promoting immunopotentiation |
ZA91463B (en) | 1990-01-25 | 1992-09-30 | Bristol Myers Squibb Co | Method of activating cytolytic activity of lymphocytes using anti-cd28 antibody |
US5470730A (en) * | 1990-09-28 | 1995-11-28 | Immunex | Method for producing TH -independent cytotoxic T lymphocytes |
US6197298B1 (en) * | 1991-04-19 | 2001-03-06 | Tanox, Inc. | Modified binding molecules specific for T lymphocytes and their use as in vivo immune modulators in animals |
US6129916A (en) * | 1991-04-19 | 2000-10-10 | Tanox, Inc. | Method of Increasing activation on proliferation of T cells using antibody-microbead conjugates |
GB9120508D0 (en) * | 1991-09-26 | 1991-11-06 | Nycomed As | Diagnostic agents |
GB9125768D0 (en) * | 1991-12-04 | 1992-02-05 | Hale Geoffrey | Therapeutic method |
EP0563485A1 (en) | 1992-03-30 | 1993-10-06 | Schering-Plough | In vitro generation of human dendritic cells and uses thereof |
AU2593192A (en) * | 1992-09-14 | 1994-04-12 | Oystein Fodstad | Detection of specific target cells in specialized or mixed cell population and solutions containing mixed cell populations |
US5837477A (en) * | 1993-01-15 | 1998-11-17 | The United States Of America As Represented By The Department Of Health And Human Services | T cell receptor ligands and methods of using same |
AU6827094A (en) * | 1993-05-07 | 1994-12-12 | Immunex Corporation | Cytokine designated 4-1bb ligand and human receptor that binds thereto |
US7211259B1 (en) * | 1993-05-07 | 2007-05-01 | Immunex Corporation | 4-1BB polypeptides and DNA encoding 4-1BB polypeptides |
ES2240962T3 (es) * | 1993-06-04 | 2005-10-16 | The United States Of America As Represented By The Secretary Of The Navy | Metodo para estimular selectivamente la proliferacion de celulas t. |
US5942607A (en) * | 1993-07-26 | 1999-08-24 | Dana-Farber Cancer Institute | B7-2: a CTLA4/CD28 ligand |
EP0938893B1 (en) * | 1993-08-10 | 2004-08-25 | W.L. Gore & Associates, Inc. | Cell encapsulating device |
US5672505A (en) * | 1993-09-27 | 1997-09-30 | Becton, Dickinson And Company | Insert for a issue culture vessel |
FR2717080B1 (fr) * | 1994-03-09 | 1996-12-13 | Synthelabo | Utilisation de l'éliprodil et de ses énantiomères pour la préparation de médicaments utiles dans le traitement des neuropathies périphériques et des maladies neurodégénératives centrales. |
EP0774252A4 (en) * | 1994-05-06 | 2000-04-26 | Kanebo Ltd | CYTOKIN POTENTIATOR AND MEDICINE AGAINST DISEAS IN WHICH THE CYTOKIN ACTIVITY IS REDUCED |
GB9416657D0 (en) * | 1994-08-17 | 1994-10-12 | Biocine Spa | T cell activation |
FR2729570A1 (fr) * | 1995-01-24 | 1996-07-26 | Idm Immuno Designed Molecules | Procede de preparation de macrophages actives, trousses et compositions pour la mise en oeuvre de ce procede |
US5985653A (en) * | 1995-06-07 | 1999-11-16 | Aastrom Biosciences, Inc. | Incubator apparatus for use in a system for maintaining and growing biological cells |
US6096532A (en) * | 1995-06-07 | 2000-08-01 | Aastrom Biosciences, Inc. | Processor apparatus for use in a system for maintaining and growing biological cells |
US5626561A (en) * | 1995-06-07 | 1997-05-06 | Gore Hybrid Technologies, Inc. | Implantable containment apparatus for a therapeutical device and method for loading and reloading the device therein |
