CA2431080A1 - Enhancement of anticancer immunity through inhibition of arginase - Google Patents
Enhancement of anticancer immunity through inhibition of arginase Download PDFInfo
- Publication number
- CA2431080A1 CA2431080A1 CA002431080A CA2431080A CA2431080A1 CA 2431080 A1 CA2431080 A1 CA 2431080A1 CA 002431080 A CA002431080 A CA 002431080A CA 2431080 A CA2431080 A CA 2431080A CA 2431080 A1 CA2431080 A1 CA 2431080A1
- Authority
- CA
- Canada
- Prior art keywords
- arginase
- tumor
- dendritic cell
- cells
- inhibition
- 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
- 102000004452 Arginase Human genes 0.000 title claims abstract description 81
- 108700024123 Arginases Proteins 0.000 title claims abstract description 81
- 230000005764 inhibitory process Effects 0.000 title claims description 59
- 230000005904 anticancer immunity Effects 0.000 title 1
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 116
- 210000004443 dendritic cell Anatomy 0.000 claims abstract description 116
- 201000011510 cancer Diseases 0.000 claims abstract description 30
- 239000000427 antigen Substances 0.000 claims abstract description 12
- 108091007433 antigens Proteins 0.000 claims abstract description 12
- 102000036639 antigens Human genes 0.000 claims abstract description 12
- 230000037361 pathway Effects 0.000 claims abstract description 11
- 230000006028 immune-suppresssive effect Effects 0.000 claims abstract description 7
- 230000000903 blocking effect Effects 0.000 claims abstract description 6
- 241000124008 Mammalia Species 0.000 claims abstract description 3
- 230000036039 immunity Effects 0.000 claims abstract description 3
- 210000004027 cell Anatomy 0.000 claims description 33
- 230000000694 effects Effects 0.000 claims description 17
- 230000001629 suppression Effects 0.000 claims description 17
- 230000006907 apoptotic process Effects 0.000 claims description 14
- 238000000034 method Methods 0.000 claims description 9
- 229940022399 cancer vaccine Drugs 0.000 claims description 8
- 238000009566 cancer vaccine Methods 0.000 claims description 8
- 229940080328 Arginase inhibitor Drugs 0.000 claims description 7
- 239000003112 inhibitor Substances 0.000 claims description 6
- 210000001519 tissue Anatomy 0.000 claims description 6
- 229960005486 vaccine Drugs 0.000 claims description 6
- VLCYCQAOQCDTCN-UHFFFAOYSA-N eflornithine Chemical compound NCCCC(N)(C(F)F)C(O)=O VLCYCQAOQCDTCN-UHFFFAOYSA-N 0.000 claims description 5
- 229920000768 polyamine Polymers 0.000 claims description 5
- 239000004475 Arginine Substances 0.000 claims description 4
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 claims description 4
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 claims description 4
- 210000001185 bone marrow Anatomy 0.000 claims description 4
- 239000003550 marker Substances 0.000 claims description 4
- 108090000978 Interleukin-4 Proteins 0.000 claims description 3
- 102000004388 Interleukin-4 Human genes 0.000 claims description 3
- 108020004459 Small interfering RNA Proteins 0.000 claims description 3
- 108090000623 proteins and genes Proteins 0.000 claims description 3
- 108010071942 Colony-Stimulating Factors Proteins 0.000 claims description 2
- 229940079593 drug Drugs 0.000 claims description 2
- 239000003814 drug Substances 0.000 claims description 2
- 239000000463 material Substances 0.000 claims 4
- 239000000203 mixture Substances 0.000 claims 4
- 229940030156 cell vaccine Drugs 0.000 claims 3
- 102100021079 Ornithine decarboxylase Human genes 0.000 claims 2
- 239000004055 small Interfering RNA Substances 0.000 claims 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 claims 1
- 102100024003 Arf-GAP with SH3 domain, ANK repeat and PH domain-containing protein 1 Human genes 0.000 claims 1
- 102100035526 B melanoma antigen 1 Human genes 0.000 claims 1
- 108010010803 Gelatin Proteins 0.000 claims 1
- 101000874316 Homo sapiens B melanoma antigen 1 Proteins 0.000 claims 1
- 101000914324 Homo sapiens Carcinoembryonic antigen-related cell adhesion molecule 5 Proteins 0.000 claims 1
- 101000914321 Homo sapiens Carcinoembryonic antigen-related cell adhesion molecule 7 Proteins 0.000 claims 1
- 101000617725 Homo sapiens Pregnancy-specific beta-1-glycoprotein 2 Proteins 0.000 claims 1
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 claims 1
- 102100022297 Integrin alpha-X Human genes 0.000 claims 1
- 102000000440 Melanoma-associated antigen Human genes 0.000 claims 1
- 108050008953 Melanoma-associated antigen Proteins 0.000 claims 1
- 102100034256 Mucin-1 Human genes 0.000 claims 1
- 108010008707 Mucin-1 Proteins 0.000 claims 1
- 108091034117 Oligonucleotide Proteins 0.000 claims 1
- 102100021768 Phosphoserine aminotransferase Human genes 0.000 claims 1
- 229920000954 Polyglycolide Polymers 0.000 claims 1
- 102100022019 Pregnancy-specific beta-1-glycoprotein 2 Human genes 0.000 claims 1
- 108010072866 Prostate-Specific Antigen Proteins 0.000 claims 1
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 claims 1
- 102100039094 Tyrosinase Human genes 0.000 claims 1
- 108060008724 Tyrosinase Proteins 0.000 claims 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 claims 1
- 239000004744 fabric Substances 0.000 claims 1
- 229920000159 gelatin Polymers 0.000 claims 1
- 239000008273 gelatin Substances 0.000 claims 1
- 235000019322 gelatine Nutrition 0.000 claims 1
- 235000011852 gelatine desserts Nutrition 0.000 claims 1
- 238000011534 incubation Methods 0.000 claims 1
- 229940028885 interleukin-4 Drugs 0.000 claims 1
- 238000012737 microarray-based gene expression Methods 0.000 claims 1
- 238000012243 multiplex automated genomic engineering Methods 0.000 claims 1
- 239000013612 plasmid Substances 0.000 claims 1
- 229920001432 poly(L-lactide) Polymers 0.000 claims 1
- 229920001420 poly(caprolactone-co-lactic acid) Polymers 0.000 claims 1
- 239000004633 polyglycolic acid Substances 0.000 claims 1
- 108040000983 polyphosphate:AMP phosphotransferase activity proteins Proteins 0.000 claims 1
- 230000009290 primary effect Effects 0.000 claims 1
- 230000009291 secondary effect Effects 0.000 claims 1
- 230000004936 stimulating effect Effects 0.000 abstract description 4
- 230000005867 T cell response Effects 0.000 abstract description 3
- 230000004044 response Effects 0.000 abstract description 3
- 230000000638 stimulation Effects 0.000 abstract description 3
- 210000002540 macrophage Anatomy 0.000 description 36
- 239000006228 supernatant Substances 0.000 description 26
- 102000052812 Ornithine decarboxylases Human genes 0.000 description 24
- 108700005126 Ornithine decarboxylases Proteins 0.000 description 24
- 102000003814 Interleukin-10 Human genes 0.000 description 21
- 108090000174 Interleukin-10 Proteins 0.000 description 21
- 210000001744 T-lymphocyte Anatomy 0.000 description 15
- 208000006552 Lewis Lung Carcinoma Diseases 0.000 description 14
- 210000003289 regulatory T cell Anatomy 0.000 description 12
- 210000004881 tumor cell Anatomy 0.000 description 11
- 230000002187 allostimulatory effect Effects 0.000 description 10
- 230000014509 gene expression Effects 0.000 description 10
- 102000004127 Cytokines Human genes 0.000 description 9
- 108090000695 Cytokines Proteins 0.000 description 9
- 230000006870 function Effects 0.000 description 9
- 238000004519 manufacturing process Methods 0.000 description 8
- 230000028993 immune response Effects 0.000 description 7
- 230000008629 immune suppression Effects 0.000 description 7
- 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
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 6
- AHLPHDHHMVZTML-BYPYZUCNSA-N L-Ornithine Chemical compound NCCC[C@H](N)C(O)=O AHLPHDHHMVZTML-BYPYZUCNSA-N 0.000 description 6
- 241000699670 Mus sp. Species 0.000 description 6
- MWUXSHHQAYIFBG-UHFFFAOYSA-N Nitric oxide Chemical compound O=[N] MWUXSHHQAYIFBG-UHFFFAOYSA-N 0.000 description 6
- 230000002401 inhibitory effect Effects 0.000 description 6
- 230000019734 interleukin-12 production Effects 0.000 description 6
- PFNFFQXMRSDOHW-UHFFFAOYSA-N spermine Chemical compound NCCCNCCCCNCCCN PFNFFQXMRSDOHW-UHFFFAOYSA-N 0.000 description 6
- 230000007246 mechanism Effects 0.000 description 5
- 230000001404 mediated effect Effects 0.000 description 5
- 238000007799 mixed lymphocyte reaction assay Methods 0.000 description 5
- 230000004614 tumor growth Effects 0.000 description 5
- 206010009944 Colon cancer Diseases 0.000 description 4
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 4
- AHLPHDHHMVZTML-UHFFFAOYSA-N Orn-delta-NH2 Natural products NCCCC(N)C(O)=O AHLPHDHHMVZTML-UHFFFAOYSA-N 0.000 description 4
- 239000001963 growth medium Substances 0.000 description 4
- 210000000987 immune system Anatomy 0.000 description 4
- 229960003104 ornithine Drugs 0.000 description 4
- 230000003389 potentiating effect Effects 0.000 description 4
- 230000028327 secretion Effects 0.000 description 4
- 210000002966 serum Anatomy 0.000 description 4
- 238000011740 C57BL/6 mouse Methods 0.000 description 3
- 102000004031 Carboxy-Lyases Human genes 0.000 description 3
- 108090000489 Carboxy-Lyases Proteins 0.000 description 3
- 206010060862 Prostate cancer Diseases 0.000 description 3
- 230000006044 T cell activation Effects 0.000 description 3
- VYMCYRPQICLHKC-WCCKRBBISA-N acetic acid;(2s)-2-amino-5-[[amino-(hydroxyamino)methylidene]amino]pentanoic acid Chemical compound CC(O)=O.OC(=O)[C@@H](N)CCCN=C(N)NO VYMCYRPQICLHKC-WCCKRBBISA-N 0.000 description 3
- 230000004913 activation Effects 0.000 description 3
- 230000006023 anti-tumor response Effects 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 230000005784 autoimmunity Effects 0.000 description 3
- 230000033228 biological regulation Effects 0.000 description 3
- 230000015572 biosynthetic process Effects 0.000 description 3
- 230000017188 evasion or tolerance of host immune response Effects 0.000 description 3
- 238000002474 experimental method Methods 0.000 description 3
- 230000008004 immune attack Effects 0.000 description 3
- 230000001965 increasing effect Effects 0.000 description 3
- 230000006698 induction Effects 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 229940063675 spermine Drugs 0.000 description 3
- 230000003827 upregulation Effects 0.000 description 3
- NHBKXEKEPDILRR-UHFFFAOYSA-N 2,3-bis(butanoylsulfanyl)propyl butanoate Chemical compound CCCC(=O)OCC(SC(=O)CCC)CSC(=O)CCC NHBKXEKEPDILRR-UHFFFAOYSA-N 0.000 description 2
- 108090000672 Annexin A5 Proteins 0.000 description 2
- 102000004121 Annexin A5 Human genes 0.000 description 2
- 208000023275 Autoimmune disease Diseases 0.000 description 2
- 206010004446 Benign prostatic hyperplasia Diseases 0.000 description 2
- 206010006187 Breast cancer Diseases 0.000 description 2
- 208000026310 Breast neoplasm Diseases 0.000 description 2
- 102000053642 Catalytic RNA Human genes 0.000 description 2
- 108090000994 Catalytic RNA Proteins 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 2
- UTJLXEIPEHZYQJ-UHFFFAOYSA-N Ornithine Natural products OC(=O)C(C)CCCN UTJLXEIPEHZYQJ-UHFFFAOYSA-N 0.000 description 2
- 208000004403 Prostatic Hyperplasia Diseases 0.000 description 2
- 241000251131 Sphyrna Species 0.000 description 2
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 2
- 210000004241 Th2 cell Anatomy 0.000 description 2
- 230000033115 angiogenesis Effects 0.000 description 2
- 230000001093 anti-cancer Effects 0.000 description 2
- 230000000259 anti-tumor effect Effects 0.000 description 2
- 235000009697 arginine Nutrition 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 201000008275 breast carcinoma Diseases 0.000 description 2
- 201000010989 colorectal carcinoma Diseases 0.000 description 2
- 230000002950 deficient Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 230000004069 differentiation Effects 0.000 description 2
- 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 2
- 201000010099 disease Diseases 0.000 description 2
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- 210000002950 fibroblast Anatomy 0.000 description 2
- 238000000684 flow cytometry Methods 0.000 description 2
- 239000012737 fresh medium Substances 0.000 description 2
- 150000002270 gangliosides Chemical class 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- 230000001506 immunosuppresive effect Effects 0.000 description 2
- 108010014723 immunosuppressive acidic protein Proteins 0.000 description 2
- 239000003018 immunosuppressive agent Substances 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 230000003993 interaction Effects 0.000 description 2
- 210000004698 lymphocyte Anatomy 0.000 description 2
- 230000003211 malignant effect Effects 0.000 description 2
- 230000035800 maturation Effects 0.000 description 2
- 201000001441 melanoma Diseases 0.000 description 2
- 230000004060 metabolic process Effects 0.000 description 2
- 125000004573 morpholin-4-yl group Chemical group N1(CCOCC1)* 0.000 description 2
- 210000000822 natural killer cell Anatomy 0.000 description 2
- 230000001613 neoplastic effect Effects 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- 229940094443 oxytocics prostaglandins Drugs 0.000 description 2
- 210000005259 peripheral blood Anatomy 0.000 description 2
- 239000011886 peripheral blood Substances 0.000 description 2
- 239000002953 phosphate buffered saline Substances 0.000 description 2
- 238000004393 prognosis Methods 0.000 description 2
- 230000000770 proinflammatory effect Effects 0.000 description 2
- 150000003180 prostaglandins Chemical class 0.000 description 2
- 230000001177 retroviral effect Effects 0.000 description 2
- 108091092562 ribozyme Proteins 0.000 description 2
- ATHGHQPFGPMSJY-UHFFFAOYSA-N spermidine Chemical compound NCCCCNCCCN ATHGHQPFGPMSJY-UHFFFAOYSA-N 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 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 2
