16 results on '"Anand Kornepati"'
Search Results
2. 298 Tumor cell-intrinsic mTORC1 signaling through raptor makes melanoma and ovarian cancer immunotherapy resistant by regulating interferon-gamma responsiveness and promoting tumor-initiating cells
- Author
-
Tyler Curiel, Xinyue Zhang, Haiyan Bai, Harshita Gupta, Anand Kornepati, Suresh Kari, Emily Salinas, Erica Osta, Yidong Chen, and Ratna Vadlamudi
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
3. 242 Pharmacologic tumor PD-L1 depletion with chlorambucil treats ovarian cancer and melanomas in a tumor PD-L1-dependent manner and renders αPD-L1-resistant tumors αPD-L1-sensitive
- Author
-
Tyler Curiel, Matthew Hart, Yilun Deng, Ryan Reyes, Myrna Garcia, Haiyan Bai, Alvaro Padron, Harshita Gupta, Niannian Ji, Anand Kornepati, Clare Murray, Srikanth Polusani, and Suresh Kari
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
4. 900 Depleting non-canonical, cell-intrinsic PD-L1 signals induces synthetic lethality to small molecule DNA damage response inhibitors in an immune independent and dependent manner
- Author
-
Tyler Curiel, Lauren Byers, Yilun Deng, Alvaro Padron, Harshita Gupta, Anand Kornepati, Clare Murray, Ratna Vadlamudi, Barbara Avalos, Cody Rogers, Kavya Ramkumar, Zexuan Liu, Eloise Dray, Weixing Zhao, and Patrick Sung
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
5. 719 CD122-directed interleukin-2 complexes and αPD-L1 differentially require innate and adaptive immunity to treat local and metastatic bladder cancer
- Author
-
Tyler Curiel, Yilun Deng, Ryan Reyes, Aravind Kancharla, Myrna Garcia, Harshita Gupta, Deyi Zhang, Niannian Ji, Neelam Mukherjee, Karen Wheeler, Anand Kornepati, and Robert Svatek
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
6. 421 Specific cephalosporin antibiotics deplete tumor PD-L1 to inhibit DNA damage sensing and sensitize to Chk1 inhibitors in vivo
- Author
-
Clare Murray, Anand Kornepati, Alvaro Padron, Yilun Deng, Myrna Garcia, Haiyan Bai, and Tyler Curiel
- Subjects
Medicine - Abstract
OBJECTIVES/GOALS: Tumor PDL1 signals to immune cells for tumor immune evasion but has cell-intrinsic signals that promote tumor virulence. We identify novel tumor PDL1 depleting drugs (PDDs) to interrupt tumor-intrinsic PDL1 signals and sensitize tumors to targeted therapy in vitro and in vivo. METHODS/STUDY POPULATION: We screened the Prestwick and LOPAC libraries for FDA-approved drugs reducing B16 melanoma PDL1 > 2.6-fold. ?-lactam antibiotics were used at 80 ?M and Chk1 inhibitor rabusertib as indicated in T24 human bladder cancer, and murine ID8agg ovarian cancer, 4T1 breast cancer and B16. Genetic PDL1 KO was by CRISPR or shRNA and re-expression by lentivirus. Viability was by MTT and protein by immunoblot. We challenged 5 NSG mice/group with 2x106 T24 (SQ) cells and 5 BALB/c mice/group with 5x105 4T1 cells (mammary fat pad) and treated with cefepime (200 mg/kg), rabusertib (2.5 mg/kg), vehicle, or combo daily from day 3. RESULTS/ANTICIPATED RESULTS: Structurally-related ?-lactam antibiotics cefepime and ceftazidime are tumor PDDs. Cefepime or ceftazidime reduced tumor PD-L1 and thus its cell-intrinsic signals to deplete the DNA damage sensing Chk2 protein and promote rabusertib synthetic lethality in vitro and in vivo in a tumor PDL1-dependent manner independent of immunity. Structurally distinct ?-lactam antibiotics did not sensitize tumor cells to rabusertib, suggesting ?-lactam antimicrobial functions did not promote PDL1 depletion or rabusertib treatment effects in vivo. Although rabusertib effects were immune-independent, both PDDs induced immunogenic tumor STING signaling, suggesting they can improve tumor immunotherapy. DISCUSSION/SIGNIFICANCE: We show a rapidly translatable way to deplete detrimental tumor-intrinsic PDL1 signals, and sensitize tumors to rabusertib. We are testing PDD structure activity relationships to improve PDD effects and testing PDD effects on other treatments, e.g., PARP inhibitors, immunotherapy.