DE69630266T2 (de) * | 1995-06-07 | 2004-09-09 | W.L. Gore & Associates, Inc., Newark | Implantierbare aufnahmevorrichtung für ein therapeutisches gerät |
WO1997001304A1 (en) | 1995-06-29 | 1997-01-16 | Mallinckrodt Medical, Inc. | Radiolabeled apatite particles containing a paramagnetic ion |
CA2227327A1 (en) | 1995-07-25 | 1997-02-13 | Celltherapy, Inc. | Autologous immune cell therapy: cell compositions, methods and applications to treatment of human disease |
US20020182730A1 (en) * | 1995-07-26 | 2002-12-05 | Micheal L. Gruenberg | Autologous immune cell therapy: cell compositions, methods and applications to treatment of human disease |
US5627070A (en) | 1995-07-26 | 1997-05-06 | Celltherapy, Inc. | Cell growing device for in vitro cell population expansion |
WO1997010807A1 (en) * | 1995-09-22 | 1997-03-27 | Gore Hybrid Technologies, Inc. | Improved cell encapsulation device |
WO1997022349A1 (en) * | 1995-12-20 | 1997-06-26 | The Board Of Trustees Of The Leland Stanford Junior University | Methods for in vivo t cell activation by antigen-pulsed dendritic cells |
PT879282E (pt) * | 1996-01-17 | 2003-11-28 | Imp College Innovations Ltd | Imunoterapia utilizando linfocitos t citotoxicos (ctl) |
DE69739951D1 (de) * | 1996-03-04 | 2010-09-16 | Calyx Bio Ventures Inc | Modifizierte schnellvermehrungsmethode ('modified-rem') zur in vitro vermehrung von t-lymphozyten |
DE19608753C1 (de) * | 1996-03-06 | 1997-06-26 | Medigene Gmbh | Transduktionssystem und seine Verwendung |
US5972721A (en) * | 1996-03-14 | 1999-10-26 | The United States Of America As Represented By The Secretary Of The Air Force | Immunomagnetic assay system for clinical diagnosis and other purposes |
KR20000064752A (ko) | 1996-03-22 | 2000-11-06 | 더 제네랄 호스피탈 코포레이션 | 중추신경계허혈또는외상의발현후폴리펩티드성장인자를투여하는방법 |
AU3036597A (en) | 1996-05-21 | 1997-12-09 | Institut National De La Sante Et De La Recherche Medicale (Inserm) | Method for using peptide complexes/major histocompatibility complex to obtain or purify antigen-specific t cells and to stimulate t cells |
US5962319A (en) * | 1997-05-19 | 1999-10-05 | Bml, Inc. | Human-Th1-specific protein, gene encoding the protein, transformants, recombinant vectors, and antibodies related to the gene |
US20010031253A1 (en) * | 1996-07-24 | 2001-10-18 | Gruenberg Micheal L. | Autologous immune cell therapy: cell compositions, methods and applications to treatment of human disease |
US5962318A (en) * | 1996-11-15 | 1999-10-05 | St. Jude Children's Research Hospital | Cytotoxic T lymphocyte-mediated immunotherapy |
US5766944A (en) * | 1996-12-31 | 1998-06-16 | Ruiz; Margaret Eileen | T cell differentiation of CD34+ stem cells in cultured thymic epithelial fragments |
JP3531712B2 (ja) * | 1997-06-04 | 2004-05-31 | トヨタ自動車株式会社 | 車両用操舵装置 |
DE69840600D1 (de) * | 1997-09-16 | 2009-04-09 | Univ Oregon Health & Science | Rekombinante mhc-moleküle welche nützlich sind für die manipulation von antigen-spezifischen t-zellen |
US6225118B1 (en) * | 1997-10-01 | 2001-05-01 | Biocure Limited | Multicellular in vitro assay of angiogenesis |
