Try a new search

Format these results:

Searched for:

in-biosketch:true

person:milleg05

Total Results:

154


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

Rethinking T Cells in Pancreas Cancer [Comment]

Medina, Benjamin D; Miller, George
Patients with pancreatic ductal adenocarcinoma do not benefit from checkpoint blockade. However, human tumors harbor evidence of adaptive immunity in clonally expanded T-cell populations. Immune intact modeling of human tumors identifies stromal sequestration as a mechanism of immune escape. Targeting the stroma combined with checkpoint blockade unleashes antitumor immunity.See related article by Seo et al., p. 3934.
PMID: 31072844
ISSN: 1078-0432
CID: 4002022

Identification of a RIP1 Kinase Inhibitor Clinical Candidate (GSK3145095) for the Treatment of Pancreatic Cancer

Harris, Philip A; Marinis, Jill M; Lich, John D; Berger, Scott B; Chirala, Anirudh; Cox, Julie A; Eidam, Patrick M; Finger, Joshua N; Gough, Peter J; Jeong, Jae U; Kang, James; Kasparcova, Viera; Leister, Lara K; Mahajan, Mukesh K; Miller, George; Nagilla, Rakesh; Ouellette, Michael T; Reilly, Michael A; Rendina, Alan R; Rivera, Elizabeth J; Sun, Helen H; Thorpe, James H; Totoritis, Rachel D; Wang, Wei; Wu, Dongling; Zhang, Daohua; Bertin, John; Marquis, Robert W
RIP1 regulates cell death and inflammation and is believed to play an important role in contributing to a variety of human pathologies, including immune-mediated inflammatory diseases and cancer. While small-molecule inhibitors of RIP1 kinase have been advanced to the clinic for inflammatory diseases and CNS indications, RIP1 inhibitors for oncology indications have yet to be described. Herein we report on the discovery and profile of GSK3145095 (compound 6). Compound 6 potently binds to RIP1 with exquisite kinase specificity and has excellent activity in blocking RIP1 kinase-dependent cellular responses. Highlighting its potential as a novel cancer therapy, the inhibitor was also able to promote a tumor suppressive T cell phenotype in pancreatic adenocarcinoma organ cultures. Compound 6 is currently in phase 1 clinical studies for pancreatic adenocarcinoma and other selected solid tumors.
PMCID:6580371
PMID: 31223438
ISSN: 1948-5875
CID: 3939432

Virome and bacteriome: two sides of the same coin

Stern, Jonathan; Miller, George; Li, Xin; Saxena, Deepak
Although bacterial dysbiosis has been previously associated with carcinogenesis and HIV infection, the impact of the virome and these disease states has been less well studied. In this review, we will summarize what is known about the interplay between both the bacterial and the viral components of the microbiome on cancer and HIV pathogenesis. Bacterial dysbiosis has been associated with carcinogenesis such as colorectal cancer (CRC), hepatocellular carcinoma (HCC), lung cancer, breast cancer, and gastric cancer. The dysbiotic pathogenesis may be species-based or community-based and can have varying mechanisms of carcinogenesis. The human virome was also associated with certain cancers. Viruses, such as cytomegalovirus (CMV), Human herpesvirus 8 (HHV-8), human papilloma virus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV), and Epstein-Barr virus (EBV), all had associations with cancers. It was also reported that an altered bacteriophage community may lead to carcinogenesis by allowing opportunistic, oncogenic bacteria to proliferate in a gastrointestinal biofilm. This mechanism shows the importance of analyzing the bacteriome and the virome concurrently as their interactions can provide insight into new mechanisms in the pathogenesis of not only cancer, but other diseases as well. The enteric bacteriome was shown to be distinctly altered in immunocompromised HIV-infected individuals, and highly active antiretroviral therapy (HAART) was shown to at least partially reverse the alterations that HIV causes in the bacteriome. Studies have shown that the progression to HIV is associated with changes in the plasma concentration of commensal viruses. HIV also acts synergistically with multiple other viruses, such as HPV, EBV, varicella zoster virus (VZV), and HHV-8. Although it has been shown that HIV infection leads to enteric virome expansion in humans, most of the research on HIV's effect on the virome was conducted in non-human primates, and there is a lack of research on the effect of HAART on the virome. Virome-wide analysis is necessary for identifying novel viral etiologies. There is currently a wealth of information on the bacteriome and its associations with cancer and HIV, but more research should be conducted on the virome's associations and reaction to HAART as well as the bacteriome-virome interactions that may play a major role in pathogenesis and recovery.
PMID: 31177014
ISSN: 1879-6265
CID: 3929642