- 239000000126 substance Substances 0.000 description 2
- 230000009885 systemic effect Effects 0.000 description 2
- 210000004981 tumor-associated macrophage Anatomy 0.000 description 2
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 2
- WQFIAVZQVYASHZ-BYPYZUCNSA-N (2s)-5-(diaminomethylideneamino)-2-(hydroxyamino)pentanoic acid Chemical compound NC(=N)NCCC[C@H](NO)C(O)=O WQFIAVZQVYASHZ-BYPYZUCNSA-N 0.000 description 1
- 102000016893 Amine Oxidase (Copper-Containing) Human genes 0.000 description 1
- 108010028700 Amine Oxidase (Copper-Containing) Proteins 0.000 description 1
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 1
- 241000024127 Argina Species 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 208000003174 Brain Neoplasms Diseases 0.000 description 1
- 102000008203 CTLA-4 Antigen Human genes 0.000 description 1
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 1
- 201000009030 Carcinoma Diseases 0.000 description 1
- 206010010144 Completed suicide Diseases 0.000 description 1
- 206010010356 Congenital anomaly Diseases 0.000 description 1
- 241000766026 Coregonus nasus Species 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- 206010063045 Effusion Diseases 0.000 description 1
- 206010018338 Glioma Diseases 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- 206010062016 Immunosuppression Diseases 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 102100037850 Interferon gamma Human genes 0.000 description 1
- 108010074328 Interferon-gamma Proteins 0.000 description 1
- 102000000589 Interleukin-1 Human genes 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-N L-arginine Chemical compound OC(=O)[C@@H](N)CCCN=C(N)N ODKSFYDXXFIFQN-BYPYZUCNSA-N 0.000 description 1
- 229930064664 L-arginine Natural products 0.000 description 1
- 235000014852 L-arginine Nutrition 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
- 241000222732 Leishmania major Species 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 210000004322 M2 macrophage Anatomy 0.000 description 1
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 description 1
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 101001002703 Mus musculus Interleukin-4 Proteins 0.000 description 1
- 102000008299 Nitric Oxide Synthase Human genes 0.000 description 1
- 108010021487 Nitric Oxide Synthase Proteins 0.000 description 1
- 102100029438 Nitric oxide synthase, inducible Human genes 0.000 description 1
- 101710089543 Nitric oxide synthase, inducible Proteins 0.000 description 1
- 239000004677 Nylon Substances 0.000 description 1
- 229940122060 Ornithine decarboxylase inhibitor Drugs 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- 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 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 108010029485 Protein Isoforms Proteins 0.000 description 1
- 102000001708 Protein Isoforms Human genes 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 230000006052 T cell proliferation Effects 0.000 description 1
- 108091008874 T cell receptors Proteins 0.000 description 1
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 1
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 102000046299 Transforming Growth Factor beta1 Human genes 0.000 description 1
- 102000011117 Transforming Growth Factor beta2 Human genes 0.000 description 1
- 101800002279 Transforming growth factor beta-1 Proteins 0.000 description 1
- 101800000304 Transforming growth factor beta-2 Proteins 0.000 description 1
- 241000223107 Trypanosoma congolense Species 0.000 description 1
- 102100031988 Tumor necrosis factor ligand superfamily member 6 Human genes 0.000 description 1
- 108050002568 Tumor necrosis factor ligand superfamily member 6 Proteins 0.000 description 1
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 1
- XSQUKJJJFZCRTK-UHFFFAOYSA-N Urea Chemical compound NC(N)=O XSQUKJJJFZCRTK-UHFFFAOYSA-N 0.000 description 1
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 1
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- 230000007815 allergy Effects 0.000 description 1
- 230000000735 allogeneic effect 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
- 230000002491 angiogenic effect Effects 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 230000005809 anti-tumor immunity Effects 0.000 description 1
- 239000000074 antisense oligonucleotide Substances 0.000 description 1
- 238000012230 antisense oligonucleotides Methods 0.000 description 1
- 230000001640 apoptogenic effect Effects 0.000 description 1
- 230000006470 autoimmune attack Effects 0.000 description 1
- IQFYYKKMVGJFEH-UHFFFAOYSA-N beta-L-thymidine Natural products O=C1NC(=O)C(C)=CN1C1OC(CO)C(O)C1 IQFYYKKMVGJFEH-UHFFFAOYSA-N 0.000 description 1
- 210000002798 bone marrow cell Anatomy 0.000 description 1
- 239000006227 byproduct Substances 0.000 description 1
- 230000000711 cancerogenic effect Effects 0.000 description 1
- 239000004202 carbamide Substances 0.000 description 1
- 231100000357 carcinogen Toxicity 0.000 description 1
- 239000003183 carcinogenic agent Substances 0.000 description 1
- 239000006143 cell culture medium Substances 0.000 description 1
- 230000036755 cellular response Effects 0.000 description 1
- 229940106189 ceramide Drugs 0.000 description 1
- 150000001783 ceramides Chemical class 0.000 description 1
- 208000019065 cervical carcinoma Diseases 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 210000003690 classically activated macrophage Anatomy 0.000 description 1
- 206010009887 colitis Diseases 0.000 description 1
- 210000001072 colon Anatomy 0.000 description 1
- 230000002860 competitive effect Effects 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 230000004940 costimulation Effects 0.000 description 1
- 230000001517 counterregulatory effect Effects 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 230000010406 dendritic cell apoptotic process Effects 0.000 description 1
- 230000003831 deregulation Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 235000020805 dietary restrictions Nutrition 0.000 description 1
- 230000009699 differential effect Effects 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 238000011124 ex vivo culture Methods 0.000 description 1
- 239000012894 fetal calf serum Substances 0.000 description 1
- 239000003365 glass fiber Substances 0.000 description 1
- 210000003714 granulocyte Anatomy 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 208000029824 high grade glioma Diseases 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 230000004046 hyporesponsiveness Effects 0.000 description 1
- 230000005934 immune activation Effects 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000036737 immune function Effects 0.000 description 1
- 230000017555 immunoglobulin mediated immune response Effects 0.000 description 1
- 230000004957 immunoregulator effect Effects 0.000 description 1
- 230000001024 immunotherapeutic effect Effects 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 229940076144 interleukin-10 Drugs 0.000 description 1
- 208000030776 invasive breast carcinoma Diseases 0.000 description 1
- 238000011835 investigation Methods 0.000 description 1
- 230000021633 leukocyte mediated immunity Effects 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 230000007108 local immune response Effects 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 208000020816 lung neoplasm Diseases 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 201000011614 malignant glioma Diseases 0.000 description 1
- 239000002609 medium Substances 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 239000002207 metabolite Substances 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 210000005087 mononuclear cell Anatomy 0.000 description 1
- 210000002864 mononuclear phagocyte Anatomy 0.000 description 1
- 229920001778 nylon Polymers 0.000 description 1
- 239000002818 ornithine decarboxylase inhibitor Substances 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 210000004303 peritoneum Anatomy 0.000 description 1
- 239000012660 pharmacological inhibitor Substances 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 210000003281 pleural cavity Anatomy 0.000 description 1
- 230000010287 polarization Effects 0.000 description 1
- 230000001855 preneoplastic effect Effects 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 208000023958 prostate neoplasm Diseases 0.000 description 1
- 235000018102 proteins Nutrition 0.000 description 1
- 102000004169 proteins and genes Human genes 0.000 description 1
- 230000006884 regulation of angiogenesis Effects 0.000 description 1
- 238000009877 rendering Methods 0.000 description 1
- 238000003118 sandwich ELISA Methods 0.000 description 1
- 210000000582 semen Anatomy 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 229940063673 spermidine Drugs 0.000 description 1
- 210000004988 splenocyte Anatomy 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 239000010902 straw Substances 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 230000009469 supplementation Effects 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 238000003786 synthesis reaction Methods 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
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 210000002303 tibia Anatomy 0.000 description 1
- 239000003104 tissue culture media Substances 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 229940099456 transforming growth factor beta 1 Drugs 0.000 description 1
- 229940072041 transforming growth factor beta 2 Drugs 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 230000005751 tumor progression Effects 0.000 description 1
- 230000001875 tumorinhibitory effect Effects 0.000 description 1
- 230000005760 tumorsuppression Effects 0.000 description 1
- 210000000689 upper leg Anatomy 0.000 description 1
- 230000029663 wound healing Effects 0.000 description 1
- 108010065816 zeta chain antigen T cell receptor Proteins 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- 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
- A61K35/30—Nerves; Brain; Eyes; Corneal cells; Cerebrospinal fluid; Neuronal stem cells; Neuronal precursor cells; Glial cells; Oligodendrocytes; Schwann cells; Astroglia; Astrocytes; Choroid plexus; Spinal cord tissue
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/10—Cellular immunotherapy characterised by the cell type used
- A61K40/19—Dendritic cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/20—Cellular immunotherapy characterised by the effect or the function of the cells
- A61K40/24—Antigen-presenting cells [APC]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/40—Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
- A61K40/41—Vertebrate antigens
- A61K40/42—Cancer antigens
- A61K40/4274—Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; Prostatic acid phosphatase [PAP]; Prostate-specific G-protein-coupled receptor [PSGR]
- A61K40/4275—Prostate specific antigen [PSA]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- 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
- C12N5/0639—Dendritic cells, e.g. Langherhans cells in the epidermis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/46—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the cancer treated
- A61K2239/55—Lung
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/46—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the cancer treated
- A61K2239/58—Prostate
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/50—Cellular immunotherapy characterised by the use of allogeneic cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
-
- 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
- C12N2501/20—Cytokines; Chemokines
- C12N2501/22—Colony stimulating factors (G-CSF, GM-CSF)
-
- 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
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
-
- 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
- C12N2501/70—Enzymes
-
- 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
- C12N2510/00—Genetically modified 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
- C12N2533/00—Supports or coatings for cell culture, characterised by material
- C12N2533/30—Synthetic polymers
- C12N2533/40—Polyhydroxyacids, e.g. polymers of glycolic or lactic acid (PGA, PLA, PLGA); Bioresorbable polymers
-
- 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
- C12N2533/00—Supports or coatings for cell culture, characterised by material
- C12N2533/50—Proteins
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Biomedical Technology (AREA)
- Epidemiology (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Immunology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Wood Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Cell Biology (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Hematology (AREA)
- General Engineering & Computer Science (AREA)
- Biochemistry (AREA)
- Developmental Biology & Embryology (AREA)
- Microbiology (AREA)
- Neurology (AREA)
- Virology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Neurosurgery (AREA)
- Ophthalmology & Optometry (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Strategies are disclosed for generating dendritic cells that possess immune stimulatory activity despite immune suppressive factors elaborated by the tumor. An aspect of the invention includes blocking the arginase pathway in dendritic cells that are pulsed with tumor antigens. Alternatively, dendritic cells modified according to the invention are placed proximally near tumor tissues so as to naturally acquire tumor antigens and stimulate T cell responses. An object of this invention is to allow stimulation of Th1 immunity in cancer-bearing mammals that generally are predisposed to Th2 or anergic responses.