DE69840739D1 (de) * | 1997-10-27 | 2009-05-28 | Merix Bioscience Inc | Methode und Zusammensetzung zur Herstellung von reifen dendritischen Zellen |
EP1030674A1 (en) * | 1997-11-10 | 2000-08-30 | Arch Development Corporation | METHODS FOR TREATMENT OF TUMORS AND TUMOR CELLS USING $i(EX VIVO) ACTIVATED T CELLS |
EP1469883A2 (en) * | 1998-02-19 | 2004-10-27 | Xcyte Therapies, Inc. | Compositions and methods for regulating lymphocyte activation |
JP4601166B2 (ja) * | 1998-05-11 | 2010-12-22 | ミルテニィ バイオテック ゲーエムベーハー | 抗原特異的t細胞の直接的選択方法 |
FR2780891A1 (fr) | 1998-07-10 | 2000-01-14 | Univ Paris Curie | Echange de lymphocytes t |
WO2000023053A2 (en) * | 1998-10-20 | 2000-04-27 | Salvatore Albani | Artificial antigen-specific cells and related methods |
JP4688254B2 (ja) * | 1999-02-09 | 2011-05-25 | 独立行政法人理化学研究所 | 腫瘍ワクチン |
US6797514B2 (en) * | 2000-02-24 | 2004-09-28 | Xcyte Therapies, Inc. | Simultaneous stimulation and concentration of cells |
US20030119185A1 (en) * | 2000-02-24 | 2003-06-26 | Xcyte Therapies, Inc. | Activation and expansion of cells |
ATE371722T1 (de) | 2000-05-11 | 2007-09-15 | Baylor Res Inst | Zusammensetzungen und verfahren zur herstellung von antigen-präsentierenden zellen |
AUPR011700A0 (en) * | 2000-09-14 | 2000-10-05 | Austin Research Institute, The | Composition comprising immunogenic virus sized particles (VSP) |
CA2442682C (en) * | 2001-03-30 | 2011-08-02 | Greenville Hospital System | Monocyte-specific particulate delivery vehicle |
US20030082806A1 (en) * | 2001-04-27 | 2003-05-01 | Xcyte Therapies, Inc. | Maturation of antigen-presenting cells using activated T cells |
EP1390065A2 (en) * | 2001-05-14 | 2004-02-25 | Duotol Ab | Methods for promoting antigen presentation and modulating immune responses using cholera toxin and its b subunit |
JP2004528042A (ja) | 2001-06-01 | 2004-09-16 | エクサイト セラピーズ, インコーポレイテッド | T細胞誘導性組織修復および再生 |
DE10129912A1 (de) | 2001-06-21 | 2003-01-02 | Efmt Entwicklungs Und Forschun | Nadelelektrode |
WO2003025158A1 (en) | 2001-09-14 | 2003-03-27 | Medcell Biologics, Inc. | Oxygen enriched bioreactor and method of culturing cells |
US20030134415A1 (en) | 2001-09-19 | 2003-07-17 | Gruenberg Micheal L. | Th1 cell adoptive immunotherapy |
WO2003024312A2 (en) | 2001-09-17 | 2003-03-27 | Valeocyte Therapies Llc | Cell therapy system |
US20030194395A1 (en) * | 2001-09-17 | 2003-10-16 | Gruenberg Micheal L. | Th1 cell adoptive immunotherapy |
US20030134341A1 (en) * | 2001-09-19 | 2003-07-17 | Medcell Biologics, Llc. | Th1 cell adoptive immunotherapy |
WO2003043643A1 (en) | 2001-11-19 | 2003-05-30 | Valeocyte Therapies Llc | Closed sterile system devices and methods |
WO2003057171A2 (en) * | 2002-01-03 | 2003-07-17 | The Trustees Of The University Of Pennsylvania | Activation and expansion of t-cells using an engineered multivalent signaling platform |
US20030170238A1 (en) * | 2002-03-07 | 2003-09-11 | Gruenberg Micheal L. | Re-activated T-cells for adoptive immunotherapy |
US20030175272A1 (en) * | 2002-03-07 | 2003-09-18 | Medcell Biologics, Inc. | Re-activated T-cells for adoptive immunotherapy |
-
2003
- 2003-12-19 WO PCT/US2003/041212 patent/WO2004104185A1/en active Application Filing
