Try a new search

Format these results:

Searched for:

person:milleg05

Total Results:

157


SHP2 inhibition diminishes KRASG12C cycling and promotes tumor microenvironment remodeling

Fedele, Carmine; Li, Shuai; Teng, Kai Wen; Foster, Connor J R; Peng, David; Ran, Hao; Mita, Paolo; Geer, Mitchell J; Hattori, Takamitsu; Koide, Akiko; Wang, Yubao; Tang, Kwan Ho; Leinwand, Joshua; Wang, Wei; Diskin, Brian; Deng, Jiehui; Chen, Ting; Dolgalev, Igor; Ozerdem, Ugur; Miller, George; Koide, Shohei; Wong, Kwok-Kin; Neel, Benjamin G
KRAS is the most frequently mutated human oncogene, and KRAS inhibition has been a longtime goal. Recently, inhibitors were developed that bind KRASG12C-GDP and react with Cys-12 (G12C-Is). Using new affinity reagents to monitor KRASG12C activation and inhibitor engagement, we found that an SHP2 inhibitor (SHP2-I) increases KRAS-GDP occupancy, enhancing G12C-I efficacy. The SHP2-I abrogated RTK feedback signaling and adaptive resistance to G12C-Is in vitro, in xenografts, and in syngeneic KRASG12C-mutant pancreatic ductal adenocarcinoma (PDAC) and non-small cell lung cancer (NSCLC). SHP2-I/G12C-I combination evoked favorable but tumor site-specific changes in the immune microenvironment, decreasing myeloid suppressor cells, increasing CD8+ T cells, and sensitizing tumors to PD-1 blockade. Experiments using cells expressing inhibitor-resistant SHP2 showed that SHP2 inhibition in PDAC cells is required for PDAC regression and remodeling of the immune microenvironment but revealed direct inhibitory effects on tumor angiogenesis and vascularity. Our results demonstrate that SHP2-I/G12C-I combinations confer a substantial survival benefit in PDAC and NSCLC and identify additional potential combination strategies.
PMID: 33045063
ISSN: 1540-9538
CID: 4632492

γδ T Cells Support Pancreatic Oncogenesis by Restraining αβ T Cell Activation

Daley, Donnele; Zambirinis, Constantinos Pantelis; Seifert, Lena; Akkad, Neha; Mohan, Navyatha; Werba, Gregor; Barilla, Rocky; Torres-Hernandez, Alejandro; Hundeyin, Mautin; Kumar Mani, Vishnu Raj; Avanzi, Antonina; Tippens, Daniel; Narayanan, Rajkishen; Jang, Jung-Eun; Newman, Elliot; Pillarisetty, Venu Gopal; Dustin, Michael Loran; Bar-Sagi, Dafna; Hajdu, Cristina; Miller, George
PMID: 33186522
ISSN: 1097-4172
CID: 4672052

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

Regulation and modulation of antitumor immunity in pancreatic cancer

Leinwand, Joshua; Miller, George
Pancreatic ductal adenocarcinoma carries a dismal prognosis, and outcomes have improved little with modern therapeutics. Checkpoint-based immunotherapy has failed to elicit responses in the vast majority of patients with pancreatic cancer. Alongside tumor cell-intrinsic mechanisms associated with oncogenic KRAS-induced inflammation, the tolerogenic myeloid cell infiltrate has emerged as a critical impediment to adaptive antitumor immune responses. Furthermore, the discovery of an intratumoral microbiome and the elucidation of host-microbe interactions that curtail antitumor immunity also present opportunities for intervention. Here we review the mechanisms of immunotherapy resistance in pancreatic ductal adenocarcinoma and discuss strategies to directly augment T cell responses in parallel with myeloid cell- and microbiome-targeted approaches that may enable immune-mediated control of this malignancy.
PMID: 32807942
ISSN: 1529-2916
CID: 4565542

Epigenetic CRISPR screens identify Npm1 as a therapeutic vulnerability in non-small cell lung cancer

Li, Fei; Ng, Wai-Lung; Luster, Troy A; Hu, Hai; Sviderskiy, Vladislav O; Dowling, Catríona M; Hollinshead, Kate E R; Zouitine, Paula; Zhang, Hua; Huang, Qingyuan; Ranieri, Michela; Wang, Wei; Fang, Zhaoyuan; Chen, Ting; Deng, Jiehui; Zhao, Kai; So, Hon-Cheong; Khodadadi-Jamayran, Alireza; Xu, Mousheng; Karatza, Angeliki; Pyon, Val; Li, Shuai; Pan, Yuanwang; Labbe, Kristen; Almonte, Christina; Poirier, John T; Miller, George; Possemato, Richard; Qi, Jun; Wong, Kwok-Kin
Despite advancements in treatment options, the overall cure and survival rates for non-small cell lung cancers (NSCLC) remain low. While small-molecule inhibitors of epigenetic regulators have recently emerged as promising cancer therapeutics, their application in patients with NSCLC is limited. To exploit epigenetic regulators as novel therapeutic targets in NSCLC, we performed pooled epigenome-wide CRISPR knockout screens in vitro and in vivo and identified the histone chaperone nucleophosmin 1 (NPM1) as a potential therapeutic target. Genetic ablation of Npm1 significantly attenuated tumor progression in vitro and in vivo. Furthermore, KRAS-mutant cancer cells were more addicted to NPM1 expression. Genetic ablation of Npm1 rewired the balance of metabolism in cancer cells from predominant aerobic glycolysis to oxidative phosphorylation and reduced the population of tumor-propagating cells. Overall, our results support NPM1 as a therapeutic vulnerability in NSCLC.
PMID: 32646968
ISSN: 1538-7445
CID: 4518022