Description
DESCRIPTION
Field of the Invention The invention disclosed relates to the field of methods of treating cancers.
More specifically, the invention pertains to the field of cancer irrununotherapy.
Background of Invention It is established that tumor cells evade the immune system of the patient.
This contributes to the general failure of immunotherapeutic approaches [1-3].
Secretion of soluble factors by tumors, such as IL-10 [4-6], free TGF-(~ [7], TGF-(3 bound to immunologlobulin [8), prostaglandins [9], low molecular weight retroviral plSE-like factors [ 10], immunosuppressive acidic protein [ 11 ], soluble MUC- i [ 12], and tumor-shed gangliosides [13) contribute to suppression of various aspects of the immune responses. Tumors also protect themselves from immune attack by coercing cells of the immune system to inhibit other cells of the immune system.
CD4 T cells have the ability to differentiate along the Thl or Th2 pathway, which in turn stimulate cell-mediated, or antibody-mediated immune responses [14]. In order to protect the host from autoimmunity, CD4 T cells also possess the ability to differentiate into T regulatory (Treg) cells that have the ability to inhibit activated T
cells regardless whether they are Thl or Th2 [15]. A specific type of Treg possesses the phenotype of CD4+ CD25+. These cells are critical for protection against autoimmunity, as demonstrated by experiments in which depletion of this cell population resulted in accelerated diabetes [16], or organ-specific autoimmune diseases [17].
Furthermore, transfer of Treg cells into animals prone to autoimmunity can delay progression or outright block disease occurrence [18].
Thus while Treg cells naturally possess the function of protecting the body against harmful autoimmune attacks, it is these same Treg cells that cancer uses to further shield itself from immune attack. For example, as stated above, tumor veils secrete various immune-inhibitory factors. These factors possess the property of not only inhibiting immune attack, but also programming the differentiation of T cells into Treg cells.
CD4+ CD25+ Treg cells have been found to circulate in higher numbers in cancer patients, where they inhibit T cell and NK cell responses [19]. Depletion of Treg cells allows for greater efficacy of cancer-specific vaccines in marine models [20, 21].
Another type of immunological cell that cancer subverts for its own means is the macrophage. Classically, macrophages are known to possess anti-cancer properties [22], and have been described to spontaneously lyse or arrest proliferation of tumor cells [23, 24]. However evidence is accumulating the macrophages can actually be involved in the stimulation of tumor growth. One suggestion of this was the observation that mice lacking mature macrophages (op/op mice) due to congenital absence of M-CSF are actually more resistant to growth of implanted tumors when compared to wild-type mice [25J. Further investigation found that cancer patients possessing high number of macrophages infiltrating the tumor had a poorer prognosis than patients with low numbers of tumor-infiltrating macrophages [26]. It was thought the mechanism by which the tumor-infiltrating macrophages stimulated an aggressive cancer phenotype was through increasing angiogenesis via secretion of endothelial-promoting factors such as VEGF [27].
To parallel the example with Treg cells, macrophages with immune inhibiting fianction, also termed M-2 or alternatively activated macrophages, are found in the normal life of a mammal. The normal function of these alternatively activated macrophages is thought to be promotion of wound healing [28]. Alternatively activated macrophages possess distinct molecular markers such as the genes Ym1 and Fizzl [29]. However, as is the case for Treg, tumors can induce alternatively activated macrophages in the tumor environment [30J. These cells inhibit local immune responses, and are themselves involved in the generation of Treg cells [31 ].
Tumors themselves and alternatively activated macrophages express high levels of the arginase, an enzyme that breaks down arginine into L-ornithine and urea [32, 33]. L-ornithine is further degraded into polyamines such as spermine and spermidine by the enzyme ornithine decarboxylase. Tmmune suppression induced by arginase overexpression can occur through several pathways: a) arginine-depletion by the tumor results in destruction of important T cell signaiing molecules such as the TCR-zeta chain [34]; b) polyamines suppress T cell activation [35]; c) polyamines suppress the proinflammatory function of macrophages [36]; and d) polyamines suppress activity of NK cells [37].
Macrophages from prototypical Thl strains (C57BL/6, BlOD2) are more easily activated to produce NO with either IFN-gamma or LPS than macrophages from Th2 strains (BALB/c, DBAI2). In marked contrast, LPS stimulates 'Th2, but not 'Thl, macrophages to increase arginine metabolism to ornithine [38]. In the context. of tumors, it is known that Th2 cytokines predispose to increased tumor aggressiveness and promote immune evasion. Th2 cytokines are also known to stimulate arginase production. In macrophages incubated with T cells: Thl T cells lead to an exclusive induction of iN~S, whereas Th2 T cells up-regulated macrophage arginase without inducing iNOS. Ab blocking experiments revealed the critical importance of IL-4 and IL-10 for arginase up-regulation [39]. Furthermore, the component of macrophage supernatant that stimulated tumor growth was substantially correlated with arginase [40].
As stated above, a characteristic feature of tumors is high production of the enzyme arginase. Serum arginase is a good marker for colorectal carcinoma progression [41-43].
Values of serum arginase activity in patients with breast carcinoma were up to 4-fold higher than those found in healthy women [44]. In prostate cancer tissue arginase expression correlates with severity of disease. A study evaluating arginase activities in prostate tissues of 50 patients with benign prostatic hyperplasia and in 23 patients with prostatic carcinoma found that arginase was elevated in cancer tissues as compared to benign prostatic hyperplasia (fivefold; P < .001). A positive correlation between arginase activity and Gleason grade of the tumors was also found [45].
Although tumor-derived arginase and macrophage argina.se are believed to be associated with immune suppression in cancer, a substantial question remains: tumors themselves are poor T cell stimulators, similarly macrophages can stimulate T cells but are not optimal, since dendritic cells (DC) are the most potent stimulators of T
cells, do they express arginase? Is tumor-induced arginase expression a mechanism by which cancer cells block T cell activation?
Although inhibitors of arginase [46], and inhibitors of enzymes downstream of arginase can block tumor growth and progression, the effects of these inhibitors is still therapeutically limited. The present invention is based on our findings that 1) DC
expression of arginase causes inhibition of immune function, and 2) reversal of arginase expression on DC can over-come the inhibitory effects of the tumor on DC
function, those stimulating antitumor immunity.
This background information is provided for the purpose of making known information believed by the applicant to be of possible relevance to the present invention. hto admission is necessarily intended, nor should be construed, that any of the proceding information constitutes prior art against the present invention.
I3escription of the Invention Disclosed are methods rendering dendritic cells potent stimulators of immune activation in such a manner that tumor-secreted immune suppressants do not inhibit their function.
A specific aspect of the invention relates to treating dendritic cells with a pharmacological inhibitor of the enzyme arginase such as l~(omega)-hydroxy-L-arginine or N(omega)-hydroxy-nor-L-arginine. This treatment wot~.ld inhibit the ability of the dendritic cell to react to a variety of tumor-secreted immune suppressants, in particular cytokines such as IL-10. In the same spirit, the dendritic cell may be modulated through the use of other known inhibitors that block the function of the enzyme arginase. Said inhibitors may be small molecular peptides, chemical analogues or suicide substrates.
In another aspect, dendritic cell expression of the enzyme arginase is suppressed using known genetic inhibitory techniques such as siRlVA, antisense oligonucleotides, hammerhead ribozymes or morpholinos. The suppression of arginase may be performed in an ex vivo manner, wherein the dendritic cells, or their progenitors are extracted from the blood or bone marrow of the patient, cultured and transfected outside of the body, and subsequently reintroduced into the patient in such a manner as to stimulate immune response against the tumor. In this aspect, the dendritic cells are either pulsed with tumor-derived antigen, or alternatively, are allowed to naturally engulf tumor antigens from the tumor it self if the dendritic cells are injected intratumorally.
Another aspect of the invention is that dendritic cells whose ornithine decarboxylase is inhibited can be used for stimulating anti-tumor responses by virtue of the fact that they are not prone to tumor-induced immune suppression. Bath chemical and/or genetic (ie siRNA, antisense oligoriucleotides, hammerhead ribozymes or morpholinos) inhibition of omithine decarboxylase will cause the formation of potent immune stimulatory dendritic cells that can prime anti-tumor responses more effectively than unmanipulated dendritic cells.
Another aspect of the invention is that inhibition of both arginase and ornithine decarboxylase endow the dendritic cell with protection from tumor-induced apoptosis.
Inhibition of expression and/or function of arginase and/or ornithine decarboxylase allows for more effective immune stimulation by increasing the amount of time that a dendritic cell can live in the context of the tumor-secreted, and surface bound apoptotic factors.
Another aspect of the invention is that manipulation of arginase and ornithine activity in dendritic cells can be achieved by systemic manipulation through dietary restriction or through systemically administered drugs such as DFMO. While this approach may be less efficacious than ex vivo manipulation, it is more convenient in situations were ex vivo culture and manipulation of dendritic cells is impossible.
Description of Figures Figure 1 illustrates that arginase inhibition blocks IL-10-induced suppression of DC IL-12 production.
Figure 2 illustrations that arginase inhibition renders DC resistant to IL-10 mediated suppression of allostimulatory capacity.
Figure 3 illustrates that arginase inhibition blocks tumor supernatant-induced suppression of DC IL-12 production.
Figure 4 illustrates that arginase inhibition renders DC resistant to tumor supernatant mediated suppression of allostimulatory capacity.
Figure 5 illustrates that arginase inhibition renders DC resistant to tumor supernatant-induced apoptosis.
Figure 6 illustrates that ODC inhibition renders DC resistant to tumor supernatant-mediated suppression of IL-12 production.
Figure 7 illustrates that ODC inhibition renders DC resistant to tumor supernatant-mediated suppression of allostimulatory function.
Figure 8 illustrates that ODC inhibition renders DC resistant to tumor supernatant-induced apoptosis.
Figure 9 illustrates that arginase and ODC-inhibited DC, but not untreated DC, are able to induce tumor suppression in vivo.
I9etailed Description of Invention ~ne embodiment of the disclosed invention teaches methods of preparing a cancer vaccine using dendritic cells that are resistant to cancer-induced immune suppression. As stated in the Background section, the arginase pathway of metabolism is upregulated in cancer cells and cancer-secreted metabolites of this pathway are involved in immune suppression. However, part of the novelty of this invention resides in our findings that tumor-secreted immune suppressive factors, such as IL-10, are also potent inducers of the arginase pathway in dendritic cells. Since T cells are more potently stimulated or inhibited during their interaction with dendritic cells, as compared to their interaction with tumor cells, it would be more likely that the arginase upregulation in the dendritic cell is more important for tumor-immune suppression than arginase upregulation in the tumor itself. Furthermore, arginase pathway activation is involved in the induction of dendritic cell apoptosis. The ability of tumors to secrete cytokines such as IL-I0 which upregulate arginase activity in dendritic cells, could be a mechanism by which tumor cells induce apoptosis of dendritic cells. Therefore the inhibition of the arginase pathway in tumor cells would not only increase the resistance of dendritic cells to tumor-secreted immune suppressive factors but also protect dendritic cells from apoptosis.
For manufacturing a cancer vaccine using dendritic cells, blockade of the arginase pathway could be achieved through: a) inhibition ~f argin.ase enzymatic activity, b) inhibition of arginase gene expression, c) inhibition of onuthine decarboxylase enzymatic activity, or d) inhibition of omithine decarboxylase gene expression.