- 2003-12-19 MX MXPA05012080A patent/MXPA05012080A/es unknown
- 2003-12-19 DK DK03817044.5T patent/DK1623017T3/da active
- 2003-12-19 DE DE60334250T patent/DE60334250D1/de not_active Expired - Lifetime
- 2003-12-19 EP EP03817044A patent/EP1623017B1/en not_active Expired - Lifetime
- 2003-12-19 CA CA002525519A patent/CA2525519A1/en not_active Abandoned
- 2003-12-19 AU AU2003300359A patent/AU2003300359A1/en not_active Abandoned
- 2003-12-19 TW TW092136274A patent/TW200502391A/zh unknown
- 2003-12-19 AT AT03817044T patent/ATE481476T1/de not_active IP Right Cessation
- 2003-12-19 US US10/742,622 patent/US20040224402A1/en not_active Abandoned
- 2003-12-19 JP JP2005500047A patent/JP2006524991A/ja active Pending
-
2007
- 2007-02-13 US US11/674,304 patent/US20070212767A1/en not_active Abandoned
-
2008
- 2008-10-14 US US12/251,224 patent/US7977095B2/en not_active Expired - Lifetime
-
2010
- 2010-10-06 JP JP2010226114A patent/JP2011036263A/ja active Pending
Patent Citations (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6534055B1 (en) * | 1988-11-23 | 2003-03-18 | Genetics Institute, Inc. | Methods for selectively stimulating proliferation of T cells |
US5858358A (en) * | 1992-04-07 | 1999-01-12 | The United States Of America As Represented By The Secretary Of The Navy | Methods for selectively stimulating proliferation of T cells |
US5874307A (en) * | 1994-06-28 | 1999-02-23 | The Institute Of Physical And Chemical Research | Process for induction culture of cytotoxic T lymphocytes having killing activity against tumor cells |
US5595881A (en) * | 1994-08-09 | 1997-01-21 | Anergen, Inc. | Method for the detection of antigen presenting cells |
US5827642A (en) * | 1994-08-31 | 1998-10-27 | Fred Hutchinson Cancer Research Center | Rapid expansion method ("REM") for in vitro propagation of T lymphocytes |
US20030124122A1 (en) * | 2000-02-24 | 2003-07-03 | Xcyte Therapies, Inc. | Simultaneous stimulation and concentration of cells |
US20030235908A1 (en) * | 2000-02-24 | 2003-12-25 | Xcyte Therapies, Inc. | Activation and expansion of cells |
US6867041B2 (en) * | 2000-02-24 | 2005-03-15 | Xcyte Therapies, Inc. | Simultaneous stimulation and concentration of cells |
US6905874B2 (en) * | 2000-02-24 | 2005-06-14 | Xcyte Therapies, Inc. | Simultaneous stimulation and concentration of cells |
Cited By (103)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP1879615A4 (en) * | 2005-05-11 | 2009-05-13 | Univ Pennsylvania | PROCESS FOR THE FAST EXPANSION OF ANTIGEN-SPECIFIC T CELLS |
EP1879615A2 (en) * | 2005-05-11 | 2008-01-23 | The Trustees Of The University Of Pennsylvania | Methods for the rapid expansion of antigen specific t-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 |
US20080261307A1 (en) * | 2007-03-16 | 2008-10-23 | University Of Ulsan Foundation For Industry Cooperation | Method of isolating and proliferating autologous antigen-specific CD8+ T Cell using anti-4-1BB antibodies |
KR100882445B1 (ko) | 2007-03-16 | 2009-02-09 | 울산대학교 산학협력단 | 항―4-1bb 항체를 이용한 항원 특이적 자가유래cd8+t 세포의 분리 및 증식 방법 |
US7932045B2 (en) * | 2007-03-16 | 2011-04-26 | University Of Ulsan Foundation For Industry Cooperation | Method of isolating and proliferating autologous antigen-specific CD8+ T cell using anti-4-1BB antibodies |