- Published
- 2022
- Full Text
- View/download PDF
7. 453 Pharmacologic tumor PDL1 depletion as a translational approach to inhibit tumor-intrinsic PDL1 signals and create novel treatment vulnerabilities
- Author
-
Clare Murray, Haiyan Bai, Anand Kornepati, Carlos Ontiveros, Yilun Deng, Alvaro Padron, Eva Galvan, Myrna Garcia, and Tyler Curiel
- Published
- 2022
- Full Text
- View/download PDF
8. 1428 Melanocyte-intrinsic PDL1 promotes tumorigenesis and progression in a novel autochthonous melanoma model
- Author
-
Carlos Ontiveros, Yilun Deng, Anand Kornepati, Clare Murray, Mary Jo Turk, Harshita Gupta, Alvaro Padron, and Tyler Curiel
- Published
- 2022
- Full Text
- View/download PDF
9. Bladder cancer cell‐intrinsic PD‐L1 signals promote mTOR and autophagy activation that can be inhibited to improve cytotoxic chemotherapy
- Author
-
Ryan Reyes, Robert S. Svatek, Anand Kornepati, Suresh Kari, Ratna K. Vadlamudi, Tyler J. Curiel, Erica Osta, Yilun Deng, Bogang Wu, Deyi Zhang, Alvaro Padron, Xiujie Sun, Rong Li, Harshita Gupta, and Aravind Kancharla
- Subjects
0301 basic medicine ,Cancer Research ,Gene Expression ,mTORC1 ,Mice, SCID ,chemotherapy ,Deoxycytidine ,B7-H1 Antigen ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,Medicine ,Neoplasm Metastasis ,Phosphorylation ,Melanoma ,Original Research ,Cancer Biology ,Ovarian Neoplasms ,Antibiotics, Antineoplastic ,biology ,Chloroquine ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Oncology ,030220 oncology & carcinogenesis ,mTOR ,bladder cancer ,Female ,autophagy ,Mechanistic Target of Rapamycin Complex 1 ,lcsh:RC254-282 ,03 medical and health sciences ,PD-L1 ,Cell Line, Tumor ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Sirolimus ,business.industry ,Cell growth ,Autophagy ,Cancer ,medicine.disease ,Gemcitabine ,Mice, Inbred C57BL ,030104 developmental biology ,Urinary Bladder Neoplasms ,Drug Resistance, Neoplasm ,PD‐L1 ,Cancer research ,biology.protein ,Cisplatin ,business ,Ovarian cancer ,Proto-Oncogene Proteins c-akt - Abstract
Tumor cell‐intrinsic programmed death‐ligand 1 (PD‐L1) signals mediate immunopathologic effects in breast, colon, and ovarian cancers and in melanomas, but bladder cancer (BC) effects are unreported. We show here that BC cell‐intrinsic PD‐L1 signals in mouse MB49 and human RT4, UM‐UC3, and UM‐UC‐14 BC cells regulate important pathologic pathways and processes, including effects not reported in other cancers. α‐PD‐L1 antibodies reduced BC cell proliferation in vitro, demonstrating direct signaling effects. BC cell‐intrinsic PD‐L1 promoted mammalian target of rapamycin complex 1 (mTORC1) signals in vitro and augmented in vivo immune‐independent cell growth and metastatic cancer spread, similar to effects we reported in melanoma and ovarian cancer. BC cell‐intrinsic PD‐L1 signals also promoted basal and stress‐induced autophagy, whereas these signals inhibited autophagy in melanoma and ovarian cancer cells. BC cell‐intrinsic PD‐L1 also mediated chemotherapy resistance to the commonly used BC chemotherapy agents cis‐platinum and gemcitabine and to the mTORC1 inhibitor, rapamycin. Thus, BC cell‐intrinsic PD‐L1 signals regulate important virulence and treatment resistance pathways that suggest novel, actionable treatment targets meriting additional studies. As a proof‐of‐concept, we showed that the autophagy inhibitor chloroquine improved cis‐platinum treatment efficacy in vivo, with greater efficacy in PD‐L1 null versus PD‐L1‐replete BC., We provide evidence that tumor‐intrinsic PD‐L1 mediates pathologic signals in human and mouse bladder cancers, independent of molecular subtype. Signals include control of proliferation, mTORC1, autophagy, and chemotherapy resistance. We show that countering the pathologic cell‐intrinsic PD‐L1 signals improves chemotherapy response in vivo of mice bearing bladder cancer tumors.