In one cancer vaccine embodiment, dendritic cells are generated from peripheral blood monoclear cells purified by density gradient. Monocytic cells are purified by adherence to plastic and cultured for 7-days in GM-CSF and IL-4 at a concentration between 20-100ng/ml, preferentially, 50 ng/ml. A concentration of 200 ~,Mol 1VOAH is added to the culture every time the cells are passaged. Tissue culture media is changed once every two days. An alternative modification is that ornithine-decarboxylase can be inhibited in the DC culture by addition of the ornithine-decarboxylase inhibitor DFM~.
Although several concentrations are useful, 3mM seems ideal based on our studies. These arginase/ornithine decarboxylase-inhibited DC are subsequently injected infra-tumorally, preferably at a concentration of 2xI09 cells. Based on the endogenous ability of DC to uptake tumor antigens, these cells provide an autologous "natural" vaccine that stimulates T cell responses against the tumor antigens.
Examples Example 1: Generation of dendritic cells resistant to the tumor-secreted cytoldne This example illustrates the generation of dendritic cells that are resistant to the immune-inhibitory activities of IL-10 by pretreating said dendritic cells with the arginase inhibitor N hydroxy-L-arginine (NOHA).
DC were generated from bone marrow progenitor cells as previously described [47).
Briefly; bone marrow cells Were flushed from the femurs and tibias of C57BL/6 mice (Jackson Labs, Bar Harbor ME), washed and cultured in ?4-well plates (2 x 106 cells/ml) in 2 ml of complete medium (RPMI-1640 supplemented with 2rnM L-glutamine, 100 U/ml of penicillin, 100 ~.g of streptomycin, 50 ~.M 2-mercaptoethanol, and 10 % fetal calf serum (all from Gibco RBL)) supplemented with recombinant GM-CSF (10 ng/ml;
Peprotech, Rocky HiII, NJ) and recombinant mouse IL-4 (10 nglml; Peprotech).
All cultures were incubated at 37°C in 5% humidified C~2. Non-adherent granulocytes were removed after 48 hours of culture and fresh medium was added.
4 experimental groups were used:
a) Unmanipulated DC cultures b) DC cultured in the presence of IL-10 c) DC cultured in the presence of arginase inhibition d) DC cultured with IL-10 and arginase inhibition In group (b) IL-10 was added throughout the culture period at a final concentration of 50 ng/ml. In group (c) a 200 ~.M concentration of the arginase inhibitor NOHA
(Sigma-Aldrich, Rockville IL) was added for the whole culture period. In group (d), the combination IL-10 and the arginase inhibitor NOHA were added as described in groups (b and c).
After 7 days of culture >90°/~ of the cells expressed the characteristic DC-specific marker CDl lc as determined by FRCS. DC were washed in phosphate buffered saline (PBS) and plated in 24-well plates at a concentration of 2 x 105 cells per well.
Cells were activated for 48 hours with LPS (10 ng/ml, Sigma Aldrich; St Louis, MO) + TNF-oc (10 ng/rnl, Peprotech). Supernatants were harvested and production of the tumor-inhibitory and Thl-activating cytokine IL-12 was assessed by sandwich ELISA (R&D Systems, Minneapolis, MN). As illustrated in Figure 1, DC generated in absence of IL-10 or arginase inhibition possessed strong IL-12 producing ability (a). Addition of IL-10 in the culture media resulted in the inhibition of IL-I2 production (b). Inhibition of arginase activity induced a slight increase in activity production of IL-I2(c).
Surprisingly, arginase inhibition spared the DC of IL-10-mediated downregulation of IL-12 production.
Additionally, mixed lymphocyte reaction (MLR) was performed to assess functional allostimulatory capacity of the DC in groups a-d. For MLR, T cells were purified from BALB/c splenocytes using nylon wool columns and were used as responders (1x106~well). DC from groups a-d (5-40x103, C57BL6 origin) were used as stimulators.
72 hour MLR was performed and the cells were pulsed with 1 ~,Ci [3H]-thymidine for the last 18 hours. The cultures were harvested on to glass fiber filters (Wallac, Turku, Finland). Radioactivity was counted using a Wallac 1450 Microbeta liquid scintillation counter and the data were analyzed with IJltraTerm 3 software. As seen in Figure 2, DC
generated in the presence of IL-10 were poor stimulators of allogeneic T cell proliferation. DC raised in the presence of arginase inhibition possessed a similar allostimulatory capacity as DC raised under control conditions. When DC were generated in the presence of IL-10 under conditions of arginase inhibition, the allostimulatory effects were preserved. This suggests that arginase inhibition can act at the level of the dendritic cell to protect from immune suppressive activities of IL-10.
Example 2: Arginase inhibition allows normal DC maturation despite presence of tumor-secreted inhibitory factors.
It is reported that generating DC in the presence of supernatants from a variety of tumors results in the production of immature DC with poor allostimulatory capacity.
t~lthough IL-10 is a known tumor-secreted factor responsible for the inhibition of DC
maturation, other factors such as ceramides, gangliosides, TGF-b and prostaglandins have also been reported. We therefore assessed whether inhibition of arginase activity could render DC
resistant to the inhibitory effects of tumor -supernatant in a manner similar to IL-10.
DC were generated as described in Example 1 with the modification that IL-10 was replaced with 20% volume of day-2 supernatant from the marine Lewis lung carcinoma (3LL) cell line (American Type Culture Collection, lVlanassas VA). Arginase inhibition was performed by supplementation of the cell culture media with a 200 p.M
concentration of the arginase inhibitor NOHA as in Example 1.
Experimental groups consisted of a) Unmanipulated DC cultures b) DC cultured in the presence of 20% 3LL supernatant c) DC cultured in the presence of arginase inhibition d) DC cultured with 20% 3LL supernatant and arginase inhibition Using the experimental conditions of Example 1, it was demonstrated that arginase inhibition was able to block the 3LL-induced inhibition of IL-12 production from DC
(Figure 3).
The conditions of Example 1 were also used to assess whether arginase inhibition could block 3LL-induced suppression of DC allostirnulatory capacity. As seen in Figure 4, arginase inhibition was successful at preserving DC allostimulatory ability despite the presence of 3LL supernatant.
Example 3 Arginase inhibition protects DC from tumor-induced apoptosis It is reported that supernatants from a variety of tumor cells can induce apoptosis in DC.
A method of protecting the DC from such apoptosis would theoretically result in a more efficacious tumor vaccine, as well as protect the immune responsive ability of the cancer patient.
Dendritic cells were generated as described in Example 1. At day 7 of culture the cells were divided into 4 groups a) Control: media from the 3T3 fibroblast cell line was added.
b) Arginase inhibition: a 200 ~,1~ concentration of the arginase inhibitor N~HA was added.
c) Tumor supernatant: B-16 supernatant was added at 20% of tissue culture volume.
d) Arginase inhibition in the presence of tumor supernatant. Both b and c were added.
At day 9 apoptosis was assessed using Annexin-V staining and analyzed by flow cytometry. As indicated in Figure 5 apoptosis was induced by the presence of tumor-supernatant while arginase inhibition was able to reduce this effect.
Example 4 Inhibition of ornithine decarboxylase (ODC) protects DC from tumor-induced immune suppression DC were cultured as described in Example 1. In some groups media was replaced with ~0% fresh media and 20% volume of day-2 culture supernatant from the marine Lewis lung carcinoma (3LL) cell line.
Experimental groups consisted of a) Unmanipulated DC cultures b) DC cultured in the presence of 20°/~ 3LL supernatant c) DC cultured in the presence of ODC inhibition d) DC cultured with 20% 3LL supernatant and ODC inhibition ODC inhibition was achieved by administration of 3 mM DL-alpha-difluoromethylornithine (DFMO) into the culture media for the whole Culture period.
Using the experimental conditions of Example 1, it was demonstrated that ODC
inhibition was able to block the 3LL-induced inhibition of IL-12 production from DC
(Figure 6).
The conditions of Example 1 were also used to assess whether ODC inhibition could block 3LL-induced suppression of DC allostimulatory capacity. As seen in Figure 7, ODC inhibition was successful at preserving DC allostimulatory ability despite the presence of 3LL supernatant.
Example 5 ~DC inhibition protects DC from tumor-induced apoptosis Dendritic cells were generated as described in Example 1. At day 7 of culture the cells were divided into 4 groups a) Control: media from the 3T3 fibroblast cell line was added.
b) ODC inhibition: a concentration of 3 mM DL-alpha-difluoromethylornithine (DFMO) was added to the culture media.
c) Tumor supernatant: B-16 supernatant was added at 20% of tissue culture volume.
d) ODC inhibition in the presence of tumor supernatant. Both b and c were added.
At day 9 apoptosis was assessed using Annexin-V staining and analyzed by flow cytometry. As indicated in Figure 8 apoptosis was induced by the presence of tumor-' supernatant while arginase inhibition was able to reduce this effect.
Example 6 Arginase-inhibited and ODC-inhibited DC induce ante-tumor responses in vivo Male 6-8 week old C57BL/6 mice were injected with RM-I marine prostate tumor cells (20,000 cells/100 ~,l) in the right flank. Tumor size was determined using Vernier caliper. DC (20,000/100 ~ul) raised using the conditions below were co-injected with tumor cells or tumor cells were injected alone:
DC generated as described in Example i in and raised under the following conditions:
a) Fed every other day control media as described in Example 1 b) Arginase was inhibited by addition of 200 p,M NOAH
c) ODC was inhibited by addition of 3 mM DMFO
mice/group were used. Tumor size was evaluated on day 20 was evaluated. As seen in Figure 9, tumor cells injected alone, or with control DC grew to a much greater extend than tumors injected with DC raised in the presence of arginase or ODC
inhibition. As repetition of this experiment in nu/nu on C57~L6 backgrounds resulted in equivalent tumor growth in all groups (data not shown), we postulate that relieving the DC of tumor-induced immune suppression by arginase inhibition or ODC inhibition, is a valuable strategy for induction of antitumor responses.
The foregoing invention has been described in such a manner to enable one skilled in the art to practice it. However, it will be readily apparent to one skilled in the art that after reading the teachings of this invention certain changes and modifications may be made without departing from the spirit or scope of the appended claims.
Furthermore, the examples illustrated are by no means binding but provide a guide for practicing certain embodiments and aspects of the invention disclosed.
1. Whiteside, T.L., 22. Immune responses to malignancies. J Allergy Clin Immunol, 2003. 111(2 Supply: p. 5677-86.
2. Igney, F.H. and P.H. Krammer, Immune escape of tumors: apopt~sis resistance and tumor counterattack. J Leukoc Biol, 2002. 71(6): p. 907-20.
3. Bodey, B., et al., Failure of cancer vaccines: the significant limitations of this approach to immunotherapy. Anticancer Res, 2000. 20(4): p. 2665-76.
4_ Kurzrock, R., Cytokine deregulation in cancer. Biomed Pharmacother, 2001.
55(9-10): p. 543-7.
5_ Mocellin, S., E. 'Wang, and F.M. Marincola, Cytokines and Immune Response ire the Tumor Microenvironment. J Immunother, 2001. 24(5): p. 392-407.
6. Salazar-Onfray, F., Interleukin-10: a cytokine used by tumors to escape immunosurveillance. Med Oncol, 1999. 16(2): p. 86-94.
7. Sasaki, A., et al., Secretion of transforming growth factor-beta 1 and -beta 2 by malignant glioma cells. Neurol Med Chir (Tokyo), 1995. 35(7): p. 423-30.
8. Beck, C., H. Schreiber, and I~. Rowley, Role of TGF-beta in immune-evasion ~f cancer. Microsc Res Tech, 2001. 52(4): p. 387-95.
9. Botti, C., et al.; Immunosuppressive factors: role in cancer development and progression. Int J Biol Markers, 1998. 13(2): p. 51-69.
10. Scheeren, R.A., et aL, Distribution of retroviral pISE-related proteins in neoplastic and non-neoplastic human tissues, and their role in the regulation of the immune response. Clin Exp Immunol, 1992. $J(1): p. 94-9.
IS
11. Kikuchi, K., H. Gotoh, and M. Kowada, A correlation between serum immunosuppressive acidic protein and altered immunocompetence in patients with brain tumours. Acta Neurochir (Wien), 1990.103(1-2): p. 52-61.
12. Chars, A.K., et al., Soluble tl~UCl secreted by human epithelial cancer cells mediates immune suppression by blocking T cell activation. Int J Cancer, 1999.
82(5): p. 721-6.
