Try a new search

Format these results:

Searched for:

person:adeego01

in-biosketch:yes

Total Results:

25


Single-cell analysis of localized prostate cancer patients links high Gleason score with an immunosuppressive profile

Adorno Febles, Victor R; Hao, Yuan; Ahsan, Aarif; Wu, Jiansheng; Qian, Yingzhi; Zhong, Hua; Loeb, Stacy; Makarov, Danil V; Lepor, Herbert; Wysock, James; Taneja, Samir S; Huang, William C; Becker, Daniel J; Balar, Arjun V; Melamed, Jonathan; Deng, Fang-Ming; Ren, Qinghu; Kufe, Donald; Wong, Kwok-Kin; Adeegbe, Dennis O; Deng, Jiehui; Wise, David R
BACKGROUND:Evading immune surveillance is a hallmark for the development of multiple cancer types. Whether immune evasion contributes to the pathogenesis of high-grade prostate cancer (HGPCa) remains an area of active inquiry. METHODS:Through single-cell RNA sequencing and multicolor flow cytometry of freshly isolated prostatectomy specimens and matched peripheral blood, we aimed to characterize the tumor immune microenvironment (TME) of localized prostate cancer (PCa), including HGPCa and low-grade prostate cancer (LGPCa). RESULTS: TILs. The PCa TME was infiltrated by macrophages but these did not clearly cluster by M1 and M2 markers. CONCLUSIONS:T cell exhaustion in localized PCa, a finding enriched in HGPCa relative to LGPCa. These studies suggest a possible link between the clinical-pathologic risk of PCa and the associated TME. Our results have implications for our understanding of the immunologic mechanisms of PCa pathogenesis and the implementation of immunotherapy for localized PCa.
PMID: 36988342
ISSN: 1097-0045
CID: 5463282

RIP1 Kinase Drives Macrophage-Mediated Adaptive Immune Tolerance in Pancreatic Cancer

Wang, Wei; Marinis, Jill M; Beal, Allison M; Savadkar, Shivraj; Wu, Yue; Khan, Mohammed; Taunk, Pardeep S; Wu, Nan; Su, Wenyu; Wu, Jingjing; Ahsan, Aarif; Kurz, Emma; Chen, Ting; Yaboh, Inedouye; Li, Fei; Gutierrez, Johana; Diskin, Brian; Hundeyin, Mautin; Reilly, Michael; Lich, John D; Harris, Philip A; Mahajan, Mukesh K; Thorpe, James H; Nassau, Pamela; Mosley, Julie E; Leinwand, Joshua; Kochen Rossi, Juan A; Mishra, Ankita; Aykut, Berk; Glacken, Michael; Ochi, Atsuo; Verma, Narendra; Kim, Jacqueline I; Vasudevaraja, Varshini; Adeegbe, Dennis; Almonte, Christina; Bagdatlioglu, Ece; Cohen, Deirdre J; Wong, Kwok-Kin; Bertin, John; Miller, George
PMID: 33049209
ISSN: 1878-3686
CID: 4632692

Innate αβ T cells Mediate Antitumor Immunity by Orchestrating Immunogenic Macrophage Programming

Hundeyin, Mautin; Kurz, Emma; Mishra, Ankita; Kochen Rossi, Juan Andres; Liudahl, Shannon M; Leis, Kenna R; Mehrotra, Harshita; Kim, Mirhee; Torres, Luisana E; Ogunsakin, Adesola; Link, Jason; Sears, Rosalie C; Sivagnanam, Shamilene; Goecks, Jeremy; Islam, Km Sadeq; Dolgalev, Igor; Savadkar, Shivraj; Wang, Wei; Aykut, Berk; Leinwand, Joshua; Diskin, Brian; Adam, Salma; Israr, Muhammad; Gelas, Maeliss; Lish, Justin; Chin, Kathryn; Farooq, Mohammad Saad; Wadowski, Benjamin; Wu, Jingjing; Shah, Suhagi; Adeegbe, Dennis O; Pushalkar, Smruti; Vasudevaraja, Varshini; Saxena, Deepak; Wong, Kwok-Kin; Coussens, Lisa M; Miller, George
Unconventional T lymphocyte populations are emerging as important regulators of tumor immunity. Despite this, the role of TCRαβ+CD4-CD8-NK1.1- innate αβ T-cells (iαβTs) in pancreatic ductal adenocarcinoma (PDA) has not been explored. We found that iαβTs represent ~10% of T-lymphocytes infiltrating PDA in mice and humans. Intra-tumoral iαβTs express a distinct TCR-repertoire and profoundly immunogenic phenotype compared to their peripheral counterparts and conventional lymphocytes. iαβTs comprised ~75% of the total intra-tumoral IL-17+ cells. Moreover, iαβT cell adoptive transfer is protective in both murine models of PDA and human organotypic systems. We show iαβT cells induce a CCR5-dependent immunogenic macrophage reprogramming, thereby enabling marked CD4+ and CD8+ T cell expansion/activation and tumor protection. Collectively, iαβTs govern fundamental intra-tumoral crosstalk between innate and adaptive immune populations and are attractive therapeutic targets.
PMID: 31266770
ISSN: 2159-8290
CID: 3968062

