WO2022109227A1 - Combined cancer therapy of b7-h3 and cd47 immune checkpoint inhbitior and methods of use - Google Patents
Combined cancer therapy of b7-h3 and cd47 immune checkpoint inhbitior and methods of use Download PDFInfo
- Publication number
- WO2022109227A1 WO2022109227A1 PCT/US2021/060025 US2021060025W WO2022109227A1 WO 2022109227 A1 WO2022109227 A1 WO 2022109227A1 US 2021060025 W US2021060025 W US 2021060025W WO 2022109227 A1 WO2022109227 A1 WO 2022109227A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- inhibitor
- composition
- antibody
- sirpa
- cells
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims abstract description 173
- 102000037982 Immune checkpoint proteins Human genes 0.000 title description 4
- 108091008036 Immune checkpoint proteins Proteins 0.000 title description 4
- 238000011275 oncology therapy Methods 0.000 title description 2
- 239000003112 inhibitor Substances 0.000 claims abstract description 288
- 102100038078 CD276 antigen Human genes 0.000 claims abstract description 226
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 225
- 101710185679 CD276 antigen Proteins 0.000 claims abstract description 222
- 239000000203 mixture Substances 0.000 claims abstract description 124
- 208000037966 cold tumor Diseases 0.000 claims abstract description 30
- 101000884279 Homo sapiens CD276 antigen Proteins 0.000 claims abstract description 4
- 210000004027 cell Anatomy 0.000 claims description 152
- 101000868279 Homo sapiens Leukocyte surface antigen CD47 Proteins 0.000 claims description 60
- 102100032913 Leukocyte surface antigen CD47 Human genes 0.000 claims description 60
- 201000001441 melanoma Diseases 0.000 claims description 59
- 230000014509 gene expression Effects 0.000 claims description 56
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 41
- 102000039446 nucleic acids Human genes 0.000 claims description 39
- 108020004707 nucleic acids Proteins 0.000 claims description 39
- 150000007523 nucleic acids Chemical class 0.000 claims description 39
- 101150036449 SIRPA gene Proteins 0.000 claims description 37
- 239000003446 ligand Substances 0.000 claims description 37
- 238000002560 therapeutic procedure Methods 0.000 claims description 28
- 230000011664 signaling Effects 0.000 claims description 26
- 108010021625 Immunoglobulin Fragments Proteins 0.000 claims description 23
- 102000008394 Immunoglobulin Fragments Human genes 0.000 claims description 23
- 108091033319 polynucleotide Proteins 0.000 claims description 22
- 102000040430 polynucleotide Human genes 0.000 claims description 22
- 239000002157 polynucleotide Substances 0.000 claims description 22
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 22
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 claims description 21
- 150000003384 small molecules Chemical class 0.000 claims description 20
- 206010009944 Colon cancer Diseases 0.000 claims description 17
- 208000001333 Colorectal Neoplasms Diseases 0.000 claims description 17
- 201000008129 pancreatic ductal adenocarcinoma Diseases 0.000 claims description 17
- 210000005007 innate immune system Anatomy 0.000 claims description 16
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 14
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 claims description 11
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 claims description 10
- 238000003782 apoptosis assay Methods 0.000 claims description 10
- 230000002496 gastric effect Effects 0.000 claims description 9
- 230000036210 malignancy Effects 0.000 claims description 9
- JODKFOVZURLVTG-UHFFFAOYSA-N 2-bromo-1-(3,3-dinitroazetidin-1-yl)ethanone Chemical compound [O-][N+](=O)C1([N+]([O-])=O)CN(C(=O)CBr)C1 JODKFOVZURLVTG-UHFFFAOYSA-N 0.000 claims description 8
- DROLGCKUCMGNSF-UHFFFAOYSA-N 3,3-dinitroazetidine Chemical class [O-][N+](=O)C1([N+]([O-])=O)CNC1 DROLGCKUCMGNSF-UHFFFAOYSA-N 0.000 claims description 8
- 229940125979 ALX148 Drugs 0.000 claims description 8
- 108700001691 ALX148 Proteins 0.000 claims description 8
- 206010006187 Breast cancer Diseases 0.000 claims description 8
- 208000026310 Breast neoplasm Diseases 0.000 claims description 8
- 102000014150 Interferons Human genes 0.000 claims description 8
- 108010050904 Interferons Proteins 0.000 claims description 8
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 8
- 206010033128 Ovarian cancer Diseases 0.000 claims description 8
- 206010061535 Ovarian neoplasm Diseases 0.000 claims description 8
- 229940126302 TTI-621 Drugs 0.000 claims description 8
- 208000008385 Urogenital Neoplasms Diseases 0.000 claims description 8
- 229950004270 enoblituzumab Drugs 0.000 claims description 8
- 201000010536 head and neck cancer Diseases 0.000 claims description 8
- 208000014829 head and neck neoplasm Diseases 0.000 claims description 8
- 229940079322 interferon Drugs 0.000 claims description 8
- 201000005202 lung cancer Diseases 0.000 claims description 8
- 208000020816 lung neoplasm Diseases 0.000 claims description 8
- 229940121476 omburtamab Drugs 0.000 claims description 8
- 102100032912 CD44 antigen Human genes 0.000 claims description 7
- 101000868273 Homo sapiens CD44 antigen Proteins 0.000 claims description 7
- 230000035772 mutation Effects 0.000 claims description 7
- 208000011932 ovarian sarcoma Diseases 0.000 claims description 7
- 101001117317 Homo sapiens Programmed cell death 1 ligand 1 Proteins 0.000 claims description 6
- 108010011536 PTEN Phosphohydrolase Proteins 0.000 claims description 6
- 230000002018 overexpression Effects 0.000 claims description 6
- 230000005522 programmed cell death Effects 0.000 claims description 5
- 108060008682 Tumor Necrosis Factor Proteins 0.000 claims description 3
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 claims description 3
- 239000002689 soil Substances 0.000 claims description 2
- 102000014160 PTEN Phosphohydrolase Human genes 0.000 claims 1
- 238000011282 treatment Methods 0.000 description 67
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 53
- 210000002540 macrophage Anatomy 0.000 description 40
- 239000008194 pharmaceutical composition Substances 0.000 description 40
- 238000002648 combination therapy Methods 0.000 description 37
- 108090000623 proteins and genes Proteins 0.000 description 37
- 201000010099 disease Diseases 0.000 description 36
- 201000011510 cancer Diseases 0.000 description 30
- 241000699670 Mus sp. Species 0.000 description 26
- 210000002865 immune cell Anatomy 0.000 description 23
- 210000004443 dendritic cell Anatomy 0.000 description 18
- 230000005764 inhibitory process Effects 0.000 description 18
- 230000004614 tumor growth Effects 0.000 description 18
- 208000035475 disorder Diseases 0.000 description 17
- 208000024891 symptom Diseases 0.000 description 17
- 206010061289 metastatic neoplasm Diseases 0.000 description 16
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 15
- 229920002477 rna polymer Polymers 0.000 description 15
- 238000003559 RNA-seq method Methods 0.000 description 14
- 230000001394 metastastic effect Effects 0.000 description 14
- 102000004169 proteins and genes Human genes 0.000 description 14
- 230000001225 therapeutic effect Effects 0.000 description 14
- 241001529936 Murinae Species 0.000 description 13
- 230000006870 function Effects 0.000 description 13
- 235000018102 proteins Nutrition 0.000 description 13
- 210000001519 tissue Anatomy 0.000 description 13
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 12
- 108020004459 Small interfering RNA Proteins 0.000 description 12
- 238000011161 development Methods 0.000 description 12
- 230000018109 developmental process Effects 0.000 description 12
- 239000012634 fragment Substances 0.000 description 12
- 210000000822 natural killer cell Anatomy 0.000 description 12
- 230000000694 effects Effects 0.000 description 11
- 239000013598 vector Substances 0.000 description 11
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 description 10
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 description 10
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 10
- 239000000427 antigen Substances 0.000 description 10
- 108091007433 antigens Proteins 0.000 description 10
- 102000036639 antigens Human genes 0.000 description 10
- 230000006041 cell recruitment Effects 0.000 description 10
- 108020004999 messenger RNA Proteins 0.000 description 10
- 238000004806 packaging method and process Methods 0.000 description 10
- 239000000523 sample Substances 0.000 description 10
- 238000011125 single therapy Methods 0.000 description 10
- 239000000243 solution Substances 0.000 description 10
- 230000002401 inhibitory effect Effects 0.000 description 9
- 101001037759 Homo sapiens Heat shock 70 kDa protein 1A Proteins 0.000 description 8
- 229940076838 Immune checkpoint inhibitor Drugs 0.000 description 8
- 238000011529 RT qPCR Methods 0.000 description 8
- 230000000259 anti-tumor effect Effects 0.000 description 8
- 210000003719 b-lymphocyte Anatomy 0.000 description 8
- 230000004663 cell proliferation Effects 0.000 description 8
- 239000012636 effector Substances 0.000 description 8
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 description 8
- 230000001105 regulatory effect Effects 0.000 description 8
- 102100030385 Granzyme B Human genes 0.000 description 7
- 102100040352 Heat shock 70 kDa protein 1A Human genes 0.000 description 7
- 101001009599 Homo sapiens Granzyme A Proteins 0.000 description 7
- 101001009603 Homo sapiens Granzyme B Proteins 0.000 description 7
- 101000979599 Homo sapiens Protein NKG7 Proteins 0.000 description 7
- 102100023370 Protein NKG7 Human genes 0.000 description 7
- 239000005557 antagonist Substances 0.000 description 7
- 230000008901 benefit Effects 0.000 description 7
- 239000000839 emulsion Substances 0.000 description 7
- 238000002474 experimental method Methods 0.000 description 7
- 238000003364 immunohistochemistry Methods 0.000 description 7
- 238000001990 intravenous administration Methods 0.000 description 7
- 230000003902 lesion Effects 0.000 description 7
- 230000007246 mechanism Effects 0.000 description 7
- 230000037361 pathway Effects 0.000 description 7
- 102000005962 receptors Human genes 0.000 description 7
- 108020003175 receptors Proteins 0.000 description 7
- 102000053642 Catalytic RNA Human genes 0.000 description 6
- 108090000994 Catalytic RNA Proteins 0.000 description 6
- 108091006020 Fc-tagged proteins Proteins 0.000 description 6
- 102100030386 Granzyme A Human genes 0.000 description 6
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 6
- 101000997835 Homo sapiens Tyrosine-protein kinase JAK1 Proteins 0.000 description 6
- 206010027480 Metastatic malignant melanoma Diseases 0.000 description 6
- 108700011259 MicroRNAs Proteins 0.000 description 6
- 241000699666 Mus <mouse, genus> Species 0.000 description 6
- 108091030071 RNAI Proteins 0.000 description 6
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 6
- 102100033438 Tyrosine-protein kinase JAK1 Human genes 0.000 description 6
- 239000004480 active ingredient Substances 0.000 description 6
- 238000009472 formulation Methods 0.000 description 6
- 230000009368 gene silencing by RNA Effects 0.000 description 6
- 238000009169 immunotherapy Methods 0.000 description 6
- 238000002347 injection Methods 0.000 description 6
- 239000007924 injection Substances 0.000 description 6
- 238000007912 intraperitoneal administration Methods 0.000 description 6
- 239000002502 liposome Substances 0.000 description 6
- 210000001165 lymph node Anatomy 0.000 description 6
- 239000000463 material Substances 0.000 description 6
- 210000002752 melanocyte Anatomy 0.000 description 6
- 208000021039 metastatic melanoma Diseases 0.000 description 6
- 239000002679 microRNA Substances 0.000 description 6
- 230000004044 response Effects 0.000 description 6
- 108091092562 ribozyme Proteins 0.000 description 6
- 238000012163 sequencing technique Methods 0.000 description 6
- 238000011746 C57BL/6J (JAX™ mouse strain) Methods 0.000 description 5
- 101000834898 Homo sapiens Alpha-synuclein Proteins 0.000 description 5
- 101000611936 Homo sapiens Programmed cell death protein 1 Proteins 0.000 description 5
- 101000652359 Homo sapiens Spermatogenesis-associated protein 2 Proteins 0.000 description 5
- 241000124008 Mammalia Species 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 102100032543 Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN Human genes 0.000 description 5
- 230000004913 activation Effects 0.000 description 5
- 239000002299 complementary DNA Substances 0.000 description 5
- 231100000135 cytotoxicity Toxicity 0.000 description 5
- 230000003013 cytotoxicity Effects 0.000 description 5
- 230000005860 defense response to virus Effects 0.000 description 5
- 239000002552 dosage form Substances 0.000 description 5
- 230000007717 exclusion Effects 0.000 description 5
- 210000002950 fibroblast Anatomy 0.000 description 5
- 238000003119 immunoblot Methods 0.000 description 5
- 230000008595 infiltration Effects 0.000 description 5
- 238000001764 infiltration Methods 0.000 description 5
- 238000001802 infusion Methods 0.000 description 5
- 210000000056 organ Anatomy 0.000 description 5
- -1 poly(2-hydroxyethyl-methacrylate) Polymers 0.000 description 5
- 230000002441 reversible effect Effects 0.000 description 5
- 239000003826 tablet Substances 0.000 description 5
- 210000004881 tumor cell Anatomy 0.000 description 5
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 5
- 102100032367 C-C motif chemokine 5 Human genes 0.000 description 4
- 108010055166 Chemokine CCL5 Proteins 0.000 description 4
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 4
- 101000617130 Homo sapiens Stromal cell-derived factor 1 Proteins 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- 102100021669 Stromal cell-derived factor 1 Human genes 0.000 description 4
- 239000000654 additive Substances 0.000 description 4
- 230000000996 additive effect Effects 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 4
- 238000009826 distribution Methods 0.000 description 4
- 210000003979 eosinophil Anatomy 0.000 description 4
- 230000030279 gene silencing Effects 0.000 description 4
- 239000012528 membrane Substances 0.000 description 4
- 238000002493 microarray Methods 0.000 description 4
- 239000003094 microcapsule Substances 0.000 description 4
- 238000010172 mouse model Methods 0.000 description 4
- 238000001543 one-way ANOVA Methods 0.000 description 4
- 239000006187 pill Substances 0.000 description 4
- 230000009467 reduction Effects 0.000 description 4
- 239000007787 solid Substances 0.000 description 4
- 238000010254 subcutaneous injection Methods 0.000 description 4
- 239000007929 subcutaneous injection Substances 0.000 description 4
- 239000004094 surface-active agent Substances 0.000 description 4
- 108010074708 B7-H1 Antigen Proteins 0.000 description 3
- WVDDGKGOMKODPV-UHFFFAOYSA-N Benzyl alcohol Chemical compound OCC1=CC=CC=C1 WVDDGKGOMKODPV-UHFFFAOYSA-N 0.000 description 3
- 238000011740 C57BL/6 mouse Methods 0.000 description 3
- 229940045513 CTLA4 antagonist Drugs 0.000 description 3
- 102000004127 Cytokines Human genes 0.000 description 3
- 108090000695 Cytokines Proteins 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- 101001037968 Homo sapiens Heat shock 70 kDa protein 1B Proteins 0.000 description 3
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 3
- 102000037984 Inhibitory immune checkpoint proteins Human genes 0.000 description 3
- 108091008026 Inhibitory immune checkpoint proteins Proteins 0.000 description 3
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 3
- 239000012270 PD-1 inhibitor Substances 0.000 description 3
- 239000012668 PD-1-inhibitor Substances 0.000 description 3
- 239000002202 Polyethylene glycol Substances 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 238000011319 anticancer therapy Methods 0.000 description 3
- 238000001574 biopsy Methods 0.000 description 3
- 230000000903 blocking effect Effects 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 239000000969 carrier Substances 0.000 description 3
- 229920001577 copolymer Polymers 0.000 description 3
- 239000003937 drug carrier Substances 0.000 description 3
- 239000008103 glucose Substances 0.000 description 3
- 230000002519 immonomodulatory effect Effects 0.000 description 3
- 230000001506 immunosuppresive effect Effects 0.000 description 3
- 239000004615 ingredient Substances 0.000 description 3
- 238000007918 intramuscular administration Methods 0.000 description 3
- 238000009533 lab test Methods 0.000 description 3
- 239000010410 layer Substances 0.000 description 3
- 239000011159 matrix material Substances 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 239000003921 oil Substances 0.000 description 3
- 235000019198 oils Nutrition 0.000 description 3
- 230000036961 partial effect Effects 0.000 description 3
- 229940121655 pd-1 inhibitor Drugs 0.000 description 3
- 150000003904 phospholipids Chemical class 0.000 description 3
- 230000004962 physiological condition Effects 0.000 description 3
- 229920001223 polyethylene glycol Polymers 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 238000011002 quantification Methods 0.000 description 3
- 230000007115 recruitment Effects 0.000 description 3
- 230000002829 reductive effect Effects 0.000 description 3
- 210000003289 regulatory T cell Anatomy 0.000 description 3
- 238000003757 reverse transcription PCR Methods 0.000 description 3
- 238000003860 storage Methods 0.000 description 3
- 238000001356 surgical procedure Methods 0.000 description 3
- 239000000725 suspension Substances 0.000 description 3
- 238000013268 sustained release Methods 0.000 description 3
- 239000012730 sustained-release form Substances 0.000 description 3
- 230000002195 synergetic effect Effects 0.000 description 3
- 238000009966 trimming Methods 0.000 description 3
- 239000003656 tris buffered saline Substances 0.000 description 3
- 239000013603 viral vector Substances 0.000 description 3
- 102100035473 2'-5'-oligoadenylate synthase-like protein Human genes 0.000 description 2
- 206010067484 Adverse reaction Diseases 0.000 description 2
- 102000035485 Allograft inflammatory factor 1 Human genes 0.000 description 2
- 108091010877 Allograft inflammatory factor 1 Proteins 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 102100024263 CD160 antigen Human genes 0.000 description 2
- 241000282472 Canis lupus familiaris Species 0.000 description 2
- 206010057250 Cell-mediated cytotoxicity Diseases 0.000 description 2
- 108010012236 Chemokines Proteins 0.000 description 2
- 102000019034 Chemokines Human genes 0.000 description 2
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 2
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 2
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- 102000053602 DNA Human genes 0.000 description 2
- 241000283086 Equidae Species 0.000 description 2
- 241000282326 Felis catus Species 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 102100040407 Heat shock 70 kDa protein 1B Human genes 0.000 description 2
- 101000597360 Homo sapiens 2'-5'-oligoadenylate synthase-like protein Proteins 0.000 description 2
- 101000761938 Homo sapiens CD160 antigen Proteins 0.000 description 2
- 101000852865 Homo sapiens Interferon alpha/beta receptor 2 Proteins 0.000 description 2
- 101001001420 Homo sapiens Interferon gamma receptor 1 Proteins 0.000 description 2
- 101001082058 Homo sapiens Interferon-induced protein with tetratricopeptide repeats 2 Proteins 0.000 description 2
- 101000775749 Homo sapiens Proto-oncogene vav Proteins 0.000 description 2
- 101000657037 Homo sapiens Radical S-adenosyl methionine domain-containing protein 2 Proteins 0.000 description 2
- 101000942626 Homo sapiens UMP-CMP kinase 2, mitochondrial Proteins 0.000 description 2
- 206010061218 Inflammation Diseases 0.000 description 2
- 102100036718 Interferon alpha/beta receptor 2 Human genes 0.000 description 2
- 102100035678 Interferon gamma receptor 1 Human genes 0.000 description 2
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 2
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 2
- 108010000817 Leuprolide Proteins 0.000 description 2
- 210000004322 M2 macrophage Anatomy 0.000 description 2
- 229930195725 Mannitol Natural products 0.000 description 2
- 108091034117 Oligonucleotide Proteins 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Natural products OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 241000288906 Primates Species 0.000 description 2
- 102100032190 Proto-oncogene vav Human genes 0.000 description 2
- 102100033749 Radical S-adenosyl methionine domain-containing protein 2 Human genes 0.000 description 2
- 108010044012 STAT1 Transcription Factor Proteins 0.000 description 2
- 108010011005 STAT6 Transcription Factor Proteins 0.000 description 2
- 102100029904 Signal transducer and activator of transcription 1-alpha/beta Human genes 0.000 description 2
- 102100023980 Signal transducer and activator of transcription 6 Human genes 0.000 description 2
- 206010041067 Small cell lung cancer Diseases 0.000 description 2
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- 229930006000 Sucrose Natural products 0.000 description 2
- 102100032947 UMP-CMP kinase 2, mitochondrial Human genes 0.000 description 2
- 108050003627 Wnt Proteins 0.000 description 2
- 102000013814 Wnt Human genes 0.000 description 2
- 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 description 2
- 230000001594 aberrant effect Effects 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 150000007513 acids Chemical class 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- 230000006838 adverse reaction Effects 0.000 description 2
- 239000000556 agonist Substances 0.000 description 2
- 230000000340 anti-metabolite Effects 0.000 description 2
- 230000030741 antigen processing and presentation Effects 0.000 description 2
- 229940100197 antimetabolite Drugs 0.000 description 2
- 239000002256 antimetabolite Substances 0.000 description 2
- 239000002246 antineoplastic agent Substances 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- 210000003651 basophil Anatomy 0.000 description 2
- 239000012472 biological sample Substances 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- YCIMNLLNPGFGHC-UHFFFAOYSA-N catechol Chemical compound OC1=CC=CC=C1O YCIMNLLNPGFGHC-UHFFFAOYSA-N 0.000 description 2
- 239000013592 cell lysate Substances 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 238000002512 chemotherapy Methods 0.000 description 2
- 208000006990 cholangiocarcinoma Diseases 0.000 description 2
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 2
- 238000011284 combination treatment Methods 0.000 description 2
- 238000007796 conventional method Methods 0.000 description 2
- 210000004748 cultured cell Anatomy 0.000 description 2
- 230000001461 cytolytic effect Effects 0.000 description 2
- 230000001934 delay Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 230000004064 dysfunction Effects 0.000 description 2
- 210000002889 endothelial cell Anatomy 0.000 description 2
- 239000012055 enteric layer Substances 0.000 description 2
- 210000002615 epidermis Anatomy 0.000 description 2
- LZCLXQDLBQLTDK-UHFFFAOYSA-N ethyl 2-hydroxypropanoate Chemical compound CCOC(=O)C(C)O LZCLXQDLBQLTDK-UHFFFAOYSA-N 0.000 description 2
- 238000011990 functional testing Methods 0.000 description 2
- 230000004547 gene signature Effects 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- BXWNKGSJHAJOGX-UHFFFAOYSA-N hexadecan-1-ol Chemical compound CCCCCCCCCCCCCCCCO BXWNKGSJHAJOGX-UHFFFAOYSA-N 0.000 description 2
- 230000001900 immune effect Effects 0.000 description 2
- 210000000987 immune system Anatomy 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 230000002757 inflammatory effect Effects 0.000 description 2
- 230000004054 inflammatory process Effects 0.000 description 2
- 238000001361 intraarterial administration Methods 0.000 description 2
- 238000007919 intrasynovial administration Methods 0.000 description 2
- 238000007913 intrathecal administration Methods 0.000 description 2
- 230000002601 intratumoral effect Effects 0.000 description 2
- RGLRXNKKBLIBQS-XNHQSDQCSA-N leuprolide acetate Chemical compound CC(O)=O.CCNC(=O)[C@@H]1CCCN1C(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H]1NC(=O)CC1)CC1=CC=C(O)C=C1 RGLRXNKKBLIBQS-XNHQSDQCSA-N 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- RLSSMJSEOOYNOY-UHFFFAOYSA-N m-cresol Chemical compound CC1=CC=CC(O)=C1 RLSSMJSEOOYNOY-UHFFFAOYSA-N 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 239000000594 mannitol Substances 0.000 description 2
- 235000010355 mannitol Nutrition 0.000 description 2
- 230000001404 mediated effect Effects 0.000 description 2
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 2
- 239000004005 microsphere Substances 0.000 description 2
- 238000002156 mixing Methods 0.000 description 2
- 210000001616 monocyte Anatomy 0.000 description 2
- 230000031942 natural killer cell mediated cytotoxicity Effects 0.000 description 2
- 210000000440 neutrophil Anatomy 0.000 description 2
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 2
- 231100000252 nontoxic Toxicity 0.000 description 2
- 230000003000 nontoxic effect Effects 0.000 description 2
- 238000010606 normalization Methods 0.000 description 2
- 239000012188 paraffin wax Substances 0.000 description 2
- AQIXEPGDORPWBJ-UHFFFAOYSA-N pentan-3-ol Chemical compound CCC(O)CC AQIXEPGDORPWBJ-UHFFFAOYSA-N 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- 230000002035 prolonged effect Effects 0.000 description 2
- QELSKZZBTMNZEB-UHFFFAOYSA-N propylparaben Chemical compound CCCOC(=O)C1=CC=C(O)C=C1 QELSKZZBTMNZEB-UHFFFAOYSA-N 0.000 description 2
- 238000003498 protein array Methods 0.000 description 2
- 238000001959 radiotherapy Methods 0.000 description 2
- GHMLBKRAJCXXBS-UHFFFAOYSA-N resorcinol Chemical compound OC1=CC=CC(O)=C1 GHMLBKRAJCXXBS-UHFFFAOYSA-N 0.000 description 2
- 150000003839 salts Chemical class 0.000 description 2
- 238000007493 shaping process Methods 0.000 description 2
- 238000012174 single-cell RNA sequencing Methods 0.000 description 2
- 210000003491 skin Anatomy 0.000 description 2
- 229910052708 sodium Inorganic materials 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- 239000008247 solid mixture Substances 0.000 description 2
- 239000000600 sorbitol Substances 0.000 description 2
- 235000010356 sorbitol Nutrition 0.000 description 2
- 239000003549 soybean oil Substances 0.000 description 2
- 235000012424 soybean oil Nutrition 0.000 description 2
- 239000003381 stabilizer Substances 0.000 description 2
- 210000000130 stem cell Anatomy 0.000 description 2
- 230000004936 stimulating effect Effects 0.000 description 2
- 210000002536 stromal cell Anatomy 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- WYWHKKSPHMUBEB-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 0.000 description 2
- 230000000699 topical effect Effects 0.000 description 2
- 230000032258 transport Effects 0.000 description 2
- 210000004981 tumor-associated macrophage Anatomy 0.000 description 2
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 description 2
- 238000002604 ultrasonography Methods 0.000 description 2
- 230000003612 virological effect Effects 0.000 description 2
- HDTRYLNUVZCQOY-UHFFFAOYSA-N α-D-glucopyranosyl-α-D-glucopyranoside Natural products OC1C(O)C(O)C(CO)OC1OC1C(O)C(O)C(O)C(CO)O1 HDTRYLNUVZCQOY-UHFFFAOYSA-N 0.000 description 1
- XMQUEQJCYRFIQS-YFKPBYRVSA-N (2s)-2-amino-5-ethoxy-5-oxopentanoic acid Chemical compound CCOC(=O)CC[C@H](N)C(O)=O XMQUEQJCYRFIQS-YFKPBYRVSA-N 0.000 description 1
- FPVKHBSQESCIEP-UHFFFAOYSA-N (8S)-3-(2-deoxy-beta-D-erythro-pentofuranosyl)-3,6,7,8-tetrahydroimidazo[4,5-d][1,3]diazepin-8-ol Natural products C1C(O)C(CO)OC1N1C(NC=NCC2O)=C2N=C1 FPVKHBSQESCIEP-UHFFFAOYSA-N 0.000 description 1
- GUAHPAJOXVYFON-ZETCQYMHSA-N (8S)-8-amino-7-oxononanoic acid zwitterion Chemical compound C[C@H](N)C(=O)CCCCCC(O)=O GUAHPAJOXVYFON-ZETCQYMHSA-N 0.000 description 1
- JLPULHDHAOZNQI-ZTIMHPMXSA-N 1-hexadecanoyl-2-(9Z,12Z-octadecadienoyl)-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCC\C=C/C\C=C/CCCCC JLPULHDHAOZNQI-ZTIMHPMXSA-N 0.000 description 1
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 1
- HIQIXEFWDLTDED-UHFFFAOYSA-N 4-hydroxy-1-piperidin-4-ylpyrrolidin-2-one Chemical compound O=C1CC(O)CN1C1CCNCC1 HIQIXEFWDLTDED-UHFFFAOYSA-N 0.000 description 1
- 229940080778 Adenosine deaminase inhibitor Drugs 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- 235000019489 Almond oil Nutrition 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 241000972773 Aulopiformes Species 0.000 description 1
- 102100029822 B- and T-lymphocyte attenuator Human genes 0.000 description 1
- 102000015735 Beta-catenin Human genes 0.000 description 1
- 108060000903 Beta-catenin Proteins 0.000 description 1
- 229920002799 BoPET Polymers 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 101001011741 Bos taurus Insulin Proteins 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 102100031172 C-C chemokine receptor type 1 Human genes 0.000 description 1
- 101710149814 C-C chemokine receptor type 1 Proteins 0.000 description 1
- 101150041972 CDKN2A gene Proteins 0.000 description 1
- 101150050673 CHK1 gene Proteins 0.000 description 1
- 101100463133 Caenorhabditis elegans pdl-1 gene Proteins 0.000 description 1
- GAGWJHPBXLXJQN-UORFTKCHSA-N Capecitabine Chemical compound C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1[C@H]1[C@H](O)[C@H](O)[C@@H](C)O1 GAGWJHPBXLXJQN-UORFTKCHSA-N 0.000 description 1
- GAGWJHPBXLXJQN-UHFFFAOYSA-N Capecitabine Natural products C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1C1C(O)C(O)C(C)O1 GAGWJHPBXLXJQN-UHFFFAOYSA-N 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 102000016362 Catenins Human genes 0.000 description 1
- 108010067316 Catenins Proteins 0.000 description 1
- KRKNYBCHXYNGOX-UHFFFAOYSA-K Citrate Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 1
- PTOAARAWEBMLNO-KVQBGUIXSA-N Cladribine Chemical compound C1=NC=2C(N)=NC(Cl)=NC=2N1[C@H]1C[C@H](O)[C@@H](CO)O1 PTOAARAWEBMLNO-KVQBGUIXSA-N 0.000 description 1
- 108010035532 Collagen Proteins 0.000 description 1
- 102000008186 Collagen Human genes 0.000 description 1
- 241000938605 Crocodylia Species 0.000 description 1
- UHDGCWIWMRVCDJ-CCXZUQQUSA-N Cytarabine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 UHDGCWIWMRVCDJ-CCXZUQQUSA-N 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 235000019739 Dicalciumphosphate Nutrition 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- SNRUBQQJIBEYMU-UHFFFAOYSA-N Dodecane Natural products CCCCCCCCCCCC SNRUBQQJIBEYMU-UHFFFAOYSA-N 0.000 description 1
- 101100310856 Drosophila melanogaster spri gene Proteins 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 208000000461 Esophageal Neoplasms Diseases 0.000 description 1
- 108700039887 Essential Genes Proteins 0.000 description 1
- 208000001382 Experimental Melanoma Diseases 0.000 description 1
- 102100028065 Fibulin-5 Human genes 0.000 description 1
- 240000008168 Ficus benjamina Species 0.000 description 1
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 1
- 102100031351 Galectin-9 Human genes 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 101100229077 Gallus gallus GAL9 gene Proteins 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 208000031448 Genomic Instability Diseases 0.000 description 1
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 1
- JZNWSCPGTDBMEW-UHFFFAOYSA-N Glycerophosphorylethanolamin Natural products NCCOP(O)(=O)OCC(O)CO JZNWSCPGTDBMEW-UHFFFAOYSA-N 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 102100034458 Hepatitis A virus cellular receptor 2 Human genes 0.000 description 1
- 206010073073 Hepatobiliary cancer Diseases 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000864344 Homo sapiens B- and T-lymphocyte attenuator Proteins 0.000 description 1