US20110229448A1 (en) * | 2007-09-20 | 2011-09-22 | St Vincent's Hospital Sydney Limited | method for identifying antigen-specific regulatory t cells |
US8975069B2 (en) * | 2007-09-20 | 2015-03-10 | St. Vincent's Hospital Sydney Limited | Method for identifying antigen-specific regulatory T cells |
US20100254958A1 (en) * | 2007-10-24 | 2010-10-07 | Anne Letsch | Antigen-Specific T-Cell Preparations from Bone Marrow |
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 |
EP2635305A1 (en) * | 2010-11-02 | 2013-09-11 | Baylor Research Institute | Enhancement of pathogen-specific memory th17 cell responses |
EP2635305A4 (en) * | 2010-11-02 | 2014-05-28 | Baylor Res Inst | IMPROVEMENT OF PATHOGENIC TH17 MEMORY LANGUAGES |
US11000596B2 (en) | 2010-11-12 | 2021-05-11 | UTI Limited Parttiership | Compositions and methods for the prevention and treatment of cancer |
US20130181333A1 (en) * | 2012-01-17 | 2013-07-18 | Chipmos Technologies Inc. | Semiconductor package structure and manufacturing method thereof |
US9053968B2 (en) * | 2012-01-17 | 2015-06-09 | Chipmos Technologies Inc. | Semiconductor package structure and manufacturing method thereof |
US10988516B2 (en) | 2012-03-26 | 2021-04-27 | Uti Limited Partnership | Methods and compositions for treating inflammation |
CN110241086A (zh) * | 2012-06-11 | 2019-09-17 | 威尔逊沃夫制造公司 | 用于过继细胞疗法的改进的细胞培养方法 |
US10953048B2 (en) | 2012-07-20 | 2021-03-23 | Taiga Biotechnologies, Inc. | Enhanced reconstitution and autoreconstitution of the hematopoietic compartment |
EP3495382A1 (en) * | 2012-10-11 | 2019-06-12 | UTI Limited Partnership | Methods and compositions for treating multiple sclerosis and related disorders |
US10905773B2 (en) | 2012-10-11 | 2021-02-02 | Uti Limited Partnership | Methods and compositions for treating multiple sclerosis and related disorders |
US10786534B2 (en) | 2013-03-11 | 2020-09-29 | Taiga Biotechnologies, Inc. | Production and use of red blood cells |
US11939595B2 (en) | 2013-03-14 | 2024-03-26 | The Johns Hopkins University | Nanoscale artificial antigen presenting cells |
KR20150141962A (ko) * | 2013-03-14 | 2015-12-21 | 더 존스 홉킨스 유니버시티 | 나노스케일 인공 항원 제시 세포 |
KR102164405B1 (ko) * | 2013-03-14 | 2020-10-12 | 더 존스 홉킨스 유니버시티 | 나노스케일 인공 항원 제시 세포 |
US11493428B2 (en) | 2013-03-15 | 2022-11-08 | Gpb Scientific, Inc. | On-chip microfluidic processing of particles |
US11486802B2 (en) | 2013-03-15 | 2022-11-01 | University Of Maryland, Baltimore | Methods and devices for high throughput purification |
US10324011B2 (en) | 2013-03-15 | 2019-06-18 | The Trustees Of Princeton University | Methods and devices for high throughput purification |
US11142746B2 (en) | 2013-03-15 | 2021-10-12 | University Of Maryland, Baltimore | High efficiency microfluidic purification of stem cells to improve transplants |
US10852220B2 (en) | 2013-03-15 | 2020-12-01 | The Trustees Of Princeton University | Methods and devices for high throughput purification |
US11338024B2 (en) | 2013-11-04 | 2022-05-24 | Uti Limited Partnership | Methods and compositions for sustained immunotherapy |
KR101503341B1 (ko) | 2014-03-12 | 2015-03-18 | 국립암센터 | 자가암항원 특이적 cd8+ t 세포의 분리 및 증식방법 |