Intron retention is a source of neoepitopes in cancer

Smart, Alicia C; Margolis, Claire A; Pimentel, Harold; He, Meng Xiao; Miao, Diana; Adeegbe, Dennis; Fugmann, Tim; Wong, Kwok-Kin; Van Allen, Eliezer M
We present an in silico approach to identifying neoepitopes derived from intron retention events in tumor transcriptomes. Using mass spectrometry immunopeptidome analysis, we show that retained intron neoepitopes are processed and presented on MHC I on the surface of cancer cell lines. RNA-derived neoepitopes should be considered for prospective personalized cancer vaccine development.
PMID: 30114007
ISSN: 1546-1696
CID: 3241392

RIP1 Kinase Drives Macrophage-Mediated Adaptive Immune Tolerance in Pancreatic Cancer

Wang, Wei; Marinis, Jill M; Beal, Allison M; Savadkar, Shivraj; Wu, Yue; Khan, Mohammed; Taunk, Pardeep S; Wu, Nan; Su, Wenyu; Wu, Jingjing; Ahsan, Aarif; Kurz, Emma; Chen, Ting; Yaboh, Inedouye; Li, Fei; Gutierrez, Johana; Diskin, Brian; Hundeyin, Mautin; Reilly, Michael; Lich, John D; Harris, Philip A; Mahajan, Mukesh K; Thorpe, James H; Nassau, Pamela; Mosley, Julie E; Leinwand, Joshua; Kochen Rossi, Juan A; Mishra, Ankita; Aykut, Berk; Glacken, Michael; Ochi, Atsuo; Verma, Narendra; Kim, Jacqueline I; Vasudevaraja, Varshini; Adeegbe, Dennis; Almonte, Christina; Bagdatlioglu, Ece; Cohen, Deirdre J; Wong, Kwok-Kin; Bertin, John; Miller, George
Pancreatic ductal adenocarcinoma (PDA) is characterized by immune tolerance and immunotherapeutic resistance. We discovered upregulation of receptor-interacting serine/threonine protein kinase 1 (RIP1) in tumor-associated macrophages (TAMs) in PDA. To study its role in oncogenic progression, we developed a selective small-molecule RIP1 inhibitor with high in vivo exposure. Targeting RIP1 reprogrammed TAMs toward an MHCIIhiTNFα+IFNγ+ immunogenic phenotype in a STAT1-dependent manner. RIP1 inhibition in TAMs resulted in cytotoxic T cell activation and T helper cell differentiation toward a mixed Th1/Th17 phenotype, leading to tumor immunity in mice and in organotypic models of human PDA. Targeting RIP1 synergized with PD1-and inducible co-stimulator-based immunotherapies. Tumor-promoting effects of RIP1 were independent of its co-association with RIP3. Collectively, our work describes RIP1 as a checkpoint kinase governing tumor immunity.
PMID: 30423296
ISSN: 1878-3686
CID: 3457042

Assessing Therapeutic Efficacy of MEK Inhibition in a KRAS G12C-Driven Mouse Model of Lung Cancer