- 101001060252 Homo sapiens Fibulin-5 Proteins 0.000 description 1
- 101001068133 Homo sapiens Hepatitis A virus cellular receptor 2 Proteins 0.000 description 1
- 101001078133 Homo sapiens Integrin alpha-2 Proteins 0.000 description 1
- 101001055222 Homo sapiens Interleukin-8 Proteins 0.000 description 1
- 101001137987 Homo sapiens Lymphocyte activation gene 3 protein Proteins 0.000 description 1
- 101001117312 Homo sapiens Programmed cell death 1 ligand 2 Proteins 0.000 description 1
- 101001133941 Homo sapiens Prolyl 3-hydroxylase 1 Proteins 0.000 description 1
- 101000744536 Homo sapiens Ras-related protein Rab-27B Proteins 0.000 description 1
- 101001100101 Homo sapiens Retinoic acid-induced protein 3 Proteins 0.000 description 1
- 101000777277 Homo sapiens Serine/threonine-protein kinase Chk2 Proteins 0.000 description 1
- 101000669447 Homo sapiens Toll-like receptor 4 Proteins 0.000 description 1
- 101000666896 Homo sapiens V-type immunoglobulin domain-containing suppressor of T-cell activation Proteins 0.000 description 1
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical class C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 108060003951 Immunoglobulin Proteins 0.000 description 1
- 102100025305 Integrin alpha-2 Human genes 0.000 description 1
- 238000012695 Interfacial polymerization Methods 0.000 description 1
- 102100027303 Interferon-induced protein with tetratricopeptide repeats 2 Human genes 0.000 description 1
- 108090001007 Interleukin-8 Proteins 0.000 description 1
- 102000004890 Interleukin-8 Human genes 0.000 description 1
- 102100026236 Interleukin-8 Human genes 0.000 description 1
- 102000002698 KIR Receptors Human genes 0.000 description 1
- 108010043610 KIR Receptors Proteins 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- 229930182816 L-glutamine Natural products 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- 102000017578 LAG3 Human genes 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 241000713666 Lentivirus Species 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 102000043129 MHC class I family Human genes 0.000 description 1
- 108091054437 MHC class I family Proteins 0.000 description 1
- 239000004907 Macro-emulsion Substances 0.000 description 1
- 206010025652 Malignant melanoma in situ Diseases 0.000 description 1
- 102000002274 Matrix Metalloproteinases Human genes 0.000 description 1
- 108010000684 Matrix Metalloproteinases Proteins 0.000 description 1
- 108010061593 Member 14 Tumor Necrosis Factor Receptors Proteins 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 229940122255 Microtubule inhibitor Drugs 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- 239000005041 Mylar™ Substances 0.000 description 1
- FXHOOIRPVKKKFG-UHFFFAOYSA-N N,N-Dimethylacetamide Chemical compound CN(C)C(C)=O FXHOOIRPVKKKFG-UHFFFAOYSA-N 0.000 description 1
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 1
- 206010030155 Oesophageal carcinoma Diseases 0.000 description 1
- 238000009004 PCR Kit Methods 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 101150012195 PREB gene Proteins 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 206010057249 Phagocytosis Diseases 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 229920001214 Polysorbate 60 Polymers 0.000 description 1
- 101710094000 Programmed cell death 1 ligand 1 Proteins 0.000 description 1
- 102100024213 Programmed cell death 1 ligand 2 Human genes 0.000 description 1
- 102100034144 Prolyl 3-hydroxylase 1 Human genes 0.000 description 1
- 108010029485 Protein Isoforms Proteins 0.000 description 1
- 102000001708 Protein Isoforms Human genes 0.000 description 1
- 102100039765 Ras-related protein Rab-27B Human genes 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 102100038453 Retinoic acid-induced protein 3 Human genes 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 235000019485 Safflower oil Nutrition 0.000 description 1
- 206010039491 Sarcoma Diseases 0.000 description 1
- 102100031075 Serine/threonine-protein kinase Chk2 Human genes 0.000 description 1
- 108010071390 Serum Albumin Proteins 0.000 description 1
- 102000007562 Serum Albumin Human genes 0.000 description 1
- 229920001800 Shellac Polymers 0.000 description 1
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 1
- 244000258044 Solanum gilo Species 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 208000005718 Stomach Neoplasms Diseases 0.000 description 1
- 108010090804 Streptavidin Proteins 0.000 description 1
- 102100039360 Toll-like receptor 4 Human genes 0.000 description 1
- HDTRYLNUVZCQOY-WSWWMNSNSA-N Trehalose Natural products O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-WSWWMNSNSA-N 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 102100028785 Tumor necrosis factor receptor superfamily member 14 Human genes 0.000 description 1
- 108010079206 V-Set Domain-Containing T-Cell Activation Inhibitor 1 Proteins 0.000 description 1
- 102100038929 V-set domain-containing T-cell activation inhibitor 1 Human genes 0.000 description 1
- 102100038282 V-type immunoglobulin domain-containing suppressor of T-cell activation Human genes 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- 230000004156 Wnt signaling pathway Effects 0.000 description 1
- 229940081735 acetylcellulose Drugs 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 239000002487 adenosine deaminase inhibitor Substances 0.000 description 1
- 125000000217 alkyl group Chemical group 0.000 description 1
- 239000008168 almond oil Substances 0.000 description 1
- HDTRYLNUVZCQOY-LIZSDCNHSA-N alpha,alpha-trehalose Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-LIZSDCNHSA-N 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 229940024606 amino acid Drugs 0.000 description 1
- 235000001014 amino acid Nutrition 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 229940124650 anti-cancer therapies Drugs 0.000 description 1
- 230000003095 anti-phagocytic effect Effects 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 235000006708 antioxidants Nutrition 0.000 description 1
- 230000005975 antitumor immune response Effects 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 239000003125 aqueous solvent Substances 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 238000003491 array Methods 0.000 description 1
- 210000003567 ascitic fluid Anatomy 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 229960000686 benzalkonium chloride Drugs 0.000 description 1
- 229960001950 benzethonium chloride Drugs 0.000 description 1
- UREZNYTWGJKWBI-UHFFFAOYSA-M benzethonium chloride Chemical compound [Cl-].C1=CC(C(C)(C)CC(C)(C)C)=CC=C1OCCOCC[N+](C)(C)CC1=CC=CC=C1 UREZNYTWGJKWBI-UHFFFAOYSA-M 0.000 description 1
- 235000019445 benzyl alcohol Nutrition 0.000 description 1
- CADWTSSKOVRVJC-UHFFFAOYSA-N benzyl(dimethyl)azanium;chloride Chemical compound [Cl-].C[NH+](C)CC1=CC=CC=C1 CADWTSSKOVRVJC-UHFFFAOYSA-N 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- HUTDDBSSHVOYJR-UHFFFAOYSA-H bis[(2-oxo-1,3,2$l^{5},4$l^{2}-dioxaphosphaplumbetan-2-yl)oxy]lead Chemical compound [Pb+2].[Pb+2].[Pb+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O HUTDDBSSHVOYJR-UHFFFAOYSA-H 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- IXIBAKNTJSCKJM-BUBXBXGNSA-N bovine insulin Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@H]1CSSC[C@H]2C(=O)N[C@@H](C)C(=O)N[C@@H](CO)C(=O)N[C@H](C(=O)N[C@H](C(N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3C=CC(O)=CC=3)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3NC=NC=3)NC(=O)[C@H](CO)NC(=O)CNC1=O)C(=O)NCC(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(O)=O)C(=O)N[C@@H](CC(N)=O)C(O)=O)=O)CSSC[C@@H](C(N2)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@@H](NC(=O)CN)[C@@H](C)CC)C(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@@H](NC(=O)[C@@H](N)CC=1C=CC=CC=1)C(C)C)C1=CN=CN1 IXIBAKNTJSCKJM-BUBXBXGNSA-N 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- UDSAIICHUKSCKT-UHFFFAOYSA-N bromophenol blue Chemical compound C1=C(Br)C(O)=C(Br)C=C1C1(C=2C=C(Br)C(O)=C(Br)C=2)C2=CC=CC=C2S(=O)(=O)O1 UDSAIICHUKSCKT-UHFFFAOYSA-N 0.000 description 1
- DQXBYHZEEUGOBF-UHFFFAOYSA-N but-3-enoic acid;ethene Chemical compound C=C.OC(=O)CC=C DQXBYHZEEUGOBF-UHFFFAOYSA-N 0.000 description 1
- LRHPLDYGYMQRHN-UHFFFAOYSA-N butyl alcohol Substances CCCCO LRHPLDYGYMQRHN-UHFFFAOYSA-N 0.000 description 1
- 125000000484 butyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 229960004117 capecitabine Drugs 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000022534 cell killing Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 230000008614 cellular interaction Effects 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- 229920002301 cellulose acetate Polymers 0.000 description 1
- 229960000541 cetyl alcohol Drugs 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 235000013330 chicken meat Nutrition 0.000 description 1
- 108700010039 chimeric receptor Proteins 0.000 description 1
- 235000012000 cholesterol Nutrition 0.000 description 1
- 229960002436 cladribine Drugs 0.000 description 1
- 238000005354 coacervation Methods 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 229920001436 collagen Polymers 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 238000002591 computed tomography Methods 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 230000000875 corresponding effect Effects 0.000 description 1
- 230000000139 costimulatory effect Effects 0.000 description 1
- 235000012343 cottonseed oil Nutrition 0.000 description 1
- 239000002385 cottonseed oil Substances 0.000 description 1
- HPXRVTGHNJAIIH-UHFFFAOYSA-N cyclohexanol Chemical compound OC1CCCCC1 HPXRVTGHNJAIIH-UHFFFAOYSA-N 0.000 description 1
- 229960000684 cytarabine Drugs 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 210000003770 dc.pdc.8+ Anatomy 0.000 description 1
- 239000003405 delayed action preparation Substances 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 238000002405 diagnostic procedure Methods 0.000 description 1
- NEFBYIFKOOEVPA-UHFFFAOYSA-K dicalcium phosphate Chemical compound [Ca+2].[Ca+2].[O-]P([O-])([O-])=O NEFBYIFKOOEVPA-UHFFFAOYSA-K 0.000 description 1
- 229940038472 dicalcium phosphate Drugs 0.000 description 1
- 229910000390 dicalcium phosphate Inorganic materials 0.000 description 1
- 150000002016 disaccharides Chemical class 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 238000012377 drug delivery Methods 0.000 description 1
- 210000001198 duodenum Anatomy 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 239000002702 enteric coating Substances 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 229940116333 ethyl lactate Drugs 0.000 description 1
- DNJIEGIFACGWOD-UHFFFAOYSA-N ethyl mercaptane Natural products CCS DNJIEGIFACGWOD-UHFFFAOYSA-N 0.000 description 1
- 239000005038 ethylene vinyl acetate Substances 0.000 description 1
- 238000001704 evaporation Methods 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 239000013604 expression vector Substances 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 210000003754 fetus Anatomy 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 238000009459 flexible packaging Methods 0.000 description 1
- 229960000390 fludarabine Drugs 0.000 description 1
- GIUYCYHIANZCFB-FJFJXFQQSA-N fludarabine phosphate Chemical compound C1=NC=2C(N)=NC(F)=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@@H]1O GIUYCYHIANZCFB-FJFJXFQQSA-N 0.000 description 1
- 229960002949 fluorouracil Drugs 0.000 description 1
- IJJVMEJXYNJXOJ-UHFFFAOYSA-N fluquinconazole Chemical compound C=1C=C(Cl)C=C(Cl)C=1N1C(=O)C2=CC(F)=CC=C2N=C1N1C=NC=N1 IJJVMEJXYNJXOJ-UHFFFAOYSA-N 0.000 description 1
- 239000004052 folic acid antagonist Substances 0.000 description 1
- 239000007789 gas Substances 0.000 description 1
- 206010017758 gastric cancer Diseases 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 229960005277 gemcitabine Drugs 0.000 description 1
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 1
- 238000010199 gene set enrichment analysis Methods 0.000 description 1
- 230000004077 genetic alteration Effects 0.000 description 1
- 231100000118 genetic alteration Toxicity 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 239000010931 gold Substances 0.000 description 1
- 229910052737 gold Inorganic materials 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 230000007773 growth pattern Effects 0.000 description 1
- 229940093915 gynecological organic acid Drugs 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 239000008240 homogeneous mixture Substances 0.000 description 1
- 208000037967 hot tumor Diseases 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 239000000017 hydrogel Substances 0.000 description 1
- 229920001477 hydrophilic polymer Polymers 0.000 description 1
- 229920001600 hydrophobic polymer Polymers 0.000 description 1
- 229920003063 hydroxymethyl cellulose Polymers 0.000 description 1
- 229940031574 hydroxymethyl cellulose Drugs 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 230000005931 immune cell recruitment Effects 0.000 description 1
- 229940126546 immune checkpoint molecule Drugs 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 230000037451 immune surveillance Effects 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 102000018358 immunoglobulin Human genes 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 238000012744 immunostaining Methods 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 239000007972 injectable composition Substances 0.000 description 1
- 102000006495 integrins Human genes 0.000 description 1
- 108010044426 integrins Proteins 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000013152 interventional procedure Methods 0.000 description 1
- 230000004068 intracellular signaling Effects 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 229960004338 leuprorelin Drugs 0.000 description 1
- 108020001756 ligand binding domains Proteins 0.000 description 1
- 239000002960 lipid emulsion Substances 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 244000144972 livestock Species 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 229940087857 lupron Drugs 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 238000002595 magnetic resonance imaging Methods 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 238000013507 mapping Methods 0.000 description 1
- 229960001428 mercaptopurine Drugs 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Chemical class 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- 239000004292 methyl p-hydroxybenzoate Substances 0.000 description 1
- 229960002216 methylparaben Drugs 0.000 description 1
- 239000004530 micro-emulsion Substances 0.000 description 1
- 231100000782 microtubule inhibitor Toxicity 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 238000005065 mining Methods 0.000 description 1
- 210000003470 mitochondria Anatomy 0.000 description 1
- 238000001565 modulated differential scanning calorimetry Methods 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- CQDGTJPVBWZJAZ-UHFFFAOYSA-N monoethyl carbonate Chemical compound CCOC(O)=O CQDGTJPVBWZJAZ-UHFFFAOYSA-N 0.000 description 1
- 150000002772 monosaccharides Chemical class 0.000 description 1
- 230000000869 mutational effect Effects 0.000 description 1
- 210000000066 myeloid cell Anatomy 0.000 description 1
- 239000002088 nanocapsule Substances 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 239000002736 nonionic surfactant Substances 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- 238000002515 oligonucleotide synthesis Methods 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- LXCFILQKKLGQFO-UHFFFAOYSA-N p-hydroxybenzoic acid methyl ester Natural products COC(=O)C1=CC=C(O)C=C1 LXCFILQKKLGQFO-UHFFFAOYSA-N 0.000 description 1
- 239000005022 packaging material Substances 0.000 description 1
- 230000000242 pagocytic effect Effects 0.000 description 1
- 238000003068 pathway analysis Methods 0.000 description 1
- 229960002340 pentostatin Drugs 0.000 description 1
- FPVKHBSQESCIEP-JQCXWYLXSA-N pentostatin Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(N=CNC[C@H]2O)=C2N=C1 FPVKHBSQESCIEP-JQCXWYLXSA-N 0.000 description 1
- 230000008782 phagocytosis Effects 0.000 description 1
- 239000008024 pharmaceutical diluent Substances 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 238000009520 phase I clinical trial Methods 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- WTJKGGKOPKCXLL-RRHRGVEJSA-N phosphatidylcholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCC=CCCCCCCCC WTJKGGKOPKCXLL-RRHRGVEJSA-N 0.000 description 1
- 150000008104 phosphatidylethanolamines Chemical class 0.000 description 1
- 230000001766 physiological effect Effects 0.000 description 1
- 210000002381 plasma Anatomy 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920001200 poly(ethylene-vinyl acetate) Polymers 0.000 description 1
- 229920000747 poly(lactic acid) Polymers 0.000 description 1
- 229920002401 polyacrylamide Polymers 0.000 description 1
- 229920000728 polyester Polymers 0.000 description 1
- 229920002338 polyhydroxyethylmethacrylate Polymers 0.000 description 1
- 238000003752 polymerase chain reaction Methods 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 229920002451 polyvinyl alcohol Polymers 0.000 description 1
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 238000010837 poor prognosis Methods 0.000 description 1
- 239000011148 porous material Substances 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 208000037920 primary disease Diseases 0.000 description 1
- 238000000513 principal component analysis Methods 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 230000000770 proinflammatory effect Effects 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 238000011321 prophylaxis Methods 0.000 description 1
- 235000010232 propyl p-hydroxybenzoate Nutrition 0.000 description 1
- 239000004405 propyl p-hydroxybenzoate Substances 0.000 description 1
- 229960003415 propylparaben Drugs 0.000 description 1
- 239000000649 purine antagonist Substances 0.000 description 1
- 239000003790 pyrimidine antagonist Substances 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 108091006082 receptor inhibitors Proteins 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 208000037922 refractory disease Diseases 0.000 description 1
- 230000009711 regulatory function Effects 0.000 description 1
- 230000003252 repetitive effect Effects 0.000 description 1
- 230000008672 reprogramming Effects 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 230000029058 respiratory gaseous exchange Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- 108020004418 ribosomal RNA Proteins 0.000 description 1
- 235000005713 safflower oil Nutrition 0.000 description 1
- 239000003813 safflower oil Substances 0.000 description 1
- 235000019515 salmon Nutrition 0.000 description 1
- 239000012723 sample buffer Substances 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- ZLGIYFNHBLSMPS-ATJNOEHPSA-N shellac Chemical compound OCCCCCC(O)C(O)CCCCCCCC(O)=O.C1C23[C@H](C(O)=O)CCC2[C@](C)(CO)[C@@H]1C(C(O)=O)=C[C@@H]3O ZLGIYFNHBLSMPS-ATJNOEHPSA-N 0.000 description 1
- 229940113147 shellac Drugs 0.000 description 1
- 239000004208 shellac Substances 0.000 description 1
- 235000013874 shellac Nutrition 0.000 description 1
- 230000035939 shock Effects 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 208000000587 small cell lung carcinoma Diseases 0.000 description 1
- 235000017557 sodium bicarbonate Nutrition 0.000 description 1
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 229940083466 soybean lecithin Drugs 0.000 description 1
- 239000008347 soybean phospholipid Substances 0.000 description 1
- 125000006850 spacer group Chemical group 0.000 description 1
- 230000002269 spontaneous effect Effects 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 238000011272 standard treatment Methods 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- SFVFIFLLYFPGHH-UHFFFAOYSA-M stearalkonium chloride Chemical compound [Cl-].CCCCCCCCCCCCCCCCCC[N+](C)(C)CC1=CC=CC=C1 SFVFIFLLYFPGHH-UHFFFAOYSA-M 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 238000011146 sterile filtration Methods 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 201000011549 stomach cancer Diseases 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 239000001797 sucrose acetate isobutyrate Substances 0.000 description 1
- 235000010983 sucrose acetate isobutyrate Nutrition 0.000 description 1
- UVGUPMLLGBCFEJ-SWTLDUCYSA-N sucrose acetate isobutyrate Chemical compound CC(C)C(=O)O[C@H]1[C@H](OC(=O)C(C)C)[C@@H](COC(=O)C(C)C)O[C@@]1(COC(C)=O)O[C@@H]1[C@H](OC(=O)C(C)C)[C@@H](OC(=O)C(C)C)[C@H](OC(=O)C(C)C)[C@@H](COC(C)=O)O1 UVGUPMLLGBCFEJ-SWTLDUCYSA-N 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 238000001847 surface plasmon resonance imaging Methods 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 235000012222 talc Nutrition 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 229960003087 tioguanine Drugs 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 238000012800 visualization Methods 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 239000008215 water for injection Substances 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- 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
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2818—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2827—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
- A61K2039/507—Comprising a combination of two or more separate antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
Definitions
- the present disclosure generally relates to compositions and methods for treating a solid tumor in a subject, wherein administering compositions having a combination of an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa can inflame a cold tumor microenvironment.
- an inhibitor of B7-H3 CD276
- an inhibitor of CD47-SIRPa can inflame a cold tumor microenvironment.
- Immunotherapies have shown to be effective for treating a variety of immune-related diseases, including cancers.
- cancers appear to be resistant to such therapies due to a number of immunological obstacles. These can include the ability of tumors to foster a tolerant microenvironment and the activation of a plethora of immunosuppressive mechanisms, which may act in concert to counteract effective immune responses. For instance, tumor-associated macrophages, marrow-derived suppressor cells, tumor-associated neutrophils, cancer-associated fibroblasts, and regulatory T cell interactions actively promote tumorigenesis.
- compositions herein may be pharmaceutical compositions comprising an inhibitor of B7-H3 (CD276), an inhibitor of CD47- SIRPa, and/or at least one pharmaceutically acceptable excipient.
- a subject in need thereof can be non-responsive to anti-programmed cell death 1/programmed cell death ligand 1 (PD1/PDL1) therapy.
- PD1/PDL1 anti-programmed cell death 1/programmed cell death ligand 1
- a subject can be a human patient having a solid tumor selected from the group of pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, sarcomas, or any combination thereof.
- a subject can be a human patient having a melanoma.
- methods disclosed herein can include administration of a composition and/or pharmaceutical composition herein wherein the effective amount of the composition and/or pharmaceutical composition herein may be sufficient to activate the innate immune system.
- methods disclosed herein can include administration of a composition and/or pharmaceutical composition herein wherein the effective amount of the composition and/or pharmaceutical composition herein may be sufficient to activate at least one population of innate immune system cells.
- populations of innate immune system cells activated by the methods herein can be macrophages, dendritic cells (DCs) and natural killer (NK) cells.
- methods disclosed herein can include administration of a composition and/or pharmaceutical composition herein wherein the effective amount of the composition and/or pharmaceutical composition herein may be sufficient to reduce tumor volume.
- methods herein can include administration of an inhibitor of B7-H3.
- methods herein can include administration of an inhibitor of B7- H3 wherein the inhibitor of B7-H3 can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment, or any combination thereof.
- an inhibitor of B7-H3 can be an antibody.
- an inhibitor of B7-H3 can be a full-length antibody or an antigen-binding fragment thereof.
- an inhibitor of B7-H3 can be a human antibody or a humanized antibody.
- an inhibitor of B7-H3 can be an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- an inhibitor of B7-H3 can be enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL-015, or any combination thereof.
- an inhibitor of B7-H3 can be a B7-H3-specific chimeric antigen receptor T (CAR-T) cell.
- methods herein can include administration of an inhibitor of CD47-SIRPa.
- an inhibitor of CD47-SIRPa can be a CD47-ligand inhibitor, a SIRPa-receptor inhibitor, or any combination thereof.
- methods herein can include administration of a CD47-ligand inhibitor.
- methods herein can include administration of a CD47-ligand inhibitor wherein the CD47-ligand inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- a CD47-ligand inhibitor can be an antibody.
- a CD47-ligand inhibitor can be a full-length antibody or an antigen-binding fragment thereof. In some embodiments, a CD47-ligand inhibitor can be a human antibody or a humanized antibody. In some embodiments, a CD47-ligand inhibitor can be an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody. In some embodiments, a CD47-ligand inhibitor can be a CD47-specific chimeric antigen receptor T (CAR- T) cell. In some embodiments, methods herein can include administration of a SIRPa-receptor inhibitor.
- methods herein can include administration of a SIRPa-receptor inhibitor wherein the SIRPa-receptor inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- a SIRPa-receptor inhibitor can be an antibody.
- a SIRPa-receptor inhibitor can be a full-length antibody or an antigen-binding fragment thereof.
- a SIRPa-receptor inhibitor can be a human antibody or a humanized antibody.
- a SIRPa-receptor inhibitor can be a SIRPa -specific chimeric antigen receptor T (CAR-T) cell.
- an inhibitor of CD47-SIRPa can be RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC-90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or any combination thereof.
- a method of inflaming a cold tumor microenvironment as disclosed herein can include administering to the cold tumor microenvironment a composition having an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa.
- a tumor having a cold tumor microenvironment can be a solid tumor.
- a tumor having a cold tumor microenvironment can be pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, and/or sarcoma.
- a tumor having a cold tumor microenvironment can be melanoma.
- a tumor having a cold tumor microenvironment can have at least one of B7-H3 overexpression, overactive P-catenin signaling, or any combination thereof.
- a tumor having a cold tumor microenvironment can have at least one genetic mutation comprising BRAF V600E , loss of PTEN, or a combination thereof.
- methods of inflaming a cold tumor microenvironment disclosed herein can increase of at least one interferon, increase of CD8+ T cells, increase of CD44 expression in CD4+ cells, in CD8+ cells, or both, increased TNFalpha expression in T cells, or any combination thereof.
- compositions for treating a solid tumor in a subject can include an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa.
- pharmaceutical compositions disclosed herein can further include at least one pharmaceutically acceptable excipient.
- pharmaceutical compositions disclosed herein can include an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa in a dosage amount that may restore CD8+ T cell effector function in a cold tumor microenvironment.
- kits are provided for transport, storage and use in treating or reducing the size of, reducing expansion of a tumor, and/or inflaming a cold tumor microenvironment as disclosed herein.
- Figs. 1A-1K are images depicting that B7-H3 was overexpressed in melanomas and regulated that tumor cell-mediated signals that influenced the tumor microenvironment.
- Fig. ID shows a representative image of positive immunostaining of B7-H3 protein expression in a tissue within the melanoma tissue array; Scale bar: 60 pm.
- Fig. IE is a graph showing that siRNA knockdown was confirmed in two human metastatic melanoma cell lines, Hs. 688(A)T and Hs. 839T, that were transfected with scrambled or B7-H3-specific siRNAs in triplicates using quantitative real-time PCR to assay for two isoforms of B7-H3.
- Fig. IF is an image of a heatmap of gene expression in Hs. 688(A)T and Hs. 839T cells that were transfected with scrambled or B7-H3-specific siRNAs, where RNA sequencing identified differentially expressing genes between the control and the silenced groups that were consistently up- or down- regulated in both cell lines.
- Fig. 1G is a representative immunoblot for B7-H3 showing B7-H3 protein expression in a panel of murine melanoma cell lines and immortalized murine melanocytes (melan-a).
- Fig. IF is an image of a heatmap of gene expression in Hs. 688(A)T and Hs. 839T cells that were transfected with scrambled or B7-H3-specific siRNAs, where RNA sequencing identified differentially expressing genes between the control and the silenced groups that were consistently up- or down- regulated in both cell lines.
- Fig. 1G is a representative immunoblot for B7
- FIG. 1H is a graph showing mRNA levels of B7-H3 as analyzed in a panel of murine melanoma cell lines and immortalized murine melanocytes (melan-a) by quantitative real-time PCR.
- Figs. II and 1J show that B7-H3 or P-catenin was silenced by gene-specific siRNAs in YUMM2.1 cells, silencing was confirmed by immunoblot (Fig. 1H), and mRNAs obtained from these cells were processed for B7-H3 downstream targets (shown in Fig. IF) in triplicates using quantitative real-time PCR (Fig. 1J).
- Fig. II and 1J show that B7-H3 or P-catenin was silenced by gene-specific siRNAs in YUMM2.1 cells, silencing was confirmed by immunoblot (Fig. 1H), and mRNAs obtained from these cells were processed for B7-H3 downstream targets (shown in Fig. IF) in triplicates using quantitative real-time
- RPPA Reverse Phase Protein Array
- CCLE Cancer Cell Line Encyclopedia
- FIG. 2A-2E are images depicting that B7-H3 inhibition led to an inflamed tumor microenvironment in P-catenin activated tumors.
- FIG. 2A is a graph showing tumor growth curves from tumor-bearing C57BL/6J mice that were subcutaneously injected with YUMM2.1 cells (1 x 10 6 ), harboring the Braf V600E/wt /Pten' / 7Cdkn2 +/ '/Bcat loxex3/wt genotype, to generate tumors where the mice were challenged with isotype control, anti-B7-H3 (600 pg, three times per week), anti-PD-1 (300 pg, every five days), or both (anti-B7-H3 and anti-PD-1) when tumor size reached 20-50 mm 3 (day 9).
- Fig. 2B is an image showing immune cell subgroups across treatment groups in RNA from the tumor (bulk) in triplicates after RNA was subjected to RNA sequencing. Enriched immune cell subgroups across treatment groups are shown.
- Fig. 2C is an image of a heatmap depicting selected genes and innate immune signals differentially expressed between the control and anti-B7-H3 treatment group.
- FIG. 2D is an image showing innate immune signals identified as enriched by RNA sequencing in the anti-B7-H3 treatment group where the tumors were harvested from tumor-bearing C57BL/6J mice that were subcutaneously injected with either YLTMM2.1 cells or YUMMERl .7 cells to generate tumors.