- Published
- 2021
10. Pharmacologic Tumor PDL1 Depletion with Cefepime or Ceftazidime Promotes DNA Damage and Sensitivity to DNA-Damaging Agents
- Author
-
Clare Murray, Eva Galvan, Carlos Ontiveros, Yilun Deng, Haiyan Bai, Alvaro Souto Padron, Kathryn Hinchee-Rodriguez, Myrna G. Garcia, Anand Kornepati, Jose Conejo-Garcia, and Tyler J. Curiel
- Subjects
PDL1 ,immunotherapy ,DNA damage ,drug repurposing ,β-lactam antibiotics ,Organic Chemistry ,General Medicine ,Ceftazidime ,Catalysis ,B7-H1 Antigen ,Computer Science Applications ,Inorganic Chemistry ,Mice ,Animals ,Physical and Theoretical Chemistry ,Cefepime ,Molecular Biology ,Melanoma ,Spectroscopy ,DNA Damage - Abstract
The interaction between tumor surface-expressed PDL1 and immune cell PD1 for the evasion of antitumor immunity is well established and is targeted by FDA-approved anti-PDL1 and anti-PD1 antibodies. Nonetheless, recent studies highlight the immunopathogenicity of tumor-intrinsic PDL1 signals that can contribute to the resistance to targeted small molecules, cytotoxic chemotherapy, and αPD1 immunotherapy. As genetic PDL1 depletion is not currently clinically tractable, we screened FDA-approved drugs to identify those that significantly deplete tumor PDL1. Among the candidates, we identified the β-lactam cephalosporin antibiotic cefepime as a tumor PDL1-depleting drug (PDD) that increases tumor DNA damage and sensitivity to DNA-damaging agents in vitro in distinct aggressive mouse and human cancer lines, including glioblastoma multiforme, ovarian cancer, bladder cancer, and melanoma. Cefepime reduced tumor PDL1 post-translationally through ubiquitination, improved DNA-damaging-agent treatment efficacy in vivo in immune-deficient and -proficient mice, activated immunogenic tumor STING signals, and phenocopied specific genetic PDL1 depletion effects. The β-lactam ring and its antibiotic properties did not appear contributory to PDL1 depletion or to these treatment effects, and the related cephalosporin ceftazidime produced similar effects. Our findings highlight the rapidly translated potential for PDDs to inhibit tumor-intrinsic PDL1 signals and improve DNA-damaging agents and immunotherapy efficacy.
- Published
- 2022
11. 298 Tumor cell-intrinsic mTORC1 signaling through raptor makes melanoma and ovarian cancer immunotherapy resistant by regulating interferon-gamma responsiveness and promoting tumor-initiating cells
- Author
-
Haiyan Bai, Yi Chen, Anand Kornepati, Suresh Kari, Juan Wang, Xinyue Zhang, Ratna K. Vadlamudi, Erica Osta, Emily Salinas, Tyler J. Curiel, and Harshita Gupta
- Subjects
Pharmacology ,Cancer Research ,business.industry ,medicine.medical_treatment ,Melanoma ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Tumor cells ,Immunotherapy ,medicine.disease ,Tumor Initiating Cells ,Oncology ,Mtorc1 signaling ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Interferon gamma ,Ovarian cancer ,business ,RC254-282 ,medicine.drug - Abstract