US10570371B2 (en) | 2014-03-12 | 2020-02-25 | Eutilex Co., Ltd. | Methods for isolating and proliferating autologous cancer antigen-specific CD8+T cells |
US10801011B2 (en) | 2014-03-12 | 2020-10-13 | National Cancer Center | Methods for isolating and proliferating autologous cancer antigen-specific CD8+ T cells |
CN105473731A (zh) * | 2014-03-12 | 2016-04-06 | 国立癌Center | 自身癌抗原特异性cd8+t细胞的分离及增殖方法 |
WO2015137724A1 (ko) * | 2014-03-12 | 2015-09-17 | 국립암센터 | 자가암항원 특이적 cd8+ t 세포의 분리 및 증식방법 |
CN107148285A (zh) * | 2014-11-25 | 2017-09-08 | Adc治疗股份有限公司 | 吡咯并苯并二氮杂*‑抗体缀合物 |
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 |
US12011480B2 (en) | 2015-05-06 | 2024-06-18 | Uti Limited Partnership | Nanoparticle compositions for sustained therapy |
US10927344B2 (en) | 2015-05-28 | 2021-02-23 | Cytiva Sweden Ab | Semi-static cell culture |
WO2016189159A1 (en) * | 2015-05-28 | 2016-12-01 | Ge Healthcare Bio-Sciences Ab | Semi-static cell culture |
US10976232B2 (en) | 2015-08-24 | 2021-04-13 | Gpb Scientific, Inc. | Methods and devices for multi-step cell purification and concentration |
WO2017069958A2 (en) | 2015-10-09 | 2017-04-27 | The Brigham And Women's Hospital, Inc. | Modulation of novel immune checkpoint targets |
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 |
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 |
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 |
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 |
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 |
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 |
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 |
US11512288B2 (en) | 2016-08-24 | 2022-11-29 | Global Life Sciences Solutions Usa Llc | Methods and kits for cell activation |
US10294454B2 (en) | 2016-08-24 | 2019-05-21 | General Electric Company | Methods and kits for cell activation |
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 |
CN110381961A (zh) * | 2016-10-24 | 2019-10-25 | Gpb科学有限责任公司 | 用于治疗用途的细胞和组合物的制备中的确定性侧向位移 |
WO2018080997A1 (en) * | 2016-10-24 | 2018-05-03 | Gpb Scientific, Llc | Deterministic lateral displacement in the preparation of cells and compositions for therapeutic uses |
US11116796B2 (en) | 2016-12-02 | 2021-09-14 | Taiga Biotechnologies, Inc. | Nanoparticle formulations |
US11859004B2 (en) | 2017-01-06 | 2024-01-02 | Eutilex Co., Ltd. | Anti-human 4-1BB antibodies and uses thereof |
US10774151B2 (en) | 2017-01-06 | 2020-09-15 | Eutilex Co., Ltd. | Anti-human 4-1BB antibodies and uses thereof |
US10919972B2 (en) | 2017-01-06 | 2021-02-16 | Eutilex Co., Ltd. | Anti-human 4-1BB antibodies 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 |
KR102713342B1 (ko) * | 2017-06-22 | 2024-10-02 | 네오갭 테라퓨틱스 아베 | T-세포 증식 방법 및 용도 |
CN110785487A (zh) * | 2017-06-22 | 2020-02-11 | Tcer公司 | T细胞扩增方法及用途 |
US12049643B2 (en) | 2017-07-14 | 2024-07-30 | The Broad Institute, Inc. | Methods and compositions for modulating cytotoxic lymphocyte activity |
CN111107871A (zh) * | 2017-07-21 | 2020-05-05 | 伯克利之光生命科技公司 | 抗原呈递合成表面、共价官能化表面、活化t细胞及其用途 |
US10149898B2 (en) | 2017-08-03 | 2018-12-11 | Taiga Biotechnologies, Inc. | Methods and compositions for the treatment of melanoma |
