Try a new search

Format these results:

Searched for:

person:milleg05

Total Results:

157


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

Regulatory T Cells Keep Pancreatic Cancer at Bay [Comment]

Aykut, Berk; Chen, Ruonan; Miller, George
Although CD4+ FOXP3+ T regulatory (Treg) cells are well-known mediators of immunologic tolerance, their influences in the tumor microenviroment are incompletely understood. Writing in this issue of Cancer Discovery, Zhang and colleagues demonstrate that in pancreatic cancer, Treg cells promote the differentiation of tumor-restraining myofibroblastic cancer-associated fibroblasts, challenging the existing notion that Treg cells enable tumor progression.See related article by Zhang et al., p. 422.
PMID: 32127405
ISSN: 2159-8290
CID: 4340642

γδ T cells Promote Steatohepatitis by Orchestrating Innate and Adaptive Immune Programming

Torres-Hernandez, Alejandro; Wang, Wei; Nikiforov, Yuri; Tejada, Karla; Torres, Luisana; Kalabin, Aleksandr; Adam, Salma; Wu, Jingjing; Lu, Lu; Chen, Ruonan; Lemmer, Aaron; Camargo, Jimmy; Hundeyin, Mautin; Diskin, Brian; Aykut, Berk; Kurz, Emma; Kochen Rossi, Juan A; Khan, Mohammed; Liria, Miguel; Sanchez, Gustavo; Wu, Nan; Su, Wenyu; Adams, Steven; Israr Ul Haq, Muhammad; Saad Farooq, Mohammad; Vasudevaraja, Varshini; Leinwand, Joshua; Miller, George
The recruitment and activation of inflammatory cells in the liver delineates the transition from hepatic steatosis to steatohepatitis. We found that in steatohepatitis, γδT cells are recruited to the liver by CCR2, CCR5, and NOD2 signaling and are skewed towards an IL-17A+ phenotype in an ICOS-ICOSL dependent manner. γδT cells exhibit a distinct Vγ4+ , PD1+ , Ly6C+ CD44+ phenotype in steatohepatitis. Moreover, γδT cells upregulate both CD1d, which is necessary for lipid-based antigens presentation, and the free fatty acid receptor CD36. γδT cells are stimulated to express IL-17A by palmitic acid and CD1d ligation. Deletion, depletion, and targeted interruption of γδT cell recruitment protects against diet-induced steatohepatitis and accelerates disease resolution. We demonstrate that hepatic γδT cells exacerbate steatohepatitis, independent of IL-17 expression, by mitigating conventional CD4+ T cell expansion and modulating their inflammatory program via CD1d-dependent VEGF expression.
PMID: 31529720
ISSN: 1527-3350
CID: 4089142

In vivo epigenetic CRISPR screen identifies Asf1a as an immunotherapeutic target in Kras-mutant lung adenocarcinoma

Li, Fei; Huang, Qingyuan; Luster, Troy A; Hu, Hai; Zhang, Hua; Ng, Wai-Lung; Khodadadi-Jamayran, Alireza; Wang, Wei; Chen, Ting; Deng, Jiehui; Ranieri, Michela; Fang, Zhaoyuan; Pyon, Val; Dowling, Catriona M; Bagdatlioglu, Ece; Almonte, Christina; Labbe, Kristen; Silver, Heather; Rabin, Alexandra R; Jani, Kandarp; Tsirigos, Aristotelis; Papagiannakopoulos, Thales; Hammerman, Peter S; Velcheti, Vamsidhar; Freeman, Gordon J; Qi, Jun; Miller, George; Wong, Kwok-Kin
Despite substantial progress in lung cancer immunotherapy, the overall response rate in KRAS-mutant lung adenocarcinoma (ADC) patients remains low. Combining standard immunotherapy with adjuvant approaches that enhance adaptive immune responses-such as epigenetic modulation of anti-tumor immunity-is therefore an attractive strategy. To identify epigenetic regulators of tumor immunity, we constructed an epigenetic-focused sgRNA library, and performed an in vivo CRISPR screen in a KrasG12D/P53-/- (KP) lung ADC model. Our data showed that loss of the histone chaperone Asf1a in tumor cells sensitizes tumors to anti-PD-1 treatment. Mechanistic studies revealed that tumor cell-intrinsic Asf1a deficiency induced immunogenic macrophage differentiation in the tumor microenvironment by upregulating GM-CSF expression and potentiated T cell activation in combination with anti-PD-1. Our results provide rationale for a novel combination therapy consisting of ASF1A inhibition and anti-PD-1 immunotherapy.
PMID: 31744829
ISSN: 2159-8290
CID: 4208912