- Fig. 2E is an image of a heatmap depicting selected genes assessed in CD45+ immune cells that were sorted and subjected to ultra low RNA sequencing in triplicates in YLTMM2.1 tumors treated with anti-B7-H3, anti-PD-1, or control.
- Figs. 3A-3I depict images showing that B7-H3 inhibition resulted in macrophage infiltration and activation.
- Figs. 3A-3C are representative images showing IBA-1 (Allograft Inflammatory Factor 1) immunohistochemistry of tumors in treatment groups with IgG control (Fig. 3A), anti-B7-H3 (Fig. 3B), or anti-PD-1 (Fig. 3C) showing activated macrophages within the tissue.
- Figs. 3D-3F are representative images showing CD47 expression of the tumor cells by immunohistochemistry in treatment groups with IgG control (Fig. 3D), anti-B7-H3 (Fig. 3E), or anti-PD-1 (Fig. 3F).
- 3G-3I are representative images showing SIRPa expression, a receptor for CD47 in immune subpopulations such as macrophages and DCs, in treatment groups with IgG control (Fig. 3G), anti-B7-H3 (Fig. 3H), or anti-PD-1 (Fig. 31).
- FIGs. 4A-4F depict images showing that therapeutic synergy was achieved when anti-
- FIG. 4B is a graph showing tumor growth curves of tumors that were generated with YUMM2.1 cells from mice treated with isotype control, anti-PD-1 (300 pg, every five days), anti-CD47 (200 pg, three times per week), or both (anti-PD- 1 and anti-CD47).
- 4F is a graph showing tumor growth curves of tumors that were generated with YUMMER1.7 cells from mice treated with IgG, anti- B7-H3, anti-PD-1, anti-CD47, or combination of anti-B7-H3 and anti-CD47, or anti-PD-1 and anti-CD47.
- FIGs. 5A-5E depict images showing that therapeutic synergy was achieved when anti-
- FIG. 5B is a graph showing UMAP of immune cell distribution for CD45+ sorted cells subjected to single cell RNA sequencing (scRNA-sequencing) in the tumors described in Fig. 5A.
- Fig. 5C is a graph showing proportions of major immune cell subsets in each sample as analyzed by scRNA-sequencing.
- Fig. 5D is a heatmap depicting immune cell subtypes for each treatment group. The key for the x-axis is; I . DC (DC.103+1 IB-), 2. T cells (T.4); 3. Stromal cells (ST); 4. ILC (LIV.NK.DX5+); 5. B cells (B.Fo); 6. Macrophages (MFI05.II-); 7.
- ILC ILC
- NKT NKT
- NKT NKT
- B cells B.CD19CONTROL
- T cells T.8MEM
- T cells T.8SP
- T cells T.4SP
- Macrophages MFI05.II+
- 17 Stem cells SC
- NKT NKT
- NKT NKT
- T.44- 19. ILC (ILC1.CD127+); 20. Macrophages (MF.II+480LO); 21 . T cells (T.8NVE); 22. T cells (T.4.Pa); 23. Macrophages (MF. l 1C-11B+); 24.
- DC DC (DC.PDC.8- ); 25. B cells (B.GC); 26. Macrophages (MF.103-11B+); 27. Epithelial cells (Ep); 28. Macrophages (MF.II-480HI); 29. Fibroblasts (FI), 30. DC (DC.8+); 31. NKT (NKT.4+); 32. NK cells (NK.49CI+); 33. Macrophages (MF.103CLOSER); 34. T cells (T.4FP3-); 35. NK cells (NK.49C1-); 36 T cells (T.Tregs); 37. DC (DC.11B-); 38. Basophils (BA); 39. Monocytes (MO); 40. DC (DC.8-); 41.
- ILC ILC
- NK cells NK.B2M-
- Fig. 5E is a beatmap showing differentially expressed genes (x-axis) between the treatment groups (y-axis).
- Figs. 6A-6T depict images showing UMAP of differentially expressed genes GZMA (Figs. 6A-6D), GZMB (Figs. 6F-6I), HSPA1A (Figs. 6K-6N), HSPA1B (Figs. 6P-6S), and expression levels of GZMA (Fig. 6E), GZMB (Fig. 6J), HSPA1A (Fig. 60), an . HSPA 1B (Fig. 6T) in immune cell subsets. Significant P values are indicated.
- Figs. 7A-7O depict images showing UMAP of differentially expressed genes CCL5 (Figs. 7A-7D), CXCR4 (Figs. 7F-7I), NKG7 (Figs. 7K-7N) and expression levels of CCL5 (Fig. 7E), CXCR4 (Fig. 7J), and NKG7 (Fig. 70) in immune cell subsets. Significant P values are indicated.
- Figs. 8A-8E depict images showing enriched chemokine signaling upon combining anti-B7-H3 and anti-CD47.
- Immunotherapy using immune checkpoint inhibitors can be effective at treating cancer in a subset of patients having solid tumors, such as melanoma. While durable and complete responses are observed in patients, many do not respond or develop resistance.
- Objective response rate using immune checkpoint inhibitors such as anti-PD-1 (B7-H1) and/or anti-CTLA-4, can range from 10% to 40%.
- the present disclosure provides, in part, a unique combination therapy of immune checkpoint inhibitors with higher response rates that likely act by exerting distinct immune regulatory functions to restore the innate immune system surrounding the tumor.
- B7-H3 (CD276) is an immune regulatory molecule that belongs to the B7 superfamily. It is expressed on the membrane of the cancer cell, but its receptor is unknown.
- B7-H3 Aberrant protein expression of B7-H3 is associated with poor prognosis in many cancers including melanoma. Monoclonal antibodies against B7-H3 are tested in preclinical and phase I clinical studies. Despite the importance of immune checkpoint molecules in the cancer field, B7-H3 is less well studied than other checkpoints and its functions remain less well delineated.
- CD47 functions through a mechanism by which the cancer cell evades innate immune surveillance (macrophage immune checkpoint).
- CD47 is expressed on the membrane of the cancer cell.
- CD47 binds and activates SIRPa, an inhibitory protein expressed on the surface of myeloid cells - macrophages -, and functions as an anti-phagocytic or “don’t eat me signal”.
- SIRPa an inhibitory protein expressed on the surface of myeloid cells - macrophages -, and functions as an anti-phagocytic or “don’t eat me signal”.
- SIRPa an inhibitory protein expressed on the surface of myeloid cells - macrophages -, and functions as an anti-phagocytic or “don’t eat me signal”.
- SIRPa an inhibitory protein expressed on the surface of myeloid cells - macrophages -, and functions as an anti-phagocytic or “don’t eat me signal”.
- Activation of SIRPa inhibit
- compositions that include an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa which are capable of treating a solid tumor in a subject, and uses thereof for activating at least one population of innate immune system cells in a cold tumor microenvironment.
- articles “a” and “an” are used herein to refer to one or to more than one (i.e., at least one) of the grammatical object of the article.
- an element means at least one element and can include more than one element.
- “About” is used to provide flexibility to a numerical range endpoint by providing that a given value may be “slightly above” or “slightly below” the endpoint without affecting the desired result.
- the term “about” in association with a numerical value means that the numerical value can vary plus or minus by 5% or less of the numerical value.
- the transitional phrase “consisting essentially of’ (and grammatical variants) is to be interpreted as encompassing the recited materials or steps “and those that do not materially affect the basic and novel characteristic(s)” of the claimed invention.
- the term “consisting essentially of’ as used herein should not be interpreted as equivalent to “comprising.”
- the present disclosure also contemplates that in some embodiments, any feature or combination of features set forth herein can be excluded or omitted. To illustrate, if the specification states that a complex comprises components A, B and C, it is specifically intended that any of A, B or C, or a combination thereof, can be omitted and disclaimed singularly or in any combination.
- treatment refers to the clinical intervention made in response to a disease, disorder or physiological condition manifested by a patient or to which a patient may be susceptible.
- the aim of treatment includes the alleviation or prevention of symptoms, slowing or stopping the progression or worsening of a disease, disorder, or condition and/or the remission of the disease, disorder or condition.
- prevent refers to eliminating or delaying the onset of a particular disease, disorder or physiological condition, or to the reduction of the degree of severity of a particular disease, disorder or physiological condition, relative to the time and/or degree of onset or severity in the absence of intervention.
- an effective amount or “therapeutically effective amount” refers to an amount sufficient to effect beneficial or desirable biological and/or clinical results.
- “individual”, “subject”, “host”, and “patient” can be used interchangeably herein and refer to any mammalian subject for whom diagnosis, treatment, prophylaxis or therapy is desired, for example, humans, pets, livestock, horses or other animals.
- the term “subject” and “patient” are used interchangeably herein and refer to both human and nonhuman animals.
- the term “nonhuman animals” of the disclosure includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dog, cat, horse, cow, chickens, amphibians, reptiles, and the like.
- the subject can be a human.
- the subject can be a human in need of treating a solid tumor (e.g., melanoma).
- polynucleotide include deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), mRNA, oligonucleotides, and the like.
- vector refers to a nucleic acid used to introduce polynucleotides into a cell that has regulatory elements to provide expression of the heterologous nucleic acids in the cell.
- Vectors include but are not limited to plasmid, minicircles, yeast, and/or viral genomes. In some alternatives, the vectors are plasmid, minicircles, or viral genomes. In some alternatives, the vector is a viral vector. In some embodiments, the viral vector is a lentivirus. In some alternatives, the vector is a lentiviral vector. In some embodiments, the vector is a foamy viral vector, adenoviral vectors, retroviral vectors or lentiviral vectors.
- chimeric antigen receptor or “CAR” or “chimeric T cell receptor” refers herein to a synthetically designed receptor having a ligand binding domain of an antibody or another peptide sequence that binds to a molecule associated with the disease or disorder and is linked via a spacer domain to one or more intracellular signaling domains of a T cell or other receptors, such as a costimulatory domain.
- Chimeric receptor can also be referred to as artificial T cell receptors, chimeric T cell receptors, chimeric immunoreceptors, and chimeric antigen receptors (CARs).
- compositions encompassing an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa.
- Compositions herein can also be pharmaceutical compositions. a. B7-H3 Inhibitors
- compositions herein can include at least one inhibitor of B7- H3 (CD276).
- B7-H3 activity can be reduced through the use of an antagonist of the receptor, a partial antagonist or an antibody that either competes with the natural agonist, blocks the activity or encourages uptake of the B7-H3 molecule.
- a B7-H3 inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- a B7-H3 inhibitor can be an anti- B7-H3 antibody or fragment thereof capable of binding to B7-H3 and inhibiting or reducing B7-H3 activity.
- other B7-H3 inhibitors can include, but are not limited to, a B7-H3-specific siRNA, RNAi, microRNA or ribozyme.
- an inhibitor of B7-H3 can be an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- B7-H3 inhibitors and/or antagonists can include anti-B7-H3 specific antibody fragments (F(ab')2 or Fab'), single chain Fv, and Fc- fusion protein of B7-H3 ligands, e.g. Fc fusion proteins of 2B4 or CD2.
- an anti-B7-H3 antibody used herein can be a polyclonal or monoclonal antibody.
- a “humanized” anti-B7-H3 antibody can be used if needed in order to avoid any potential use incompatibilities (e.g. adverse reactions when introducing the ILC2 cells exposed to such an antibody, if needed).
- an inhibitor of B7-H3 can be a B7-H3-specific chimeric antigen receptor T (CAR-T) cell.
- an inhibitor of B7-H3 suitable for use herein can be, but is not limited to, enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL- 015, or a combination thereof.
- CD47-SIRPa Inhibitors [0044]
- compositions herein can include at least one inhibitor CD47- SIRPa.
- CD47-activity can be reduced through the use of an antagonist of the receptor (SIRPa), a partial antagonist or an antibody that either competes with the natural agonist, blocks the activity or encourages uptake of the CD47 molecule.
- SIRPa antagonist of the receptor
- a CD47-SIRPa inhibitor can be a CD47-ligand inhibitor.
- a CD47-SIRPa inhibitor can be a SIRPa-receptor inhibitor.
- a CD47 inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- a CD47 inhibitor can be an anti-CD47 antibody or fragment thereof capable of binding to CD47 and inhibiting or reducing CD47 activity.
- other CD47 inhibitors can include, but are not limited to, a CD47-specific siRNA, RNAi, microRNA or ribozyme.
- an inhibitor of CD47 can be an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- CD47 inhibitors and/or antagonists can include anti-CD47 specific antibody fragments (F(ab')2 or Fab'), single chain Fv, and Fc-fusion protein of B7-H3 ligands, e.g. Fc fusion proteins of 2B4 or CD2.
- an anti- CD47 antibody used herein can be a polyclonal or monoclonal antibody.
- a “humanized” anti-CD47 antibody can be used if needed in order to avoid any potential use incompatibilities.
- an inhibitor of CD47 can be a CD47-specific chimeric antigen receptor T (CAR-T) cell.
- a SIRPa inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- a SIRPa inhibitor can be an anti-SIRPa antibody or fragment thereof capable of binding to SIRPa and inhibiting or reducing SIRPa activity.
- other SIRPa inhibitors can include, but are not limited to, a SIRPa-specific siRNA, RNAi, microRNA or ribozyme.
- an inhibitor of SIRPa can be an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- SIRPa inhibitors and/or antagonists can include anti- SIRPa specific antibody fragments (F(ab')2 or Fab'), single chain Fv, and Fc-fusion protein of SIRPa, e.g. Fc fusion proteins of 2B4 or CD2.
- an anti-SIRPa antibody used herein can be a polyclonal or monoclonal antibody.
- a “humanized” anti-SIRPa antibody can be used if needed in order to avoid any potential use incompatibilities (e.g. adverse reactions when introducing the ILC2 cells exposed to such an antibody, if needed).
- an inhibitor of SIRPa can be a SIRPa -specific chimeric antigen receptor T (CAR-T) cell.
- an inhibitor of CD47-SIRPa suitable for use herein can be, but is not limited to, RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC-90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or a combination thereof.
- compositions herein may encompass an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa.
- compositions herein may encompass an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa, wherein a B7-H3 inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof and a CD47-SIRPa, can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- compositions herein may encompass a B7-H3-specific siRNA, RNAi, microRNA or ribozyme and a CD47-specific siRNA, RNAi, microRNA or ribozyme and/or a SIRPa-specific siRNA, RNAi, microRNA or ribozyme.
- compositions herein may encompass an anti-B7-H3 antibody and an anti-CD47 antibody and/or an anti-SIRPa antibody.
- compositions herein may encompass: enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL- 015, or any combination thereof; and RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC- 90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or any combination thereof.
- compositions herein may encompass: enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL- 015, or any combination thereof; and RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC- 90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M
- the B7-H3 inhibitors and/or CD47-SIRPa inhibitors, as described herein can be mixed with a pharmaceutically acceptable carrier (excipient) to form a pharmaceutical composition for use in treating a target disease.
- a pharmaceutically acceptable carrier excipient
- “Acceptable” means that the carrier must be compatible with the active ingredient of the composition (and preferably, capable of stabilizing the active ingredient) and not deleterious to the subject to be treated.
- compositions to be used in the present methods can comprise pharmaceutically acceptable carriers, excipients, or stabilizers in the form of lyophilized formulations or aqueous solutions.
- pharmaceutically acceptable carriers excipients, or stabilizers in the form of lyophilized formulations or aqueous solutions.
- Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations used, and comprise buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
- the pharmaceutical composition described herein comprises liposomes containing the antibodies (or the encoding nucleic acids) which can be prepared by methods known in the art, such as described in Epstein, et al., Proc. Natl. Acad. Sci. USA 82:3688 (1985); Hwang, et al., Proc. Natl. Acad. Sci. USA 77:4030 (1980); and U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556.
- Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
- PEG-PE PEG-derivatized phosphatidylethanolamine
- one or more of the B7-H3 inhibitors and/or CD47-SIRPa inhibitors can be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
- colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
- macroemulsions for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
- compositions described herein can be formulated in sustained-release format.
- sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the inhibitors, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
- sustained- release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinyl alcohol)), polylactides (U.S. Pat. No.
- copolymers ofL-glutamic acid and 7 ethyl-L-glutamate copolymers ofL-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid- glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(-)-3- hydroxybutyric acid.
- LUPRON DEPOTTM injectable microspheres composed of lactic acid- glycolic acid copolymer and leuprolide acetate
- sucrose acetate isobutyrate sucrose acetate isobutyrate
- poly-D-(-)-3- hydroxybutyric acid poly-D-(-)-3- hydroxybutyric acid.
- compositions to be used for in vivo administration must be sterile. This may be readily accomplished by, for example, filtration through sterile filtration membranes. Therapeutic compositions disclosed herein can be placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
- compositions described herein can be in unit dosage forms such as tablets, pills, capsules, powders, granules, solutions or suspensions, or suppositories, for oral, parenteral or rectal administration, or administration by inhalation or insufflation.
- the principal active ingredient can be mixed with a pharmaceutical carrier, e.g., conventional tableting ingredients such as com starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g., water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a non-toxic pharmaceutically acceptable salt thereof.
- a pharmaceutical carrier e.g., conventional tableting ingredients such as com starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g., water
- preformulation compositions when referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
- This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 900 mg of the active ingredient of the present invention.
- the tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
- the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
- the two components can be separated by an enteric layer that serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release.
- enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
- Suitable surface-active agents include, in particular, non-ionic agents, such as poly oxy ethylenesorbitans (e.g., TweenTM 20, 40, 60, 80 or 85) and other sorbitans (e.g., SpanTM 20, 40, 60, 80 or 85).
- Compositions with a surface-active agent are conveniently comprise between 0.05 and 5% surfaceactive agent, and can be between 0.1 and 2.5%. It are be appreciated that other ingredients may be added, for example mannitol or other pharmaceutically acceptable vehicles, if necessary.
- Suitable emulsions may be prepared using commercially available fat emulsions, such as IntralipidTM, LiposynTM, InfonutrolTM, LipofundinTM and LipiphysanTM.
- the active ingredient may be either dissolved in a pre-mixed emulsion composition or alternatively it may be dissolved in an oil (e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, com oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e.g. egg phospholipids, soybean phospholipids or soybean lecithin) and water.
- an oil e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, com oil or almond oil
- a phospholipid e.g. egg phospholipids, soybean phospholipids or soybean lecithin
- Suitable emulsions are typically contain up to 20% oil, for example, between 5 and 20%.
- the fat emulsion can comprise fat droplets between 0.1 and 1.0 .im, particularly 0.1 and 0.5 .im, and have a pH in the range of 5.5 to 8.0.
- the emulsion compositions can be those prepared by mixing an inhibitor with IntralipidTM or the components thereof (soybean oil, egg phospholipids, glycerol and water).
- compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
- the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above.
- the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
- Compositions in preferably sterile pharmaceutically acceptable solvents may be nebulized by use of gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device may be attached to a face mask, tent or intermittent positive pressure breathing machine. Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
- pharmaceutical formulations herein may comprise an anti-B7- H3 antibody at a suitable concentration, for example, about 200 pg to about 1000 pg (e.g., about 200 pg, about 250 pg, about 300 pg, about 350 pg, about 400 pg, about 450 pg, about 500 pg, about 550 pg, about 600 pg, about 650 pg, about 700 pg, about 750 pg, about 800 pg, about 850 pg, about 900 pg, about 950 pg, about 1000 pg).
- a suitable concentration for example, about 200 pg to about 1000 pg (e.g., about 200 pg, about 250 pg, about 300 pg, about 350 pg, about 400 pg, about 450 pg, about 500 pg, about 550 pg, about 600 pg, about 650 pg, about 700 pg, about 750 pg, about
- pharmaceutical formulations herein may comprise an anti-CD47 antibody at a suitable concentration, for example, about 50 pg to about 800 pg (e.g., about 50 pg, about 100 pg, about 150 pg, about 200 pg, about 250 pg, about 300 pg, about 350 pg, about 400 pg, about 450 pg, about 500 pg, about 550 pg, about 600 pg, about 650 pg, about 700 pg, about 750 pg, about 800 pg).
- pharmaceutical formulations herein may compromise both an anti-B7-H3 antibody and an anti- CD47 antibody.
- pharmaceutical formulations herein may comprise an anti- B7-H3 antibody at about 200 pg to about 1000 pg (e.g., about 200 pg, about 250 pg, about 300 pg, about 350 pg, about 400 pg, about 450 pg, about 500 pg, about 550 pg, about 600 pg, about 650 pg, about 700 pg, about 750 pg, about 800 pg, about 850 pg, about 900 pg, about 950 pg, about 1000 pg) and an anti-CD47 antibody at about 50 pg to about 800 pg (e.g., about 50 pg, about 100 pg, about 150 pg, about 200 pg, about 250 pg, about 300 pg, about 350 pg, about 400 pg, about 450 pg, about 500 pg, about 550 pg, about 600 pg, about 650 pg, about
- compositions disclosed herein may be packaged.
- packaging of a composition may be for storage, shipment, display for sale, or a combination thereof.
- compositions may be packaged using one or more suitable materials known in the art.
- compositions may be packaged using one or more suitable methods known in the art.
- the choice of packaging material and/or packaging method is dependent on the dosage form of a composition disclosed herein to be packaged.
- compositions disclosed herein may be packaged wherein packaging increases the length of time a composition can be stored.
- the “shelflife” of a composition is the length of time after formulation that a composition can maintain one or more physiological effects following administration to a subject as detailed herein.
- compositions disclosed herein may be packaged wherein packaging increases the shelf-life of a composition by about 1 week, about 1 month, or about 6 months.
- compositions disclosed herein may be packaged wherein packaging increases the length of time a composition can be stored at about -85°C to about -75°C.
- compositions disclosed herein may be packaged wherein packaging of at least one composition component increases the length of time that composition component can be stored at room temperature by about 1 week, about 1 month, or about 6 months. In some embodiments, compositions disclosed herein may be packaged wherein packaging of at least one composition component increases the length of time that composition component can be stored at about -85°C to about -75°C by about 1 week, about 1 month, or about 6 months.
- the present disclosure provides methods for treating solid tumors.
- the present disclosure provides methods for treating solid tumors (e.g., melanoma) that are not responsive to one or more anti-cancer therapies.
- the present disclosure provides methods for treating solid tumors (e.g., melanoma) that are not responsive to anti-programmed cell death 1/programmed cell death ligand 1 (PD1/PDL1) therapy using any of the compositions having an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa as disclosed herein.
- Also provided in the present disclosure are methods of inflaming a cold tumor microenvironment using any of the compositions having an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa as disclosed herein.
- a solid tumor can be pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, and sarcomas.
- PDA pancreatic ductal adenocarcinoma
- CRC colorectal cancer
- melanoma melanoma
- breast cancer lung cancer
- upper and lower gastrointestinal malignancies squamous cell head and neck cancer
- genitourinary cancer ovarian cancer
- sarcomas a solid tumor is a melanoma.
- a solid tumor to be treated by methods disclosed herein can have B7-H3 overexpression, overactive P-catenin signaling, or a combination thereof.
- a solid tumor to be treated by methods disclosed herein can have at least one genetic mutation.
- a solid tumor can have a genetic mutation of BRAF V600E .
- a solid tumor can have a genetic mutation resulting in the loss of PTEN.
- Certain embodiments herein can provide methods for treating solid tumors (e.g., melanoma) in a subject in need thereof.
- the subject to be treated by the methods described herein can be a mammal, more preferably a human. Mammals include, but are not limited to, farm animals, sport animals, pets, primates, horses, dogs, cats, mice and rats.
- a human subject who needs the treatment may be a human patient having, at risk for, or suspected of having a target disease/disorder, such as a solid tumor, including a melanoma.
- a subject to any of the methods herein can be any subject for whom treatment or therapy is desired.
- a subject can have or can be suspected of having cancer, a tumor, or any combination thereof.
- a subject can have or can be suspected of having one or more primary tumors, one or more metastatic tumors such as solid tumors or any combination thereof.
- a subject can be a mammal.
- a subject can be a human patient.
- a human patient such as an adult, child, adolescent, toddler, young adult or infant or fetus who is in need of the methods herein can be identified by routine medical examination, e.g., laboratory tests, biopsy, magnetic resonance imaging (MRI) scans, ultrasound exams, and the like.
- routine medical examination e.g., laboratory tests, biopsy, magnetic resonance imaging (MRI) scans, ultrasound exams, and the like.
- MRI magnetic resonance imaging
- a subject to be treated by the methods described herein can be a human patient having, suspected of having, or a risk for developing a solid tumor.
- a subject to be treated by the methods described herein can have a tumor wherein the tumor has at least one B7H3 + cell.
- a subject to be treated by the methods described herein can have a tumor wherein the tumor has at least one CD47 + cell.
- a subject to be treated by the methods described herein can have a tumor wherein the tumor has at least one CD47 + cell/B7H3 + cell.
- a human subject who needs the treatment may be a human patient having, at risk for, or suspected of having melanoma.
- Methods herein can be used to treat a human patient having a Stage 0, Stage I, Stage II, Stage III, or Stage IV melanoma.
- a Stage 0 melanoma (melanoma in situ) has not grown deeper than the top layer of the skin (the epidermis).
- a melanoma can be staged according to the Ameri can Joint Committee on Cancer (AJCC) TNM staging system.
- Stage 0 The cancer is confined to the epidermis, the outermost skin layer and has not spread to nearby lymph nodes or to distant parts of the body;
- Stage I The tumor is no more than 2 mm (2/25 of an inch) thick and might or might not be ulcerated and has not spread to nearby lymph nodes or to distant parts of the body;
- Stage II The tumor is more than 1 mm thick and may be thicker than 4 mm, it might or might not be ulcerated, and the cancer has not spread to nearby lymph nodes or to distant parts of the body;
- Stage IV - The tumor can be any thickness and might or might not be ulcerated, the cancer might or might not have spread to nearby lymph nodes, and it has spread to distant lymph nodes or to organs such as the
- the subject is a human patient who is in need of enhancing immunity.
- the human patient may have a solid tumor.
- solid tumor cancers include melanoma, pancreatic duct adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, cholangiocarcinoma, breast cancer, lung cancer (for example, non-small cell lung cancer, NSCLC, and small cell lung cancer, SCLC), upper and lower gastrointestinal malignancies (including, but not limited to, esophageal, gastric, and hepatobiliary cancer), squamous cell head and neck cancer, genitourinary cancers, ovarian cancer, and sarcomas.
- a subject having a solid tumor can be identified by routine medical examination, e.g., laboratory tests, organ functional tests, CT scans, or ultrasounds.
- the subject to be treated by the method described herein may be a human cancer patient who has undergone or is subjecting to an anticancer therapy, for example, chemotherapy, radiotherapy, immunotherapy, or surgery.
- treating refers to the application or administration of a composition including one or more active agents to a subject, who has a target disease or disorder, a symptom of the disease/disorder, or a predisposition toward the disease/disorder, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect the disorder, a symptom of the disease or disorder, or the predisposition toward the disease or disorder.
- Alleviating a target disease/disorder includes delaying the development or progression of the disease, or reducing disease severity or prolonging survival. Alleviating the disease or prolonging survival does not necessarily require curative results. As used therein, "delaying" the development of a target disease or disorder means to defer, hinder, slow, retard, stabilize, and/or postpone progression of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individuals being treated.
- a method that “delays” or alleviates the development of a disease, or delays the onset of the disease is a method that reduces probability of developing one or more symptoms of the disease in a given time frame and/or reduces extent of the symptoms in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a number of subjects sufficient to give a statistically significant result.
- “Development” or “progression” of a disease means initial manifestations and/or ensuing progression of the disease. Development of the disease can be detectable and assessed using standard clinical techniques as well known in the art. However, development also refers to progression that may be undetectable. For purpose of this disclosure, development or progression refers to the biological course of the symptoms. “Development” includes occurrence, recurrence, and onset. As used herein “onset” or “occurrence” of a target disease or disorder includes initial onset and/or recurrence.
- the present disclosure provides methods for reducing, ameliorating, or eliminating one or more symptom(s) associated with a solid tumor.
- the treatment methods disclosed herein involve the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa.
- the present disclosure provides methods of administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa for reducing, ameliorating, or eliminating one or more symptom(s) associated with a solid tumor.
- the present disclosure provides methods of administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa for reducing, ameliorating, or eliminating one or more symptom(s) associated with a solid tumor wherein administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as disclosed herein synergistically reduces, ameliorates, and/or eliminates one or more symptom(s) associated with a solid tumor.
- methods of administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa reduces, ameliorates, and/or eliminates one or more symptom(s) associated with a solid tumor at a faster rate compared to the rate after administering a single therapy of an inhibitor of B7-H3 or a single therapy of an inhibitor of CD47-SIRPa to a subject.
- methods of administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa herein reduces, ameliorates, and/or eliminates one or more symptom(s) associated with a solid tumor at a rate that can be at least about 10% faster, at least about 20% faster, at least about 30% faster, at least about 40% faster, or at least about 50% faster compared to the rate after administering a single therapy of an inhibitor of B7-H3 or a single therapy of an inhibitor of CD47-SIRPa to a subject.
- methods of administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa herein reduces, ameliorates, and/or eliminates one or more symptom(s) associated with a solid tumor at a rate that can be about 10% to about 99% faster, about 15% to about 95% faster, or about 20% to about 90% faster compared to the rate after administering a single therapy of an inhibitor of B7- H3 or a single therapy of an inhibitor of CD47-SIRPa to a subject.
- an effective amount of the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa can be given to a subject having a solid tumor e.g., melanoma), wherein the subject is on a treatment involving the one or more chemotherapeutics.
- an effective amount of the one or more chemotherapeutics are given to a subject having a solid tumor (e.g., melanoma), wherein the subject is on a treatment involving the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa.
- an effective amount of the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa and an effective amount of the one or more chemotherapeutics are given to the subject, concurrently or sequentially.
- the methods of the present disclosure increase anti-tumor activity (e.g., reduce cell proliferation, tumor growth, tumor volume, tumor burden, tumor load, and/or number of metastatic lesions over time) as compared to levels prior to treatment or in a control subject.