US10864259B2 (en) | 2017-08-03 | 2020-12-15 | 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 |
WO2019027465A1 (en) | 2017-08-03 | 2019-02-07 | Taiga Biotechnologies, Inc. | PHARMACEUTICAL METHODS AND COMPOSITIONS FOR THE TREATMENT OF MELANOMA |
CN111182918A (zh) * | 2017-08-04 | 2020-05-19 | 特鲁提诺生物科学公司 | 用于激活免疫细胞的方法 |
US11149251B2 (en) | 2017-09-01 | 2021-10-19 | Gpb Scientific, Inc. | Methods for preparing therapeutically active cells using microfluidics |
US11306288B2 (en) | 2017-09-01 | 2022-04-19 | Gpb Scientific, Inc. | Methods for preparing therapeutically active cells using microfluidics |
WO2019046052A1 (en) * | 2017-09-01 | 2019-03-07 | Gpb Scientific, Llc | METHODS FOR PREPARING THERAPEUTICALLY ACTIVE CELLS USING MICROFLUIDIC |
US10844353B2 (en) | 2017-09-01 | 2020-11-24 | Gpb Scientific, Inc. | Methods for preparing therapeutically active cells using microfluidics |
US10988734B2 (en) | 2017-09-01 | 2021-04-27 | Gpb Scientific, Inc. | Methods for preparing therapeutically active cells using microfluidics |
US12043870B2 (en) | 2017-10-02 | 2024-07-23 | The Broad Institute, Inc. | Methods and compositions for detecting and modulating an immunotherapy resistance gene signature in cancer |
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 |
US12018080B2 (en) | 2017-11-13 | 2024-06-25 | The Broad Institute, Inc. | Methods and compositions for treating cancer by targeting the CLEC2D-KLRB1 pathway |
WO2019094983A1 (en) | 2017-11-13 | 2019-05-16 | The Broad Institute, Inc. | Methods and compositions for treating cancer by targeting the clec2d-klrb1 pathway |
US11994512B2 (en) | 2018-01-04 | 2024-05-28 | Massachusetts Institute Of Technology | Single-cell genomic methods to generate ex vivo cell systems that recapitulate in vivo biology with improved fidelity |
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 |
US12036240B2 (en) | 2018-06-14 | 2024-07-16 | The Broad Institute, Inc. | Compositions and methods targeting complement component 3 for inhibiting tumor growth |
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 |
WO2020131586A2 (en) | 2018-12-17 | 2020-06-25 | The Broad Institute, Inc. | Methods for identifying neoantigens |
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 |
US11981922B2 (en) | 2019-10-03 | 2024-05-14 | Dana-Farber Cancer Institute, Inc. | Methods and compositions for the modulation of cell interactions and signaling in the tumor microenvironment |
US11793787B2 (en) | 2019-10-07 | 2023-10-24 | The Broad Institute, Inc. | Methods and compositions for enhancing anti-tumor immunity by targeting steroidogenesis |
US11865168B2 (en) | 2019-12-30 | 2024-01-09 | Massachusetts Institute Of Technology | Compositions and methods for treating bacterial infections |
WO2024124044A1 (en) | 2022-12-07 | 2024-06-13 | The Brigham And Women’S Hospital, Inc. | Compositions and methods targeting sat1 for enhancing anti¬ tumor immunity during tumor progression |
Also Published As
Publication number | Publication date |
---|---|
US20090137017A1 (en) | 2009-05-28 |
JP2011036263A (ja) | 2011-02-24 |
DE60334250D1 (de) | 2010-10-28 |
JP2006524991A (ja) | 2006-11-09 |
TW200502391A (en) | 2005-01-16 |