CDK7 Inhibition Potentiates Genome Instability Triggering Anti-tumor Immunity in Small Cell Lung Cancer

Zhang, Hua; Christensen, Camilla L; Dries, Ruben; Oser, Matthew G; Deng, Jiehui; Diskin, Brian; Li, Fei; Pan, Yuanwang; Zhang, Xuzhu; Yin, Yandong; Papadopoulos, Eleni; Pyon, Val; Thakurdin, Cassandra; Kwiatkowski, Nicholas; Jani, Kandarp; Rabin, Alexandra R; Castro, Dayanne M; Chen, Ting; Silver, Heather; Huang, Qingyuan; Bulatovic, Mirna; Dowling, Catríona M; Sundberg, Belen; Leggett, Alan; Ranieri, Michela; Han, Han; Li, Shuai; Yang, Annan; Labbe, Kristen E; Almonte, Christina; Sviderskiy, Vladislav O; Quinn, Max; Donaghue, Jack; Wang, Eric S; Zhang, Tinghu; He, Zhixiang; Velcheti, Vamsidhar; Hammerman, Peter S; Freeman, Gordon J; Bonneau, Richard; Kaelin, William G; Sutherland, Kate D; Kersbergen, Ariena; Aguirre, Andrew J; Yuan, Guo-Cheng; Rothenberg, Eli; Miller, George; Gray, Nathanael S; Wong, Kwok-Kin
Cyclin-dependent kinase 7 (CDK7) is a central regulator of the cell cycle and gene transcription. However, little is known about its impact on genomic instability and cancer immunity. Using a selective CDK7 inhibitor, YKL-5-124, we demonstrated that CDK7 inhibition predominately disrupts cell-cycle progression and induces DNA replication stress and genome instability in small cell lung cancer (SCLC) while simultaneously triggering immune-response signaling. These tumor-intrinsic events provoke a robust immune surveillance program elicited by T cells, which is further enhanced by the addition of immune-checkpoint blockade. Combining YKL-5-124 with anti-PD-1 offers significant survival benefit in multiple highly aggressive murine models of SCLC, providing a rationale for new combination regimens consisting of CDK7 inhibitors and immunotherapies.
PMID: 31883968
ISSN: 1878-3686
CID: 4251032

Targeting the interleukin-17 immune axis for cancer immunotherapy

Vitiello, Gerardo A; Miller, George
The role of IL-17 in cancer remains controversial. Emerging evidence suggests that during early oncogenesis IL-17 supports tumor growth, whereas in established tumors IL-17 production by γδ and Th17 cells potentiates antitumor immunity. Consequently, γδ and Th17 cells are attractive targets for immunotherapy in the IL-17 immune axis. To optimize IL-17-based immunotherapy, a deeper understanding of the cytokines dictating IL-17 production and the polarity of γδ and Th17 cells is critical. Here, we delve into the dichotomous roles of IL-17 in cancer and provide insight into the tumor microenvironment conducive for successful IL-17-based γδ and Th17 cell immunotherapy.
PMID: 31727783
ISSN: 1540-9538
CID: 4187022

Progress Toward Identifying Exact Proxies for Predicting Response to Immunotherapies