BackgroundAlthough immunotherapy can induce durable anti-tumor response in multiple cancers, immune checkpoint blockade (ICB) therapy resistance in ovarian cancer and melanoma remains problematic. Here, we report that tumor cell-intrinsic mTORC1 regulates ICB response through mTORC1 defining subunit Raptor (Rptor) by modulating interferon-gamma (IFNg) resistance and tumor-initiating cell (TIC) virulence.MethodsWe knocked down two distinct mTORC1 signaling components: Rptor (Rptorlo, aids in mTORC1 assembly) and Lamtor1 (Ltor1lo, docks mTORC1 on lysosomes) in murine ovarian cancer ID8agg and melanoma B16 cells. PD-L1 was CRISPR knocked out in B16 and human ovarian cancer line ES2. Mice with tumors were treated with a-PD-L1± a-CD8 antibody. TICs were estimated by flow-cytometry.1ResultsRptorlo B16 and ID8agg, but not Ltor1lo B16 tumors grew slower and were a-PD-L1 responsive unlike control (ctrl) in WT mice. We noted that ctrl and Rptorlo B16 and ID8agg cells expressed similar surface PD-L1 in vitro. Thus, Rptor suppresses a-PD-L1 response in ICB-resistant tumors. Tumor immune analysis revealed increased CD8+ T cell% and a trend to increased IFNg+CD8+ T cells in a-PD-L1 treated Rptorlo, but not ctrl B16. Rptorlo a-PD-L1 efficacy was lost with a-CD8 and in IFNg knockout mice. In vitro, IFNg suppressed Rptorlo ID8agg proliferation, unlike ctrl. These data suggested that lack of Rptor makes tumors ICB responsive, possibly by making tumors IFNg-sensitive and increasing IFNg+CD8+ T cells. Further, tumor and draining lymph node (DLN) TCF1+PD-1+ T cell stem cells (critical for aPD-L1/PD-1 success2 3) were significantly higher in a-PD-L1 treated Rptorlo tumors. Thus, tumor Rptor status could regulate tumor microenvironment and distal DLN immune landscape on a-PD-L1 treatment.We previously published that mTORC1 promotes PD-L1-dependent tumor proliferation, TIC virulence1 4 PD-L1KO B16 and ES2 cells expressed similar total Rptor protein. However, lower levels of Rptor were loaded in mTOR complex in absence of PD-L1, as assessed by a-mTOR immunoprecipitation, suggesting that pro-tumorigenic Rptor functions were downstream of, and dependent on PD-L1. Successful Rptorlo aPD-L1 treatment reduced TIC in vivo, an effect reversed in absence of CD8+ T cells or host IFNg. Inhibiting ID8agg mTORC1 with rapamycin reduced stemness genes oct4, nanog expression by QPCR. Further, ID8agg Rptorlo TIC formed significantly smaller tumors versus ctrl TIC in immune-compromised NSG mice, confirming their reduced virulence. Rptor, but not Ltor1, expression inversely correlated with tumor CD8+ infiltrate in IMvigor210 trial, and strongly with TIC gene signature in ovarian cancer patients.5 6ConclusionsTumor-cell intrinsic Rptor modulates ICB resistance, IFNg responsiveness, immune microenvironment, and TIC virulence.AcknowledgementsN/ATrial RegistrationN/AReferencesGupta HB, et al. Tumor cell-intrinsic PD-L1 promotes tumor-initiating cell generation and functions in melanoma and ovarian cancer. Signal Transduct Target Ther 2016;1,Article number:16030.Im SJ, et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016;537:417–421.Dammeijer F, et al. The PD-1/PD-L1-checkpoint restrains T cell immunity in tumor-draining lymph nodes. Cancer Cell 2020;38:685–700.Clark CA, et al. Tumor-intrinsic PD-L1 signals regulate cell growth, pathogenesis, and autophagy in ovarian cancer and melanoma. Cancer Res 2016;76:6964–6974.Smith BA, et al. A human adult stem cell signature marks aggressive variants across epithelial cancers. Cell Rep 2018;24:3353–3366.Hoffman-Censits JH, et al. IMvigor 210, a phase II trial of atezolizumab (MPDL3280A) in platinum-treated locally advanced or metastatic urothelial carcinoma (mUC). J Clin Oncol 2016;34, no.2_suppl:355–355.