13. Takahashi, K., et al., Escape mechanisms of melanoma from immune system by soluble melanoma antigen. J Irnmunol, 1988. 140(9): p. 3244-8.
14. Mosmann, T.R. and R.L. Coffman, THd and TH2 cells: different patterns of lymphokine secretion lead to d~erent functional properties. Anna Rev Immunol, 1989. 7: p. 145-73.
15. Bluestone, J.A. and A.K. Abbas, Natural versus adaptive regulatory T
cells. Nat Rev Immunol, 2003. 3(3): p. 253-7.
16. Salomon, B., et al., p7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes.
Immunity, 2000. 12(4): p. 431-40.
17. Bagavant, H., et al., Differential effect of neonatal tlaymectomy on systemic and organ-specific autoimmune disease. Int Immunol, 2002. 14(12): p. 1397-406.
18. Xu, D., et al., CD4+CD25+ regulatory T cells suppress differentiation and functions of Thl and Th2 cells, Leishmania major infection, and colitis in mice. J
Immunol, 2003. 170(1): p. 394-9.
19. Wolf, A.M., et al., Increase of regulatory T cells in the peripheral blood of cancer patients. Clin Cancer Res, 2003. g(2): p. 606-12.
20: Golgher, D., et al., Depletion of CD25+ regulatory cells uncovers immune responses to shared marine tumor rejection antigens. Eur J Immunol, 2002.
32(11): p. 3267-75.
21. Sutmuller, R.P., et al., Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T
lymphocyte responses. J Exp Med, 2001. 194(6): p. 823-32.
22. Bhaumik, S., et al., Activated macrophages migrate to the subcutaneous tumor site via the peritoneum: a novel route of cell trafficking. E~p Cell Res, 2001.
266(1): p. 44-52.
23. Kimura, S., et al., Antitumour potential of pleural cavity macrophages in lung cancer patients without malignant effusion. Br J Cancer, 1989. 59(4): p. 535-9.
24. Lavnikova, N., et al., Macrophage and interleukin-I induced nitric oxide production and cytostasis in hamster tumor cells varying in malignant potential. J
Leukoc Biol, 1997. 61(4): p. 452-8.
25. Nowicki, A., et al., Impaired tumor growth in colony-stimulatingfactor I
(CSF
1)-deficient, macrophage-deficient oplop mouse: evidence for a role of CSF I-dependent macrophages in formation of tumor stroma. Int J Cancer, 1996. 65{1):
p. 112-9.
26. Leek, R.D., et al., Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res, 1996. 56(20): p. 4625-9.
27. Lewis, C.E., et al., Cytokine regulation of angiogenesis in breast cancer:
the role of tumor-associated macrophages. J Leukoc Biol, 1995. 57(5): p. 747-51.
28. Kodelja, V., et al., Differences in angiogenic potential of classically vs alternatively activated macrophages. Immunobiology, 1997. 197(5): p. 478-93.
29. Nair, M.G., D.W. Cochrane, and J.E. Allen, Macrophages in chronic type 2 inflammation have a novel phenotype characterized by the abundant expression of Yml and Fizzl that can be partly replicated in vitro. Immunol Lett, 2003.
85(2):
p. 173-80.
30. Mantovani, A., et al., Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. 'Trends Immunol, 2002. 23(11): p. 549-55.
31. 'Tzachanis, D., et al., Blockade of B7/CD28 in mixed lymphocyte reaction cultures results in the generation of alternatively activated .macrophages, which suppress T cell responses. Blood, 2002. 99(4): p. 1465-73.
32. Namangala, B., et al., Alternative versus classical macrophage activation during experimental African trypanosomosis. J Leukoc Bi.ol, 2001. 69(3): p. 387-96.
33. Noel, W., et al., Infection stage-dependent modulation of macrophage activation in Trypanosoma congolense-resistant and -susceptible mice. Infect Immure, 2002.
70(11): p. 6180-7.
34. Rodriguez, P.C., et al., Regulation of T cell receptor CD3zeta chairs expression by L-arginine. J Biol Chem, 2002. 277(24): p. 21123-9.
35. Quan, C.P., et al., Delineation between T and B suppressive molecules ji~om human seminal plasma: II. Spermine is the major suppressor of ?=lymphocytes in vitro. Am J Reprod Immunol, 1990. 22(1-2): p. 64-9.
36. Zhang, M., et al.; Spermine inhibits proinflammatory cytokine synthesis in human mononuclear cells: a counterregulatory mechanism that restrains the immune response. J Exp Med, 1997. 185(10): p. 1759-68.
37. Evans, C.H., T.S. Lee, and A.A. Flugelman, Spermine-directed immunosuppression of cervical carcinoma cell sensitivity to a majority of lymphokine-activated killer lymphocyte cytotoxicity. Nat Immure, 1995. 14(3):
p.
157-63.
38. Mills, C.D., et al., M 1/M 2 macrophages and the ThllTh2 paradigm. 3 Immunol, 2000. 164( 12): p. 6166-73.
39. Munder, M., K. Eichmann, and M. Modolell, Alternative metabolic states in murine macrophages reflected by the nitric oxide synthaselarginase balance:
competitive regulation by CD4+ T cells correlates with ThllTh.~ phenotype. J
Immunol, 1998. 160(11): p. 5347-54.
40. Currie, G.A., Promotion of f brosarcoma cell gromth by products of syngeneic host macrophages. Br J Cancer, 1981. 44(4): p. 506-13.
41. Porembska, Z., et al., Serum arginase activity in postsurgical monitoring of patients with colorectal carcinoma. Cancer, 2002. 94(11): p. 2930-4.
42. del Ara, R.M., et al., Diagnostic performance of arginase activity in colorectal cancer. Clin Exp Med, 2002. 2(1): p. 53-7.
43. Porembska, Z., et al., Arginase isoforms in human colorectal cancer. Clin Chim Acta, 2001. 305(1-2): p. 157-65.
44. Straws, B., I. Cepelak, and G. Festa, Arginase, a new marker of mammary carcinoma. Clin Chim Acta, 1992. 210(1-2): p. 5-12.
1g 45. Keskinege, A., S. Elgun, and E. Yilinaz, Possible implications of arginase and diamine oxidase in prostatic carcinoma. Cancer Detect Prev, 2001. 25(1): p. 76-9.
46. Schleiffer, R., et al., Nitric oxide synthase inhibition promotes carcinogen-induced preneoplastic changes in the colon of rats. Nitric Oxide, 2000. 4(6):
p.
583-9.
47. Min, W.P., et al., Dendritic cells genetically engineered to express Fas ligand induce donor-specific hyporesponsiveness and prolong allograft survival. J
Immunol, 2000. 164(I): p. 161-7.
Field of the Invention The invention disclosed relates to the field of methods of treating cancers.
More specifically, the invention pertains to the field of cancer irrununotherapy.
Background of Invention It is established that tumor cells evade the immune system of the patient.
This contributes to the general failure of immunotherapeutic approaches [1-3].
Secretion of soluble factors by tumors, such as IL-10 [4-6], free TGF-(~ [7], TGF-(3 bound to immunologlobulin [8), prostaglandins [9], low molecular weight retroviral plSE-like factors [ 10], immunosuppressive acidic protein [ 11 ], soluble MUC- i [ 12], and tumor-shed gangliosides [13) contribute to suppression of various aspects of the immune responses. Tumors also protect themselves from immune attack by coercing cells of the immune system to inhibit other cells of the immune system.
CD4 T cells have the ability to differentiate along the Thl or Th2 pathway, which in turn stimulate cell-mediated, or antibody-mediated immune responses [14]. In order to protect the host from autoimmunity, CD4 T cells also possess the ability to differentiate into T regulatory (Treg) cells that have the ability to inhibit activated T
cells regardless whether they are Thl or Th2 [15]. A specific type of Treg possesses the phenotype of CD4+ CD25+. These cells are critical for protection against autoimmunity, as demonstrated by experiments in which depletion of this cell population resulted in accelerated diabetes [16], or organ-specific autoimmune diseases [17].
Furthermore, transfer of Treg cells into animals prone to autoimmunity can delay progression or outright block disease occurrence [18].
Thus while Treg cells naturally possess the function of protecting the body against harmful autoimmune attacks, it is these same Treg cells that cancer uses to further shield itself from immune attack. For example, as stated above, tumor veils secrete various immune-inhibitory factors. These factors possess the property of not only inhibiting immune attack, but also programming the differentiation of T cells into Treg cells.
CD4+ CD25+ Treg cells have been found to circulate in higher numbers in cancer patients, where they inhibit T cell and NK cell responses [19]. Depletion of Treg cells allows for greater efficacy of cancer-specific vaccines in marine models [20, 21].
Another type of immunological cell that cancer subverts for its own means is the macrophage. Classically, macrophages are known to possess anti-cancer properties [22], and have been described to spontaneously lyse or arrest proliferation of tumor cells [23, 24]. However evidence is accumulating the macrophages can actually be involved in the stimulation of tumor growth. One suggestion of this was the observation that mice lacking mature macrophages (op/op mice) due to congenital absence of M-CSF are actually more resistant to growth of implanted tumors when compared to wild-type mice [25J. Further investigation found that cancer patients possessing high number of macrophages infiltrating the tumor had a poorer prognosis than patients with low numbers of tumor-infiltrating macrophages [26]. It was thought the mechanism by which the tumor-infiltrating macrophages stimulated an aggressive cancer phenotype was through increasing angiogenesis via secretion of endothelial-promoting factors such as VEGF [27].
To parallel the example with Treg cells, macrophages with immune inhibiting fianction, also termed M-2 or alternatively activated macrophages, are found in the normal life of a mammal. The normal function of these alternatively activated macrophages is thought to be promotion of wound healing [28]. Alternatively activated macrophages possess distinct molecular markers such as the genes Ym1 and Fizzl [29]. However, as is the case for Treg, tumors can induce alternatively activated macrophages in the tumor environment [30J. These cells inhibit local immune responses, and are themselves involved in the generation of Treg cells [31 ].
Tumors themselves and alternatively activated macrophages express high levels of the arginase, an enzyme that breaks down arginine into L-ornithine and urea [32, 33]. L-ornithine is further degraded into polyamines such as spermine and spermidine by the enzyme ornithine decarboxylase. Tmmune suppression induced by arginase overexpression can occur through several pathways: a) arginine-depletion by the tumor results in destruction of important T cell signaiing molecules such as the TCR-zeta chain [34]; b) polyamines suppress T cell activation [35]; c) polyamines suppress the proinflammatory function of macrophages [36]; and d) polyamines suppress activity of NK cells [37].
Macrophages from prototypical Thl strains (C57BL/6, BlOD2) are more easily activated to produce NO with either IFN-gamma or LPS than macrophages from Th2 strains (BALB/c, DBAI2). In marked contrast, LPS stimulates 'Th2, but not 'Thl, macrophages to increase arginine metabolism to ornithine [38]. In the context. of tumors, it is known that Th2 cytokines predispose to increased tumor aggressiveness and promote immune evasion. Th2 cytokines are also known to stimulate arginase production. In macrophages incubated with T cells: Thl T cells lead to an exclusive induction of iN~S, whereas Th2 T cells up-regulated macrophage arginase without inducing iNOS. Ab blocking experiments revealed the critical importance of IL-4 and IL-10 for arginase up-regulation [39]. Furthermore, the component of macrophage supernatant that stimulated tumor growth was substantially correlated with arginase [40].
As stated above, a characteristic feature of tumors is high production of the enzyme arginase. Serum arginase is a good marker for colorectal carcinoma progression [41-43].
Values of serum arginase activity in patients with breast carcinoma were up to 4-fold higher than those found in healthy women [44]. In prostate cancer tissue arginase expression correlates with severity of disease. A study evaluating arginase activities in prostate tissues of 50 patients with benign prostatic hyperplasia and in 23 patients with prostatic carcinoma found that arginase was elevated in cancer tissues as compared to benign prostatic hyperplasia (fivefold; P < .001). A positive correlation between arginase activity and Gleason grade of the tumors was also found [45].
Although tumor-derived arginase and macrophage argina.se are believed to be associated with immune suppression in cancer, a substantial question remains: tumors themselves are poor T cell stimulators, similarly macrophages can stimulate T cells but are not optimal, since dendritic cells (DC) are the most potent stimulators of T
cells, do they express arginase? Is tumor-induced arginase expression a mechanism by which cancer cells block T cell activation?