Epigenetic silencing of the ubiquitin ligase subunit FBXL7 impairs c-SRC degradation and promotes epithelial-to-mesenchymal transition and metastasis

Moro, Loredana; Simoneschi, Daniele; Kurz, Emma; Arbini, Arnaldo A; Jang, Shaowen; Guaragnella, Nicoletta; Giannattasio, Sergio; Wang, Wei; Chen, Yu-An; Pires, Geoffrey; Dang, Andrew; Hernandez, Elizabeth; Kapur, Payal; Mishra, Ankita; Tsirigos, Aristotelis; Miller, George; Hsieh, Jer-Tsong; Pagano, Michele
Epigenetic plasticity is a pivotal factor that drives metastasis. Here, we show that the promoter of the gene that encodes the ubiquitin ligase subunit FBXL7 is hypermethylated in advanced prostate and pancreatic cancers, correlating with decreased FBXL7 mRNA and protein levels. Low FBXL7 mRNA levels are predictive of poor survival in patients with pancreatic and prostatic cancers. FBXL7 mediates the ubiquitylation and proteasomal degradation of active c-SRC after its phosphorylation at Ser 104. The DNA-demethylating agent decitabine recovers FBXL7 expression and limits epithelial-to-mesenchymal transition and cell invasion in a c-SRC-dependent manner. In vivo, FBXL7-depleted cancer cells form tumours with a high metastatic burden. Silencing of c-SRC or treatment with the c-SRC inhibitor dasatinib together with FBXL7 depletion prevents metastases. Furthermore, decitabine reduces metastases derived from prostate and pancreatic cancer cells in a FBXL7-dependent manner. Collectively, this research implicates FBXL7 as a metastasis-suppressor gene and suggests therapeutic strategies to counteract metastatic dissemination of pancreatic and prostatic cancer cells.
PMID: 32839549
ISSN: 1476-4679
CID: 4574162

Lung-derived HMGB1 is detrimental for vascular remodeling of metabolically imbalanced arterial macrophages

Boytard, Ludovic; Hadi, Tarik; Silvestro, Michele; Qu, Hengdong; Kumpfbeck, Andrew; Sleiman, Rayan; Fils, Kissinger Hyppolite; Alebrahim, Dornazsadat; Boccalatte, Francesco; Kugler, Matthias; Corsica, Annanina; Gelb, Bruce E; Jacobowitz, Glenn; Miller, George; Bellini, Chiara; Oakes, Jessica; Silvestre, Jean-Sébastien; Zangi, Lior; Ramkhelawon, Bhama
Pulmonary disease increases the risk of developing abdominal aortic aneurysms (AAA). However, the mechanism underlying the pathological dialogue between the lungs and aorta is undefined. Here, we find that inflicting acute lung injury (ALI) to mice doubles their incidence of AAA and accelerates macrophage-driven proteolytic damage of the aortic wall. ALI-induced HMGB1 leaks and is captured by arterial macrophages thereby altering their mitochondrial metabolism through RIPK3. RIPK3 promotes mitochondrial fission leading to elevated oxidative stress via DRP1. This triggers MMP12 to lyse arterial matrix, thereby stimulating AAA. Administration of recombinant HMGB1 to WT, but not Ripk3-/- mice, recapitulates ALI-induced proteolytic collapse of arterial architecture. Deletion of RIPK3 in myeloid cells, DRP1 or MMP12 suppression in ALI-inflicted mice repress arterial stress and brake MMP12 release by transmural macrophages thereby maintaining a strengthened arterial framework refractory to AAA. Our results establish an inter-organ circuitry that alerts arterial macrophages to regulate vascular remodeling.
PMID: 32855420
ISSN: 2041-1723
CID: 4575922