- Published
- 2021
12. 719 CD122-directed interleukin-2 complexes and αPD-L1 differentially require innate and adaptive immunity to treat local and metastatic bladder cancer
- Author
-
Robert S. Svatek, Karen Wheeler, Myrna Garcia, Deyi Zhang, Anand Kornepati, Niannian Ji, Neelam Mukherjee, Tyler J. Curiel, Harshita Gupta, Aravind Kancharla, Ryan Reyes, and Yilun Deng
- Subjects
Bladder cancer ,business.industry ,T cell ,medicine.medical_treatment ,Immunotherapy ,medicine.disease ,Acquired immune system ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Immune checkpoint ,medicine.anatomical_structure ,Immune system ,Cancer immunotherapy ,medicine ,Cancer research ,business ,CD8 - Abstract
Background αPD-L1 bladder cancer (BC) immunotherapy is effective in Methods We used PD-L1+ mouse BC cell lines MB49 and MBT-2, for orthotopic, intravesical (i.e., in bladder) and intravenous challenge studies of local versus lung metastatic BC. Results αPD-L1 or IL-2c alone reduced tumor burden and extended survival in local MB49 and MBT-2. Using in vivo cell depletions, we found that γδ T cells and NK cells, but strikingly not CD8+ T cells, were necessary for IL-2c efficacy in bladder. We confirmed γδ T cell requirements for IL-2c, but not αPD-L1 efficacy in γδ T cell-null TCRδKO mice. TCRβKO conventional T cell-null mice exhibited IL-2c, but not αPD-L1 responsiveness for orthotopic BC treatment. Neither agent alone treated lung metastatic MB49 or MBT-2 but the drug combination improved survival in both tumor models. Combination treatment effects in lungs were distinct from bladder, requiring CD8+ T and NK cells, but not γδ T cells. Conclusions BC immunotherapy effects differ by anatomic compartment and use distinct mechanisms to treat primary and metastatic BC. CD122-directed IL-2 is a promising BC immunotherapy strategy, and IL-2c is a candidate mediator through innate immune effects. αPD-L1 could improve IL-2c efficacy by engagement of adaptive immune responses including to improve metastatic disease treatment efficacy. Ethics Approval All procedures involving animals in this study were approved by the UT Health San Antonio Institutional Animal Care and Use Committee (IACUC) and conducted in accordance with UT Health San Antonio Department of Laboratory Animal Resources standards. References Shah AY, Gao J, Siefker-Radtke AO. Five new therapies or just one new treatment? A critical look at immune checkpoint inhibition in urothelial cancer: Future Medicine, 2017. Arenas-Ramirez N, Zou C, Popp S, et al. Improved cancer immunotherapy by a CD25-mimobody conferring selectivity to human interleukin-2. Science translational medicine 2016;8(367):367ra166-367ra166.
- Published
- 2020
13. CD122-directed interleukin-2 treatment mechanisms in bladder cancer differ from αPD-L1 and include tissue-selective γδ T cell activation
- Author
-
Ryan Reyes, Chenghao Zhang, Karen Wheeler, Alvaro Padron, Neelam Mukherjee, Deyi Zhang, Harshita Gupta, Aravind Kancharla, Jose R. Conejo-Garcia, Anand Kornepati, Niannian Ji, Myrna Garcia, Robert S. Svatek, Tyler J. Curiel, Yilun Deng, Onur Boyman, University of Zurich, and Curiel, Tyler J
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Lung Neoplasms ,medicine.medical_treatment ,B7-H1 Antigen ,Mice ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Tumor Microenvironment ,preclinical ,Immunology and Allergy ,1306 Cancer Research ,Molecular Targeted Therapy ,Immune Checkpoint Inhibitors ,Intraepithelial Lymphocytes ,RC254-282 ,Clinical/Translational Cancer Immunotherapy ,Mice, Inbred C3H ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Acquired immune system ,Tumor Burden ,3004 Pharmacology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,2723 Immunology and Allergy ,Molecular Medicine ,2730 Oncology ,immunotherapy ,Signal Transduction ,medicine.drug ,Interleukin 2 ,T cell ,Immunology ,610 Medicine & health ,Mice, Transgenic ,03 medical and health sciences ,drug evaluation ,Cell Line, Tumor ,medicine ,Animals ,lymphocyte activation ,Pharmacology ,2403 Immunology ,Tumor microenvironment ,Bladder cancer ,business.industry ,Immunotherapy ,medicine.disease ,Interleukin-2 Receptor beta Subunit ,Mice, Inbred C57BL ,030104 developmental biology ,Urinary Bladder Neoplasms ,1313 Molecular Medicine ,10033 Clinic for Immunology ,Cancer research ,Interleukin-2 ,Ovarian cancer ,business ,CD8 - Abstract