Although inhibitors of arginase [46], and inhibitors of enzymes downstream of arginase can block tumor growth and progression, the effects of these inhibitors is still therapeutically limited. The present invention is based on our findings that 1) DC
expression of arginase causes inhibition of immune function, and 2) reversal of arginase expression on DC can over-come the inhibitory effects of the tumor on DC
function, those stimulating antitumor immunity.
This background information is provided for the purpose of making known information believed by the applicant to be of possible relevance to the present invention. hto admission is necessarily intended, nor should be construed, that any of the proceding information constitutes prior art against the present invention.
I3escription of the Invention Disclosed are methods rendering dendritic cells potent stimulators of immune activation in such a manner that tumor-secreted immune suppressants do not inhibit their function.
A specific aspect of the invention relates to treating dendritic cells with a pharmacological inhibitor of the enzyme arginase such as l~(omega)-hydroxy-L-arginine or N(omega)-hydroxy-nor-L-arginine. This treatment wot~.ld inhibit the ability of the dendritic cell to react to a variety of tumor-secreted immune suppressants, in particular cytokines such as IL-10. In the same spirit, the dendritic cell may be modulated through the use of other known inhibitors that block the function of the enzyme arginase. Said inhibitors may be small molecular peptides, chemical analogues or suicide substrates.
In another aspect, dendritic cell expression of the enzyme arginase is suppressed using known genetic inhibitory techniques such as siRlVA, antisense oligonucleotides, hammerhead ribozymes or morpholinos. The suppression of arginase may be performed in an ex vivo manner, wherein the dendritic cells, or their progenitors are extracted from the blood or bone marrow of the patient, cultured and transfected outside of the body, and subsequently reintroduced into the patient in such a manner as to stimulate immune response against the tumor. In this aspect, the dendritic cells are either pulsed with tumor-derived antigen, or alternatively, are allowed to naturally engulf tumor antigens from the tumor it self if the dendritic cells are injected intratumorally.
Another aspect of the invention is that dendritic cells whose ornithine decarboxylase is inhibited can be used for stimulating anti-tumor responses by virtue of the fact that they are not prone to tumor-induced immune suppression. Bath chemical and/or genetic (ie siRNA, antisense oligoriucleotides, hammerhead ribozymes or morpholinos) inhibition of omithine decarboxylase will cause the formation of potent immune stimulatory dendritic cells that can prime anti-tumor responses more effectively than unmanipulated dendritic cells.
Another aspect of the invention is that inhibition of both arginase and ornithine decarboxylase endow the dendritic cell with protection from tumor-induced apoptosis.
Inhibition of expression and/or function of arginase and/or ornithine decarboxylase allows for more effective immune stimulation by increasing the amount of time that a dendritic cell can live in the context of the tumor-secreted, and surface bound apoptotic factors.
Another aspect of the invention is that manipulation of arginase and ornithine activity in dendritic cells can be achieved by systemic manipulation through dietary restriction or through systemically administered drugs such as DFMO. While this approach may be less efficacious than ex vivo manipulation, it is more convenient in situations were ex vivo culture and manipulation of dendritic cells is impossible.
Description of Figures Figure 1 illustrates that arginase inhibition blocks IL-10-induced suppression of DC IL-12 production.
Figure 2 illustrations that arginase inhibition renders DC resistant to IL-10 mediated suppression of allostimulatory capacity.
Figure 3 illustrates that arginase inhibition blocks tumor supernatant-induced suppression of DC IL-12 production.
Figure 4 illustrates that arginase inhibition renders DC resistant to tumor supernatant mediated suppression of allostimulatory capacity.
Figure 5 illustrates that arginase inhibition renders DC resistant to tumor supernatant-induced apoptosis.
Figure 6 illustrates that ODC inhibition renders DC resistant to tumor supernatant-mediated suppression of IL-12 production.
Figure 7 illustrates that ODC inhibition renders DC resistant to tumor supernatant-mediated suppression of allostimulatory function.
Figure 8 illustrates that ODC inhibition renders DC resistant to tumor supernatant-induced apoptosis.
Figure 9 illustrates that arginase and ODC-inhibited DC, but not untreated DC, are able to induce tumor suppression in vivo.
I9etailed Description of Invention ~ne embodiment of the disclosed invention teaches methods of preparing a cancer vaccine using dendritic cells that are resistant to cancer-induced immune suppression. As stated in the Background section, the arginase pathway of metabolism is upregulated in cancer cells and cancer-secreted metabolites of this pathway are involved in immune suppression. However, part of the novelty of this invention resides in our findings that tumor-secreted immune suppressive factors, such as IL-10, are also potent inducers of the arginase pathway in dendritic cells. Since T cells are more potently stimulated or inhibited during their interaction with dendritic cells, as compared to their interaction with tumor cells, it would be more likely that the arginase upregulation in the dendritic cell is more important for tumor-immune suppression than arginase upregulation in the tumor itself. Furthermore, arginase pathway activation is involved in the induction of dendritic cell apoptosis. The ability of tumors to secrete cytokines such as IL-I0 which upregulate arginase activity in dendritic cells, could be a mechanism by which tumor cells induce apoptosis of dendritic cells. Therefore the inhibition of the arginase pathway in tumor cells would not only increase the resistance of dendritic cells to tumor-secreted immune suppressive factors but also protect dendritic cells from apoptosis.
For manufacturing a cancer vaccine using dendritic cells, blockade of the arginase pathway could be achieved through: a) inhibition ~f argin.ase enzymatic activity, b) inhibition of arginase gene expression, c) inhibition of onuthine decarboxylase enzymatic activity, or d) inhibition of omithine decarboxylase gene expression.
In one cancer vaccine embodiment, dendritic cells are generated from peripheral blood monoclear cells purified by density gradient. Monocytic cells are purified by adherence to plastic and cultured for 7-days in GM-CSF and IL-4 at a concentration between 20-100ng/ml, preferentially, 50 ng/ml. A concentration of 200 ~,Mol 1VOAH is added to the culture every time the cells are passaged. Tissue culture media is changed once every two days. An alternative modification is that ornithine-decarboxylase can be inhibited in the DC culture by addition of the ornithine-decarboxylase inhibitor DFM~.
Although several concentrations are useful, 3mM seems ideal based on our studies. These arginase/ornithine decarboxylase-inhibited DC are subsequently injected infra-tumorally, preferably at a concentration of 2xI09 cells. Based on the endogenous ability of DC to uptake tumor antigens, these cells provide an autologous "natural" vaccine that stimulates T cell responses against the tumor antigens.
Examples Example 1: Generation of dendritic cells resistant to the tumor-secreted cytoldne This example illustrates the generation of dendritic cells that are resistant to the immune-inhibitory activities of IL-10 by pretreating said dendritic cells with the arginase inhibitor N hydroxy-L-arginine (NOHA).
DC were generated from bone marrow progenitor cells as previously described [47).
Briefly; bone marrow cells Were flushed from the femurs and tibias of C57BL/6 mice (Jackson Labs, Bar Harbor ME), washed and cultured in ?4-well plates (2 x 106 cells/ml) in 2 ml of complete medium (RPMI-1640 supplemented with 2rnM L-glutamine, 100 U/ml of penicillin, 100 ~.g of streptomycin, 50 ~.M 2-mercaptoethanol, and 10 % fetal calf serum (all from Gibco RBL)) supplemented with recombinant GM-CSF (10 ng/ml;
Peprotech, Rocky HiII, NJ) and recombinant mouse IL-4 (10 nglml; Peprotech).
All cultures were incubated at 37°C in 5% humidified C~2. Non-adherent granulocytes were removed after 48 hours of culture and fresh medium was added.
4 experimental groups were used:
a) Unmanipulated DC cultures b) DC cultured in the presence of IL-10 c) DC cultured in the presence of arginase inhibition d) DC cultured with IL-10 and arginase inhibition In group (b) IL-10 was added throughout the culture period at a final concentration of 50 ng/ml. In group (c) a 200 ~.M concentration of the arginase inhibitor NOHA
(Sigma-Aldrich, Rockville IL) was added for the whole culture period. In group (d), the combination IL-10 and the arginase inhibitor NOHA were added as described in groups (b and c).
After 7 days of culture >90°/~ of the cells expressed the characteristic DC-specific marker CDl lc as determined by FRCS. DC were washed in phosphate buffered saline (PBS) and plated in 24-well plates at a concentration of 2 x 105 cells per well.
Cells were activated for 48 hours with LPS (10 ng/ml, Sigma Aldrich; St Louis, MO) + TNF-oc (10 ng/rnl, Peprotech). Supernatants were harvested and production of the tumor-inhibitory and Thl-activating cytokine IL-12 was assessed by sandwich ELISA (R&D Systems, Minneapolis, MN). As illustrated in Figure 1, DC generated in absence of IL-10 or arginase inhibition possessed strong IL-12 producing ability (a). Addition of IL-10 in the culture media resulted in the inhibition of IL-I2 production (b). Inhibition of arginase activity induced a slight increase in activity production of IL-I2(c).
Surprisingly, arginase inhibition spared the DC of IL-10-mediated downregulation of IL-12 production.
Additionally, mixed lymphocyte reaction (MLR) was performed to assess functional allostimulatory capacity of the DC in groups a-d. For MLR, T cells were purified from BALB/c splenocytes using nylon wool columns and were used as responders (1x106~well). DC from groups a-d (5-40x103, C57BL6 origin) were used as stimulators.
72 hour MLR was performed and the cells were pulsed with 1 ~,Ci [3H]-thymidine for the last 18 hours. The cultures were harvested on to glass fiber filters (Wallac, Turku, Finland). Radioactivity was counted using a Wallac 1450 Microbeta liquid scintillation counter and the data were analyzed with IJltraTerm 3 software. As seen in Figure 2, DC
generated in the presence of IL-10 were poor stimulators of allogeneic T cell proliferation. DC raised in the presence of arginase inhibition possessed a similar allostimulatory capacity as DC raised under control conditions. When DC were generated in the presence of IL-10 under conditions of arginase inhibition, the allostimulatory effects were preserved. This suggests that arginase inhibition can act at the level of the dendritic cell to protect from immune suppressive activities of IL-10.
Example 2: Arginase inhibition allows normal DC maturation despite presence of tumor-secreted inhibitory factors.
It is reported that generating DC in the presence of supernatants from a variety of tumors results in the production of immature DC with poor allostimulatory capacity.
t~lthough IL-10 is a known tumor-secreted factor responsible for the inhibition of DC
maturation, other factors such as ceramides, gangliosides, TGF-b and prostaglandins have also been reported. We therefore assessed whether inhibition of arginase activity could render DC
resistant to the inhibitory effects of tumor -supernatant in a manner similar to IL-10.
DC were generated as described in Example 1 with the modification that IL-10 was replaced with 20% volume of day-2 supernatant from the marine Lewis lung carcinoma (3LL) cell line (American Type Culture Collection, lVlanassas VA). Arginase inhibition was performed by supplementation of the cell culture media with a 200 p.M
concentration of the arginase inhibitor NOHA as in Example 1.
Experimental groups consisted of a) Unmanipulated DC cultures b) DC cultured in the presence of 20% 3LL supernatant c) DC cultured in the presence of arginase inhibition d) DC cultured with 20% 3LL supernatant and arginase inhibition Using the experimental conditions of Example 1, it was demonstrated that arginase inhibition was able to block the 3LL-induced inhibition of IL-12 production from DC
(Figure 3).
The conditions of Example 1 were also used to assess whether arginase inhibition could block 3LL-induced suppression of DC allostirnulatory capacity. As seen in Figure 4, arginase inhibition was successful at preserving DC allostimulatory ability despite the presence of 3LL supernatant.
Example 3 Arginase inhibition protects DC from tumor-induced apoptosis It is reported that supernatants from a variety of tumor cells can induce apoptosis in DC.
A method of protecting the DC from such apoptosis would theoretically result in a more efficacious tumor vaccine, as well as protect the immune responsive ability of the cancer patient.
Dendritic cells were generated as described in Example 1. At day 7 of culture the cells were divided into 4 groups a) Control: media from the 3T3 fibroblast cell line was added.
b) Arginase inhibition: a 200 ~,1~ concentration of the arginase inhibitor N~HA was added.
c) Tumor supernatant: B-16 supernatant was added at 20% of tissue culture volume.
d) Arginase inhibition in the presence of tumor supernatant. Both b and c were added.