- anti-tumor activity e.g., reduce cell proliferation, tumor growth, tumor volume, tumor burden, tumor load, and/or number of metastatic lesions over time
- the methods of the present disclosure increase anti-tumor activity (e.g., reduce cell proliferation, tumor growth, tumor volume, tumor burden, tumor load, and/or number of metastatic lesions over time) by at least about 10%, least about 20%, least about 25%, least about 30%, least about 40%, least about 50%, least about 60%, least about 70%, least about 75%, least about 80%, least about 85%, least about 90%, least about 95%, or more as compared to levels prior to treatment or in a control subject.
- anti-tumor activity e.g., reduce cell proliferation, tumor growth, tumor volume, tumor burden, tumor load, and/or number of metastatic lesions over time
- the methods of the present disclosure increase anti-tumor activity (e.g., reduce cell proliferation, tumor growth, tumor volume, tumor burden, tumor load, and/or number of metastatic lesions over time) by at about 5% to about 99%, about 10% to about 95%, or about 15% to about 90% as compared to levels prior to treatment or in a control subject.
- the methods of the present disclosure synergistically increase anti-tumor activity (e.g., reduce cell proliferation, tumor growth, tumor volume, tumor burden, tumor load, and/or number of metastatic lesions over time) as compared to levels after treatment of a subject with a single therapy of an inhibitor of B7-H3 or a single therapy of an inhibitor of CD47-SIRPa.
- reduction may be measured by comparing cell proliferation, tumor growth, and/or tumor volume in a subject before and after administration of composition disclosed herein.
- a method of treating or ameliorating a cancer in a subject herein can ablate, reverse, and/or attenuate one or more symptoms of the cancer.
- a method of treating or ameliorating a cancer in a subject herein can allow one or more symptoms of the cancer to improve.
- a method of treating or ameliorating a cancer in a subject herein can allow one or more symptoms of the cancer to improve by at least about 10%, least about 20%, least about 30%, least about 40%, least about 50%, least about 60%, least about 70%, least about 80%, least about 90%, least about 95%, or more as compared to levels prior to treatment.
- a method of treating or ameliorating a cancer in a subject herein can allow one or more symptoms of the cancer to improve by at about 5% to about 99%, about 10% to about 95%, or about 15% to about 90% as compared to levels prior to treatment.
- cancerous cells and/or biomarkers in a subject can be measured in a biological sample, such as blood, serum, plasma, urine, peritoneal fluid, and/or a biopsy from a tissue or organ.
- methods herein can include administration of a composition herein to reduce tumor volume, size, load, and/or burden in a subject to an undetectable size, or less than the subject's tumor volume, size, load, and/or burden prior to treatment.
- methods herein can include administration of a composition herein to reduce tumor volume, size, load, and/or burden in a subject to an undetectable size, or to less than about 1%, less than about 2%, less than about 5%, less than about 10%, less than about 20%, less than about 25%, less than about 30%, less than about 40%, less than about 50%, less than about 60%, less than about 70%, less than about 75%, less than about 80%, or less than about 90% of the subject's tumor volume, size, load, and/or burden prior to treatment.
- methods herein can include administration of a composition herein to reduce the cell proliferation rate and/or tumor growth rate in a subject to an undetectable rate. In some embodiments, methods herein can include administration of a composition herein to reduce the cell proliferation rate and/or tumor growth rate in a subject to a rate less than the rate prior to treatment.
- methods herein can include administration of a composition herein to reduce the cell proliferation rate and/or tumor growth rate in a subject to a rate less than about 1%, less than about 2%, less than about 5%, less than about 10%, less than about 20%, less than about 25%, less than about 30%, less than about 40%, less than about 50%, less than about 60%, less than about 70%, less than about 75%, less than about 80%, or less than about 90% of the rate prior to treatment.
- methods herein can include administration of a composition herein to reduce the development of and/or the number or size of metastatic lesions in a subject to an undetectable rate. In some embodiments, methods herein can include administration of a composition herein to reduce the development of and/or the number or size of metastatic lesions in a subject to a rate less than the rate prior to treatment.
- methods herein can include administration of a composition herein to reduce the development of and/or the number or size of metastatic lesions in a subject to a rate less than about 1%, less than about 2%, less than about 5%, less than about 10%, less than about 20%, less than about 25%, less than about 30%, less than about 40%, less than about 50%, less than about 60%, less than about 70%, less than about 75%, less than about 80%, or less than about 90% of the rate prior to treatment.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to activate the innate immune system.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to activate the innate immune system in a tumor microenvironment by at least about 20% (e.g., least about 20%, least about 30%, least about 40%, least about 50%, least about 60%, least about 70%, least about 80%, least about 90% or greater).
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to activate the innate immune system in a tumor microenvironment by about 20% to about 99%, by about 25% to about 95%, or by about 30% to about 90%.
- the methods of the present disclosure synergistically activate the innate immune system in a tumor microenvironment compared to activation after treatment of a subject with a single therapy of an inhibitor of B7-H3 or a single therapy of an inhibitor of CD47- SIRPa.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to activate at least one population of innate immune system cells in a tumor microenvironment by at least about 20% (e.g., at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or greater).
- the combined therapy of an inhibitor of B7- H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to activate at least one population of innate immune system cells in a tumor microenvironment by about 20% to about 99%, about 25% to about 95%, or about 30% to about 90%.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may activate macrophages, dendritic cells (DCs), natural killer (NK) cells, or any combination thereof.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may activate macrophages, dendritic cells (DCs), natural killer (NK) cells, or any combination thereof by at least about 20% (e.g., at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or greater).
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47- SIRPa as described herein may activate macrophages, dendritic cells (DCs), natural killer (NK) cells, or any combination thereof.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may activate macrophages, dendritic cells (DCs), natural killer (NK) cells, or any combination thereof by about 20% to about 99%, about 25% to about 95%, or about 30% to about 90%.
- DCs dendritic cells
- NK natural killer
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to inflame a cold tumor microenvironment.
- hot and cold are routinely used to refer to T cell-infiltrated, inflamed but non-infiltrated, and non-inflamed tumors. Characteristics of hot tumors can include, but are not limited to the presence of tumor-infiltrating lymphocytes (TILs), expression of anti-programmed death-ligand 1 (PD-L1) on tumor-associated immune cells, possible genomic instability and the presence of a preexisting anti-tumor immune response.
- TILs tumor-infiltrating lymphocytes
- PD-L1 anti-programmed death-ligand 1
- Characteristics of cold tumors can include, but are not limited to poorly infiltrated with T cells, immunologically unaware (scarcely expressing PD-L1), high proliferation with low mutational burden (low expression of neoantigens), and low expression of antigen presentation machinery markers such as major histocompatibility complex class I (MHC I).
- MHC I major histocompatibility complex class I
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to inflame a cold tumor microenvironment by at least about 20% (e.g., at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or greater) in vivo.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to inflame a cold tumor microenvironment by about 20% to about 99%, about 25% to about 95%, or about 30% to about 90%.
- inflaming a cold tumor microenvironment using the methods disclosed herein can increase of at least one interferon, increase of CD8+ T cells, increase of CD44 expression in CD4+ cells, in CD8+ cells, or both, increased TNFalpha expression in T cells, or a combination thereof.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to restore CD8+ T cell effector function in a cold tumor microenvironment.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to initiate immune-cell mediated cytotoxicity.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to initiate CTL-cell mediated cytotoxicity.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to initiate NK-cell mediated cytotoxicity.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to modulate gene expression of GZMA, GZMB, NKG7, IL2R, HSPA1A, HSPA1, RANTES, CCL5, CCR5, CXCR4, CXCL12, or any combination thereof.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to increase gene expression of GZMA, GZMB, NKG7, IL2R, HSPA1A, HSPAJ, RANTES, CCL5, CCR5, CXCR4, CXCL12, or any combination thereof
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to increase cytokine signaling in a subject.
- the combined therapy of an inhibitor of B7- H3 and an inhibitor of CD47-SIRPa as described herein may be administered to increase cytokine signaling CCL5 (RANTES, CCL5/CCR5) signaling in a subject.
- CCL5 cytokine signaling CCL5
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to increase chemokine signaling in a subject.
- the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to increase CXCR4 (CXCR4/CXCL12) signaling in a subject.
- compositions herein Conventional methods, known to those of ordinary skill in the art of medicine, can be used to administer the compositions herein to a subject, depending upon the type of disease to be treated or the site of the disease.
- the combined therapy of B7-H3 inhibitors and CD47-SIRPa inhibitors can be administered to a subject by intravenous infusion.
- injectable compositions may contain various carriers such as vegetable oils, dimethylactamide, dimethyformamide, ethyl lactate, ethyl carbonate, isopropyl myristate, ethanol, and polyols (glycerol, propylene glycol, liquid polyethylene glycol, and the like).
- water soluble antibodies can be administered by the drip method, whereby a pharmaceutical formulation containing the B7-H3 inhibitor and/or CD47-SIRPa inhibitor and a physiologically acceptable excipient can be infused.
- Physiologically acceptable excipients may include, for example, 5% dextrose, 0.9% saline, Ringer’s solution or other suitable excipients.
- Intramuscular preparations e.g., a sterile formulation of a suitable soluble salt form of the antibody, can be dissolved and administered in a pharmaceutical excipient such as Water-for-Inj ection, 0.9% saline, or 5% glucose solution.
- the combined B7-H3 inhibitor and CD47-SIRPa inhibitor therapy herein can be administered concurrently with the one or more chemotherapeutics.
- the combined B7-H3 inhibitor and CD47-SIRPa inhibitor therapy can be administered before or after the one or more chemotherapeutics.
- the B7- H3 inhibitor and CD47-SIRPa inhibitor therapy can be administered after administration of a combined therapy of the B7-H3 inhibitor and CD47-SIRPa inhibitor therapy and one or more chemotherapeutics.
- the B7-H3 inhibitor therapy can be administered after administration of a combined therapy of the B7-H3 inhibitor and CD47-SIRPa inhibitor therapy.
- the CD47-SIRPa inhibitor therapy can be administered after administration of a combined therapy of the B7-H3 inhibitor and CD47-SIRPa inhibitor therapy.
- the one or more chemotherapeutics can be administered systemically. In some embodiments, the one or more chemotherapeutics is administered locally. In some embodiments, the one or more chemotherapeutics can be administered by intravenous administration, e.g., as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerebrospinal, subcutaneous, intra-arterial, intra-articular, intrasynovial, intrathecal, intratumoral, oral, inhalation or topical routes.
- the one or more chemotherapeutics is administered to the subject by intravenous infusion.
- the one or more chemotherapeutics may include an antimetabolite, a microtubule inhibitor, or a combination thereof.
- Antimetabolites include, for example, folic acid antagonist (e.g., methotrexate) and nucleotide analogs such as pyrimidine antagonist (e.g., 5-fluorouracil, foxuridine, cytarabine, capecitabine, and gemcitabine), purine antagonist (e.g., 6-mercaptopurine and 6-thioguanine), and adenosine deaminase inhibitor (e.g., cladribine, fludarabine and pentostatin).
- folic acid antagonist e.g., methotrexate
- nucleotide analogs such as pyrimidine antagonist (e.g., 5-fluorouracil, foxuridine, cytarabine, capecita
- the methods are provided, the combined B7-H3 inhibitor and CD47-SIRPa inhibitor therapy can be administered prior to, concurrently, or subsequent administration of a checkpoint inhibitor that is not a B7-H3 inhibitor or a CD47-SIRPa inhibitor.
- checkpoint inhibitors that are not a B7-H3 inhibitor or a CD47-SIRPa inhibitor can include a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof capable of inhibiting one or more checkpoint proteins.
- Non-limiting examples of checkpoint proteins that can be the target of a checkpoint inhibitor that is not a B7-H3 inhibitor or a CD47-SIRPa inhibitor can include CTLA- 4, PDL1, PDL2, PD1, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4 (belongs to the CD2 family of molecules and is expressed on all NK, y5, and memory CD8 + (aP) T cells), CD160 (also referred to as BY55), CGEN-15049, CHK 1 and CHK2 kinases, A2aR and various B-7 family ligands.
- An effective amount of the pharmaceutical composition described herein can be administered to a subject (e.g., a human) in need of the treatment via a suitable route, systemically or locally.
- a B7-H3 inhibitor and CD47-SIRPa inhibitor therapy may be administered by intravenous administration, e.g., as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerebrospinal, subcutaneous, intra-arterial, intraarticular, intrasynovial, intrathecal, intratumoral, oral, inhalation or topical routes.
- Empirical considerations such as the half-life, generally contribute to the determination of the dosage.
- the combined B7-H3 inhibitor and CD47-SIRPa inhibitor therapy includes anti-B7-H3 and anti-CD47/SIRPa antibodies that are compatible with the human immune system, such as humanized antibodies or fully human antibodies
- the half-life of the antibody may be prolonged.
- Frequency of administration may be determined and adjusted over the course of therapy, and is generally, but not necessarily, based on treatment and/or suppression and/or amelioration and/or delay of a target disease/disorder.
- sustained continuous release formulations of an antibody may be appropriate.
- Various formulations and devices for achieving sustained release are known in the art.
- a subject having a target solid tumor as disclosed herein can be identified by routine medical examination, e.g., laboratory tests, organ functional tests, genetic tests, interventional procedure (biopsy, surgery) any and all relevant imaging modalities,.
- the subject to be treated by the method described herein is a human cancer patient who has undergone or is subjecting to an anti-cancer therapy, for example, chemotherapy, radiotherapy, immunotherapy, or surgery.
- subjects may have received prior immuno-modulatory anti-tumor agents.
- Non-limiting examples of such immuno-modulatory agents include, but are not limited to as anti-PDl, anti-PD-Ll, anti-CTLA-4, anti-OX40, anti- CD137, and the like.
- a subject shows disease progression through the treatment.
- a subject is resistant to the treatment (either de novo or acquired).
- such a subject is demonstrated as having advanced malignancies (e.g., inoperable or metastatic).
- the subject has no standard therapeutic options available or ineligible for standard treatment options, which refer to therapies commonly used in clinical settings for treating the corresponding solid tumor.
- the subject herein may be a human patient having a refractory disease, for example, a refractory melanoma.
- refractory refers to the tumor that does not respond to or becomes resistant to a treatment.
- the subject herein may have a tumor that is resistant to at least one immune-modulatory anti-tumor agents.
- the subject herein may have a tumor that is resistant to anti-PDl, anti-PD-Ll, or a combination thereof.
- the subject may be a human patient having a relapsed disease, for example, a relapsed melanoma.
- relapsed or “relapses” refers to a tumor that returns or progresses following a period of improvement (e.g., a partial or complete response) with treatment.
- kits for use in treating or alleviating a solid tumor for example, melanoma, PDA, CRC, HCC, or cholangiocarcinoma, and others described herein.
- kits can include one or more containers having an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa.
- a kit provided herein can optionally have one or more chemotherapeutics.
- the kit can have instructions for use in accordance with any of the methods described herein.
- the included instructions can include a description of administration of the inhibitor of B7-H3 and/or inhibitor of CD47-SIRPa to treat, delay the onset, or alleviate a target disease as those described herein.
- the kit can further comprise a description of selecting an individual suitable for treatment based on identifying whether that individual has the target disease, e.g., applying the diagnostic method as described herein.
- the instructions comprise a description of administering an antibody to an individual at risk of the target disease.
- the instructions relating to the use of an inhibitor of B7-H3 and/or inhibitor of CD47- SIRPa can generally include information as to dosage, dosing schedule, and route of administration for the intended treatment.
- the containers may be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses.
- Instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit), but machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk) are also acceptable.
- the label or package insert indicates that the composition is used for treating, delaying the onset and/or alleviating the solid tumor.
- instructions are provided for practicing any of the methods described herein.
- kits of this invention are in suitable packaging.
- suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like.
- packages for use in combination with a specific device such as an inhaler, nasal administration device (e.g., an atomizer) or an infusion device such as a minipump.
- a kit has a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
- the container also has a sterile access port (for example the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
- a sterile access port for example the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle.
- At least one active agent in the composition is an inhibitor of B7-H3 and/or inhibitor of CD47- SIRPa such as those described herein.
- Kits may optionally provide additional components such as buffers and interpretive information.
- the kit comprises a container and a label or package insert(s) on or associated with the container.
- the invention provides articles of manufacture comprising contents of the kits described above.
- Example 1 B7-H3 was overexpressed in melanoma and regulated tumor cell intrinsic signals that affected the tumor microenvironment
- Proteins were electrophoretically transferred onto polyvinylidene difluoride membranes for 1 hour, washed in Tris-buffered saline (TBS; 100 mmol/L Tris-HCl, pH 7.5, and 150 mmol/L NaCl), before being blocked for 1 hour in TBS containing 0.1% Tween-20 and 5% milk (TBST-milk).
- TBS Tris-buffered saline
- Primary antibody (anti-human B7-H3) incubations were performed overnight at 4°C in TBST-milk followed by washing and then a 1-hour incubation with either anti-rabbit or anti-mouse horseradish peroxidase-conjugated secondary antibody diluted in TBST-milk.
- RNA Integrity Number of each sample was determined using the 2100 Bioanalyzer instrument (Agilent Technologies). Samples were selected with stringent criteria based on RNA Integrity Number of 7.0 or greater, and a minimum of 500 ng of total RNA.
- RNA was reverse transcribed to cDNA using RNA to cDNA EcoDryTM Premix (Random Hexamers) (Takara Bio).
- the cDNA was next assayed in quantitative real-time PCR (qPCR) using the QuantiNovaTM SYBR® Green PCR Kit (Qiagen) on the CFX384TM Real-Time System (Bio-Rad).
- qPCR quantitative real-time PCR
- Qiagen QuantiNovaTM SYBR® Green PCR Kit
- Bio-Rad Real-Time System
- Metastatic cell lines showed significantly higher levels of B7-H3 expression both at the protein (Fig. 1A) and mRNA level (Fig. IB) as compared to primary melanoma cells, suggesting a role for B7-H3 during progression from primary disease to metastasis.
- Figs. 1C and ID show that high levels of membranous and cytoplasmic expression of B7-H3 were found in a significant portion of the cases - 61%.
- RNA extracted from the samples was subjected to ribosomal RNA depletion and further used as input for library construction by the Illumina TruSeq Standard Total RNA library Pre Gold kit (Illumina).
- the libraries were then sequenced on the Illumina HiSeq 2500 system (Illumina) using 2 x 100 bp paired-end protocol to a minimum mean coverage of 50x.
- Raw reads were quality filtered by trimming the read ends; bases with quality ⁇ 20 were removed.
- the quality at the 20th percentile was computed. The entire read was discarded if the quality at the 20th percentile was less than 15. Reads shorter than 40 bases after trimming were discarded as well. If one or more reads in the pair failed the quality check both reads were discarded.
- Paired-end RNA sequencing reads were mapped to the human reference genome (Ensembl annotation, build 37) and to sequences from the Repbase database of human repetitive elements (release 19) using the STAR aligner in a manner similar to that described in Dobin et al., Bioinformatics. 2013;29(l): 15-21, the disclosure of which is incorporated herein in its entirety. Aligned reads were assigned to genes using the featureCounts function of the Rsubread package via the external Ensembl annotation. This procedure generated the raw read counts for each gene.
- Expression data were used in conjunction with the weights computed by the voom transformation based on the mean-variance relationship of log read counts to calculate moderated t-statistics using empirical Bayes in a manner similar to that described in Law et al., Genome Biol. 2014;15(2):R29,the disclosure of which is incorporated herein in its entirety.
- mRNA protein-coding RNA
- the gene signature differentiating each pair of subtypes was identified. For this analysis, only genes with counts per million reads greater than 10 in at least 2 samples were considered.
- B7-H3 downstream effectors are modulators of the tumor microenvironment -MMP3 (matrix metalloproteinase family), P3H1 and FBLN5 (collagen family), ITGA2 (integrin family), CXCL8 (IL8, inflammation), and interferon stimulating genes IFIT2 (antiviral responses), CMPK2 (antiviral responses), OASL (antiviral responses), and RSAD2 (antiviral responses).
- IFIT2 antiviral responses
- CMPK2 antiviral responses
- OASL antiviral responses
- RSAD2 antiviral responses
- B7-H3 was also notable in YUMM1.7/YUMMER1.7 cells (Braf V600E/wt /Pten' / 7Cdkn2a' / ‘), but cells with activated P-catenin, YUMM2.1, had the highest level.
- B7-H3 or P-catenin was silenced using gene-specific siRNAs.
- Fig. II shows that silencing of B7-H3 had no effect on P-catenin, but knocking down P-catenin lowered B7-H3 protein levels, suggesting that B7-H3 was in part modulated by P-catenin in YUMM2.1 cells.
- B7-H3 overexpression co-occurred with activated P-catenin signaling in YUMM2.1 cells and a subset of human cancers.
- YUMM2.1 (as well as YLnMM1.7/YUMMER1.7) preclinical model represents a suitable system to study the local microenvironment regulated by B7-H3.
- Example 2 Blockade of B7-H3, but not PD-1, resulted in an inflamed tumor microenvironment in a preclinical model of activated P-catenin signaling
- RNA sequencing similar to methods described in example 1. Using the resulting sequencing data, first assessed was the quality of paired-end reads with FASTQC (vO.l 1.8). Next, reads were filtered with BBDUK from BBTOOLS (v37.53) to remove adapters, known artefacts, and quality trimmed (PHRED quality score ⁇ 10). Reads that became too short after trimming (N ⁇ 60 bp) were discarded. Singleton reads (i.e. reads whose mate has been discarded) were not retained.
- the transcript-level quantification of cleaned data was estimated using SALMON (vl.0.0) by a quasi -mapping on Mus musculus GRCm38.
- immune cell lineage-specific transcript signatures in particular innate (macrophages, DCs, and NK cells) populations of the immune system, were significantly upregulated in mice treated with anti-B7-H3 antibody as compared to the isotype control, or anti-PD-1, and to a lesser extent with the combination of both antibodies.
- CCR5 immune cell recruitment
- IL4R inflammatory
- JAK1/3, and STAT6 macrophage inhibitory signals
- CD47 macrophage inhibitory signals
- IFN type I IFN
- IFNGR1/2, JAK1/2 type II IFN
- mice were injected with 5 x IO 5 YUMMER1.7 murine melanoma cells subcutaneously into the flanks of each mouse. Following injection of the YUMMER1.7 cells, mice were administered the same treatments in the same manner as that described above for the YUMM2.1 tumor model mice.
- Macrophage signature in particul r m crophage inhibitory signals (CD47, SIRPa) was confirmed using CD45+ RNA sequencing (Fig. 2E). Macrophage signature was also confirmed in harvested tumor tissue by immunohistochemistry using methods described in the examples herein.
- Figs. 3A-3C show IBA-1 (Allograft Inflammatory Factor 1) immunohistochemistry of tumors in treatment groups with IgG control, anti-B7-H3, or anti-PD-1 showing activated macrophages within the tissue whereas Figs. 3D-3F show CD47 expression of the tumor cells and Figs.
- IBA-1 Allograft Inflammatory Factor 1
- 3G-3I show that SIRPa expression, a receptor for CD47 in immune subpopulations such as macrophages and DCs, was also indicated.
- CD47/Sirpa macrophage checkpoint activation
- CCR5 enrichment of myeloid recruitment signals
- IL4R pro-inflammatory signals
- mice were pretreated with 200 pg of anti-NKl .1 (clone PK136, BioXCell) via i.p. on days 1 and 3 prior to the murine melanoma cell challenge, and then received treatment of 150 pg twice a week.
- anti-CD8a clone YTS169.4 BioXcell 250 pg via i.p. three times per week was used. Tumor volumes were calculated using the following equation: 0.5 x 1 x w 2 .
- NK cell and CD8+ T cell depletion experiments showed dependency to both antitumor effector mechanisms upon anti- B7-H3 treatment (Figs. 4C and 4D). It was next sought to determine therapeutic benefit of blocking B7-H3 and PD-1, B7-H3 and CD47, or PD-1 and CD47 in a different preclinical melanoma model, YUMMER1.7, driven by Braf 600E/wt /Pten’ / 7Cdkn2a' / ' and high levels of B7- H3. Experiments did not demonstrate any additive benefit of CD47 combination, nor by combining B7-H3 and PD-1 inhibitors together (Figs. 4E and 4F).
- scRNA sequencing single cell RNA sequencing was performed on CD45+ sorted cells.
- Single cell suspensions were stained with diluted hashtag antibodies according to the New York Genome Center hashing protocol, similar to the methods described in Stoeckius et al., Genome Biol 19, 224 (2018), the disclosure of which is incorporated herein in its entirety. Hashed samples were pooled in equal amounts of live cells to achieve a target of 2 million cells/ml.
- the hashed sample pool was then loaded onto two lanes of the lOx Genomics NextGen 5’vl.l assay according to the manufacturer’s instructions with a targeted cell recovery of 20,000 cells.
- Gene expression libraries were made as per the lOx Genomics protocol.
- hashtag oligonucleotides HTO
- HTO hashtag oligonucleotides
- the PCR product was isolated from the mRNA-derived cDNA via SPRI Select size selection and used for making HTO libraries according to the New York Genome Center hashing protocol. All libraries were quantified via Agilent 2100 hsDNA Bioanalyzer and KAPA library quantification kit (Roche).
- HTO libraries were sequenced at a targeted depth of 25,000 reads per cells, and HTO libraries were sequenced at a targeted read depth of 1,000 reads per cell. All libraries were sequenced on the Illumina NovaSeq S2 100 cycle kit using run parameters set to 28x8x0x60 (Rlxi7xi5xR2).
- BCL files were first base-called and demultiplexed using cellranger mkfastq v5.0.1. Alignment to mouse reference mm 10 and feature counting were performed using cellranger count v5.0.1. Then, HTO barcodes were mapped to samples of origin. Briefly, a “key” matrix with biological samples as rows and HTO features as columns was created.
- a cell was populated with a value of “ 1 ” if the sample was supposed to be positive for the hashtag, “0” otherwise.
- pairwise distances with a cosine similarity metric were computed from the key matrix to generate a cosine similarity matrix.
- a barcode was initially assigned to a sample based on the largest cosine similarity to a sample.
- a signal to noise ratio (sn) was calculated by subtracting its highest distance metric from its second highest distance metric. The sn ratio for each initially assigned sample usually followed a bi-modal distribution. In order to identify the local minimum of this distribution, four standard deviations were calculated from the mode of the right most identified local maximum.
- NK.49CI+ NK cells
- NKT NKT
- DCs ((DC.8-, DC.11B-, DC.8+, DC.PCD.8-) was observed together with macrophages, Tregs, and eosinophils.
- T cells T.CD8, T.CD4, T.4EFF, T.4.Pa, T.4SP, T.DN, T.4PLN, T4. FP3+25, and T. ISP
- T.CD8 T.CD8, T.CD4, T.4EFF, T.4.Pa, T.4SP, T.DN, T.4PLN, T4. FP3+25, and T. ISP
- macrophages B cells
- DCs and eosinophils Fig. 5D
- CTL- and NK-cell mediated cytotoxicity IgG vs. B7-H3/CD47
- GZMA GZMA
- GZMB NKG7
- IL2R heat shock genes
- HSPA1A and HSPA1B B7-H3 vs. B7-H3/CD47, CD47 vs. B7-H3/CD47
- CCL5 cytokine signaling CCL5
- CXCR4 CXCR4/CXCL12
- HSPA1A and HSPA1B genes not only expressed at high levels during tumor cytotoxicity (IgG vs. B7-H3/CD47, Figs. 6K-6T), but they expressed across most of the immune cell types ubiquitously, whereas GZMA and GZMB was mostly expressed in macrophages, NK cells, and T cells (Figs. 6A-6J). Expression details for CCL5, CXCR4, and NKG7 were also mapped out (Figs. 7A-7O).
- Example 4 Therapeutic synergy was dependent on CCR5 and IL4R signaling [00123] Since TAM-associated inflammatory signals were consistently upregulated upon B7- H3 inhibition, the antitumor effects of co-targeting B7-H3 and CD47 were examined to determine if there was dependency on CCR5 or IL4R signaling. C57BL/6 CCR5-/- or IL4R-/- mice were used in the exemplary methods. Tumors were generated as described herein and the mice were treated with IgG control, B7-H3 inhibitor, and CD47 inhibitor alone or in combination according to methods described herein. As shown in Fig. 8A, the therapeutic synergy was robust but dissipated when these signaling axes were absent (Figs.
- RNA-sequencing data (illuminahiseq_maseqv2-RSEM_genes, version 01/28/2016) and clinical phenotype (merge clinical, version, version 01/28/2016) were downloaded from firebrowse of Broad Institute.
- B7-H3 correlated significantly with suppressive signals (M2 macrophage, myeloid derived suppressive cell (MDSC), T cell exclusion, and cancer associated fibroblast (CAF) signature), whereas it significantly anti-correlated with PD-1, and moderately with PD-L1, and IL-15 (Figs. 8D and 8E).
- M2 macrophage, myeloid derived suppressive cell (MDSC), T cell exclusion, and cancer associated fibroblast (CAF) signature M2 macrophage, myeloid derived suppressive cell (MDSC), T cell exclusion, and cancer associated fibroblast (CAF) signature
- a method for treating a solid tumor in a subject comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition, the pharmaceutical composition comprising an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa, wherein the subject is not responsive to anti-programmed cell death 1/programmed cell death ligand 1 (PD1/PDL1) therapy.
- a pharmaceutical composition comprising an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa, wherein the subject is not responsive to anti-programmed cell death 1/programmed cell death ligand 1 (PD1/PDL1) therapy.
- the at least one population of innate immune system cells is selected from the group consisting of macrophages, dendritic cells (DCs) and natural killer (NK) cells.
- [00135] 8 The method of any one of embodiments 1-7, wherein the inhibitor of B7-H3 comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- SIRPa is a CD47-ligand inhibitor, a SIRPa-receptor inhibitor, or a combination thereof.
- CD47-ligand inhibitor comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- CD47-ligand inhibitor is an antibody.
- the antibody is a full-length antibody or an antigen-binding fragment thereof.
- CD47-ligand inhibitor is an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- CD47-ligand inhibitor is a CD47-specific chimeric antigen receptor T (CAR-T) cell.
- SIRPa-receptor inhibitor comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- SIRPa-receptor inhibitor is an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- a method of inflaming a cold tumor microenvironment comprising administering to the cold tumor microenvironment a composition comprising an inhibitor of B7- H3 (CD276) and an inhibitor of CD47-SIRPa.