WO2004104185A1 (en) | 2004-12-02 |
MXPA05012080A (es) | 2006-02-22 |
EP1623017A4 (en) | 2006-07-05 |
AU2003300359A1 (en) | 2004-12-13 |
US7977095B2 (en) | 2011-07-12 |
EP1623017A1 (en) | 2006-02-08 |
US20070212767A1 (en) | 2007-09-13 |
ATE481476T1 (de) | 2010-10-15 |
DK1623017T3 (da) | 2011-01-10 |
EP1623017B1 (en) | 2010-09-15 |
CA2525519A1 (en) | 2004-12-02 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US7977095B2 (en) | Generation and isolation of antigen-specific T cells | |
US20200071663A1 (en) | Methods for eliminating at least a substantial portion of a clonal antigen-specific memory t cell subpopulation | |
US20030082806A1 (en) | Maturation of antigen-presenting cells using activated T cells | |
US8637307B2 (en) | Activation and expansion of T-cells using an engineered multivalent signaling platform as a research tool | |
US7638326B2 (en) | Activation and expansion of T-cells using an engineered multivalent signaling platform | |
US7435592B2 (en) | Compositions for allogeneic cell therapy | |
US20070274974A1 (en) | Compositions and methods for restoring immune responsiveness in patients with immunological defects | |
JP2006516197A (ja) | 細胞の活性化および拡大の方法 | |
WO2006133398A2 (en) | In vitro activated donor t-cells to promote transplant engraftment | |
US20080267972A1 (en) | Donor Lymphocyte Infusion of T Cells For the Treatment of Cancer | |
US20040175373A1 (en) | Compositions and methods for eliminating undesired subpopulations of T cells in patients with immunological defects related to autoimmunity and organ or hematopoietic stem cell transplantation | |
AU2002305287A1 (en) | Maturation of antigen-presenting cells using activated T cells |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AS | Assignment |
Owner name: XCYTE THERAPIES, INC., WASHINGTON Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BONYAHADI, MARK;KALAMASZ, DALE;REEL/FRAME:014684/0971;SIGNING DATES FROM 20040426 TO 20040428 |
|
AS | Assignment |
Owner name: XCYTE THERAPIES, INC., WASHINGTON Free format text: ARTICLES OF INCORPORATION;ASSIGNOR:XCYTE THERAPIES, INC.;REEL/FRAME:016536/0334 Effective date: 20040319 |
|
AS | Assignment |
Owner name: INVITROGEN CORPORATION, CALIFORNIA Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:XCYTE THERAPIES, INC.;REEL/FRAME:018303/0117 Effective date: 20051214 |
|
STCB | Information on status: application discontinuation |
Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION |
|
AS | Assignment |
Owner name: LIFE TECHNOLOGIES CORPORATION,CALIFORNIA Free format text: MERGER;ASSIGNOR:INVITROGEN CORPORATION;REEL/FRAME:023882/0551 Effective date: 20081121 Owner name: LIFE TECHNOLOGIES CORPORATION, CALIFORNIA Free format text: MERGER;ASSIGNOR:INVITROGEN CORPORATION;REEL/FRAME:023882/0551 Effective date: 20081121 |
|
AS | Assignment |
Owner name: LIFE TECHNOLOGIES CORPORATION, CALIFORNIA Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE APPLICATION NO 09452626 PREVIOUSLY RECORDED ON REEL 023882 FRAME 0551. ASSIGNOR(S) HEREBY CONFIRMS THE MERGER SHOULD NOT HAVE BEEN RECORDED AGAINST THIS PATENT APPLICATION NUMBER;ASSIGNOR:INVITROGEN CORPORATION;REEL/FRAME:034217/0490 Effective date: 20081121 |