At day 9 apoptosis was assessed using Annexin-V staining and analyzed by flow cytometry. As indicated in Figure 5 apoptosis was induced by the presence of tumor-supernatant while arginase inhibition was able to reduce this effect.
Example 4 Inhibition of ornithine decarboxylase (ODC) protects DC from tumor-induced immune suppression DC were cultured as described in Example 1. In some groups media was replaced with ~0% fresh media and 20% volume of day-2 culture supernatant from the marine Lewis lung carcinoma (3LL) cell line.
Experimental groups consisted of a) Unmanipulated DC cultures b) DC cultured in the presence of 20°/~ 3LL supernatant c) DC cultured in the presence of ODC inhibition d) DC cultured with 20% 3LL supernatant and ODC inhibition ODC inhibition was achieved by administration of 3 mM DL-alpha-difluoromethylornithine (DFMO) into the culture media for the whole Culture period.
Using the experimental conditions of Example 1, it was demonstrated that ODC
inhibition was able to block the 3LL-induced inhibition of IL-12 production from DC
(Figure 6).
The conditions of Example 1 were also used to assess whether ODC inhibition could block 3LL-induced suppression of DC allostimulatory capacity. As seen in Figure 7, ODC inhibition was successful at preserving DC allostimulatory ability despite the presence of 3LL supernatant.
Example 5 ~DC inhibition protects DC from tumor-induced apoptosis Dendritic cells were generated as described in Example 1. At day 7 of culture the cells were divided into 4 groups a) Control: media from the 3T3 fibroblast cell line was added.
b) ODC inhibition: a concentration of 3 mM DL-alpha-difluoromethylornithine (DFMO) was added to the culture media.
c) Tumor supernatant: B-16 supernatant was added at 20% of tissue culture volume.
d) ODC inhibition in the presence of tumor supernatant. Both b and c were added.
At day 9 apoptosis was assessed using Annexin-V staining and analyzed by flow cytometry. As indicated in Figure 8 apoptosis was induced by the presence of tumor-' supernatant while arginase inhibition was able to reduce this effect.
Example 6 Arginase-inhibited and ODC-inhibited DC induce ante-tumor responses in vivo Male 6-8 week old C57BL/6 mice were injected with RM-I marine prostate tumor cells (20,000 cells/100 ~,l) in the right flank. Tumor size was determined using Vernier caliper. DC (20,000/100 ~ul) raised using the conditions below were co-injected with tumor cells or tumor cells were injected alone:
DC generated as described in Example i in and raised under the following conditions:
a) Fed every other day control media as described in Example 1 b) Arginase was inhibited by addition of 200 p,M NOAH
c) ODC was inhibited by addition of 3 mM DMFO
mice/group were used. Tumor size was evaluated on day 20 was evaluated. As seen in Figure 9, tumor cells injected alone, or with control DC grew to a much greater extend than tumors injected with DC raised in the presence of arginase or ODC
inhibition. As repetition of this experiment in nu/nu on C57~L6 backgrounds resulted in equivalent tumor growth in all groups (data not shown), we postulate that relieving the DC of tumor-induced immune suppression by arginase inhibition or ODC inhibition, is a valuable strategy for induction of antitumor responses.
The foregoing invention has been described in such a manner to enable one skilled in the art to practice it. However, it will be readily apparent to one skilled in the art that after reading the teachings of this invention certain changes and modifications may be made without departing from the spirit or scope of the appended claims.
Furthermore, the examples illustrated are by no means binding but provide a guide for practicing certain embodiments and aspects of the invention disclosed.
1. Whiteside, T.L., 22. Immune responses to malignancies. J Allergy Clin Immunol, 2003. 111(2 Supply: p. 5677-86.
2. Igney, F.H. and P.H. Krammer, Immune escape of tumors: apopt~sis resistance and tumor counterattack. J Leukoc Biol, 2002. 71(6): p. 907-20.
3. Bodey, B., et al., Failure of cancer vaccines: the significant limitations of this approach to immunotherapy. Anticancer Res, 2000. 20(4): p. 2665-76.
4_ Kurzrock, R., Cytokine deregulation in cancer. Biomed Pharmacother, 2001.
55(9-10): p. 543-7.
5_ Mocellin, S., E. 'Wang, and F.M. Marincola, Cytokines and Immune Response ire the Tumor Microenvironment. J Immunother, 2001. 24(5): p. 392-407.
6. Salazar-Onfray, F., Interleukin-10: a cytokine used by tumors to escape immunosurveillance. Med Oncol, 1999. 16(2): p. 86-94.
7. Sasaki, A., et al., Secretion of transforming growth factor-beta 1 and -beta 2 by malignant glioma cells. Neurol Med Chir (Tokyo), 1995. 35(7): p. 423-30.
8. Beck, C., H. Schreiber, and I~. Rowley, Role of TGF-beta in immune-evasion ~f cancer. Microsc Res Tech, 2001. 52(4): p. 387-95.
9. Botti, C., et al.; Immunosuppressive factors: role in cancer development and progression. Int J Biol Markers, 1998. 13(2): p. 51-69.
10. Scheeren, R.A., et aL, Distribution of retroviral pISE-related proteins in neoplastic and non-neoplastic human tissues, and their role in the regulation of the immune response. Clin Exp Immunol, 1992. $J(1): p. 94-9.
IS
11. Kikuchi, K., H. Gotoh, and M. Kowada, A correlation between serum immunosuppressive acidic protein and altered immunocompetence in patients with brain tumours. Acta Neurochir (Wien), 1990.103(1-2): p. 52-61.
12. Chars, A.K., et al., Soluble tl~UCl secreted by human epithelial cancer cells mediates immune suppression by blocking T cell activation. Int J Cancer, 1999.
82(5): p. 721-6.
13. Takahashi, K., et al., Escape mechanisms of melanoma from immune system by soluble melanoma antigen. J Irnmunol, 1988. 140(9): p. 3244-8.
14. Mosmann, T.R. and R.L. Coffman, THd and TH2 cells: different patterns of lymphokine secretion lead to d~erent functional properties. Anna Rev Immunol, 1989. 7: p. 145-73.
15. Bluestone, J.A. and A.K. Abbas, Natural versus adaptive regulatory T
cells. Nat Rev Immunol, 2003. 3(3): p. 253-7.
16. Salomon, B., et al., p7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes.
Immunity, 2000. 12(4): p. 431-40.
17. Bagavant, H., et al., Differential effect of neonatal tlaymectomy on systemic and organ-specific autoimmune disease. Int Immunol, 2002. 14(12): p. 1397-406.
18. Xu, D., et al., CD4+CD25+ regulatory T cells suppress differentiation and functions of Thl and Th2 cells, Leishmania major infection, and colitis in mice. J
Immunol, 2003. 170(1): p. 394-9.
19. Wolf, A.M., et al., Increase of regulatory T cells in the peripheral blood of cancer patients. Clin Cancer Res, 2003. g(2): p. 606-12.
20: Golgher, D., et al., Depletion of CD25+ regulatory cells uncovers immune responses to shared marine tumor rejection antigens. Eur J Immunol, 2002.
32(11): p. 3267-75.
21. Sutmuller, R.P., et al., Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T
lymphocyte responses. J Exp Med, 2001. 194(6): p. 823-32.
22. Bhaumik, S., et al., Activated macrophages migrate to the subcutaneous tumor site via the peritoneum: a novel route of cell trafficking. E~p Cell Res, 2001.
266(1): p. 44-52.
23. Kimura, S., et al., Antitumour potential of pleural cavity macrophages in lung cancer patients without malignant effusion. Br J Cancer, 1989. 59(4): p. 535-9.
24. Lavnikova, N., et al., Macrophage and interleukin-I induced nitric oxide production and cytostasis in hamster tumor cells varying in malignant potential. J
Leukoc Biol, 1997. 61(4): p. 452-8.
25. Nowicki, A., et al., Impaired tumor growth in colony-stimulatingfactor I
(CSF
1)-deficient, macrophage-deficient oplop mouse: evidence for a role of CSF I-dependent macrophages in formation of tumor stroma. Int J Cancer, 1996. 65{1):
p. 112-9.
26. Leek, R.D., et al., Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res, 1996. 56(20): p. 4625-9.
27. Lewis, C.E., et al., Cytokine regulation of angiogenesis in breast cancer:
the role of tumor-associated macrophages. J Leukoc Biol, 1995. 57(5): p. 747-51.
28. Kodelja, V., et al., Differences in angiogenic potential of classically vs alternatively activated macrophages. Immunobiology, 1997. 197(5): p. 478-93.
29. Nair, M.G., D.W. Cochrane, and J.E. Allen, Macrophages in chronic type 2 inflammation have a novel phenotype characterized by the abundant expression of Yml and Fizzl that can be partly replicated in vitro. Immunol Lett, 2003.
85(2):
p. 173-80.
30. Mantovani, A., et al., Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. 'Trends Immunol, 2002. 23(11): p. 549-55.
31. 'Tzachanis, D., et al., Blockade of B7/CD28 in mixed lymphocyte reaction cultures results in the generation of alternatively activated .macrophages, which suppress T cell responses. Blood, 2002. 99(4): p. 1465-73.
32. Namangala, B., et al., Alternative versus classical macrophage activation during experimental African trypanosomosis. J Leukoc Bi.ol, 2001. 69(3): p. 387-96.
33. Noel, W., et al., Infection stage-dependent modulation of macrophage activation in Trypanosoma congolense-resistant and -susceptible mice. Infect Immure, 2002.
70(11): p. 6180-7.
34. Rodriguez, P.C., et al., Regulation of T cell receptor CD3zeta chairs expression by L-arginine. J Biol Chem, 2002. 277(24): p. 21123-9.
35. Quan, C.P., et al., Delineation between T and B suppressive molecules ji~om human seminal plasma: II. Spermine is the major suppressor of ?=lymphocytes in vitro. Am J Reprod Immunol, 1990. 22(1-2): p. 64-9.
36. Zhang, M., et al.; Spermine inhibits proinflammatory cytokine synthesis in human mononuclear cells: a counterregulatory mechanism that restrains the immune response. J Exp Med, 1997. 185(10): p. 1759-68.
37. Evans, C.H., T.S. Lee, and A.A. Flugelman, Spermine-directed immunosuppression of cervical carcinoma cell sensitivity to a majority of lymphokine-activated killer lymphocyte cytotoxicity. Nat Immure, 1995. 14(3):
p.
157-63.
38. Mills, C.D., et al., M 1/M 2 macrophages and the ThllTh2 paradigm. 3 Immunol, 2000. 164( 12): p. 6166-73.
39. Munder, M., K. Eichmann, and M. Modolell, Alternative metabolic states in murine macrophages reflected by the nitric oxide synthaselarginase balance:
competitive regulation by CD4+ T cells correlates with ThllTh.~ phenotype. J
Immunol, 1998. 160(11): p. 5347-54.
40. Currie, G.A., Promotion of f brosarcoma cell gromth by products of syngeneic host macrophages. Br J Cancer, 1981. 44(4): p. 506-13.
41. Porembska, Z., et al., Serum arginase activity in postsurgical monitoring of patients with colorectal carcinoma. Cancer, 2002. 94(11): p. 2930-4.
42. del Ara, R.M., et al., Diagnostic performance of arginase activity in colorectal cancer. Clin Exp Med, 2002. 2(1): p. 53-7.
43. Porembska, Z., et al., Arginase isoforms in human colorectal cancer. Clin Chim Acta, 2001. 305(1-2): p. 157-65.
44. Straws, B., I. Cepelak, and G. Festa, Arginase, a new marker of mammary carcinoma. Clin Chim Acta, 1992. 210(1-2): p. 5-12.
1g 45. Keskinege, A., S. Elgun, and E. Yilinaz, Possible implications of arginase and diamine oxidase in prostatic carcinoma. Cancer Detect Prev, 2001. 25(1): p. 76-9.
46. Schleiffer, R., et al., Nitric oxide synthase inhibition promotes carcinogen-induced preneoplastic changes in the colon of rats. Nitric Oxide, 2000. 4(6):
p.
583-9.
47. Min, W.P., et al., Dendritic cells genetically engineered to express Fas ligand induce donor-specific hyporesponsiveness and prolong allograft survival. J
Immunol, 2000. 164(I): p. 161-7.
Claims (18)
1. A cancer vaccine comprised of;
a) a dendritic cell, b) a tumor antigen, c) an arginase inhibitor.
a) a dendritic cell, b) a tumor antigen, c) an arginase inhibitor.