- the solid tumor is selected from the group consisting of pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, and sarcomas.
- PDA pancreatic ductal adenocarcinoma
- CRC colorectal cancer
- melanoma melanoma
- breast cancer lung cancer
- upper and lower gastrointestinal malignancies squamous cell head and neck cancer
- genitourinary cancer ovarian cancer
- sarcomas sarcomas.
- inflaming a cold tumor microenvironment comprises increase of at least one interferon, increase of CD8+ T cells, increase of CD44 expression in CD4+ cells, in CD8+ cells, or both, increased TNF alpha expression in T cells, or a combination thereof.
- [00162] 35 The method of any one of embodiments 28-34, wherein the inhibitor of B7-H3 comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- [00163] 36 The method of embodiment 35, wherein the inhibitor of B7-H3 is an antibody.
- inhibitor of B7-H3 is an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- inhibitor of B7-H3 comprises at least one of enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL-015, or a combination thereof.
- SIRPa is a CD47-ligand inhibitor, a SIRPa-receptor inhibitor, or a combination thereof.
- the CD47-ligand inhibitor comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- [00173] 46 The method of embodiment 44, wherein the antibody is a human antibody or a humanized antibody.
- CD47-ligand inhibitor is an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- CD47-ligand inhibitor is a CD47-specific chimeric antigen receptor T (CAR-T) cell.
- SIRPa-receptor inhibitor comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- SIRPa-receptor inhibitor is an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- the inhibitor of CD47-SIRPa comprises at least one of RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC-90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or a combination thereof.
- a pharmaceutical composition for treating a solid tumor in a subject comprising an inhibitor of B7-H3 (CD276) and an inhibitor of CD47- SIRPa.
- the pharmaceutical composition of embodiment 55 further comprising at least one pharmaceutically acceptable excipient.
- composition of embodiment 55 or embodiment 56, wherein the inhibitor of B7-H3 (CD276) and the inhibitor of CD47-SIRPa. comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
- any one of embodiments 55-58, wherein the inhibitor of CD47-SIRPa comprises at least one of RRx-001, a dinitroazetidine derivative, Hu5F9- G4, CC-90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or a combination thereof.
- composition for treating a solid tumor in a subject comprising at least one inhibitor of B7-H3 (CD276) and at least one inhibitor of CD47-SIRPa
- composition of embodiment 61 further comprising at least one pharmaceutically acceptable excipient.
- composition of either embodiment 61 or embodiment 62, wherein the at least one inhibitor of B7-H3 comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment, or any combination thereof.
- composition of embodiment 63, wherein the at least one inhibitor of B7-H3 comprises an antibody.
- composition of embodiment 63, wherein the at least one inhibitor of B7-H3 comprises an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- the at least one inhibitor of B7-H3 comprises a B7-H3-specific chimeric antigen receptor T (CAR-T) cell.
- CAR-T chimeric antigen receptor T
- 67 The composition of embodiment 63, wherein the at least one inhibitor of B7-H3 comprises enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL-015, or any combination thereof.
- composition of either embodiment 61 or embodiment 62, wherein the at least one inhibitor of CD47-SIRPa comprises at least one CD47-ligand inhibitor, at least one SIRPa- receptor inhibitor, or any combination thereof.
- SIRPa comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment, or any combination thereof.
- composition of embodiment 68, wherein the at least one CD47-ligand inhibitor comprises an antibody.
- composition of embodiment 68, wherein the at least one CD47-ligand inhibitor comprises an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- composition of embodiment 68, wherein the at least one CD47-ligand inhibitor comprises a CD47-specific chimeric antigen receptor T (CAR-T) cell.
- CAR-T CD47-specific chimeric antigen receptor T
- composition of embodiment 68, wherein the at least one SIRPa-receptor inhibitor comprises an antibody.
- composition of embodiment 68, wherein the at least one SIRPa-receptor inhibitor comprises an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
- SIRPa comprises RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC-90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or any combination thereof.
- a method of preventing, treating, or ameliorating a soil tumor in a subject in need thereof comprising administering an effective amount of a composition of any one of embodiments 61-75, wherein the subject in need thereof has or is suspected of having a solid tumor that is not responsive to at least one anti-programmed cell death 1/programmed cell death ligand 1 (PD1/PDL1) therapy.
- PD1/PDL1 anti-programmed cell death 1/programmed cell death ligand 1
- the subject in need thereof is a human patient having or suspected of having a solid tumor selected from the group consisting of pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, and sarcomas.
- a solid tumor selected from the group consisting of pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, and sarcomas.
- kits comprising a pharmaceutical composition according to any one of embodiments 55-75 and at least one container.
- kits of embodiment 83 further comprising one or more chemotherapeutics.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Biophysics (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Genetics & Genomics (AREA)
- Biochemistry (AREA)
- Veterinary Medicine (AREA)
- General Chemical & Material Sciences (AREA)
- Public Health (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present disclosure generally relates to compositions and methods for treating a solid tumor in a subject, wherein administering compositions having a combination of an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPα can inflame a cold tumor microenvironment.
Description
COMBINED CANCER THERAPY OF B7-H3 AND CD47 IMMUNE CHECKPOINT INHBITIOR AND METHODS OF USE
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Application 63/115,699 filed November 19, 2020, the disclosure of which is hereby incorporated by reference in its entirety.
GOVERNMENTAL RIGHTS
[0002] This invention was made with government support under CAI 77940 awarded by the National Institutes of Health. The government has certain rights in the invention.
FIELD
[0003] The present disclosure generally relates to compositions and methods for treating a solid tumor in a subject, wherein administering compositions having a combination of an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa can inflame a cold tumor microenvironment.
BACKGROUND
[0004] Immunotherapies have shown to be effective for treating a variety of immune-related diseases, including cancers. However, many cancers appear to be resistant to such therapies due to a number of immunological obstacles. These can include the ability of tumors to foster a tolerant microenvironment and the activation of a plethora of immunosuppressive mechanisms, which may act in concert to counteract effective immune responses. For instance, tumor-associated macrophages, marrow-derived suppressor cells, tumor-associated neutrophils, cancer-associated fibroblasts, and regulatory T cell interactions actively promote tumorigenesis.
[0005] Significant advances in checkpoint inhibitor development and use have indicated that such immunotherapies could be a hopeful therapeutic option in for treating solid cancerous tumors. Whereas some subjects positively respond to such therapies, a large percentage of cancer patients never demonstrate a clinical response or stabilized disease after treatment with one or more checkpoint inhibitors. As such, reversing the immunosuppressed microenvironment and switching to an inflamed and immune infiltrated phenotype, identifying immune signaling axes that
mechanistically function via divergent pathways, and ultimately finding effective therapeutic combinations are of interest in the cancer field. Accordingly, there is a need in the field to develop methods of treating solid tumors, such as melanomas, that are resistant to currently available immunotherapies.
SUMMARY
[0006] An aspect of the present disclosure provides methods for treating a solid tumor in a subject. In some embodiments, methods can include administering to a subject in need thereof an effective amount a composition, the composition having an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa. In accordance with these embodiments, compositions herein may be pharmaceutical compositions comprising an inhibitor of B7-H3 (CD276), an inhibitor of CD47- SIRPa, and/or at least one pharmaceutically acceptable excipient. In some embodiments, a subject in need thereof can be non-responsive to anti-programmed cell death 1/programmed cell death ligand 1 (PD1/PDL1) therapy. In some embodiments, a subject can be a human patient having a solid tumor selected from the group of pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, sarcomas, or any combination thereof. In some embodiments, a subject can be a human patient having a melanoma. [0007] In certain embodiments, methods disclosed herein can include administration of a composition and/or pharmaceutical composition herein wherein the effective amount of the composition and/or pharmaceutical composition herein may be sufficient to activate the innate immune system. In some embodiments, methods disclosed herein can include administration of a composition and/or pharmaceutical composition herein wherein the effective amount of the composition and/or pharmaceutical composition herein may be sufficient to activate at least one population of innate immune system cells. In some embodiments, populations of innate immune system cells activated by the methods herein can be macrophages, dendritic cells (DCs) and natural killer (NK) cells. In some embodiments, methods disclosed herein can include administration of a composition and/or pharmaceutical composition herein wherein the effective amount of the composition and/or pharmaceutical composition herein may be sufficient to reduce tumor volume.
[0008] In certain embodiments, methods herein can include administration of an inhibitor of B7-H3. In some embodiments, methods herein can include administration of an inhibitor of B7- H3 wherein the inhibitor of B7-H3 can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment, or any combination thereof. In some embodiments, an inhibitor of B7-H3 can be an antibody. In some embodiments, an inhibitor of B7-H3 can be a full-length antibody or an antigen-binding fragment thereof. In some embodiments, an inhibitor of B7-H3 can be a human antibody or a humanized antibody. In some embodiments, an inhibitor of B7-H3 can be an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody. In some embodiments, an inhibitor of B7-H3 can be enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL-015, or any combination thereof. In some embodiments, an inhibitor of B7-H3 can be a B7-H3-specific chimeric antigen receptor T (CAR-T) cell.
[0009] In certain embodiments, methods herein can include administration of an inhibitor of CD47-SIRPa. In some embodiments, an inhibitor of CD47-SIRPa can be a CD47-ligand inhibitor, a SIRPa-receptor inhibitor, or any combination thereof. In some embodiments, methods herein can include administration of a CD47-ligand inhibitor. In some embodiments, methods herein can include administration of a CD47-ligand inhibitor wherein the CD47-ligand inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof. In some embodiments, a CD47-ligand inhibitor can be an antibody. In some embodiments, a CD47-ligand inhibitor can be a full-length antibody or an antigen-binding fragment thereof. In some embodiments, a CD47-ligand inhibitor can be a human antibody or a humanized antibody. In some embodiments, a CD47-ligand inhibitor can be an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody. In some embodiments, a CD47-ligand inhibitor can be a CD47-specific chimeric antigen receptor T (CAR- T) cell. In some embodiments, methods herein can include administration of a SIRPa-receptor inhibitor. In some embodiments, methods herein can include administration of a SIRPa-receptor inhibitor wherein the SIRPa-receptor inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof. In some embodiments, a SIRPa-receptor inhibitor can be an antibody. In some embodiments, a SIRPa-receptor inhibitor can be a full-length antibody or an antigen-binding
fragment thereof. In some embodiments, a SIRPa-receptor inhibitor can be a human antibody or a humanized antibody. In some embodiments, a SIRPa-receptor inhibitor can be a SIRPa -specific chimeric antigen receptor T (CAR-T) cell. In some embodiments, an inhibitor of CD47-SIRPa can be RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC-90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or any combination thereof.
[0010] Another aspect of the present disclosure provides methods for inflaming a cold tumor microenvironment. In certain embodiments, a method of inflaming a cold tumor microenvironment as disclosed herein can include administering to the cold tumor microenvironment a composition having an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa. In some embodiments, a tumor having a cold tumor microenvironment can be a solid tumor. In some embodiments, a tumor having a cold tumor microenvironment can be pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, and/or sarcoma. In some embodiments, a tumor having a cold tumor microenvironment can be melanoma. In some embodiments, a tumor having a cold tumor microenvironment can have at least one of B7-H3 overexpression, overactive P-catenin signaling, or any combination thereof. In some embodiments, a tumor having a cold tumor microenvironment can have at least one genetic mutation comprising BRAFV600E, loss of PTEN, or a combination thereof.
[0011] In certain embodiments, methods of inflaming a cold tumor microenvironment disclosed herein can increase of at least one interferon, increase of CD8+ T cells, increase of CD44 expression in CD4+ cells, in CD8+ cells, or both, increased TNFalpha expression in T cells, or any combination thereof.
[0012] Another aspect of the present disclosure provides pharmaceutical compositions for treating a solid tumor in a subject. In some embodiments, pharmaceutical compositions for treating a solid tumor in a subject as disclosed herein can include an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa. In some embodiments, pharmaceutical compositions disclosed herein can further include at least one pharmaceutically acceptable excipient. In some embodiments, pharmaceutical compositions disclosed herein can include an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa in a dosage amount that may restore CD8+ T cell effector function in a cold tumor microenvironment.
[0013] In certain embodiments, kits are provided for transport, storage and use in treating or reducing the size of, reducing expansion of a tumor, and/or inflaming a cold tumor microenvironment as disclosed herein.
BRIEF DESCRIPTION OF THE FIGURES
[0014] The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present disclosure, which can be better understood by reference to the drawing in combination with the detailed description of specific embodiments presented herein.
[0015] Figs. 1A-1K are images depicting that B7-H3 was overexpressed in melanomas and regulated that tumor cell-mediated signals that influenced the tumor microenvironment. Fig. 1A shows a representative image of an immunoblot detection of B7-H3 protein expression in a panel of human primary (n = 12) and metastatic (n = 15) melanoma cell lines, and human melanocytes (n = 1). Fig. IB shows a graph that quantifies B7-H3 mRNA expression in a panel of human primary (n = 12) and metastatic (n = 15) melanoma cell lines, and human melanocytes (n = 1) as assessed by quantitative real-time PCR (n = 28). Fig. 1C shows a graph of B7-H3 protein expression in a melanoma tissue arrays consisting of metastatic melanomas (n = 46) examined for B7-H3 protein expression by immunohistochemistry. Fig. ID shows a representative image of positive immunostaining of B7-H3 protein expression in a tissue within the melanoma tissue array; Scale bar: 60 pm. Fig. IE is a graph showing that siRNA knockdown was confirmed in two human metastatic melanoma cell lines, Hs. 688(A)T and Hs. 839T, that were transfected with scrambled or B7-H3-specific siRNAs in triplicates using quantitative real-time PCR to assay for two isoforms of B7-H3. Fig. IF is an image of a heatmap of gene expression in Hs. 688(A)T and Hs. 839T cells that were transfected with scrambled or B7-H3-specific siRNAs, where RNA sequencing identified differentially expressing genes between the control and the silenced groups that were consistently up- or down- regulated in both cell lines. Fig. 1G is a representative immunoblot for B7-H3 showing B7-H3 protein expression in a panel of murine melanoma cell lines and immortalized murine melanocytes (melan-a). Fig. 1H is a graph showing mRNA levels of B7-H3 as analyzed in a panel of murine melanoma cell lines and immortalized murine melanocytes (melan-a) by quantitative real-time PCR. Figs. II and 1J show that B7-H3 or P-catenin was
silenced by gene-specific siRNAs in YUMM2.1 cells, silencing was confirmed by immunoblot (Fig. 1H), and mRNAs obtained from these cells were processed for B7-H3 downstream targets (shown in Fig. IF) in triplicates using quantitative real-time PCR (Fig. 1J). Fig. II is an image showing Reverse Phase Protein Array (RPPA) data for 899 cancer cell lines from 24 organs, including 50 melanoma cell lines, downloaded from Cancer Cell Line Encyclopedia (CCLE) where each row indicates a differentially expressed gene and each column represents a tumor sample and is color-coded on the basis of median-centered log2 gene expression levels. Fig. IK is an image showing a heatmap depicting a high-level correlation between WNT-signaling genes and B7-H3, and anti-correlation with VAV1.
[0016] Figs. 2A-2E are images depicting that B7-H3 inhibition led to an inflamed tumor microenvironment in P-catenin activated tumors. FIG. 2A is a graph showing tumor growth curves from tumor-bearing C57BL/6J mice that were subcutaneously injected with YUMM2.1 cells (1 x 106), harboring the BrafV600E/wt/Pten'/7Cdkn2+/'/Bcatloxex3/wt genotype, to generate tumors where the mice were challenged with isotype control, anti-B7-H3 (600 pg, three times per week), anti-PD-1 (300 pg, every five days), or both (anti-B7-H3 and anti-PD-1) when tumor size reached 20-50 mm3 (day 9). Treatments were continued until the end of the experiment. Tumor growth curves were determined by measuring tumor volume three times a week. The arrow indicates the first day when the drug or control was injected into mice. Fig. 2B is an image showing immune cell subgroups across treatment groups in RNA from the tumor (bulk) in triplicates after RNA was subjected to RNA sequencing. Enriched immune cell subgroups across treatment groups are shown. Fig. 2C is an image of a heatmap depicting selected genes and innate immune signals differentially expressed between the control and anti-B7-H3 treatment group. Fig. 2D is an image showing innate immune signals identified as enriched by RNA sequencing in the anti-B7-H3 treatment group where the tumors were harvested from tumor-bearing C57BL/6J mice that were subcutaneously injected with either YLTMM2.1 cells or YUMMERl .7 cells to generate tumors. Fig. 2E is an image of a heatmap depicting selected genes assessed in CD45+ immune cells that were sorted and subjected to ultra low RNA sequencing in triplicates in YLTMM2.1 tumors treated with anti-B7-H3, anti-PD-1, or control.
[0017] Figs. 3A-3I depict images showing that B7-H3 inhibition resulted in macrophage infiltration and activation. Figs. 3A-3C are representative images showing IBA-1 (Allograft
Inflammatory Factor 1) immunohistochemistry of tumors in treatment groups with IgG control (Fig. 3A), anti-B7-H3 (Fig. 3B), or anti-PD-1 (Fig. 3C) showing activated macrophages within the tissue. Figs. 3D-3F are representative images showing CD47 expression of the tumor cells by immunohistochemistry in treatment groups with IgG control (Fig. 3D), anti-B7-H3 (Fig. 3E), or anti-PD-1 (Fig. 3F). Figs. 3G-3I are representative images showing SIRPa expression, a receptor for CD47 in immune subpopulations such as macrophages and DCs, in treatment groups with IgG control (Fig. 3G), anti-B7-H3 (Fig. 3H), or anti-PD-1 (Fig. 31).
[0018] Figs. 4A-4F depict images showing that therapeutic synergy was achieved when anti-
B7-H3 (or anti-PD-1) was combined with anti-CD47. Fig. 4A is a graph showing tumor growth curves of tumors that were generated with YUMM2.1 cells (IxlO6) from mice treated with isotype control, anti-B7-H3 (600 pg, three times per week), anti-CD47 (200 pg, three times per week), or both (anti-B7-H3 and anti-CD47) (n = 6 mice per group). Fig. 4B is a graph showing tumor growth curves of tumors that were generated with YUMM2.1 cells from mice treated with isotype control, anti-PD-1 (300 pg, every five days), anti-CD47 (200 pg, three times per week), or both (anti-PD- 1 and anti-CD47). Fig. 4C is a graph showing tumor growth curves of tumors that were generated with YUMM2.1 cells from NK cell depleted-mice, where the mice were pretreated with anti NK1.1 (200 pg, days 1 and 3) followed by 150 pg twice a week (n = 6 mice per group). Fig. 4D is a graph showing tumor growth curves of tumors that were generated with YUMM2.1 cells from CD8+ T cell depleted-mice, where the mice were pretreated with anti-CD8a 250 pg three times per week was used (n = 6 mice per group). Fig. 4E is a graph showing tumor growth curves of tumors that were generated with YUMMER1.7 cells (5 x 105) harboring the BrafV600E/wt/Pten'/7Cdkn2'/' genotype, from mice treated with anti-B7-H3, anti-PD-1, or both (anti-B7-H3 and anti-PD-1) once the tumor size reached 20-50 mm3 (n = rl mice per group). Fig. 4F is a graph showing tumor growth curves of tumors that were generated with YUMMER1.7 cells from mice treated with IgG, anti- B7-H3, anti-PD-1, anti-CD47, or combination of anti-B7-H3 and anti-CD47, or anti-PD-1 and anti-CD47.
[0019] Figs. 5A-5E depict images showing that therapeutic synergy was achieved when anti-
B7-H3 (or anti-PD-1) was combined with anti-CD47 in P-catenin activated tumors. Fig. 5A is a graph showing tumor growth curves for tumors generated by subcutaneous injection with YUMM2.1 cells (IxlO6) and treated with isotype control, anti-B7-H3 (600 pg, three times per
week), anti-CD47 (200 pg, three times per week), or both (anti-B7-H3 and anti-CD47) (n = 6 mice per group). Quantitative results were analyzed by one-way ANOVA. Fig. 5B is a graph showing UMAP of immune cell distribution for CD45+ sorted cells subjected to single cell RNA sequencing (scRNA-sequencing) in the tumors described in Fig. 5A. Fig. 5C is a graph showing proportions of major immune cell subsets in each sample as analyzed by scRNA-sequencing. Fig. 5D is a heatmap depicting immune cell subtypes for each treatment group. The key for the x-axis is; I . DC (DC.103+1 IB-), 2. T cells (T.4); 3. Stromal cells (ST); 4. ILC (LIV.NK.DX5+); 5. B cells (B.Fo); 6. Macrophages (MFI05.II-); 7. ILC (ILC3); 8. NKT (NKT.4-); 9. Macrophages (MF.F480HI); 10. DC (DC.8-4-1 IB-); 11. NKT (NKT.44+); 12. B cells (B.CD19CONTROL); 13. T cells (T.8MEM), 14. T cells (T.8SP); 15. T cells (T.4SP), 16. Macrophages (MFI05.II+); 17 Stem cells (SC); 18. NKT (NKT.44-); 19. ILC (ILC1.CD127+); 20. Macrophages (MF.II+480LO); 21 . T cells (T.8NVE); 22. T cells (T.4.Pa); 23. Macrophages (MF. l 1C-11B+); 24. DC (DC.PDC.8- ); 25. B cells (B.GC); 26. Macrophages (MF.103-11B+); 27. Epithelial cells (Ep); 28. Macrophages (MF.II-480HI); 29. Fibroblasts (FI), 30. DC (DC.8+); 31. NKT (NKT.4+); 32. NK cells (NK.49CI+); 33. Macrophages (MF.103CLOSER); 34. T cells (T.4FP3-); 35. NK cells (NK.49C1-); 36 T cells (T.Tregs); 37. DC (DC.11B-); 38. Basophils (BA); 39. Monocytes (MO); 40. DC (DC.8-); 41. ILC (ILC2); 42. NK cells (NK.B2M-); 43. Macrophages (MF); 44 B cells (B.FrF); 45. T cells (T.4MEM); 46. Macrophages (MF.480INT.NAIVE); 47. T cells (T.4FP3+25+); 48. Endothelial cells (EC); 49. Tgd (Tgd); 50. Eosinophils (EO); 51. B cells (B.MEM); 52. T cells (T.4EFF); 53. DC (DC.PDC.8+); 54. T cells (T.ISP); 55. T cells (T.CD8); 56. T cells (T.8EFF); 57. Neutrophils (GN); 58. Macrophages (MF.l 1 CLOSER), 59. T cells (T.4.PLN); 60. T cells (T.DN); 61. B cells (preB); 62. T cells (T.CD4); 63. Macrophages (MF.169+11 CHI), 64. DC (DC.103-11B+). Fig. 5E is a beatmap showing differentially expressed genes (x-axis) between the treatment groups (y-axis).
[0020] Figs. 6A-6T depict images showing UMAP of differentially expressed genes GZMA (Figs. 6A-6D), GZMB (Figs. 6F-6I), HSPA1A (Figs. 6K-6N), HSPA1B (Figs. 6P-6S), and expression levels of GZMA (Fig. 6E), GZMB (Fig. 6J), HSPA1A (Fig. 60), an . HSPA 1B (Fig. 6T) in immune cell subsets. Significant P values are indicated.
[0021] Figs. 7A-7O depict images showing UMAP of differentially expressed genes CCL5 (Figs. 7A-7D), CXCR4 (Figs. 7F-7I), NKG7 (Figs. 7K-7N) and expression levels of CCL5 (Fig.
7E), CXCR4 (Fig. 7J), and NKG7 (Fig. 70) in immune cell subsets. Significant P values are indicated.
[0022] Figs. 8A-8E depict images showing enriched chemokine signaling upon combining anti-B7-H3 and anti-CD47. Fig. 8A shows a graph of tumor volume over time for tumors generated by subcutaneous injection of YUMM2.1 cells (IxlO6) into C57BL/6J mice and challenged with isotype control, anti-B7-H3, anti-PD-1, anti-CD47, or combination of anti-B7-H3 and anti-CD47, or anti-PD-1 and anti-CD47 (n = 6 mice per group). Quantitative results were analyzed by oneway ANOVA. Fig. 8B shows a graph of tumor volume over time for tumors generated by subcutaneous injection of YUMM2.1 cells (IxlO6) into C57BL/6 CCR5-/- mice and challenged with isotype control, anti-B7-H3, anti-PD-1, anti-CD47, or combination of anti-B7-H3 and anti- CD47, or anti-PD-1 and anti-CD47 (n = 6 mice per group). Quantitative results were analyzed by one-way ANOVA. Fig. 8C shows a graph of tumor volume over time for tumors generated by subcutaneous injection of YUMM2.1 cells (IxlO6) into C57BL/6 IL4R-/- mice and challenged with isotype control, anti-B7-H3, anti-PD-1, anti-CD47, or combination of anti-B7-H3 and anti- CD47, or anti-PD-1 and anti-CD47 (n = 5 mice per group). Quantitative results were analyzed by one-way ANOVA. Figs. 8D and 8E are RNA sequencing files of metastatic melanomas of the TCGA (n = 320) where signatures for T cell exclusion and dysfunction (M2 macrophages - M2, myeloid-derived suppressive cells -MDSC, cancer-associated fibroblasts - CAF, T cell exclusion, and T cell dysfunction) (Fig. 8D) and their correlations (Fig. 8E) with B7-H3, PD-1, PDL-1 and others were examined.
DETAILED DESCRIPTION
[0023] Immunotherapy using immune checkpoint inhibitors can be effective at treating cancer in a subset of patients having solid tumors, such as melanoma. While durable and complete responses are observed in patients, many do not respond or develop resistance. Objective response rate using immune checkpoint inhibitors, such as anti-PD-1 (B7-H1) and/or anti-CTLA-4, can range from 10% to 40%. The present disclosure provides, in part, a unique combination therapy of immune checkpoint inhibitors with higher response rates that likely act by exerting distinct immune regulatory functions to restore the innate immune system surrounding the tumor.
[0024] B7-H3 (CD276) is an immune regulatory molecule that belongs to the B7 superfamily. It is expressed on the membrane of the cancer cell, but its receptor is unknown. Aberrant protein expression of B7-H3 is associated with poor prognosis in many cancers including melanoma. Monoclonal antibodies against B7-H3 are tested in preclinical and phase I clinical studies. Despite the importance of immune checkpoint molecules in the cancer field, B7-H3 is less well studied than other checkpoints and its functions remain less well delineated.
[0025] CD47 functions through a mechanism by which the cancer cell evades innate immune surveillance (macrophage immune checkpoint). CD47 is expressed on the membrane of the cancer cell. CD47 binds and activates SIRPa, an inhibitory protein expressed on the surface of myeloid cells - macrophages -, and functions as an anti-phagocytic or “don’t eat me signal”. Activation of SIRPa inhibits the phagocytic activity of macrophages. Blocking the CD47-SIRPa interaction allows macrophages to engulf cancer cells. Monoclonal antibodies inhibiting CD47 are tested in pre-clinical models, and phase I clinical trials are either completed or ongoing.
[0026] Provided herein are compositions that include an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa which are capable of treating a solid tumor in a subject, and uses thereof for activating at least one population of innate immune system cells in a cold tumor microenvironment.
I. Definitions
[0027] For the purposes of promoting an understanding of the principles of the present disclosure, reference will now be made to preferred embodiments and specific language will be used to describe the same. It will nevertheless be understood that no limitation of the scope of the disclosure is thereby intended, such alteration and further modifications of the disclosure as illustrated herein, being contemplated as would normally occur to one skilled in the art to which the disclosure relates.
[0028] As used in the specification, articles “a” and “an” are used herein to refer to one or to more than one (i.e., at least one) of the grammatical object of the article. By way of example, “an element” means at least one element and can include more than one element.
[0029] “About” is used to provide flexibility to a numerical range endpoint by providing that a given value may be “slightly above” or “slightly below” the endpoint without affecting the
desired result. The term “about” in association with a numerical value means that the numerical value can vary plus or minus by 5% or less of the numerical value.
[0030] Throughout this specification, unless the context requires otherwise, the word “comprise” and “include” and variations (e.g., “comprises,” “comprising,” “includes,” “including”) will be understood to imply the inclusion of a stated component, feature, element, or step or group of components, features, elements or steps but not the exclusion of any other integer or step or group of integers or steps.
[0031] As used herein, “and/or” refers to and encompasses any and all possible combinations of one or more of the associated listed items, as well as the lack of combinations where interpreted in the alternative (“or”).
[0032] As used herein, the transitional phrase “consisting essentially of’ (and grammatical variants) is to be interpreted as encompassing the recited materials or steps “and those that do not materially affect the basic and novel characteristic(s)” of the claimed invention. Thus, the term “consisting essentially of’ as used herein should not be interpreted as equivalent to “comprising.” [0033] Moreover, the present disclosure also contemplates that in some embodiments, any feature or combination of features set forth herein can be excluded or omitted. To illustrate, if the specification states that a complex comprises components A, B and C, it is specifically intended that any of A, B or C, or a combination thereof, can be omitted and disclaimed singularly or in any combination.
[0034] Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise- indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. For example, if a concentration range is stated as 1% to 50%, it is intended that values such as 2% to 40%, 10% to 30%, or 1% to 3%, etc., are expressly enumerated in this specification. These are only examples of what is specifically intended, and all possible combinations of numerical values between and including the lowest value and the highest value enumerated are to be considered to be expressly stated in this disclosure.
[0035] As used herein, "treatment," "therapy" and/or "therapy regimen" refer to the clinical intervention made in response to a disease, disorder or physiological condition manifested by a patient or to which a patient may be susceptible. The aim of treatment includes the alleviation or
prevention of symptoms, slowing or stopping the progression or worsening of a disease, disorder, or condition and/or the remission of the disease, disorder or condition.
[0036] As used herein, “prevent” or “prevention” refers to eliminating or delaying the onset of a particular disease, disorder or physiological condition, or to the reduction of the degree of severity of a particular disease, disorder or physiological condition, relative to the time and/or degree of onset or severity in the absence of intervention.
[0037] The term “effective amount" or "therapeutically effective amount" refers to an amount sufficient to effect beneficial or desirable biological and/or clinical results.