BackgroundAnti-programmed death-ligand 1 (αPD-L1) immunotherapy is approved to treat bladder cancer (BC) but is effective in + antitumor T cells known to improve αPD-L1 efficacy. We hypothesized that the tumor microenvironment, including local immune cells in primary versus metastatic BC, differentially affects immunotherapy responses and that IL-2c effects could differ from, and thus complement αPD-L1.MethodsWe studied mechanisms of IL-2c and αPD-L1 efficacy using PD-L1+ mouse BC cell lines MB49 and MBT-2 in orthotopic (bladder) and metastatic (lung) sites.ResultsIL-2c reduced orthotopic tumor burden and extended survival in MB49 and MBT-2 BC models, similar to αPD-L1. Using antibody-mediated cell depletions and genetically T cell-deficient mice, we unexpectedly found that CD8+ T cells were not necessary for IL-2c efficacy against tumors in bladder, whereas γδ T cells, not reported to contribute to αPD-L1 efficacy, were indispensable for IL-2c efficacy there. αPD-L1 responsiveness in bladder required conventional T cells as expected, but not γδ T cells, altogether defining distinct mechanisms for IL-2c and αPD-L1 efficacy. γδ T cells did not improve IL-2c treatment of subcutaneously challenged BC or orthotopic (peritoneal) ovarian cancer, consistent with tissue-specific and/or tumor-specific γδ T cell contributions to IL-2c efficacy. IL-2c significantly altered bladder intratumoral γδ T cell content, activation status, and specific γδ T cell subsets with antitumor or protumor effector functions. Neither IL-2c nor αPD-L1 alone treated lung metastatic MB49 or MBT-2 BC, but their combination improved survival in both models. Combination treatment efficacy in lungs required CD8+ T cells but not γδ T cells.ConclusionsMechanistic insights into differential IL-2c and αPD-L1 treatment and tissue-dependent effects could help develop rational combination treatment strategies to improve treatment efficacy in distinct cancers. These studies also provide insights into γδ T cell contributions to immunotherapy in bladder and engagement of adaptive immunity by IL-2c plus αPD-L1 to treat refractory lung metastases.
- Published
- 2021
- Full Text
- View/download PDF
14. Abstract B10: High-dimensional (30-plex) imaging mass cytometry on tissue microarray identifies novel PD-L1-inclusive immunophenotypes associated with overall survival in stage III melanoma
- Author
-
Heather G. Hambright, Suhendan Ekmekcioglu, Dai Ogata, Anand Kornepati, Roland L. Bassett, Elizabeth A. Grimm, and Tyler J. Curiel
- Subjects
Cancer Research ,education.field_of_study ,Tumor microenvironment ,Tissue microarray ,Melanoma ,Population ,FOXP3 ,Biology ,medicine.disease ,Oncology ,Cancer research ,medicine ,Keratin 8 ,Immunohistochemistry ,Mass cytometry ,education - Abstract
We sought to determine the subcellular distribution of 30+ immune-related markers including PD-L1 in stage III melanoma tumor tissues (nuclear, cytoplasmic, surface) using novel imaging mass cytometry technology that enables simultaneous detection of over 30 immune and tumor markers. Metal-tagged antibodies were used to stain stage III melanoma tissues similarly to an IHC-based protocol, followed by laser ablation and mass cytometry using the Helios® and Hyperion® technology (Fluidigm). Markers for immune subsets included CD45, CD3, CD4, CD8, PD-1, Granzyme B, FoxP3, CD11b, CD11c, CD107, CD20, Vista, and CD68. Markers for tumor cell phenotyping included PD-L1 (intracellular and extracellular domain specific), E-cadherin, B-catenin, PD-L2, and keratin 8/18. Structural, inflammatory, survival, and signaling markers included Ki67, p53, p-tyrosine, Bcl-2, Cl-caspase 3, histone H3, collagen-1, actin, and arginase-1. We further validated subcellular distribution of PD-L1 in the same patient tumor cores using fluorochrome-labeled antibodies and 100X confocal microscopy to show difference in surface-only, cytoplasmic-only, and nuclear PD-L1. We then assessed the association between subcellular PD-L1 and immune populations in the tumor microenvironment, along