Filipovic, Aleksandra; Miller, George; Bolen, Joseph
Clinical value and utility of checkpoint inhibitors, a drug class targeting adaptive immune suppression pathways (PD-1, PDL-1, and CTLA-4), is growing rapidly and maintains status of a landmark achievement in oncology. Their efficacy has transformed life expectancy in multiple deadly cancer types (melanoma, lung cancer, renal/urothelial carcinoma, certain colorectal cancers, lymphomas, etc.). Despite significant clinical development efforts, therapeutic indication of approved checkpoint inhibitors are not as wide as the oncology community and patients would like them to be, potentially bringing into question their universal efficacy across tumor histologies. With the main goal of expanding immunotherapy applications, identifying of biomarkers to accurately predict therapeutic response and treatment related side-effects are a paramount need in the field. Specificities surrounding checkpoint inhibitors in clinic, such as unexpected tumor response patterns (pseudo- and hyper-progression), late responders, as well as specific immune mediated toxicities, complicate the management of patients. They stem from the complexities and dynamics of the tumor/host immune interactions, as well as baseline tumor biology. Search for clinically effective biomarkers therefore calls for a holistic approach, rather than implementation of a single analyte. The goal is to achieve dynamic and comprehensive acquisition, analyses and interpretation of immunological and biologic information about the tumor and the immune system, and to compute these parameters into an actionable, maximally predictive value at the individual patient level. Limitation delaying swift incorporation of validated immuno-oncology biomarkers span from standardized biospecimens acquisition and processing, selection of proficient biomarker discovery and validation methods, to establishing multidisciplinary consortiums and data sharing platforms. Multi-disciplinary efforts have already yielded some approved (PDL-1 and MSI-status) and other advanced tests (TMB, neoantigen pattern, and TIL infiltration rate). Importantly, clinical trial taskforces now recognize the imperative of the biomarker-driven trial design and execution, to enable translating biomarker discoveries into the clinical setting. This will ensure we utilize the "conspiracy" between the peripheral and intra-tumoral dynamic markers in shaping responses to checkpoint blockade, for the ultimate patient benefit.
PMCID:7092703
PMID: 32258034
ISSN: 2296-634x
CID: 4374572

Upregulation of ZIP14 and Altered Zinc Homeostasis in Muscles in Pancreatic Cancer Cachexia

Shakri, Ahmad Rushdi; Zhong, Timothy James; Ma, Wanchao; Coker, Courtney; Kim, Sean; Calluori, Stephanie; Scholze, Hanna; Szabolcs, Matthias; Caffrey, Thomas; Grandgenett, Paul M; Hollingsworth, Michael A; Tanji, Kurenai; Kluger, Michael D; Miller, George; Biswas, Anup Kumar; Acharyya, Swarnali
Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer type in which the mortality rate approaches the incidence rate. More than 85% of PDAC patients experience a profound loss of muscle mass and function, known as cachexia. PDAC patients with this condition suffer from decreased tolerance to anti-cancer therapies and often succumb to premature death due to respiratory and cardiac muscle wasting. Yet, there are no approved therapies available to alleviate cachexia. We previously found that upregulation of the metal ion transporter, Zip14, and altered zinc homeostasis are critical mediators of cachexia in metastatic colon, lung, and breast cancer models. Here, we show that a similar mechanism is likely driving the development of cachexia in PDAC. In two independent experimental metastasis models generated from the murine PDAC cell lines, Pan02 and FC1242, we observed aberrant Zip14 expression and increased zinc ion levels in cachectic muscles. Moreover, in advanced PDAC patients, high levels of ZIP14 in muscles correlated with the presence of cachexia. These studies underscore the importance of altered ZIP14 function in PDAC-associated cachexia development and highlight a potential therapeutic opportunity for improving the quality of life and prolonging survival in PDAC patients.
PMID: 31861290
ISSN: 2072-6694
CID: 4335202