[0038] As used herein, “individual”, “subject”, “host”, and “patient” can be used interchangeably herein and refer to any mammalian subject for whom diagnosis, treatment, prophylaxis or therapy is desired, for example, humans, pets, livestock, horses or other animals. As used herein, the term "subject" and "patient" are used interchangeably herein and refer to both human and nonhuman animals. The term "nonhuman animals" of the disclosure includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dog, cat, horse, cow, chickens, amphibians, reptiles, and the like. In some embodiments, the subject can be a human. In other embodiments, the subject can be a human in need of treating a solid tumor (e.g., melanoma).
[0039] As used herein, “polynucleotide,” “nucleic acid” or “nucleic acid molecule” include deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), mRNA, oligonucleotides, and the like.
[0040] As used herein, “vector”, “expression vector” or “construct” refers to a nucleic acid used to introduce polynucleotides into a cell that has regulatory elements to provide expression of the heterologous nucleic acids in the cell. Vectors include but are not limited to plasmid, minicircles, yeast, and/or viral genomes. In some alternatives, the vectors are plasmid, minicircles, or viral genomes. In some alternatives, the vector is a viral vector. In some embodiments, the viral vector is a lentivirus. In some alternatives, the vector is a lentiviral vector. In some embodiments, the vector is a foamy viral vector, adenoviral vectors, retroviral vectors or lentiviral vectors.
[0041] As used herein, “chimeric antigen receptor” or “CAR” or “chimeric T cell receptor” refers herein to a synthetically designed receptor having a ligand binding domain of an antibody or another peptide sequence that binds to a molecule associated with the disease or disorder and is linked via a spacer domain to one or more intracellular signaling domains of a T cell or other
receptors, such as a costimulatory domain. Chimeric receptor can also be referred to as artificial T cell receptors, chimeric T cell receptors, chimeric immunoreceptors, and chimeric antigen receptors (CARs).
IL Compositions
[0042] Aspects of the present disclosure include compositions encompassing an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa. Compositions herein can also be pharmaceutical compositions. a. B7-H3 Inhibitors
[0043] In certain embodiments, compositions herein can include at least one inhibitor of B7- H3 (CD276). In some embodiments, B7-H3 activity can be reduced through the use of an antagonist of the receptor, a partial antagonist or an antibody that either competes with the natural agonist, blocks the activity or encourages uptake of the B7-H3 molecule. In accordance with these embodiments, a B7-H3 inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof. In certain embodiments, a B7-H3 inhibitor can be an anti- B7-H3 antibody or fragment thereof capable of binding to B7-H3 and inhibiting or reducing B7-H3 activity. In some embodiments, other B7-H3 inhibitors can include, but are not limited to, a B7-H3-specific siRNA, RNAi, microRNA or ribozyme. In some embodiments, an inhibitor of B7-H3 can be an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody. Other B7-H3 inhibitors and/or antagonists can include anti-B7-H3 specific antibody fragments (F(ab')2 or Fab'), single chain Fv, and Fc- fusion protein of B7-H3 ligands, e.g. Fc fusion proteins of 2B4 or CD2. In some embodiments, an anti-B7-H3 antibody used herein can be a polyclonal or monoclonal antibody. A “humanized” anti-B7-H3 antibody can be used if needed in order to avoid any potential use incompatibilities (e.g. adverse reactions when introducing the ILC2 cells exposed to such an antibody, if needed). In some embodiments, an inhibitor of B7-H3 can be a B7-H3-specific chimeric antigen receptor T (CAR-T) cell. In some embodiments, an inhibitor of B7-H3 suitable for use herein can be, but is not limited to, enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL- 015, or a combination thereof. h. CD47-SIRPa Inhibitors
[0044] In certain embodiments, compositions herein can include at least one inhibitor CD47- SIRPa. In some embodiments, CD47-activity can be reduced through the use of an antagonist of the receptor (SIRPa), a partial antagonist or an antibody that either competes with the natural agonist, blocks the activity or encourages uptake of the CD47 molecule. In some embodiments, a CD47-SIRPa inhibitor can be a CD47-ligand inhibitor. In some embodiments, a CD47-SIRPa inhibitor can be a SIRPa-receptor inhibitor.
[0045] In accordance with these embodiments, a CD47 inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof. In some embodiments, a CD47 inhibitor can be an anti-CD47 antibody or fragment thereof capable of binding to CD47 and inhibiting or reducing CD47 activity. In other embodiments, other CD47 inhibitors can include, but are not limited to, a CD47-specific siRNA, RNAi, microRNA or ribozyme. In some embodiments, an inhibitor of CD47 can be an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody. Other CD47 inhibitors and/or antagonists can include anti-CD47 specific antibody fragments (F(ab')2 or Fab'), single chain Fv, and Fc-fusion protein of B7-H3 ligands, e.g. Fc fusion proteins of 2B4 or CD2. In some embodiments, an anti- CD47 antibody used herein can be a polyclonal or monoclonal antibody. A “humanized” anti-CD47 antibody can be used if needed in order to avoid any potential use incompatibilities. In some embodiments, an inhibitor of CD47 can be a CD47-specific chimeric antigen receptor T (CAR-T) cell.
[0046] In accordance with these embodiments, a SIRPa inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof. In some embodiments, a SIRPa inhibitor can be an anti-SIRPa antibody or fragment thereof capable of binding to SIRPa and inhibiting or reducing SIRPa activity. In some embodiments, other SIRPa inhibitors can include, but are not limited to, a SIRPa-specific siRNA, RNAi, microRNA or ribozyme. In some embodiments, an inhibitor of SIRPa can be an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody. Other SIRPa inhibitors and/or antagonists can include anti- SIRPa specific antibody fragments (F(ab')2 or Fab'), single chain Fv, and Fc-fusion protein of SIRPa, e.g. Fc fusion proteins of 2B4 or CD2. In some embodiments, an anti-SIRPa antibody used herein can be a polyclonal or monoclonal antibody. A “humanized” anti-SIRPa antibody can be used if needed in order to avoid any potential use
incompatibilities (e.g. adverse reactions when introducing the ILC2 cells exposed to such an antibody, if needed). In some embodiments, an inhibitor of SIRPa can be a SIRPa -specific chimeric antigen receptor T (CAR-T) cell.
[0047] In some embodiments, an inhibitor of CD47-SIRPa suitable for use herein can be, but is not limited to, RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC-90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or a combination thereof.
[0048] In certain embodiments, compositions herein may encompass an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa. In some embodiments, compositions herein may encompass an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa, wherein a B7-H3 inhibitor can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof and a CD47-SIRPa, can be a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof. In some embodiments, compositions herein may encompass a B7-H3-specific siRNA, RNAi, microRNA or ribozyme and a CD47-specific siRNA, RNAi, microRNA or ribozyme and/or a SIRPa-specific siRNA, RNAi, microRNA or ribozyme. In some embodiments, compositions herein may encompass an anti-B7-H3 antibody and an anti-CD47 antibody and/or an anti-SIRPa antibody. In some embodiments, compositions herein may encompass: enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL- 015, or any combination thereof; and RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC- 90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or any combination thereof. c. Pharmaceutical Compositions
[0049] The B7-H3 inhibitors and/or CD47-SIRPa inhibitors, as described herein can be mixed with a pharmaceutically acceptable carrier (excipient) to form a pharmaceutical composition for use in treating a target disease. “Acceptable” means that the carrier must be compatible with the active ingredient of the composition (and preferably, capable of stabilizing the active ingredient) and not deleterious to the subject to be treated. Pharmaceutically acceptable excipients (carriers) including buffers, which are well known in the art. See, e.g., Remington: The Science and Practice of Pharmacy 20th Ed. (2000) Lippincott Areiams and Wilkins, Ed. K. E. Hoover.
[0050] The pharmaceutical compositions to be used in the present methods can comprise pharmaceutically acceptable carriers, excipients, or stabilizers in the form of lyophilized formulations or aqueous solutions. (Remington: The Science and Practice of Pharmacy 20th Ed. (2000) Lippincott Areiams and Wilkins, Ed. K. E. Hoover). Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations used, and comprise buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrans; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; saltforming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or nonionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG). In some examples, the pharmaceutical composition described herein comprises liposomes containing the antibodies (or the encoding nucleic acids) which can be prepared by methods known in the art, such as described in Epstein, et al., Proc. Natl. Acad. Sci. USA 82:3688 (1985); Hwang, et al., Proc. Natl. Acad. Sci. USA 77:4030 (1980); and U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556. Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
[0051] In some embodiments, one or more of the B7-H3 inhibitors and/or CD47-SIRPa inhibitors, can be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or
in macroemulsions. Such techniques are known in the art, see, e.g., Remington, The Science and Practice of Pharmacy 20th Ed. Mack Publishing (2000).
[0052] In some embodiments, pharmaceutical compositions described herein can be formulated in sustained-release format. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the inhibitors, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained- release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinyl alcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers ofL-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT™ (injectable microspheres composed of lactic acid- glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(-)-3- hydroxybutyric acid.
[0053] The pharmaceutical compositions to be used for in vivo administration must be sterile. This may be readily accomplished by, for example, filtration through sterile filtration membranes. Therapeutic compositions disclosed herein can be placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
[0054] The pharmaceutical compositions described herein can be in unit dosage forms such as tablets, pills, capsules, powders, granules, solutions or suspensions, or suppositories, for oral, parenteral or rectal administration, or administration by inhalation or insufflation.
[0055] For preparing solid compositions such as tablets, the principal active ingredient can be mixed with a pharmaceutical carrier, e.g., conventional tableting ingredients such as com starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g., water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a non-toxic pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 900 mg of the active ingredient of
the present invention. The tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer that serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate. Suitable surface-active agents (surfactant) include, in particular, non-ionic agents, such as poly oxy ethylenesorbitans (e.g., Tween™ 20, 40, 60, 80 or 85) and other sorbitans (e.g., Span™ 20, 40, 60, 80 or 85). Compositions with a surface-active agent are conveniently comprise between 0.05 and 5% surfaceactive agent, and can be between 0.1 and 2.5%. It are be appreciated that other ingredients may be added, for example mannitol or other pharmaceutically acceptable vehicles, if necessary.
[0056] Suitable emulsions may be prepared using commercially available fat emulsions, such as Intralipid™, Liposyn™, Infonutrol™, Lipofundin™ and Lipiphysan™. The active ingredient may be either dissolved in a pre-mixed emulsion composition or alternatively it may be dissolved in an oil (e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, com oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e.g. egg phospholipids, soybean phospholipids or soybean lecithin) and water. It are be appreciated that other ingredients may be added, for example glycerol or glucose, to adjust the tonicity of the emulsion. Suitable emulsions are typically contain up to 20% oil, for example, between 5 and 20%. The fat emulsion can comprise fat droplets between 0.1 and 1.0 .im, particularly 0.1 and 0.5 .im, and have a pH in the range of 5.5 to 8.0. The emulsion compositions can be those prepared by mixing an inhibitor with Intralipid™ or the components thereof (soybean oil, egg phospholipids, glycerol and water).
[0057] Pharmaceutical compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
[0058] Compositions in preferably sterile pharmaceutically acceptable solvents may be nebulized by use of gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device may be attached to a face mask, tent or intermittent positive pressure breathing machine. Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner. [0059] In some embodiments, pharmaceutical formulations herein may comprise an anti-B7- H3 antibody at a suitable concentration, for example, about 200 pg to about 1000 pg (e.g., about 200 pg, about 250 pg, about 300 pg, about 350 pg, about 400 pg, about 450 pg, about 500 pg, about 550 pg, about 600 pg, about 650 pg, about 700 pg, about 750 pg, about 800 pg, about 850 pg, about 900 pg, about 950 pg, about 1000 pg). In some embodiments, pharmaceutical formulations herein may comprise an anti-CD47 antibody at a suitable concentration, for example, about 50 pg to about 800 pg (e.g., about 50 pg, about 100 pg, about 150 pg, about 200 pg, about 250 pg, about 300 pg, about 350 pg, about 400 pg, about 450 pg, about 500 pg, about 550 pg, about 600 pg, about 650 pg, about 700 pg, about 750 pg, about 800 pg). In some embodiments, pharmaceutical formulations herein may compromise both an anti-B7-H3 antibody and an anti- CD47 antibody. In some embodiments, pharmaceutical formulations herein may comprise an anti- B7-H3 antibody at about 200 pg to about 1000 pg (e.g., about 200 pg, about 250 pg, about 300 pg, about 350 pg, about 400 pg, about 450 pg, about 500 pg, about 550 pg, about 600 pg, about 650 pg, about 700 pg, about 750 pg, about 800 pg, about 850 pg, about 900 pg, about 950 pg, about 1000 pg) and an anti-CD47 antibody at about 50 pg to about 800 pg (e.g., about 50 pg, about 100 pg, about 150 pg, about 200 pg, about 250 pg, about 300 pg, about 350 pg, about 400 pg, about 450 pg, about 500 pg, about 550 pg, about 600 pg, about 650 pg, about 700 pg, about 750 pg, about 800 pg). In some embodiments, pharmaceutical formulations herein may compromise an anti-B7-H3 antibody and an anti-CD47 antibody in two separate units. d. Packaging
[0060] In some embodiments, compositions disclosed herein may be packaged. In some aspects, packaging of a composition may be for storage, shipment, display for sale, or a combination thereof. In some embodiments, compositions may be packaged using one or more suitable materials known in the art. In some embodiments, compositions may be packaged using one or more suitable methods known in the art. In s some embodiments, the choice of packaging
material and/or packaging method is dependent on the dosage form of a composition disclosed herein to be packaged.
[0061] In some embodiments, compositions disclosed herein may be packaged wherein packaging increases the length of time a composition can be stored. As used herein, the “shelflife” of a composition is the length of time after formulation that a composition can maintain one or more physiological effects following administration to a subject as detailed herein. In some embodiments, compositions disclosed herein may be packaged wherein packaging increases the shelf-life of a composition by about 1 week, about 1 month, or about 6 months. In some embodiments, compositions disclosed herein may be packaged wherein packaging increases the length of time a composition can be stored at about -85°C to about -75°C. In some embodiments, compositions disclosed herein may be packaged wherein packaging of at least one composition component increases the length of time that composition component can be stored at room temperature by about 1 week, about 1 month, or about 6 months. In some embodiments, compositions disclosed herein may be packaged wherein packaging of at least one composition component increases the length of time that composition component can be stored at about -85°C to about -75°C by about 1 week, about 1 month, or about 6 months.
III. Methods
[0062] The present disclosure provides methods for treating solid tumors. In some embodiments, the present disclosure provides methods for treating solid tumors (e.g., melanoma) that are not responsive to one or more anti-cancer therapies. In some embodiments, the present disclosure provides methods for treating solid tumors (e.g., melanoma) that are not responsive to anti-programmed cell death 1/programmed cell death ligand 1 (PD1/PDL1) therapy using any of the compositions having an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa as disclosed herein. Also provided in the present disclosure are methods of inflaming a cold tumor microenvironment using any of the compositions having an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa as disclosed herein.
[0063] In some embodiments, the present disclosure provides methods of treating a solid tumor. In some embodiments, a solid tumor can be pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal
malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, and sarcomas. In an exemplary example, a solid tumor is a melanoma.
[0064] In some embodiments, a solid tumor to be treated by methods disclosed herein can have B7-H3 overexpression, overactive P-catenin signaling, or a combination thereof. In some aspects, a solid tumor to be treated by methods disclosed herein can have at least one genetic mutation. In some examples, a solid tumor can have a genetic mutation of BRAFV600E. In some examples, a solid tumor can have a genetic mutation resulting in the loss of PTEN.
[0065] Certain embodiments herein can provide methods for treating solid tumors (e.g., melanoma) in a subject in need thereof. The subject to be treated by the methods described herein can be a mammal, more preferably a human. Mammals include, but are not limited to, farm animals, sport animals, pets, primates, horses, dogs, cats, mice and rats. A human subject who needs the treatment may be a human patient having, at risk for, or suspected of having a target disease/disorder, such as a solid tumor, including a melanoma.
[0066] In some embodiments, a subject to any of the methods herein can be any subject for whom treatment or therapy is desired. In some embodiments, a subject can have or can be suspected of having cancer, a tumor, or any combination thereof. In some embodiments, a subject can have or can be suspected of having one or more primary tumors, one or more metastatic tumors such as solid tumors or any combination thereof. In some embodiments, a subject can be a mammal. In some embodiments, a subject can be a human patient. In some embodiments, a human patient such as an adult, child, adolescent, toddler, young adult or infant or fetus who is in need of the methods herein can be identified by routine medical examination, e.g., laboratory tests, biopsy, magnetic resonance imaging (MRI) scans, ultrasound exams, and the like.
[0067] In some embodiments, a subject to be treated by the methods described herein can be a human patient having, suspected of having, or a risk for developing a solid tumor. In some embodiments, a subject to be treated by the methods described herein can have a tumor wherein the tumor has at least one B7H3+ cell. In some embodiments, a subject to be treated by the methods described herein can have a tumor wherein the tumor has at least one CD47+ cell. In some embodiments, a subject to be treated by the methods described herein can have a tumor wherein the tumor has at least one CD47+ cell/B7H3+ cell.
[0068] In some examples, a human subject who needs the treatment may be a human patient having, at risk for, or suspected of having melanoma. Methods herein can be used to treat a human patient having a Stage 0, Stage I, Stage II, Stage III, or Stage IV melanoma. As used herein, a Stage 0 melanoma (melanoma in situ) has not grown deeper than the top layer of the skin (the epidermis). As used herein, a melanoma can be staged according to the Ameri can Joint Committee on Cancer (AJCC) TNM staging system. Using the AJCC TNM staging system for Melanomas: Stage 0 - The cancer is confined to the epidermis, the outermost skin layer and has not spread to nearby lymph nodes or to distant parts of the body; Stage I - The tumor is no more than 2 mm (2/25 of an inch) thick and might or might not be ulcerated and has not spread to nearby lymph nodes or to distant parts of the body; Stage II - The tumor is more than 1 mm thick and may be thicker than 4 mm, it might or might not be ulcerated, and the cancer has not spread to nearby lymph nodes or to distant parts of the body; Stage III - The tumor is no more than 4 mm thick and might or might not be ulcerated, the cancer has spread to 1, 2, 3, 4 or more nearby lymph nodes, and it has not spread to distant parts of the body, and Stage IV - The tumor can be any thickness and might or might not be ulcerated, the cancer might or might not have spread to nearby lymph nodes, and it has spread to distant lymph nodes or to organs such as the lungs, liver or brain.
[0069] In some embodiments, the subject is a human patient who is in need of enhancing immunity. For example, the human patient may have a solid tumor. Examples of solid tumor cancers include melanoma, pancreatic duct adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, cholangiocarcinoma, breast cancer, lung cancer (for example, non-small cell lung cancer, NSCLC, and small cell lung cancer, SCLC), upper and lower gastrointestinal malignancies (including, but not limited to, esophageal, gastric, and hepatobiliary cancer), squamous cell head and neck cancer, genitourinary cancers, ovarian cancer, and sarcomas. A subject having a solid tumor can be identified by routine medical examination, e.g., laboratory tests, organ functional tests, CT scans, or ultrasounds. In some embodiments, the subject to be treated by the method described herein may be a human cancer patient who has undergone or is subjecting to an anticancer therapy, for example, chemotherapy, radiotherapy, immunotherapy, or surgery.
[0070] As used herein, the term “treating” refers to the application or administration of a composition including one or more active agents to a subject, who has a target disease or disorder, a symptom of the disease/disorder, or a predisposition toward the disease/disorder, with the
purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect the disorder, a symptom of the disease or disorder, or the predisposition toward the disease or disorder.
[0071] Alleviating a target disease/disorder includes delaying the development or progression of the disease, or reducing disease severity or prolonging survival. Alleviating the disease or prolonging survival does not necessarily require curative results. As used therein, "delaying" the development of a target disease or disorder means to defer, hinder, slow, retard, stabilize, and/or postpone progression of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individuals being treated. A method that “delays” or alleviates the development of a disease, or delays the onset of the disease, is a method that reduces probability of developing one or more symptoms of the disease in a given time frame and/or reduces extent of the symptoms in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a number of subjects sufficient to give a statistically significant result.
[0072] “Development” or “progression” of a disease means initial manifestations and/or ensuing progression of the disease. Development of the disease can be detectable and assessed using standard clinical techniques as well known in the art. However, development also refers to progression that may be undetectable. For purpose of this disclosure, development or progression refers to the biological course of the symptoms. “Development” includes occurrence, recurrence, and onset. As used herein “onset” or “occurrence” of a target disease or disorder includes initial onset and/or recurrence.
[0073] In some embodiments, the present disclosure provides methods for reducing, ameliorating, or eliminating one or more symptom(s) associated with a solid tumor. The treatment methods disclosed herein involve the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa. In some embodiments, the present disclosure provides methods of administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa for reducing, ameliorating, or eliminating one or more symptom(s) associated with a solid tumor. In some embodiments, the present disclosure provides methods of administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa for reducing, ameliorating, or eliminating one or more symptom(s) associated with a solid tumor wherein administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as disclosed herein synergistically reduces,
ameliorates, and/or eliminates one or more symptom(s) associated with a solid tumor. In some embodiments, methods of administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa herein reduces, ameliorates, and/or eliminates one or more symptom(s) associated with a solid tumor at a faster rate compared to the rate after administering a single therapy of an inhibitor of B7-H3 or a single therapy of an inhibitor of CD47-SIRPa to a subject. In some embodiments, methods of administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa herein reduces, ameliorates, and/or eliminates one or more symptom(s) associated with a solid tumor at a rate that can be at least about 10% faster, at least about 20% faster, at least about 30% faster, at least about 40% faster, or at least about 50% faster compared to the rate after administering a single therapy of an inhibitor of B7-H3 or a single therapy of an inhibitor of CD47-SIRPa to a subject. In some embodiments, methods of administering a combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa herein reduces, ameliorates, and/or eliminates one or more symptom(s) associated with a solid tumor at a rate that can be about 10% to about 99% faster, about 15% to about 95% faster, or about 20% to about 90% faster compared to the rate after administering a single therapy of an inhibitor of B7- H3 or a single therapy of an inhibitor of CD47-SIRPa to a subject.
[0074] In some embodiments, an effective amount of the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa can be given to a subject having a solid tumor e.g., melanoma), wherein the subject is on a treatment involving the one or more chemotherapeutics. In some embodiments, an effective amount of the one or more chemotherapeutics are given to a subject having a solid tumor (e.g., melanoma), wherein the subject is on a treatment involving the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa. In some embodiments, an effective amount of the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa and an effective amount of the one or more chemotherapeutics are given to the subject, concurrently or sequentially.
[0075] In some embodiments, the methods of the present disclosure increase anti-tumor activity (e.g., reduce cell proliferation, tumor growth, tumor volume, tumor burden, tumor load, and/or number of metastatic lesions over time) as compared to levels prior to treatment or in a control subject. In some embodiments, the methods of the present disclosure increase anti-tumor activity (e.g., reduce cell proliferation, tumor growth, tumor volume, tumor burden, tumor load,
and/or number of metastatic lesions over time) by at least about 10%, least about 20%, least about 25%, least about 30%, least about 40%, least about 50%, least about 60%, least about 70%, least about 75%, least about 80%, least about 85%, least about 90%, least about 95%, or more as compared to levels prior to treatment or in a control subject. In some embodiments, the methods of the present disclosure increase anti-tumor activity (e.g., reduce cell proliferation, tumor growth, tumor volume, tumor burden, tumor load, and/or number of metastatic lesions over time) by at about 5% to about 99%, about 10% to about 95%, or about 15% to about 90% as compared to levels prior to treatment or in a control subject. In some embodiments, the methods of the present disclosure synergistically increase anti-tumor activity (e.g., reduce cell proliferation, tumor growth, tumor volume, tumor burden, tumor load, and/or number of metastatic lesions over time) as compared to levels after treatment of a subject with a single therapy of an inhibitor of B7-H3 or a single therapy of an inhibitor of CD47-SIRPa.
[0076] In some embodiments, reduction may be measured by comparing cell proliferation, tumor growth, and/or tumor volume in a subject before and after administration of composition disclosed herein. In some embodiments, a method of treating or ameliorating a cancer in a subject herein can ablate, reverse, and/or attenuate one or more symptoms of the cancer. In some embodiments, a method of treating or ameliorating a cancer in a subject herein can allow one or more symptoms of the cancer to improve. In some embodiments, a method of treating or ameliorating a cancer in a subject herein can allow one or more symptoms of the cancer to improve by at least about 10%, least about 20%, least about 30%, least about 40%, least about 50%, least about 60%, least about 70%, least about 80%, least about 90%, least about 95%, or more as compared to levels prior to treatment. In some embodiments, a method of treating or ameliorating a cancer in a subject herein can allow one or more symptoms of the cancer to improve by at about 5% to about 99%, about 10% to about 95%, or about 15% to about 90% as compared to levels prior to treatment.
[0077] In some embodiments, before, during, and after the administration of a composition disclosed herein, cancerous cells and/or biomarkers in a subject can be measured in a biological sample, such as blood, serum, plasma, urine, peritoneal fluid, and/or a biopsy from a tissue or organ.
[0078] In some embodiments, methods herein can include administration of a composition herein to reduce tumor volume, size, load, and/or burden in a subject to an undetectable size, or less than the subject's tumor volume, size, load, and/or burden prior to treatment. In some embodiments, methods herein can include administration of a composition herein to reduce tumor volume, size, load, and/or burden in a subject to an undetectable size, or to less than about 1%, less than about 2%, less than about 5%, less than about 10%, less than about 20%, less than about 25%, less than about 30%, less than about 40%, less than about 50%, less than about 60%, less than about 70%, less than about 75%, less than about 80%, or less than about 90% of the subject's tumor volume, size, load, and/or burden prior to treatment.
[0079] In some embodiments, methods herein can include administration of a composition herein to reduce the cell proliferation rate and/or tumor growth rate in a subject to an undetectable rate. In some embodiments, methods herein can include administration of a composition herein to reduce the cell proliferation rate and/or tumor growth rate in a subject to a rate less than the rate prior to treatment. In some embodiments, methods herein can include administration of a composition herein to reduce the cell proliferation rate and/or tumor growth rate in a subject to a rate less than about 1%, less than about 2%, less than about 5%, less than about 10%, less than about 20%, less than about 25%, less than about 30%, less than about 40%, less than about 50%, less than about 60%, less than about 70%, less than about 75%, less than about 80%, or less than about 90% of the rate prior to treatment.
[0080] In some embodiments, methods herein can include administration of a composition herein to reduce the development of and/or the number or size of metastatic lesions in a subject to an undetectable rate. In some embodiments, methods herein can include administration of a composition herein to reduce the development of and/or the number or size of metastatic lesions in a subject to a rate less than the rate prior to treatment. In some embodiments, methods herein can include administration of a composition herein to reduce the development of and/or the number or size of metastatic lesions in a subject to a rate less than about 1%, less than about 2%, less than about 5%, less than about 10%, less than about 20%, less than about 25%, less than about 30%, less than about 40%, less than about 50%, less than about 60%, less than about 70%, less than about 75%, less than about 80%, or less than about 90% of the rate prior to treatment.
[0081] In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to activate the innate immune system. In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to activate the innate immune system in a tumor microenvironment by at least about 20% (e.g., least about 20%, least about 30%, least about 40%, least about 50%, least about 60%, least about 70%, least about 80%, least about 90% or greater). In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to activate the innate immune system in a tumor microenvironment by about 20% to about 99%, by about 25% to about 95%, or by about 30% to about 90%. In some embodiments, the methods of the present disclosure synergistically activate the innate immune system in a tumor microenvironment compared to activation after treatment of a subject with a single therapy of an inhibitor of B7-H3 or a single therapy of an inhibitor of CD47- SIRPa.
[0082] In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to activate at least one population of innate immune system cells in a tumor microenvironment by at least about 20% (e.g., at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or greater). In some embodiments, the combined therapy of an inhibitor of B7- H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to activate at least one population of innate immune system cells in a tumor microenvironment by about 20% to about 99%, about 25% to about 95%, or about 30% to about 90%. In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may activate macrophages, dendritic cells (DCs), natural killer (NK) cells, or any combination thereof. In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may activate macrophages, dendritic cells (DCs), natural killer (NK) cells, or any combination thereof by at least about 20% (e.g., at least about 20%, at least about 30%, at least about 40%, at least about
50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or greater). In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47- SIRPa as described herein may activate macrophages, dendritic cells (DCs), natural killer (NK) cells, or any combination thereof. In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may activate macrophages, dendritic cells (DCs), natural killer (NK) cells, or any combination thereof by about 20% to about 99%, about 25% to about 95%, or about 30% to about 90%.
[0083] In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to inflame a cold tumor microenvironment. As used herein, the terms “hot” and “cold” are routinely used to refer to T cell-infiltrated, inflamed but non-infiltrated, and non-inflamed tumors. Characteristics of hot tumors can include, but are not limited to the presence of tumor-infiltrating lymphocytes (TILs), expression of anti-programmed death-ligand 1 (PD-L1) on tumor-associated immune cells, possible genomic instability and the presence of a preexisting anti-tumor immune response. Characteristics of cold tumors can include, but are not limited to poorly infiltrated with T cells, immunologically ignorant (scarcely expressing PD-L1), high proliferation with low mutational burden (low expression of neoantigens), and low expression of antigen presentation machinery markers such as major histocompatibility complex class I (MHC I). In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to inflame a cold tumor microenvironment by at least about 20% (e.g., at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or greater) in vivo. In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to inflame a cold tumor microenvironment by about 20% to about 99%, about 25% to about 95%, or about 30% to about 90%. In some embodiments, inflaming a cold tumor microenvironment using the methods disclosed herein can increase of at least one interferon, increase of CD8+ T cells, increase of CD44 expression in CD4+ cells, in CD8+ cells, or both, increased TNFalpha expression in T cells, or a combination thereof.
[0084] In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to a subject in need of the treatment at an amount sufficient to restore CD8+ T cell effector function in a cold tumor microenvironment.