with overall and recurrence-free survival. We wrote custom algorithms for data processing to identify novel immune subsets, ran SPADE and viSNE analyses for cross-validation, and employed Imaris Bitplane software to validate our own algorithms for immune population identification. Overall, we found that intracellular-only PD-L1 in tumor tissues has the highest correlation (Pearson r) to CD11b, FoxP3, PD-1, arginase, Ki67, PD-L2 and nuclear stain iridium, while extracellular (surface) PD-L1 in tumors has the highest correlation (Pearson r) to FoxP3, PD-1, arginase, Ki67, PD-L2 and Vista. We identified two novel immunophenotypes (histone-3+/p-tyrosinehi /Vistalo / PD-L1-ICDlo / PD-L1-ECDlo and CD11bhi / PD-L1-ICDhi / PD-L1-ECDhi) that are positively associated with overall patient survival. Of note, only phospho-Tyrhi was significantly associated with RFS. This work highlights the use of cutting-edge technology that can employ the highest multiplexing available for assessing novel interactions of 30+ tumor tissue markers for identification of novel immunophenotypes and biomarker in stage III melanoma patients. Our novel study showcases a novel methodologic approach to high-throughput analysis of retrospective profiling of the melanoma tumor immune microenvironment on a single IHC slide, followed by appropriate and rigorous cross-validation using Fluidigm’s novel CyTOF imaging technology. Our study provides a groundwork for this powerful, novel technology in translational and clinical research for biomarker identification and identification of novel combinatorial markers that may predict overall and recurrence-free survival in stage III melanoma. Citation Format: Heather G. Hambright, Anand V.R. Kornepati, Dai Ogata, Roland R.L. Bassett, Suhendan Ekmekcioglu, Elizabeth A. Grimm, Tyler J. Curiel. High-dimensional (30-plex) imaging mass cytometry on tissue microarray identifies novel PD-L1-inclusive immunophenotypes associated with overall survival in stage III melanoma [abstract]. In: Proceedings of the AACR Special Conference on Melanoma: From Biology to Target; 2019 Jan 15-18; Houston, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(19 Suppl):Abstract nr B10.
- Published
- 2020
- Full Text
- View/download PDF
15. CD122-selective IL-2 complexes target γδ T and NK cells to reduce tumor-promoting Th17 effects and synergize with αPD-L1 to treat primary and metastatic bladder cancer
- Author
-
Ryan Michael Reyes, Yilun Deng, Deyi Zhang, Neelam Mukherjee, Niannian Ji, Karen Wheeler, Harshita B. Gupta, Myrna Garcia, Anand Kornepati, Robert S Svatek, and Tyler J Curiel
- Subjects
Immunology ,Immunology and Allergy - Abstract
αPD-L1 bladder cancer (BC) immunotherapy is effective in
- Published
- 2020
- Full Text
- View/download PDF
16. Tumor cell-intrinsic programmed death protein 1 expression and induction in human cancer cell lines
- Author
-
Erica Osta, Harshita B. Gupta, Deyi Zhang, Anand Kornepati, Curtis A. Clark, and Tyler J. Curiel
- Subjects
Immunology ,Immunology and Allergy - Abstract
The programmed death protein 1 (PD-1) and ligand (PD-L1) axis inhibits T cell activation and reduces anti-tumor immunity. Most studies of the PD-1/PD-L1 axis test extrinsic, immune-dependent effects. However, tumor cell-intrinsic PD-L1 regulates immune-independent tumor mTOR signals, autophagy, and tumor initiating cell generation. Melanoma cell-intrinsic PD-1 regulates melanoma growth and mTOR. We tested PD-1 expression and induction in multiple human tumor cell lines. We detected PD-1 in human cell lines in vitro from ES2 ovarian cancer, VMM122 melanoma, and RT4 bladder cancer by flow cytometry, Western blot and RT-PCR. All lines were negative for the other PD-L1 receptor, CD80, by flow cytometry. Incubating lines with IFNα or IFNγ for 48 hours further augmented PD-1 expression as detected by flow, Western blot and RT-PCR. αPD-1 antibody (50 μg/ml) reduced proliferation of RT4 bladder cancer cells in vitro, demonstrating a direct PD-1 signal effect in these cells, and αPD-L1 similarly reduced in vitro proliferation. Interestingly, anti-PD-1 effects on reducing proliferation were similar: 41.5% reduction; p
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.