Targeting Piezo1 unleashes innate immunity against cancer and infectious disease

Aykut, Berk; Chen, Ruonan; Kim, Jacqueline I; Wu, Dongling; Shadaloey, Sorin A A; Abengozar, Raquel; Preiss, Pamela; Saxena, Anjana; Pushalkar, Smruti; Leinwand, Joshua; Diskin, Brian; Wang, Wei; Werba, Gregor; Berman, Matthew; Lee, Steve Ki Buom; Khodadadi-Jamayran, Alireza; Saxena, Deepak; Coetzee, William A; Miller, George
Piezo1 is a mechanosensitive ion channel that has gained recognition for its role in regulating diverse physiological processes. However, the influence of Piezo1 in inflammatory disease, including infection and tumor immunity, is not well studied. We postulated that Piezo1 links physical forces to immune regulation in myeloid cells. We found signal transduction via Piezo1 in myeloid cells and established this channel as the primary sensor of mechanical stress in these cells. Global inhibition of Piezo1 with a peptide inhibitor was protective against both cancer and septic shock and resulted in a diminution in suppressive myeloid cells. Moreover, deletion of Piezo1 in myeloid cells protected against cancer and increased survival in polymicrobial sepsis. Mechanistically, we show that mechanical stimulation promotes Piezo1-dependent myeloid cell expansion by suppressing the retinoblastoma gene Rb1 We further show that Piezo1-mediated silencing of Rb1 is regulated via up-regulation of histone deacetylase 2. Collectively, our work uncovers Piezo1 as a targetable immune checkpoint that drives immunosuppressive myelopoiesis in cancer and infectious disease.
PMID: 32826342
ISSN: 2470-9468
CID: 4567692

PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer

Diskin, Brian; Adam, Salma; Cassini, Marcelo F; Sanchez, Gustavo; Liria, Miguel; Aykut, Berk; Buttar, Chandan; Li, Eric; Sundberg, Belen; Salas, Ruben D; Chen, Ruonan; Wang, Junjie; Kim, Mirhee; Farooq, Mohammad Saad; Nguy, Susanna; Fedele, Carmine; Tang, Kwan Ho; Chen, Ting; Wang, Wei; Hundeyin, Mautin; Rossi, Juan A Kochen; Kurz, Emma; Haq, Muhammad Israr Ul; Karlen, Jason; Kruger, Emma; Sekendiz, Zennur; Wu, Dongling; Shadaloey, Sorin A A; Baptiste, Gillian; Werba, Gregor; Selvaraj, Shanmugapriya; Loomis, Cynthia; Wong, Kwok-Kin; Leinwand, Joshua; Miller, George
Programmed cell death protein 1 (PD-1) ligation delimits immunogenic responses in T cells. However, the consequences of programmed cell death 1 ligand 1 (PD-L1) ligation in T cells are uncertain. We found that T cell expression of PD-L1 in cancer was regulated by tumor antigen and sterile inflammatory cues. PD-L1+ T cells exerted tumor-promoting tolerance via three distinct mechanisms: (1) binding of PD-L1 induced STAT3-dependent 'back-signaling' in CD4+ T cells, which prevented activation, reduced TH1-polarization and directed TH17-differentiation. PD-L1 signaling also induced an anergic T-bet-IFN-γ- phenotype in CD8+ T cells and was equally suppressive compared to PD-1 signaling; (2) PD-L1+ T cells restrained effector T cells via the canonical PD-L1-PD-1 axis and were sufficient to accelerate tumorigenesis, even in the absence of endogenous PD-L1; (3) PD-L1+ T cells engaged PD-1+ macrophages, inducing an alternative M2-like program, which had crippling effects on adaptive antitumor immunity. Collectively, we demonstrate that PD-L1+ T cells have diverse tolerogenic effects on tumor immunity.
PMID: 32152508
ISSN: 1529-2916
CID: 4349682

Detection of pancreatic ductal adenocarcinoma with galectin-9 serum levels

Seifert, Adrian M; Reiche, Charlotte; Heiduk, Max; Tannert, Anna; Meinecke, Ann-Christin; Baier, Stephanie; von Renesse, Janusz; Kahlert, Christoph; Distler, Marius; Welsch, Thilo; Reissfelder, Christoph; Aust, Daniela E; Miller, George; Weitz, Jürgen; Seifert, Lena
Pancreatic ductal adenocarcinoma (PDAC) responds poorly to checkpoint blockade, such as anti-CTLA-4 and anti-PD-1. Galectin-9, a β-galactoside-binding lectin, promotes immune suppression through T-cell inhibition, and programming of tolerogenic macrophages. Of all cancers tested, PDAC showed the highest expression of LGALS9 (galectin-9) mRNA. We analyzed formalin-fixed and paraffin-embedded specimens from 83 patients with PDAC stained for galectin-9. Using flow cytometry, we determined galectin-9 expression on immune cells from tumor and matched blood samples from 12 patients with resectable PDAC. Furthermore, we analyzed galectin-9 serum levels by enzyme-linked immunosorbent assay using serum samples from 70 patients with PDAC, from 36 individuals with benign pancreatic disease, and from 28 healthy controls. Galectin-9 was highly expressed in human PDAC compared with normal pancreas and present on both tumor and immune cells. Tumor-infiltrating immune cells, especially CD3+ T cells, showed upregulation of galectin-9 compared with immune cells from matched blood. Blood γδ T cells from PDAC patients had higher galectin-9 expression than γδ T cells from healthy individuals. Galectin-9 polarized macrophages toward a protumoral M2 phenotype leading to suppressed T-cell cytokine secretion. Furthermore, serum concentration of galectin-9 was able to discriminate PDAC from benign pancreatic disease and healthy individuals, and was prognostic for stage IV patients. Galectin-9 is a new biomarker for the detection of PDAC.
PMID: 32055023
ISSN: 1476-5594
CID: 4304602