[0085] In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to initiate immune-cell mediated cytotoxicity. In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to initiate CTL-cell mediated cytotoxicity. In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to initiate NK-cell mediated cytotoxicity. In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to modulate gene expression of GZMA, GZMB, NKG7, IL2R, HSPA1A, HSPA1, RANTES, CCL5, CCR5, CXCR4, CXCL12, or any combination thereof. In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to increase gene expression of GZMA, GZMB, NKG7, IL2R, HSPA1A, HSPAJ, RANTES, CCL5, CCR5, CXCR4, CXCL12, or any combination thereof In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to increase cytokine signaling in a subject. In some embodiments, the combined therapy of an inhibitor of B7- H3 and an inhibitor of CD47-SIRPa as described herein may be administered to increase cytokine signaling CCL5 (RANTES, CCL5/CCR5) signaling in a subject. In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to increase chemokine signaling in a subject. In some embodiments, the combined therapy of an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa as described herein may be administered to increase CXCR4 (CXCR4/CXCL12) signaling in a subject.
[0086] Conventional methods, known to those of ordinary skill in the art of medicine, can be used to administer the compositions herein to a subject, depending upon the type of disease to be treated or the site of the disease. In some embodiments, the combined therapy of B7-H3 inhibitors and CD47-SIRPa inhibitors can be administered to a subject by intravenous infusion. Injectable compositions may contain various carriers such as vegetable oils, dimethylactamide,
dimethyformamide, ethyl lactate, ethyl carbonate, isopropyl myristate, ethanol, and polyols (glycerol, propylene glycol, liquid polyethylene glycol, and the like). For intravenous injection, water soluble antibodies can be administered by the drip method, whereby a pharmaceutical formulation containing the B7-H3 inhibitor and/or CD47-SIRPa inhibitor and a physiologically acceptable excipient can be infused. Physiologically acceptable excipients may include, for example, 5% dextrose, 0.9% saline, Ringer’s solution or other suitable excipients. Intramuscular preparations, e.g., a sterile formulation of a suitable soluble salt form of the antibody, can be dissolved and administered in a pharmaceutical excipient such as Water-for-Inj ection, 0.9% saline, or 5% glucose solution.
[0087] In some embodiments, the combined B7-H3 inhibitor and CD47-SIRPa inhibitor therapy herein can be administered concurrently with the one or more chemotherapeutics. In some embodiments, the combined B7-H3 inhibitor and CD47-SIRPa inhibitor therapy can be administered before or after the one or more chemotherapeutics. In some embodiments, the B7- H3 inhibitor and CD47-SIRPa inhibitor therapy can be administered after administration of a combined therapy of the B7-H3 inhibitor and CD47-SIRPa inhibitor therapy and one or more chemotherapeutics. In some embodiments, the B7-H3 inhibitor therapy can be administered after administration of a combined therapy of the B7-H3 inhibitor and CD47-SIRPa inhibitor therapy. In some embodiments, the CD47-SIRPa inhibitor therapy can be administered after administration of a combined therapy of the B7-H3 inhibitor and CD47-SIRPa inhibitor therapy. In some embodiments, the one or more chemotherapeutics can be administered systemically. In some embodiments, the one or more chemotherapeutics is administered locally. In some embodiments, the one or more chemotherapeutics can be administered by intravenous administration, e.g., as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerebrospinal, subcutaneous, intra-arterial, intra-articular, intrasynovial, intrathecal, intratumoral, oral, inhalation or topical routes. In one embodiment, the one or more chemotherapeutics is administered to the subject by intravenous infusion. The one or more chemotherapeutics may include an antimetabolite, a microtubule inhibitor, or a combination thereof. Antimetabolites include, for example, folic acid antagonist (e.g., methotrexate) and nucleotide analogs such as pyrimidine antagonist (e.g., 5-fluorouracil, foxuridine, cytarabine,
capecitabine, and gemcitabine), purine antagonist (e.g., 6-mercaptopurine and 6-thioguanine), and adenosine deaminase inhibitor (e.g., cladribine, fludarabine and pentostatin).
[0088] In some embodiments, the methods are provided, the combined B7-H3 inhibitor and CD47-SIRPa inhibitor therapy can be administered prior to, concurrently, or subsequent administration of a checkpoint inhibitor that is not a B7-H3 inhibitor or a CD47-SIRPa inhibitor. In some embodiments, checkpoint inhibitors that are not a B7-H3 inhibitor or a CD47-SIRPa inhibitor can include a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof capable of inhibiting one or more checkpoint proteins. Non-limiting examples of checkpoint proteins that can be the target of a checkpoint inhibitor that is not a B7-H3 inhibitor or a CD47-SIRPa inhibitor can include CTLA- 4, PDL1, PDL2, PD1, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4 (belongs to the CD2 family of molecules and is expressed on all NK, y5, and memory CD8+ (aP) T cells), CD160 (also referred to as BY55), CGEN-15049, CHK 1 and CHK2 kinases, A2aR and various B-7 family ligands.
[0089] An effective amount of the pharmaceutical composition described herein can be administered to a subject (e.g., a human) in need of the treatment via a suitable route, systemically or locally. In some embodiments, a B7-H3 inhibitor and CD47-SIRPa inhibitor therapy may be administered by intravenous administration, e.g., as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerebrospinal, subcutaneous, intra-arterial, intraarticular, intrasynovial, intrathecal, intratumoral, oral, inhalation or topical routes.
[0090] Determination of whether an amount of the combined B7-H3 inhibitor and CD47- SIRPa inhibitor therapy achieved a therapeutic effect would be evident to one of skill in the art. Effective amounts vary, as recognized by those skilled in the art, depending on the particular condition being treated, the severity of the condition, the individual patient parameters including age, physical condition, size, gender and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose of the individual components or combinations thereof be used, that is, the highest safe dose according to sound medical judgment.
[0091] Empirical considerations, such as the half-life, generally contribute to the determination of the dosage. For example, where the combined B7-H3 inhibitor and CD47-SIRPa inhibitor therapy includes anti-B7-H3 and anti-CD47/SIRPa antibodies that are compatible with the human immune system, such as humanized antibodies or fully human antibodies, the half-life of the antibody may be prolonged. Frequency of administration may be determined and adjusted over the course of therapy, and is generally, but not necessarily, based on treatment and/or suppression and/or amelioration and/or delay of a target disease/disorder. Alternatively, sustained continuous release formulations of an antibody may be appropriate. Various formulations and devices for achieving sustained release are known in the art.
[0092] A subject having a target solid tumor as disclosed herein, for example, melanoma, can be identified by routine medical examination, e.g., laboratory tests, organ functional tests, genetic tests, interventional procedure (biopsy, surgery) any and all relevant imaging modalities,. In some embodiments, the subject to be treated by the method described herein is a human cancer patient who has undergone or is subjecting to an anti-cancer therapy, for example, chemotherapy, radiotherapy, immunotherapy, or surgery. In some embodiments, subjects may have received prior immuno-modulatory anti-tumor agents. Non-limiting examples of such immuno-modulatory agents include, but are not limited to as anti-PDl, anti-PD-Ll, anti-CTLA-4, anti-OX40, anti- CD137, and the like. In some embodiments, a subject shows disease progression through the treatment. In other embodiments, a subject is resistant to the treatment (either de novo or acquired). In some embodiments, such a subject is demonstrated as having advanced malignancies (e.g., inoperable or metastatic). Alternatively or in addition, in some embodiments, the subject has no standard therapeutic options available or ineligible for standard treatment options, which refer to therapies commonly used in clinical settings for treating the corresponding solid tumor.
[0093] In some embodiments, the subject herein may be a human patient having a refractory disease, for example, a refractory melanoma. As used herein, “refractory” refers to the tumor that does not respond to or becomes resistant to a treatment. In some embodiments, the subject herein may have a tumor that is resistant to at least one immune-modulatory anti-tumor agents. In some embodiments, the subject herein may have a tumor that is resistant to anti-PDl, anti-PD-Ll, or a combination thereof. In some embodiments, the subject may be a human patient having a relapsed disease, for example, a relapsed melanoma. As used herein, “relapsed” or “relapses” refers to a
tumor that returns or progresses following a period of improvement (e.g., a partial or complete response) with treatment.
IV. Kits for Use in Therapy of Solid Tumors
[0094] The present disclosure also provides kits for use in treating or alleviating a solid tumor, for example, melanoma, PDA, CRC, HCC, or cholangiocarcinoma, and others described herein. Such kits can include one or more containers having an inhibitor of B7-H3 and an inhibitor of CD47-SIRPa. A kit provided herein can optionally have one or more chemotherapeutics.
[0095] In some embodiments, the kit can have instructions for use in accordance with any of the methods described herein. The included instructions can include a description of administration of the inhibitor of B7-H3 and/or inhibitor of CD47-SIRPa to treat, delay the onset, or alleviate a target disease as those described herein. In some embodiments, the kit can further comprise a description of selecting an individual suitable for treatment based on identifying whether that individual has the target disease, e.g., applying the diagnostic method as described herein. In still other embodiments, the instructions comprise a description of administering an antibody to an individual at risk of the target disease.
[0096] The instructions relating to the use of an inhibitor of B7-H3 and/or inhibitor of CD47- SIRPa can generally include information as to dosage, dosing schedule, and route of administration for the intended treatment. The containers may be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses. Instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit), but machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk) are also acceptable.
[0097] The label or package insert indicates that the composition is used for treating, delaying the onset and/or alleviating the solid tumor. In some embodiments, instructions are provided for practicing any of the methods described herein.
[0098] The kits of this invention are in suitable packaging. Suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. Also contemplated are packages for use in combination with a specific device, such as an inhaler, nasal administration device (e.g., an atomizer) or an infusion device such as a minipump. In some embodiments, a kit has a sterile access port (for example the container may be an
intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). In some embodiments, the container also has a sterile access port (for example the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is an inhibitor of B7-H3 and/or inhibitor of CD47- SIRPa such as those described herein.
[0099] Kits may optionally provide additional components such as buffers and interpretive information. Normally, the kit comprises a container and a label or package insert(s) on or associated with the container. In some embodiments, the invention provides articles of manufacture comprising contents of the kits described above.
General Techniques
[00100] The practice of the present invention are employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art. Such techniques are explained fully in the literature, such as, Molecular Cloning: A Laboratory Manual, second edition (Sambrook, et al., 1989) Cold Spring Harbor Press; Oligonucleotide Synthesis (M. J. Gait, ed., 1984); Methods in Molecular Biology, Humana Press; Cell Biology: A Laboratory Notebook (J. E. Cellis, ed., 1998) Academic Press; Animal Cell Culture (R. I. Freshney, ed., 1987); Introduction to Cell and Tissue Culture (J. P. Mather and P. E. Roberts, 1998) Plenum Press; Cell and Tissue Culture: Laboratory Procedures (A. Doyle, J. B. Griffiths, and D. G. Newell, eds., 1993-8) J. Wiley and Sons; Methods in Enzymology (Academic Press, Inc.); Handbook of Experimental Immunology (D. M. Weir and C. C. Blackwell, eds.); Gene Transfer Vectors for Mammalian Cells (J. M. Miller and M. P. Calos, eds., 1987); Current Protocols in Molecular Biology (F. M. Ausubel, et al., eds., 1987); PCR: The Polymerase Chain Reaction, (Mullis, et al., eds., 1994); Current Protocols in Immunology (J. E. Coligan et al., eds., 1991); Short Protocols in Molecular Biology (Wiley and Sons, 1999); Immunobiology (C. A. Janeway and P. Travers, 1997); Antibodies (P. Finch, 1997); Antibodies: a practical approach (D. Catty., ed., IRL Press, 1988-1989); Monoclonal antibodies: a practical approach (P. Shepherd and C. Dean, eds., Oxford University Press, 2000); Using antibodies: a laboratory manual (E. Harlow and D. Lane (Cold Spring Harbor Laboratory Press, 1999); The Antibodies (M. Zanetti and J. D. Capra, eds., Harwood Academic Publishers, 1995).
[00101] Without further elaboration, it is believed that one skilled in the art can, based on the above description, utilize the present invention to its fullest extent. The following specific embodiments are, therefore, to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever. All publications cited herein are incorporated by reference for the purposes or subject matter referenced herein.
EXAMPLES
[00102] While the present disclosure has been described with reference to the specific embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the disclosure. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process step or steps, to the objective, spirit, and scope of the present disclosure. All such modifications are intended to be within the scope of the disclosure.
Example 1. B7-H3 was overexpressed in melanoma and regulated tumor cell intrinsic signals that affected the tumor microenvironment
[00103] To determine the expression pattern of B7-H3 in melanoma, protein (n = 13 cell lines) and mRNA (n = 28 cell lines) levels were examined in a panel of primary and metastatic human melanoma cells and human melanocytes. Human primary cell lines tested included: WM853, WM902B, WM983A, WD1341D, WM1366, and WM1862. Human metastatic melanoma cell lines tested included: MeWo, SkMel28, Htl44, MM485, and 501Mel. All cell lines were grown and maintained in 2% Melanoma Medium (4:1 MCDB153 with 1.5 g/L sodium bicarbonate and Leibovitz’s L-15 medium with 2 mM L-glutamine, 0.005 mg/ml bovine insulin, 1.68 mM CaCh, 2% fetal bovine serum). All cells were maintained at 37°C in a 5% CO2 environment.
[00104] To determine the expression pattern of B7-H3 protein in the cell lines, cultured cells were harvested and resulting cell lysates subjected to immunoblotting. In brief, cells were solubilized in 200 pl of sodium dodecyl sulfate-polyacrylamide gel electrophoresis sample buffer (4% sodium dodecyl sulfate, 60 mmol/L Tris, pH 6.8, 5% glycerol, 0.01% bromophenol blue, and 50 mmol/L mercaptoethanol), and heated to 95°C for 5 minutes. Whole cell protein extracts (20 to 40 pg) were separated on 6 to 15% sodium dodecyl sulfate-polyacrylamide gels. Proteins were electrophoretically transferred onto polyvinylidene difluoride membranes for 1 hour, washed in
Tris-buffered saline (TBS; 100 mmol/L Tris-HCl, pH 7.5, and 150 mmol/L NaCl), before being blocked for 1 hour in TBS containing 0.1% Tween-20 and 5% milk (TBST-milk). Primary antibody (anti-human B7-H3) incubations were performed overnight at 4°C in TBST-milk followed by washing and then a 1-hour incubation with either anti-rabbit or anti-mouse horseradish peroxidase-conjugated secondary antibody diluted in TBST-milk. Immunocomplexes were visualized using a chemiluminescence system and detected on photographic film. After analysis, Western blots were stripped once and reprobed (anti-P-actin) to demonstrate even protein loading. [00105] To determine the expression pattern of B7-H3 mRNA in the cell lines, cultured cells were harvested and resulting cell lysates subjected to quantitative real-time PCR (qRT-PCR). In brief, total RNA was extracted from samples using the RNeasy Protect Mini Kit (Qiagen). The RNA Integrity Number of each sample was determined using the 2100 Bioanalyzer instrument (Agilent Technologies). Samples were selected with stringent criteria based on RNA Integrity Number of 7.0 or greater, and a minimum of 500 ng of total RNA. For validating the transcript signatures in melanoma cell lines, 1 pg total RNA was reverse transcribed to cDNA using RNA to cDNA EcoDry™ Premix (Random Hexamers) (Takara Bio). The cDNA was next assayed in quantitative real-time PCR (qPCR) using the QuantiNova™ SYBR® Green PCR Kit (Qiagen) on the CFX384™ Real-Time System (Bio-Rad). GAPDH was used as a housekeeping gene for normalization.
[00106] Metastatic cell lines showed significantly higher levels of B7-H3 expression both at the protein (Fig. 1A) and mRNA level (Fig. IB) as compared to primary melanoma cells, suggesting a role for B7-H3 during progression from primary disease to metastasis.
[00107] To assess the frequency of aberrant expression in metastatic disease, we next stained a tissue microarray consisting of metastatic human melanoma samples (n = 46) with anti-B7-H3 using immunohistochemistry. Briefly, 46 approximately 2 mm cores were retrieved from human melanoma samples and embedded into a single tissue microarray paraffin block. For immunohistochemistry, 5-pm sections of the tissue microarray paraffin block were subjected to antigen retrieval and blocking using Target Retrieval Solution and Serum-Free Protein Block (Dako), sequentially. Sections were further stained with diluted antibody with Background Reducing Components and Streptavidin/HRP (Dako). Signals were developed via the SignalStain® DAB Substrate Kit (Cell Signaling). Figs. 1C and ID show that high levels of
membranous and cytoplasmic expression of B7-H3 were found in a significant portion of the cases - 61%.
[00108] To gain insight into B7-H3 functions, the downstream effectors of B7-H3 were examined. Two human metastatic melanoma cell lines were selected for the study, Hs. 688(A)T and Hs. 839T, with the highest levels of B7-H3 expression. Cells were transfected with either scrambled or B7-H3 specific siRNAs in triplicates and silencing was confirmed using quantitative RT-PCR according to the methods described in this example. The transcriptomes were also analyzed via RNA-sequencing. In brief, the total RNA extracted from the samples was subjected to ribosomal RNA depletion and further used as input for library construction by the Illumina TruSeq Standard Total RNA library Pre Gold kit (Illumina). The libraries were then sequenced on the Illumina HiSeq 2500 system (Illumina) using 2 x 100 bp paired-end protocol to a minimum mean coverage of 50x. Raw reads were quality filtered by trimming the read ends; bases with quality <20 were removed. Next, the quality at the 20th percentile was computed. The entire read was discarded if the quality at the 20th percentile was less than 15. Reads shorter than 40 bases after trimming were discarded as well. If one or more reads in the pair failed the quality check both reads were discarded. Paired-end RNA sequencing reads were mapped to the human reference genome (Ensembl annotation, build 37) and to sequences from the Repbase database of human repetitive elements (release 19) using the STAR aligner in a manner similar to that described in Dobin et al., Bioinformatics. 2013;29(l): 15-21, the disclosure of which is incorporated herein in its entirety. Aligned reads were assigned to genes using the featureCounts function of the Rsubread package via the external Ensembl annotation. This procedure generated the raw read counts for each gene. Gene expression in the form of log2 counts per million reads was computed and normalized across the samples using the TMM method as implemented in the cal cNormF actors function of edgeR package in a manner similar to that described in Robinson et al., Bioinformatics. 2010;26(l): 139-140 and Robinson et al., Genome Biol. 2010;l 1(3):R25, the disclosures of which are incorporated herein in their entirety. Differential expression analysis was performed using the limma software package. Expression data were used in conjunction with the weights computed by the voom transformation based on the mean-variance relationship of log read counts to calculate moderated t-statistics using empirical Bayes in a manner similar to that described in Law et al., Genome Biol. 2014;15(2):R29,the disclosure of which is incorporated
herein in its entirety. For protein-coding RNA (mRNA), the gene signature differentiating each pair of subtypes was identified. For this analysis, only genes with counts per million reads greater than 10 in at least 2 samples were considered. To create a comprehensive global heatmap depicting the gene signature of different subtypes, genes to be differentially expressed with a Benjamini- Hochberg-corrected P value of P < 0.005 and a fold change of >1.5 were considered. Principal component analysis of the resulting sets of differentially expressed genes confirmed effective grouping of subtypes. To determine the biological modules that are affected in our differentially expressed gene set, the Database for Annotation, Visualization and Integrated Discovery (DAVID), Ingenuity Pathway Analysis, and the Gene Ontology database were used. Pathways were examoined with a Benjamini -Hochberg-adjusted P value of P < 0.05. The analysis revealed eleven up- and six down-regulated genes upon silencing of B7-H3 that were consistent across both cell lines (Figs. 1E-1F). Surprisingly, it was found that majority of B7-H3 downstream effectors are modulators of the tumor microenvironment -MMP3 (matrix metalloproteinase family), P3H1 and FBLN5 (collagen family), ITGA2 (integrin family), CXCL8 (IL8, inflammation), and interferon stimulating genes IFIT2 (antiviral responses), CMPK2 (antiviral responses), OASL (antiviral responses), and RSAD2 (antiviral responses). These data suggested that B7-H3, when overexpressed, had key functions in shaping the tumor microenvironment by suppressing antiviral and inflammatory responses, and by regulating matricellular proteins.
[00109] To study the effects of B7-H3 on the microenvironment, a suitable model system was searched for next. Prior studies on B7-H3 utilized the B 16 spontaneous mouse melanoma model that has several limitations, as it does not reflect the genetic alterations, ultraviolet-driven Protein and mRNA expression levels of B7-H3 in a panel of murine cells was examined by immunoblotting (Fig. 1G) blotting and quantitative RT-PCR (Fig. 1H), respectively using methods described earlier in this Example. While melan-a cells - an immortalized murine melanocyte line with loss of Cdkn2a - and B16 melanoma lines showed significantly low levels of B7-H3 expression, cells obtained from genetically engineered murine models (GEMM), in particular YUMM2.1 cells with activated P-catenin signaling (BrafV600E/wt/Pten'/7Cdkn2a+/‘ /Bcatloxex3/wt) revealed significantly high levels (Figs. 1G and 1H). Expression of B7-H3 was also notable in YUMM1.7/YUMMER1.7 cells (BrafV600E/wt/Pten'/7Cdkn2a'/‘), but cells with activated P-catenin, YUMM2.1, had the highest level.
[00110] Next, B7-H3 or P-catenin was silenced using gene-specific siRNAs. Fig. II shows that silencing of B7-H3 had no effect on P-catenin, but knocking down P-catenin lowered B7-H3 protein levels, suggesting that B7-H3 was in part modulated by P-catenin in YUMM2.1 cells. The downstream effectors of B7-H3 in YUMM2.1 murine cells were compared to those found in human melanomas by quantitative RT-PCR (Fig. 1J). Majority such s RAB27B and GPRC5A, as well as interferon stimulating genes - IFIT2, CMPK2, OASL, and RSAD2 - were validated. In aggregate, it was concluded that YUMM2.1 cells represented a suitable model and offer a unique advantage to study B7-H3 and its effects on the local microenvironment.
[00111] To address the relationship between B7-H3 and Wnt/p-catenin signaling in human cells, their correlation was examined by mining the Reverse Phase Protein Array dataset from the Cancer Cell Line Encyclopedia (CCLE, n = 899). A co-occurrence of B7-H3 and Wnt signaling pathway proteins was found including P-catenin, whereas anti-correlation was noted between B7- H3 and VAV1 (Fig. IK). In sum, B7-H3 overexpression co-occurred with activated P-catenin signaling in YUMM2.1 cells and a subset of human cancers. YUMM2.1 (as well as YLnMM1.7/YUMMER1.7) preclinical model represents a suitable system to study the local microenvironment regulated by B7-H3.
Example 2. Blockade of B7-H3, but not PD-1, resulted in an inflamed tumor microenvironment in a preclinical model of activated P-catenin signaling
[00112] Clinical and preclinical studies demonstrated that melanomas driven by P-catenin, in particular those co-occurring with BRAFV600E and loss of PTEN as in YUMM2.1 model are highly metastatic and unresponsive to anti-PD-l/anti-CTLA-4, due to lack of immune cell infiltration and T-cell exclusion. This example aimed to investigate the tumor microenvironment changes driven by B7-H3 in a syngeneic tumor system (YUMM2.1) and explore therapeutic strategies by targeting B7-H3.
[00113] Immunocompetent C57BL/6J mice were injected with 1 x 106 YUMM2.1 murine melanoma cells subcutaneously into the flanks of each mouse. Nine days after the first injection, 600 pg of IgG control, 600 pg of anti-B7-H3 (clone MJ18, BioXCell), 300 pg of anti-PDl (clone RMP1-14, BioXCell), or 600 pg of anti-B7-H3 and 300 pg of anti-PDl were injected intraperitoneally (i.p.) where IgG or anti-B7-H3 was injected three times a week and anti-PDl was injected every five days. Injections were continued until the resulting tumors reached 20-50 mm3.
Antitumor activities and changes within the local milieu were then examined by RNA-sequencing. In brief, total RNA isolated from murine tumor bulk was subjected to RNA sequencing similar to methods described in example 1. Using the resulting sequencing data, first assessed was the quality of paired-end reads with FASTQC (vO.l 1.8). Next, reads were filtered with BBDUK from BBTOOLS (v37.53) to remove adapters, known artefacts, and quality trimmed (PHRED quality score < 10). Reads that became too short after trimming (N<60 bp) were discarded. Singleton reads (i.e. reads whose mate has been discarded) were not retained. The transcript-level quantification of cleaned data was estimated using SALMON (vl.0.0) by a quasi -mapping on Mus musculus GRCm38. The gene-level quantification was estimated using tximport library and converted 22,231 mouse gene expression to 15,091 ortholog human gene expression using biomaRt library. Differential expression was assessed on tumor samples under IgG treatment (n = 3), B7-H3 inhibitor treatment (n = 3), PD-1 inhibitor treatment (n = 3), and combination treatment of B7-H3 inhibitor with PD-1 inhibitor (n = 3) using R DESeq2 library (vl.81.1) similar to the methods described in Luo et al., BMC Bioinformatics. 2009; 10: 161, the disclsure of which is incoprotaed herein in its entirety. The differential expression between two conditions was called when the adjusted /J- value was at 5% and log2 fold change was more than 1. Next, gene set enrichment analysis was performed that captured pathways perturbed towards both directions simultaneously using the 15,091 ranked genes identified in our dataset and annotated in ENSEMBL (v94) against the KEGG pathways using R GAGE (v2.28.2) and Pathview (vl.18.2) libraries. Heatmap was drawn using R ComplexHeatmap library (v2.0.0). Fig. 2A shows that tumor reduction was observed with either anti-B7-H3 or anti -PD-1 therapy, but no synergy was observed when combined. Even though the anti -PD-1 versus control appeared to be responsive to this treatment, the interpretation of the data on the murine system was regarded as unresponsive or resistant based on clinical trials and preclinical work, and that any effective treatment should flatten the growth curve significantly as compared to anti-PD-1 alone group.
[00114] To identify the composition of the immune cell infiltrate within the tumor milieu, next analyzed was the major immune cell subsets and functional categories (i.e. CD4+ T cells, CD8+ T cells, NK cells, dendritic cells, B cells, and macrophages) from the dissected tumors using bulk RNA sequencing as well as CD45+ sorted cells (ultra low RNA sequencing, each treatment group in triplicates) using methods described in the examples herein. As shown in Fig. 2B,
immune cell lineage-specific transcript signatures, in particular innate (macrophages, DCs, and NK cells) populations of the immune system, were significantly upregulated in mice treated with anti-B7-H3 antibody as compared to the isotype control, or anti-PD-1, and to a lesser extent with the combination of both antibodies. It was noted that there was an upregulation of immune cell recruitment (CCR5), inflammatory (IL4R, JAK1/3, and STAT6), and macrophage inhibitory signals (CD47, SIRPa), as well as Interferon pathways (type I IFN (IFNAR2/JAK1/STAT1) and type II IFN (IFNGR1/2, JAK1/2)) upon B7-H3 inhibition in the P-catenin activated YUMM2.1 tumors (Fig. 2C).
[00115] It was next determined whether similar results were observed when YUMMER1.7 tumor model was used in an identical experiment. To generate the YUMMER1.7 tumor models, immunocompetent C57BL/6J mice were injected with 5 x IO5 YUMMER1.7 murine melanoma cells subcutaneously into the flanks of each mouse. Following injection of the YUMMER1.7 cells, mice were administered the same treatments in the same manner as that described above for the YUMM2.1 tumor model mice. While B7-H3 inhibition resulted in more immune cell infiltration in YUMMER1.7 tumors as compared to control or PD-1 inhibition, the degree of infiltration and inflammation was significantly higher in the P-catenin activated YUMM2.1 tumors in mice treated with B7-H3 inhibitor as compared to YUMMER1.7 tumors (Fig. 2D). CCR5 and IL4R (JAK1/3 and STAT6) signaling axes were elevated when B7-H3 is inhibited in both models, whereas there was increased type I IFN (IFNAR2/JAK1/STAT1) and type II IFN (IFNGR1/2, JAK1/2), CCR5/CCR1, and TLR4/RF5 signaling in the p-catenin activated YUMM2. 1 tumors. Macrophage signature, in particul r m crophage inhibitory signals (CD47, SIRPa) was confirmed using CD45+ RNA sequencing (Fig. 2E). Macrophage signature was also confirmed in harvested tumor tissue by immunohistochemistry using methods described in the examples herein. Figs. 3A-3C show IBA-1 (Allograft Inflammatory Factor 1) immunohistochemistry of tumors in treatment groups with IgG control, anti-B7-H3, or anti-PD-1 showing activated macrophages within the tissue whereas Figs. 3D-3F show CD47 expression of the tumor cells and Figs. 3G-3I show that SIRPa expression, a receptor for CD47 in immune subpopulations such as macrophages and DCs, was also indicated. An intense macrophage signal depicted by IBA-1 expression (activated macrophages) in the anti-B7-H3 group as compared to the others was noted (Figs. 3A-3C). All together, these data suggest that elevated levels of B7-H3 confer immunosuppressive signals, and
its inhibition leads to reprogramming of the tumor microenvironment in favor of macrophage infiltration and potential macrophage checkpoint activation (CD47/Sirpa), enrichment of myeloid recruitment signals (CCR5), and pro-inflammatory signals (IL4R).
Example 3. Blockade ofB7-H3 together with CD47, as well as inhibition of PD-1 and CD47, were synergistic in reducing tumor growth in a preclinical model of activated f- catenin signaling
[00116] Since CD47 and its interacting partner SIRPa were significantly upregulated in tumors upon anti-B7-H3 treatment, it was next explored whether combining and co-targeting molecules signaling distinct pathways, B7-H3 and CD47, had a synergistic therapeutic effect. In addition to treating YUMM2.1 tumor models with IgG, anti-B7-H3, mice were also injected i.p. with 200 pg of anti-CD47 (clone MIAP301, BioXCell) three times per week. Growth patterns in YUMM2.1 tumors upon treatment with IgG, anti-B7-H3, anti-CD47, or combination of the two showed similar antitumor activities with anti-B7-H3 or anti-CD47 alone, but importantly significant tumor reduction and additive benefit was noted when B7-H3 and CD47 inhibitors were combined (Fig. 4A, Fig. 5A). It was then explored whether inhibiting PD-1 was synergistic with CD47 in this model, and it was found that such additive therapeutic benefit does exist, and this combination may be a candidate in the clinical setting as well (Fig. 4B, Fig. 8A).