Li, Shuai; Liu, Shengwu; Deng, Jiehui; Akbay, Esra A; Hai, Josephine; Ambrogio, Chiara; Zhang, Long; Zhou, Fangyu; Jenkins, Russell W; Adeegbe, Dennis O; Gao, Peng; Wang, Xiaoen; Paweletz, Cloud P; Herter-Sprie, Grit S; Chen, Ting; Gutierrez Quiceno, Laura; Zhang, Yanxi; Merlino, Ashley A; Quinn, Max M; Zeng, Yu; Yu, Xiaoting; Liu, Yuting; Fan, Lichao; Aguirre, Andrew J; Barbie, David A; Yi, Xianghua; Wong, Kwok-Kin
PURPOSE/OBJECTIVE:Despite the challenge to directly target mutant KRAS due to its high GTP affinity, some agents are under development against downstream signaling pathways, such as MEK inhibitors. However, it remains controversial whether MEK inhibitors can boost current chemotherapy in KRAS-mutant lung tumors in clinic. Considering the genomic heterogeneity among lung cancer patients, it is valuable to test potential therapeutics in KRAS-mutation driven mouse models. EXPERIMENTAL DESIGN/METHODS:We first compared the pERK1/2 level in lung cancer samples with different KRAS substitutions and generated a new genetically engineered mouse model whose tumor was driven by KRAS G12C, the most common KRAS mutation in lung cancer. Next, we evaluated the efficacy of selumetinib or its combination with chemotherapy, in KRAS G12C tumors compared to KRAS G12D tumors. Moreover, we generated KRAS G12C/p53 R270H model to explore the role of a dominant negative p53 mutation detected in patients in responsiveness to MEK inhibition. RESULTS:We determined higher pERK1/2 in KRAS G12C lung tumors compared to KRAS G12D. Using mouse models, we further identified that KRAS G12C tumors are significantly more sensitive to selumetinib compared with Kras G12D tumors. MEK inhibition significantly increased chemotherapeutic efficacy and progression-free survival of KRAS G12C mice. Interestingly, p53 co-mutation rendered KRAS G12C lung tumors less sensitive to combination treatment with selumetinib and chemotherapy. CONCLUSIONS:Our data demonstrate that unique KRAS mutations and concurrent mutations in tumor-suppressor genes are important factors for lung tumor responses to MEK inhibitor. Our preclinical study supports further clinical evaluation of combined MEK inhibition and chemotherapy for lung cancer patients harboring KRAS G12C and wildtype p53 status.
PMID: 29945997
ISSN: 1078-0432
CID: 3162862

BET bromodomain inhibition cooperates with PD-1 blockade to facilitate antitumor response in Kras-mutant non-small cell lung cancer

Adeegbe, Dennis O; Liu, Shengwu; Hattersley, Maureen; Bowden, Michaela; Zhou, Chensheng W; Li, Shuai; Vlahos, Raven; Grondine, Michael; Dolgalev, Igor; Ivanova, Elena; Quinn, Max M; Gao, Peng; Hammerman, Peter S; Bradner, James E; Diehl, J Alan; Rustgi, Anil K; Bass, Adam J; Tsirigos, Aristotelis; Freeman, Gordon J; Chen, Huawei; Wong, Kwok-Kin
KRAS mutation is present in approximately 30% of human lung adenocarcinomas. Although recent advances in targeted therapy have shown great promise, effective targeting of KRAS remains elusive, and concurrent alterations in tumor suppressors render KRAS-mutant tumors even more resistant to existing therapies. Contributing to the refractoriness of KRAS-mutant tumors are immunosuppressive mechanisms, such as increased presence of suppressive regulatory T cells (Tregs) in tumors and elevated expression of the inhibitory receptor PD-1 on tumor-infiltrating T cells. Treatment with BET bromodomain inhibitors is beneficial for hematologic malignancies, and they have Treg-disruptive effects in a non-small cell lung cancer (NSCLC) model. Targeting PD-1 inhibitory signals through PD-1 antibody blockade also has substantial therapeutic impact in lung cancer, although these outcomes are limited to a minority of patients. We hypothesized that the BET bromodomain inhibitor JQ1 would synergize with PD-1 blockade to promote a robust antitumor response in lung cancer. In the present study, using Kras+/LSL-G12D; Trp53L/L (KP) mouse models of NSCLC, we identified cooperative effects between JQ1 and PD-1 antibody. The numbers of tumor-infiltrating Tregs were reduced and activation of tumor-infiltrating T cells, which had a T-helper type 1 (Th1) cytokine profile, was enhanced, underlying their improved effector function. Furthermore, lung tumor-bearing mice treated with this combination showed robust and long-lasting antitumor responses compared to either agent alone, culminating in substantial improvement in the overall survival of treated mice. Thus, combining BET bromodomain inhibition with immune checkpoint blockade offers a promising therapeutic approach for solid malignancies such as lung adenocarcinoma.
PMID: 30087114
ISSN: 2326-6074
CID: 3226582

Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors

Miao, Diana; Margolis, Claire A; Vokes, Natalie I; Liu, David; Taylor-Weiner, Amaro; Wankowicz, Stephanie M; Adeegbe, Dennis; Keliher, Daniel; Schilling, Bastian; Tracy, Adam; Manos, Michael; Chau, Nicole G; Hanna, Glenn J; Polak, Paz; Rodig, Scott J; Signoretti, Sabina; Sholl, Lynette M; Engelman, Jeffrey A; Getz, Gad; Jänne, Pasi A; Haddad, Robert I; Choueiri, Toni K; Barbie, David A; Haq, Rizwan; Awad, Mark M; Schadendorf, Dirk; Hodi, F Stephen; Bellmunt, Joaquim; Wong, Kwok-Kin; Hammerman, Peter; Van Allen, Eliezer M
Tumor mutational burden correlates with response to immune checkpoint blockade in multiple solid tumors, although in microsatellite-stable tumors this association is of uncertain clinical utility. Here we uniformly analyzed whole-exome sequencing (WES) of 249 tumors and matched normal tissue from patients with clinically annotated outcomes to immune checkpoint therapy, including radiographic response, across multiple cancer types to examine additional tumor genomic features that contribute to selective response. Our analyses identified genomic correlates of response beyond mutational burden, including somatic events in individual driver genes, certain global mutational signatures, and specific HLA-restricted neoantigens. However, these features were often interrelated, highlighting the complexity of identifying genetic driver events that generate an immunoresponsive tumor environment. This study lays a path forward in analyzing large clinical cohorts in an integrated and multifaceted manner to enhance the ability to discover clinically meaningful predictive features of response to immune checkpoint blockade.
PMCID:6119118
PMID: 30150660
ISSN: 1546-1718
CID: 3257072

NK cells mediate synergistic antitumor effects of combined inhibition of HDAC6 and BET in a SCLC preclinical model

Liu, Yan; Li, Yuyang; Liu, Shengwu; Adeegbe, Dennis O; Christensen, Camilla L; Quinn, Max M; Dries, Ruben; Han, Shiwei; Buczkowski, Kevin; Wang, Xiaoen; Chen, Ting; Gao, Peng; Zhang, Hua; Li, Fei; Hammerman, Peter S; Bradner, James E; Quayle, Steven N; Wong, Kwok-Kin
Small cell lung cancer (SCLC) has the highest malignancy among all lung cancers, exhibiting aggressive growth and early metastasis to distant sites. For 30 years, treatment options for SCLC have been limited to chemotherapy, warranting the need for more effective treatments. Frequent inactivation of TP53 and RB1 as well as histone dysmodifications in SCLC suggest that transcriptional and epigenetic regulations play a major role in SCLC disease evolution. Here we performed a synthetic lethal screen using the BET inhibitor JQ1 and an shRNA library targeting 550 epigenetic genes in treatment-refractory SCLC xenograft models and identified HDAC6 as a synthetic lethal target in combination with JQ1. Combined treatment of human and mouse SCLC cell line-derived xenograft tumors with the HDAC6 inhibitor ricolinostat (ACY-1215) and JQ1 demonstrated significant inhibition of tumor growth; this effect was abolished upon depletion of NK cells, suggesting that these innate immune lymphoid cells play a role in SCLC tumor treatment response. Collectively, these findings suggest a potential new treatment for recurrent SCLC.
PMID: 29760044
ISSN: 1538-7445
CID: 3121342

Synergistic Immunostimulatory Effects and Therapeutic Benefit of Combined Histone Deacetylase and Bromodomain Inhibition in Non-small Cell Lung Cancer

Adeegbe, Dennis; Liu, Yan; Lizotte, Patrick H; Kamihara, Yusuke; Aref, Amir R; Almonte, Christina; Dries, Ruben; Li, Yuyang; Liu, Shengwu; Wang, Xiaoen; Warner-Hatten, Tiquella; Castrillon, Jessica; Yuan, Guo-Cheng; Poudel-Neupane, Neermala; Zhang, Haikuo; Guerriero, Jennifer L; Han, Shiwei; Awad, Mark M; Barbie, David A; Ritz, Jerome; Jones, Simon S; Hammerman, Peter S; Bradner, James E; Quayle, Steven N; Wong, Kwok-Kin
Effective therapies for non-small cell lung cancer (NSCLC) remain challenging despite an increasingly comprehensive understanding of somatically altered oncogenic pathways. It is now clear that therapeutic agents with potential to impact the tumor immune microenvironment potentiate immune-orchestrated therapeutic benefit. Herein we evaluated the immunoregulatory properties of histone deacetylase (HDAC) and bromodomain inhibitors, two classes of drugs that modulate the epigenome, with a focus on key cell subsets that are engaged in an immune response. By evaluating human peripheral blood and NSCLC tumors, we show that the selective HDAC6 inhibitor ricolinostat promotes phenotypic changes that support enhanced T cell activation and improved function of antigen presenting cells. The bromodomain inhibitor JQ1 attenuated CD4+Foxp3+ T regulatory cell suppressive function and synergized with ricolinostat to facilitate immune-mediated tumor growth arrest, leading to prolonged survival of mice with lung adenocarcinomas. Collectively, our findings highlight the immunomodulatory effects of two epigenetic modifiers that, together, promote T cell-mediated anti-tumor immunity and demonstrate their therapeutic potential for treatment of NSCLC.
PMCID:5540748
PMID: 28408401
ISSN: 2159-8290
CID: 2528362