2. The vaccine of claim 1 wherein a dendritic cell encompasses a bone marrow-derived cell expressing substantial amounts of the surface marker CD11c, MHC II, CD80, and CD86.
3. The vaccine of claim 1 wherein said dendritic cell is purified from bone marrow cultures at 4-13 days of culture in interleukin-4 and granulocyte-monocyte colony stimulating factor.
4. The cancer vaccine of claim 1 wherein a tumor antigen is derived from molecules found on tumors but not found on healthy, non-tumorous tissue.
5. The cancer vaccine of claim 1 wherein tumour antigens comprise of MUC-1, MAGE, BAGE, PSA, PAP, tyrosinase, and CEA.
6. A dendritic cell whose function is not substantially inhibited by immune suppressive factors in the body of a cancer patient whereas said dendritic cell is treated with a dose of the arginase inhibitor L-NOAH effective to substantially inhibit the enzyme arginase.
7. The dendritic cell of claim 6 wherein arginase inhibition is performed by incubation of 200µ M L-NOAH with said dendritic cell.
8. A dendritic cell whose function is not substantially inhibited by immune suppressive factors in the body of a cancer patient wherein said dendritic cell is transfected with short interfering RNA oligonucleotides ranging from 19-25 base pairs that possess substantial homology with an exon from the gene encoding arginase.
9. A dendritic cell whose function is not substantially inhibited by immune suppressive factors in the body of a cancer patient wherein said dendritic cell is transfected with a plasmid encoding a sequence that when transcribed into RNA yields a hairpin loop that degrades intracellularly into short interfering RNA that possesses substantial homology with an exon from the gene encoding arginase.
10. The utility of the dendritic cell of claims 6-10 as a cancer vaccine, wherein said dendritic cell is administered a tumor antigen and subsequently introduced into the body of a mammal in need thereof at a concentration needed to stimulate tumor-specific immunity.
11. A cellular vaccine composition wherein the dendritic cell of claim 10 is admixed into a biocompatible sponge-like material and inserted into the proximity of a tumor.
12. The cellular vaccine composition of claim 11 wherein the sponge-like material is comprised of epsilon-caprolactone-co-L-lactide reinforced with knitted poly-L-lactide fabric PCLA.
13. The cellular vaccine composition of claim 11 wherein the sponge-like material is comprised mainly of gelatin.
14. The vaccine composition of claim 11 wherein the sponge-like material is comprised of polyglycolic acid.
15. A method of protecting dendritic cells from tumor-induced apoptosis through inhibition of arginase pathway.
16. A method of blocking tumor-induced DC suppression by administering an effective amount of an inhibitor of ODC such as .alpha.-difluoromethylornithine (DFMO).
17. A method of blocking tumor-induced DC suppression by restricting arginine or polyamine intake.
18. A method of blocking tumor-induced DC suppression by administering drugs whose primary or secondary effects include suppression of arginine levels, arginase activity, or ODC activity or levels.
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CA002431080A CA2431080A1 (en) | 2003-06-02 | 2003-06-02 | Enhancement of anticancer immunity through inhibition of arginase |
Applications Claiming Priority (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CA002431080A CA2431080A1 (en) | 2003-06-02 | 2003-06-02 | Enhancement of anticancer immunity through inhibition of arginase |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| CA2431080A1 true CA2431080A1 (en) | 2004-12-02 |
Family
ID=33557556
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| CA002431080A Abandoned CA2431080A1 (en) | 2003-06-02 | 2003-06-02 | Enhancement of anticancer immunity through inhibition of arginase |
Country Status (1)
| Country | Link |
|---|---|
| CA (1) | CA2431080A1 (en) |
Cited By (10)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2006058486A1 (en) * | 2004-12-03 | 2006-06-08 | Bio-Cancer Treatment International Limited | Use of arginase in combination with 5fu and other compounds for treatment of human malignancies |
| WO2006133561A1 (en) * | 2005-06-15 | 2006-12-21 | London Health Sciences Centre Research Inc. | Method of cancer treatment using sirna silencing |
| WO2018089490A1 (en) * | 2016-11-08 | 2018-05-17 | Calithera Biosciences, Inc. | Arginase inhibitor combination therapies |
| US10065974B2 (en) | 2015-10-30 | 2018-09-04 | Calithera Biosciences, Inc. | Compositions and methods for inhibiting arginase activity |
| US10143699B2 (en) | 2015-06-23 | 2018-12-04 | Calithera Biosciences, Inc. | Compositions and methods for inhibiting arginase activity |
| US10287303B2 (en) | 2016-12-22 | 2019-05-14 | Calithera Biosciences, Inc. | Compositions and methods for inhibiting arginase activity |
| WO2019145453A1 (en) * | 2018-01-28 | 2019-08-01 | Universite De Geneve | Arginase suppression for cancer treatment |
| US10494339B2 (en) | 2017-05-12 | 2019-12-03 | Calithera Biosciences, Inc. | Method of preparing (3R,4S)-3-acetamido-4-allyl-N-(tert-butyl)pyrrolidine-3-carboxamide |
| US10538537B2 (en) | 2010-04-22 | 2020-01-21 | Mars, Incorporated | Inhibitors of arginase and their therapeutic applications |
| US10603330B2 (en) | 2010-10-26 | 2020-03-31 | Mars, Incorporated | Arginase inhibitors as therapeutics |
-
2003
- 2003-06-02 CA CA002431080A patent/CA2431080A1/en not_active Abandoned
Cited By (23)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2006058486A1 (en) * | 2004-12-03 | 2006-06-08 | Bio-Cancer Treatment International Limited | Use of arginase in combination with 5fu and other compounds for treatment of human malignancies |
| WO2006133561A1 (en) * | 2005-06-15 | 2006-12-21 | London Health Sciences Centre Research Inc. | Method of cancer treatment using sirna silencing |
| EP1898936A4 (en) * | 2005-06-15 | 2009-05-06 | London Health Sci Ct Res Inc | Method of cancer treatment using sirna silencing |
| US8389708B2 (en) | 2005-06-15 | 2013-03-05 | Weiping Min | Method of cancer treatment using siRNA silencing |
| US10538537B2 (en) | 2010-04-22 | 2020-01-21 | Mars, Incorporated | Inhibitors of arginase and their therapeutic applications |
| US11389464B2 (en) | 2010-10-26 | 2022-07-19 | Mars, Incorporated | Arginase inhibitors as therapeutics |
| US10603330B2 (en) | 2010-10-26 | 2020-03-31 | Mars, Incorporated | Arginase inhibitors as therapeutics |
| US10905701B2 (en) | 2015-06-23 | 2021-02-02 | Calithera Biosciences, Inc. | Compositions and methods for inhibiting arginase activity |
| US10143699B2 (en) | 2015-06-23 | 2018-12-04 | Calithera Biosciences, Inc. | Compositions and methods for inhibiting arginase activity |
| US10398714B2 (en) | 2015-06-23 | 2019-09-03 | Calithera Biosciences, Inc. | Compositions and methods for inhibiting arginase activity |
| US10851118B2 (en) | 2015-10-30 | 2020-12-01 | Calithera Biosciences, Inc. | Compositions and methods for inhibiting arginase activity |
| US10844080B2 (en) | 2015-10-30 | 2020-11-24 | Calithera Biosciences, Inc. | Compositions and methods for inhibiting arginase activity |
| US10065974B2 (en) | 2015-10-30 | 2018-09-04 | Calithera Biosciences, Inc. | Compositions and methods for inhibiting arginase activity |
| US11291674B2 (en) | 2016-11-08 | 2022-04-05 | Calithera Biosciences, Inc. | Arginase inhibitor combination therapies |
| WO2018089490A1 (en) * | 2016-11-08 | 2018-05-17 | Calithera Biosciences, Inc. | Arginase inhibitor combination therapies |
| US10287303B2 (en) | 2016-12-22 | 2019-05-14 | Calithera Biosciences, Inc. | Compositions and methods for inhibiting arginase activity |
| US11021495B2 (en) | 2016-12-22 | 2021-06-01 | Calithera Biosciences, Inc. | Compositions and methods for inhibiting arginase activity |
| US12054501B2 (en) | 2016-12-22 | 2024-08-06 | Precision Pharmaceuticals, Inc. | Compositions and methods for inhibiting arginase activity |
| US10494339B2 (en) | 2017-05-12 | 2019-12-03 | Calithera Biosciences, Inc. | Method of preparing (3R,4S)-3-acetamido-4-allyl-N-(tert-butyl)pyrrolidine-3-carboxamide |
| US10906872B2 (en) | 2017-05-12 | 2021-02-02 | Calithera Biosciences, Inc. | Method of preparing (3R,4S)-3-acetamido-4-allyl-n-(tert-butyl)pyrrolidine-3-carboxamide |
| US11370754B2 (en) | 2017-05-12 | 2022-06-28 | Calithera Biosciences, Inc. | Method of preparing (3R,4S)-3-acetamido-4-allyl-n-(tert-butyl)pyrrolidine-3-carboxamide |
| WO2019145453A1 (en) * | 2018-01-28 | 2019-08-01 | Universite De Geneve | Arginase suppression for cancer treatment |
| CN111902532A (en) * | 2018-01-28 | 2020-11-06 | 日内瓦大学 | Arginase inhibition for cancer treatment |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Huang et al. | Antigen-loaded monocyte administration induces potent therapeutic antitumor T cell responses | |
| Lu et al. | Th9 cells promote antitumor immune responses in vivo | |
| KR100995340B1 (en) | Vaccine containing monocyte or undifferentiated myeloid cells loaded with ligand and antigen of natural killer T cells | |
| Galetto et al. | Drug-and cell-mediated antitumor cytotoxicities modulate cross-presentation of tumor antigens by myeloid dendritic cells | |
| Young | Protective mechanisms of head and neck squamous cell carcinomas from immune assault | |
| WO1998004282A1 (en) | Cancer immunotherapy using autologous tumor cells combined with allogeneic cytokine-secreting cells | |
| KR20000049096A (en) | Cancer immunotherapy using tumor cells combined with mixed lymphocytes | |
| Crittenden et al. | Intratumoral immunotherapy: using the tumour against itself | |
| Bringmann et al. | RNA vaccines in cancer treatment | |
| CN109125347A (en) | The means and method of active cell immunization therapy are carried out to cancer using dendritic cells and the tumour cell killed by high hydrostatic pressure | |
| AU2009223838A1 (en) | Allogeneic cancer cell-based immunotherapy | |
| Lopes et al. | New generation of DNA-based immunotherapy induces a potent immune response and increases the survival in different tumor models | |
| CA2431080A1 (en) | Enhancement of anticancer immunity through inhibition of arginase | |
| Shahrokhi et al. | Tumor necrosis factor-α/CD40 ligand-engineered mesenchymal stem cells greatly enhanced the antitumor immune response and lifespan in mice | |
| Velders et al. | CD4+ T cell matters in tumor immunity | |
| Yao et al. | Hollow Cu2MoS4 nanoparticles loaded with immune checkpoint inhibitors reshape the tumor microenvironment to enhance immunotherapy for pancreatic cancer | |
| KR20130060188A (en) | Methods and Compositions for Inhibition of Regulatory T Cells | |
| Rodrı́guez-Calvillo et al. | Upregulation of natural killer cells functions underlies the efficacy of intratumorally injected dendritic cells engineered to produce interleukin-12 | |
| Palumbo et al. | Polymer-mediated DNA vaccine delivery via bystander cells requires a proper balance between transfection efficiency and cytotoxicity | |
| Ishikawa et al. | Intratumoral injection of IL‐2‐activated NK cells enhances the antitumor effect of intradermally injected paraformaldehyde‐fixed tumor vaccine in a rat intracranial brain tumor model | |
| US11071753B2 (en) | Micro-RNA-155 enhances the efficacy of dendritic cell vaccine for cancer | |
| Schwaab et al. | Tumor‐related immunity in prostate cancer patients treated with human recombinant granulocyte monocyte‐colony stimulating factor (GM‐CSF) | |
| O'Neill et al. | Immunotherapeutic potential of dendritic cells generated in long term stroma-dependent cultures | |
| De Wit et al. | IL-12 stimulation but not B7 expression increases melanoma killing by patient cytotoxic T lymphocytes (CTL) | |
| Clavreul et al. | Effects of syngeneic cellular vaccinations alone or in combination with GM-CSF on the weakly immunogenic F98 glioma model |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| FZDE | Discontinued |