[00117] Next, the YUMM2.1 tumor models were subjected to NK cell and CD8+ T cell depletion experiments. In brief, for NK cell depletion experiments, mice were pretreated with 200 pg of anti-NKl .1 (clone PK136, BioXCell) via i.p. on days 1 and 3 prior to the murine melanoma cell challenge, and then received treatment of 150 pg twice a week. For the CD8+ T cell depletion experiment, anti-CD8a (clone YTS169.4 BioXcell) 250 pg via i.p. three times per week was used. Tumor volumes were calculated using the following equation: 0.5 x 1 x w2. NK cell and CD8+ T cell depletion experiments showed dependency to both antitumor effector mechanisms upon anti- B7-H3 treatment (Figs. 4C and 4D). It was next sought to determine therapeutic benefit of blocking B7-H3 and PD-1, B7-H3 and CD47, or PD-1 and CD47 in a different preclinical melanoma model, YUMMER1.7, driven by Braf 600E/wt/Pten’/7Cdkn2a'/' and high levels of B7- H3. Experiments did not demonstrate any additive benefit of CD47 combination, nor by combining B7-H3 and PD-1 inhibitors together (Figs. 4E and 4F).
[00118] To determine the mechanism of tumor cytotoxicity, single cell RNA sequencing
(scRNA sequencing) was performed on CD45+ sorted cells. In brief, CD45+ cells were sorted from murine tumor samples undergoing IgG (n = 3), B7-H3 inhibition (n = 3), CD47 inhibition ( = 3), and combination treatment with B7-H3 and CD47 inhibition (n = 3). Single cell suspensions were stained with diluted hashtag antibodies according to the New York Genome Center hashing protocol, similar to the methods described in Stoeckius et al., Genome Biol 19, 224 (2018), the disclosure of which is incorporated herein in its entirety. Hashed samples were pooled in equal amounts of live cells to achieve a target of 2 million cells/ml. The hashed sample pool was then loaded onto two lanes of the lOx Genomics NextGen 5’vl.l assay according to the manufacturer’s instructions with a targeted cell recovery of 20,000 cells. Gene expression libraries were made as per the lOx Genomics protocol. During cDNA amplification, hashtag oligonucleotides (HTO) were enriched with the addition of 3 pmol of a HTO Additive primer. The PCR product was isolated from the mRNA-derived cDNA via SPRI Select size selection and used for making HTO libraries according to the New York Genome Center hashing protocol. All libraries were quantified via Agilent 2100 hsDNA Bioanalyzer and KAPA library quantification kit (Roche). Gene expression libraries were sequenced at a targeted depth of 25,000 reads per cells, and HTO libraries were sequenced at a targeted read depth of 1,000 reads per cell. All libraries were sequenced on the Illumina NovaSeq S2 100 cycle kit using run parameters set to 28x8x0x60 (Rlxi7xi5xR2). For the scRNA sequencing data analysis, BCL files were first base-called and demultiplexed using cellranger mkfastq v5.0.1. Alignment to mouse reference mm 10 and feature counting were performed using cellranger count v5.0.1. Then, HTO barcodes were mapped to samples of origin. Briefly, a “key” matrix with biological samples as rows and HTO features as columns was created. A cell was populated with a value of “ 1 ” if the sample was supposed to be positive for the hashtag, “0” otherwise. For each barcode, pairwise distances with a cosine similarity metric were computed from the key matrix to generate a cosine similarity matrix. Thus, a barcode was initially assigned to a sample based on the largest cosine similarity to a sample. Additionally, for each barcode, a signal to noise ratio (sn) was calculated by subtracting its highest distance metric from its second highest distance metric. The sn ratio for each initially assigned sample usually followed a bi-modal distribution. In order to identify the local minimum of this distribution, four standard deviations were calculated from the mode of the right most identified local maximum. Only barcodes superior or equal to local minimum were kept as the final barcodes belonging to that sample. Data were
then read and analyzed using Bioconductor libraries including DropletUtils (vl .12.3) and scater (v.1.20.1). The cells with a high percentage of mitochondria were filtered out and library size normalization was then performed. All data was merged using R BiocSingular library (vl.8.1), renormalized to adjust for differences in depth using R batchelor library (vl.8.1), dimensionality reduced using UMAP method, and then subjected to Louvain clustering. Using R SingleR (v.1.6.1) and celldex (vl.2.0) libraries, the clusters were annotated and each cell individually using the Wilcoxon method with the fine pruned labels from the Immunological Genome Project (See Heng et al., Nat Immunol 9, 1091-1094 (2008)) which consisted of microarray profiles of pure mouse immune cells. To identify differentially expressed genes between treatments, FindMarker of R Seurat library (v4.0.2) was used. Heatmaps were drawn using the R Compl exHeatmap library (v2.0.0).
[00119] An average of 838 cells per sample were sequenced with a total of 10,054 cells after quality control. Overall, major immune subsets were represented within the groups with similar distributions across treatment categories (Figs. 5B-5C). The immune cells were then subtyped based on known gene expression signatures. In the CD47 treated group, a predominance of NK cells (NK.49CI+), NKT (NKT.4+), and DCs ((DC.8-, DC.11B-, DC.8+, DC.PCD.8-) was observed together with macrophages, Tregs, and eosinophils. In the B7-H3 treated group, there was abundance of T cells (T.CD8, T.CD4, T.4EFF, T.4.Pa, T.4SP, T.DN, T.4PLN, T4. FP3+25, and T. ISP) along with macrophages, B cells, DCs and eosinophils (Fig. 5D). These data suggested recruitment of specific immune cell subsets and utilization of different mechanisms during tumor cell killing upon CD47 and B7-H3 inhibition.
[00120] Differentially expressed genes between the treatment groups were assessed to evaluate gene expression specific signals during cytolytic activity (Fig. 5E). CTL- and NK-cell mediated cytotoxicity (IgG vs. B7-H3/CD47) such as GZMA, GZMB, NKG7, IL2R as well as heat shock genes HSPA1A and HSPA1B (B7-H3 vs. B7-H3/CD47, CD47 vs. B7-H3/CD47) were enriched. Enrichment of cytokine signaling CCL5 (RANTES, CCL5/CCR5) upon B7-H3 inhibition, and CXCR4 (CXCR4/CXCL12) upon CD47 inhibition — tboth of which are critical for hematopoietic cell trafficking — was observed. In the data, CCL5 was unique to B7-H3 inhibition, whereas CXCR4 was observed with CD47 treatment.
[00121] Leveraging scRNA-sequencing, data were dissected further and immune cells that
expressed these molecules were mapped out using UMAP as described herein (Figs. 6A-6T and Figs. 7A-7O) HSPA1A and HSPA1B genes not only expressed at high levels during tumor cytotoxicity (IgG vs. B7-H3/CD47, Figs. 6K-6T), but they expressed across most of the immune cell types ubiquitously, whereas GZMA and GZMB was mostly expressed in macrophages, NK cells, and T cells (Figs. 6A-6J). Expression details for CCL5, CXCR4, and NKG7 were also mapped out (Figs. 7A-7O).
[00122] In aggregate, these data showed that inhibition of B7-H3/CD47 in combination was effective in a well-characterized beta-catenin driven murine model. Both inhibitors utilized distinct immune cell populations during cytotoxicity. They employed known cytolytic mechanisms such as GZMA, GZMB, NKG7, HSPA1, and HSPAIB, and their expression patterns on immune cells was dissected herein.
Example 4. Therapeutic synergy was dependent on CCR5 and IL4R signaling [00123] Since TAM-associated inflammatory signals were consistently upregulated upon B7- H3 inhibition, the antitumor effects of co-targeting B7-H3 and CD47 were examined to determine if there was dependency on CCR5 or IL4R signaling. C57BL/6 CCR5-/- or IL4R-/- mice were used in the exemplary methods. Tumors were generated as described herein and the mice were treated with IgG control, B7-H3 inhibitor, and CD47 inhibitor alone or in combination according to methods described herein. As shown in Fig. 8A, the therapeutic synergy was robust but dissipated when these signaling axes were absent (Figs. 8B-8C). Without being bound by theory, the observed dissipation was likely due to a lack of tumor-associated macrophage and DC recruitment and antigen presentation. Similar results were obtained with the combination of PD-1 and CD47 inhibition (Figs. 8A-8C).
[00124] In order to determine whether B7-H3 conferred a suppressive microenvironment in human melanomas, the transcriptome data of the TCGA’s metastatic melanomas was next examined (n = 320). In brief) total RNA was isolated from human tumor tissues and subjected to RNA-sequencing. TCGA’s RNA-sequencing data (illuminahiseq_maseqv2-RSEM_genes, version 01/28/2016) and clinical phenotype (merge clinical, version, version 01/28/2016) were downloaded from firebrowse of Broad Institute. Analysis showed that B7-H3 correlated significantly with suppressive signals (M2 macrophage, myeloid derived suppressive cell (MDSC), T cell exclusion, and cancer associated fibroblast (CAF) signature), whereas it
significantly anti-correlated with PD-1, and moderately with PD-L1, and IL-15 (Figs. 8D and 8E). [00125] In aggregate, these data provided an additional layer of evidence that B7-H3 conferred immunosuppressive signals and when inhibited, modulated the microenvironment. The data suggested cooperative and non-redundant functions of B7-H3- and CD47-driven signaling and provided evidence that their blockade was efficacious therapeutically in preclinical murine models resistant to PD-1 inhibition.
[00126] In sum, the data provided in the examples herein showed that B7-H3 and CD47 drove distinct signaling pathways in shaping the tumor microenvironment and their combined blockade resulted in cooperative cytotoxicity. The data also delineated independent pathways affecting the tumor microenvironment, as combining distinct immune cell targeting mechanism (i.e. CD8+ T cell effector function, NK cell cytotoxicity, macrophage-mediated phagocytosis) was a viable therapeutic strategy.
NUMBERED EMBODIMENTS
[00127] Notwithstanding the appended claims, the following numbered embodiments are also contemplated herein and form part of the instant disclosure:
[00128] 1. A method for treating a solid tumor in a subject, the method comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition, the pharmaceutical composition comprising an inhibitor of B7-H3 (CD276) and an inhibitor of CD47-SIRPa, wherein the subject is not responsive to anti-programmed cell death 1/programmed cell death ligand 1 (PD1/PDL1) therapy.
[00129] 2. The method of embodiment 1, wherein the subject is a human patient having a solid tumor selected from the group consisting of pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, and sarcomas.
[00130] 3. The method of embodiment 2, wherein the subject is a human patient having melanoma.
[00131] 4. The method of any one of embodiments 1-3, wherein the effective amount of the pharmaceutical composition is sufficient to activate the innate immune system.
[00132] 5. The method of any one of embodiments 1-4, wherein the effective amount of the pharmaceutical composition is sufficient to activate at least one population of innate immune system cells.
[00133] 6. The method of embodiment 5, wherein the at least one population of innate immune system cells is selected from the group consisting of macrophages, dendritic cells (DCs) and natural killer (NK) cells.
[00134] 7. The method of any one of embodiments 1-6, wherein the effective amount of the pharmaceutical composition is sufficient to reduce tumor volume.
[00135] 8. The method of any one of embodiments 1-7, wherein the inhibitor of B7-H3 comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
[00136] 9. The method of embodiment 8, wherein the inhibitor of B7-H3 is an antibody.
[00137] 10. The method of embodiment 9, wherein the antibody is a full-length antibody or an antigen-binding fragment thereof.
[00138] 11. The method of embodiment 9, wherein the antibody is a human antibody or a humanized antibody.
[00139] 12. The method of embodiment 8, wherein the inhibitor of B7-H3 is an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
[00140] 13. The method of embodiment 8, wherein the inhibitor of B7-H3 comprises at least one of enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL-015, or a combination thereof.
[00141] 14. The method of any one of embodiments 1-7, wherein the inhibitor of B7-H3 comprises a B7-H3-specific chimeric antigen receptor T (CAR-T) cell.
[00142] 15. The method of any one of embodiments 1-7, wherein the inhibitor of CD47-
SIRPa is a CD47-ligand inhibitor, a SIRPa-receptor inhibitor, or a combination thereof.
[00143] 16. The method of embodiment 15, wherein the CD47-ligand inhibitor comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
[00144] 17. The method of embodiment 16, wherein the CD47-ligand inhibitor is an antibody.
[00145] 18. The method of embodiment 17, wherein the antibody is a full-length antibody or an antigen-binding fragment thereof.
[00146] 19. The method of embodiment 17, wherein the antibody is a human antibody or a humanized antibody.
[00147] 20. The method of embodiment 16, wherein the CD47-ligand inhibitor is an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
[00148] 21. The method of embodiment 15, wherein the CD47-ligand inhibitor is a CD47- specific chimeric antigen receptor T (CAR-T) cell.
[00149] 22. The method of embodiment 15, wherein the SIRPa-receptor inhibitor comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
[00150] 23. The method of embodiment 22, wherein the SIRPa-receptor inhibitor is an antibody.
[00151] 24. The method of embodiment 23, wherein the antibody is a full-length antibody or an antigen-binding fragment thereof.
[00152] 25. The method of embodiment 23, wherein the antibody is a human antibody or a humanized antibody.
[00153] 26. The method of embodiment 22, wherein the SIRPa-receptor inhibitor is an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
[00154] 27. The method of embodiment 15, wherein the inhibitor of CD47-SIRPa comprises at least one of RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC-90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or a combination thereof.
[00155] 28. A method of inflaming a cold tumor microenvironment, the method comprising administering to the cold tumor microenvironment a composition comprising an inhibitor of B7- H3 (CD276) and an inhibitor of CD47-SIRPa.
[00156] 29. The method of embodiment 28, wherein a tumor having the cold tumor microenvironment is a solid tumor.
[00157] 30. The method of embodiment 29, the solid tumor is selected from the group consisting of pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma,
breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, and sarcomas.
[00158] 31. The method of embodiment 30, wherein the solid tumor is melanoma.
[00159] 32. The method of any one of embodiments 29-31, wherein the solid tumor comprises at least one of B7-H3 overexpression, overactive P-catenin signaling, or a combination thereof.
[00160] 33. The method of any one of embodiments 29-33, wherein the solid tumor comprises at least one genetic mutation comprising BRAFV600E, loss of PTEN, or a combination thereof.
[00161] 34. The method of any one of embodiments 28-33, wherein inflaming a cold tumor microenvironment comprises increase of at least one interferon, increase of CD8+ T cells, increase of CD44 expression in CD4+ cells, in CD8+ cells, or both, increased TNF alpha expression in T cells, or a combination thereof.
[00162] 35. The method of any one of embodiments 28-34, wherein the inhibitor of B7-H3 comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
[00163] 36. The method of embodiment 35, wherein the inhibitor of B7-H3 is an antibody.
[00164] 37. The method of embodiment 36, wherein the antibody is a full-length antibody or an antigen-binding fragment thereof.
[00165] 38. The method of embodiment 36, wherein the antibody is a human antibody or a humanized antibody.
[00166] 39. The method of embodiment 35, wherein the inhibitor of B7-H3 is an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
[00167] 40. The method of embodiment 35, wherein the inhibitor of B7-H3 comprises at least one of enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL-015, or a combination thereof.
[00168] 41. The method of any one of embodiments 28-34, wherein the inhibitor of B7-H3 comprises a B7-H3-specific chimeric antigen receptor T (CAR-T) cell.
[00169] 42. The method of any one of embodiments 28-34, wherein the inhibitor of CD47-
SIRPa is a CD47-ligand inhibitor, a SIRPa-receptor inhibitor, or a combination thereof.
[00170] 43. The method of embodiment 42, wherein the CD47-ligand inhibitor comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
[00171] 44. The method of embodiment 43, wherein the CD47-ligand inhibitor is an antibody.
[00172] 45. The method of embodiment 44, wherein the antibody is a full-length antibody or an antigen-binding fragment thereof.
[00173] 46. The method of embodiment 44, wherein the antibody is a human antibody or a humanized antibody.
[00174] 47. The method of embodiment 43, wherein the CD47-ligand inhibitor is an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
[00175] 48. The method of embodiment 42, wherein the CD47-ligand inhibitor is a CD47- specific chimeric antigen receptor T (CAR-T) cell.
[00176] 49. The method of embodiment 42, wherein the SIRPa-receptor inhibitor comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
[00177] 50. The method of embodiment 49, wherein the SIRPa-receptor inhibitor is an antibody.
[00178] 51. The method of embodiment 50, wherein the antibody is a full-length antibody or an antigen-binding fragment thereof.
[00179] 52. The method of embodiment 50, wherein the antibody is a human antibody or a humanized antibody.
[00180] 53. The method of embodiment 49, wherein the SIRPa-receptor inhibitor is an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
[00181] 54. The method of embodiment 42, wherein the inhibitor of CD47-SIRPa comprises at least one of RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC-90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or a combination thereof.
[00182] 55. A pharmaceutical composition for treating a solid tumor in a subject, the pharmaceutical composition comprising an inhibitor of B7-H3 (CD276) and an inhibitor of CD47- SIRPa.
[00183] 56. The pharmaceutical composition of embodiment 55 further comprising at least one pharmaceutically acceptable excipient.
[00184] 57. The pharmaceutical composition of embodiment 55 or embodiment 56, wherein the inhibitor of B7-H3 (CD276) and the inhibitor of CD47-SIRPa.comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment or a combination thereof.
[00185] 58. The pharmaceutical composition of any one of embodiments 55-57, wherein the inhibitor of B7-H3 comprises at least one of enoblituzumab, omburtamab, orlotamab, BP- 102, DS- 7300, 8H-9, TCB-005, YBL-015, or a combination thereof.
[00186] 59. The pharmaceutical composition of any one of embodiments 55-58, wherein the inhibitor of CD47-SIRPa comprises at least one of RRx-001, a dinitroazetidine derivative, Hu5F9- G4, CC-90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or a combination thereof.
[00187] 60. The pharmaceutical composition of any one of embodiments 55-59, wherein the inhibitor of B7-H3 (CD276) and the inhibitor of CD47-SIRPa are administered in a dosage amount that restores CD8+ T cell effector function in a cold tumor microenvironment.
[00188] 61. A composition for treating a solid tumor in a subject, the composition comprising at least one inhibitor of B7-H3 (CD276) and at least one inhibitor of CD47-SIRPa
[00189] 62. The composition of embodiment 61 further comprising at least one pharmaceutically acceptable excipient.
[00190] 63. The composition of either embodiment 61 or embodiment 62, wherein the at least one inhibitor of B7-H3 comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment, or any combination thereof.
[00191] 64. The composition of embodiment 63, wherein the at least one inhibitor of B7-H3 comprises an antibody.
[00192] 65. The composition of embodiment 63, wherein the at least one inhibitor of B7-H3 comprises an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody. [00193] 66. The composition of embodiment 63, wherein the at least one inhibitor of B7-H3 comprises a B7-H3-specific chimeric antigen receptor T (CAR-T) cell.
[00194] 67. The composition of embodiment 63, wherein the at least one inhibitor of B7-H3 comprises enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL-015, or any combination thereof.
[00195] 68. The composition of either embodiment 61 or embodiment 62, wherein the at least one inhibitor of CD47-SIRPa comprises at least one CD47-ligand inhibitor, at least one SIRPa- receptor inhibitor, or any combination thereof.
[00196] 69. The composition of embodiment 68, wherein the at least one inhibitor of CD47-
SIRPa comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment, or any combination thereof.
[00197] 70. The composition of embodiment 68, wherein the at least one CD47-ligand inhibitor comprises an antibody.
[00198] 71. The composition of embodiment 68, wherein the at least one CD47-ligand inhibitor comprises an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
[00199] 72. The composition of embodiment 68, wherein the at least one CD47-ligand inhibitor comprises a CD47-specific chimeric antigen receptor T (CAR-T) cell.
[00200] 73. The composition of embodiment 68, wherein the at least one SIRPa-receptor inhibitor comprises an antibody.
[00201] 74. The composition of embodiment 68, wherein the at least one SIRPa-receptor inhibitor comprises an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
[00202] 75. The composition of embodiment 68, wherein the at least one inhibitor of CD47-
SIRPa comprises RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC-90002, TTI-621, ALX148, SRF231, SHR-1603, IBI188, ST-1901, SGNCD-47M, or any combination thereof.
[00203] 76. A method of preventing, treating, or ameliorating a soil tumor in a subject in need thereof, the method comprising administering an effective amount of a composition of any one of embodiments 61-75, wherein the subject in need thereof has or is suspected of having a solid tumor that is not responsive to at least one anti-programmed cell death 1/programmed cell death ligand 1 (PD1/PDL1) therapy.
[00204] 77. The method of embodiment 76, wherein the subject in need thereof is a human patient having or suspected of having a solid tumor selected from the group consisting of pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, and sarcomas.
[00205] 78. The method of embodiment 77, wherein the subject in need thereof is a human patient having or suspected of having melanoma.
[00206] 79. The method of any one of embodiments 76-78, wherein the solid tumor comprises at least one of B7-H3 overexpression, overactive P-catenin signaling, or any combination thereof. [00207] 80. The method of any one of embodiments 76-79, wherein the solid tumor comprises at least one genetic mutation comprising BRAFV600E, loss of PTEN, or a combination thereof.
[00208] 81. The method of any one of embodiments 76-80, wherein administering an effective amount of a composition of any one of embodiments 61-75 activates the innate immune system of the subject in need thereof.
[00209] 82. The method of any one of embodiments 76-81, wherein administering an effective amount of a composition of any one of embodiments 61-75 to the subject in need thereof inflames a cold tumor microenvironment, wherein inflaming a cold tumor microenvironment comprises an increase of at least one interferon, an increase of CD8+ T cells, an increase of CD44 expression in CD4+ cells, an increase of CD44 expression in CD8+ cells, an increase of TNF alpha expression in T cells, or any combination thereof.
[00210] 83. A kit comprising a pharmaceutical composition according to any one of embodiments 55-75 and at least one container.
[00211] 84. The kit of embodiment 83 further comprising one or more chemotherapeutics.
Claims
1. A composition for treating a solid tumor in a subject, the composition comprising at least one inhibitor of B7-H3 (CD276) and at least one inhibitor of CD47-SIRPa
2. The composition of claim 1 further comprising at least one pharmaceutically acceptable excipient.
3. The composition of either claim 1 or claim 2, wherein the at least one inhibitor of B7-H3 comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment, or any combination thereof.
4. The composition of claim 3, wherein the at least one inhibitor of B7-H3 comprises an antibody.
5. The composition of claim 3, wherein the at least one inhibitor of B7-H3 comprises an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
6. The composition of claim 3, wherein the at least one inhibitor of B7-H3 comprises a B7- H3 -specific chimeric antigen receptor T (CAR-T) cell.
7. The composition of claim 3, wherein the at least one inhibitor of B7-H3 comprises enoblituzumab, omburtamab, orlotamab, BP-102, DS-7300, 8H-9, TCB-005, YBL-015, or any combination thereof.
8. The composition of either claim 1 or claim 2, wherein the at least one inhibitor of CD47- SIRPa comprises at least one CD47-ligand inhibitor, at least one SIRPa-receptor inhibitor, or any combination thereof.
54
9. The composition of claim 8, wherein the at least one inhibitor of CD47-SIRPa comprises at least one of a small molecule, a peptide, a polynucleotide, a genetically modified cell or an antibody, an antibody fragment, or any combination thereof.
10. The composition of claim 8, wherein the at least one CD47-ligand inhibitor comprises an antibody.
11. The composition of claim 8, wherein the at least one CD47-ligand inhibitor comprises an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
12. The composition of claim 8, wherein the at least one CD47-ligand inhibitor comprises a CD47-specific chimeric antigen receptor T (CAR-T) cell.
13. The composition of claim 8, wherein the at least one SIRPa-receptor inhibitor comprises an antibody.
14. The composition of claim 8, wherein the at least one SIRPa-receptor inhibitor comprises an isolated nucleic acid or set of nucleic acids, which collectively encodes an antibody.
15. The composition of claim 8, wherein the at least one inhibitor of CD47-SIRPa comprises RRx-001, a dinitroazetidine derivative, Hu5F9-G4, CC-90002, TTI-621, ALX148, SRF231, SHR- 1603, IBI188, ST-1901, SGNCD-47M, or any combination thereof.
16. A method of preventing, treating, or ameliorating a soil tumor in a subject in need thereof, the method comprising administering an effective amount of a composition of any one of claims 1-15, wherein the subject in need thereof has or is suspected of having a solid tumor that is not responsive to at least one anti-programmed cell death 1/programmed cell death ligand 1 (PD1/PDL1) therapy.
55
17. The method of claim 16, wherein the subject in need thereof is a human patient having or suspected of having a solid tumor selected from the group consisting of pancreatic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, breast cancer, lung cancer, upper and lower gastrointestinal malignancies, squamous cell head and neck cancer, genitourinary cancer, ovarian cancer, and sarcomas.
18. The method of claim 17, wherein the subject in need thereof is a human patient having or suspected of having melanoma.
19. The method of any one of claims 16-18, wherein the solid tumor comprises at least one of B7-H3 overexpression, overactive P-catenin signaling, or any combination thereof.
20. The method of any one of claims 16-19, wherein the solid tumor comprises at least one genetic mutation comprising BRAFV600E, loss of PTEN, or a combination thereof.
21. The method of any one of claims 16-20, wherein administering an effective amount of a composition of any one of claims 1-15 activates the innate immune system of the subject in need thereof.
22. The method of any one of claims 16-21, wherein administering an effective amount of a composition of any one of claims 1-15 to the subject in need thereof inflames a cold tumor microenvironment, wherein inflaming a cold tumor microenvironment comprises an increase of at least one interferon, an increase of CD8+ T cells, an increase of CD44 expression in CD4+ cells, an increase of CD44 expression in CD8+ cells, an increase of TNFalpha expression in T cells, or any combination thereof.
56
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202063115699P | 2020-11-19 | 2020-11-19 | |
US63/115,699 | 2020-11-19 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022109227A1 true WO2022109227A1 (en) | 2022-05-27 |
Family
ID=81708101
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2021/060025 WO2022109227A1 (en) | 2020-11-19 | 2021-11-19 | Combined cancer therapy of b7-h3 and cd47 immune checkpoint inhbitior and methods of use |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2022109227A1 (en) |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20140242095A1 (en) * | 2011-10-19 | 2014-08-28 | University Health Network | Antibodies and antibody fragments targeting sirp-alpha and their use in treating hematologic cancers |
US20160257751A1 (en) * | 2015-03-04 | 2016-09-08 | Sorrento Therapeutics, Inc. | Antibody therapeutics that bind cd47 |
US20160264672A1 (en) * | 2010-03-04 | 2016-09-15 | Macrogenics, Inc. | Antibodies Reactive with B7-H3 and Uses Thereof |
US20200171028A1 (en) * | 2017-03-10 | 2020-06-04 | Viiamet Pharmaceuticals Holdings, LLC | Metalloenzyme inhibitor compounds |
WO2020128893A1 (en) * | 2018-12-21 | 2020-06-25 | Pfizer Inc. | Combination treatments of cancer comprising a tlr agonist |
-
2021
- 2021-11-19 WO PCT/US2021/060025 patent/WO2022109227A1/en active Application Filing
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20160264672A1 (en) * | 2010-03-04 | 2016-09-15 | Macrogenics, Inc. | Antibodies Reactive with B7-H3 and Uses Thereof |
US20140242095A1 (en) * | 2011-10-19 | 2014-08-28 | University Health Network | Antibodies and antibody fragments targeting sirp-alpha and their use in treating hematologic cancers |
US20160257751A1 (en) * | 2015-03-04 | 2016-09-08 | Sorrento Therapeutics, Inc. | Antibody therapeutics that bind cd47 |
US20200171028A1 (en) * | 2017-03-10 | 2020-06-04 | Viiamet Pharmaceuticals Holdings, LLC | Metalloenzyme inhibitor compounds |
WO2020128893A1 (en) * | 2018-12-21 | 2020-06-25 | Pfizer Inc. | Combination treatments of cancer comprising a tlr agonist |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Melamed et al. | Multiple sclerosis and cancer: the ying-yang effect of disease modifying therapies | |
US11705220B2 (en) | Systems and methods for identifying cancer treatments from normalized biomarker scores | |
AU2018233032B2 (en) | TEC family kinase inhibitor adjuvant therapy | |
US10584341B2 (en) | Antagonistic PDL1 aptamers and their applications in cancer therapy | |
US20230085710A1 (en) | Novel polyinosinic - polycytidylic acid compositions | |
US20220396839A1 (en) | Methods of detecting a fusion gene encoding a neoantigen | |
CN114502241A (en) | Combination cancer therapy with anti-galectin-9 antibodies and chemotherapeutic agents | |
WO2022109227A1 (en) | Combined cancer therapy of b7-h3 and cd47 immune checkpoint inhbitior and methods of use | |
US20240263240A1 (en) | Cd274 mutations for cancer treatment | |
KR102129195B1 (en) | Improved interferon treatment | |
US10533179B2 (en) | Antagonistic CTLA-4 aptamers and applications thereof in enhancing immune activity | |
TW202313107A (en) | Compositions and methods for treating lung cancer | |
CN116829953A (en) | Methods of determining resistance to checkpoint inhibitor therapy | |
US11994511B2 (en) | Biomarkers indicative of prostate cancer and treatment thereof | |
US20230366030A1 (en) | Methods of detecting an inactivating mutation of pbrm1 in meningioma | |
Arribas et al. | ERBB4-mediated signaling is a mediator of resistance to BTK and PI3K inhibitors in B cell lymphoid neoplasms | |
Murphy et al. | Overcome Prostate Cancer Resistance to Immune Checkpoint Therapy with Ketogenic Diet-Induced Epigenetic Reprogramming | |
WO2024007015A2 (en) | Ret gene fusions and uses thereof | |
WO2024168146A1 (en) | Braf gene fusions and uses thereof | |
WO2023178290A1 (en) | Use of combined cd274 copy number changes and tmb to predict response to immunotherapies | |
WO2023230444A2 (en) | Abl1 fusions and uses thereof | |
WO2023235822A1 (en) | Igf1r activation mutations and uses thereof | |
Varga et al. | Sapanisertib Inhibitor |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 21895645 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 21895645 Country of ref document: EP Kind code of ref document: A1 |