Targeting SYK signaling in myeloid cells protects against liver fibrosis and hepatocarcinogenesis

Torres-Hernandez, Alejandro; Wang, Wei; Nikiforov, Yuri; Tejada, Karla; Torres, Luisana; Kalabin, Aleksandr; Wu, Yue; Haq, Muhammad Israr Ul; Khan, Mohammed Y; Zhao, Zhen; Su, Wenyu; Camargo, Jimmy; Hundeyin, Mautin; Diskin, Brian; Adam, Salma; Rossi, Juan A Kochen; Kurz, Emma; Aykut, Berk; Shadaloey, Sorin A A; Leinwand, Joshua; Miller, George
Liver fibrosis and fibrosis-associated hepatocarcinogenesis are driven by chronic inflammation and are leading causes of morbidity and death worldwide. SYK signaling regulates critical processes in innate and adaptive immunity, as well as parenchymal cells. We discovered high SYK expression in the parenchymal hepatocyte, hepatic stellate cell (HSC), and the inflammatory compartments in the fibrotic liver. We postulated that targeting SYK would mitigate hepatic fibrosis and oncogenic progression. We found that inhibition of SYK with the selective small molecule inhibitors Piceatannol and PRT062607 markedly protected against toxin-induced hepatic fibrosis, associated hepatocellular injury and intra-hepatic inflammation, and hepatocarcinogenesis. SYK inhibition resulted in increased intra-tumoral expression of the p16 and p53 but decreased expression of Bcl-xL and SMAD4. Further, hepatic expression of genes regulating angiogenesis, apoptosis, cell cycle regulation, and cellular senescence were affected by targeting SYK. We found that SYK inhibition mitigated both HSC trans-differentiation and acquisition of an inflammatory phenotype in T cells, B cells, and myeloid cells. However, in vivo experiments employing selective targeted deletion of SYK indicated that only SYK deletion in the myeloid compartment was sufficient to confer protection against fibrogenic progression. Targeting SYK promoted myeloid cell differentiation into hepato-protective TNFαlow CD206hi phenotype downregulating mTOR, IL-8 signaling and oxidative phosphorylation. Collectively, these data suggest that SYK is an attractive target for experimental therapeutics in treating hepatic fibrosis and oncogenesis.
PMID: 30742098
ISSN: 1476-5594
CID: 3656052

The role of the microbiome in immunologic development and its implication for pancreatic cancer immunotherapy

Sethi, Vrishketan; Vitiello, Gerardo A; Saxena, Deepak; Miller, George; Dudeja, Vikas
Our understanding of the microbiome and its role in immunity, cancer initiation, and cancer progression has evolved significantly over the past century. The "germ theory of cancer" was first proposed in the early 20th century, and shortly thereafter the bacterium Helicobacter pylori and later, Fusobacterium nucleatum were implicated in the development of gastric and colorectal cancers respectively. However, with the development of reliable mouse models and affordable sequencing technologies, the most fascinating aspect of the microbiome-cancer relationship, where microbes undermine cancer immune surveillance and indirectly promote oncogenesis, has only recently been described. In this review, we highlight the essential role of the microbiome in immune system development and maturation. We review how microbe-induced immune activation promotes oncogenesis, focusing particularly on pancreatic carcinogenesis, and show that modulation of the microbiome augments the anti-cancer immune response and enables successful immunotherapy against pancreatic cancer.
PMID: 30768986
ISSN: 1528-0012
CID: 3655892

Specialized dendritic cells induce tumor-promoting IL-10+IL-17+ FoxP3neg regulatory CD4+ T cells in pancreatic carcinoma

Barilla, Rocky M; Diskin, Brian; Caso, Raul Caso; Lee, Ki Buom; Mohan, Navyatha; Buttar, Chandan; Adam, Salma; Sekendiz, Zennur; Wang, Junjie; Salas, Ruben D; Cassini, Marcelo F; Karlen, Jason; Sundberg, Belen; Akbar, Hashem; Levchenko, Dmitry; Gakhal, Inderdeep; Gutierrez, Johana; Wang, Wei; Hundeyin, Mautin; Torres-Hernandez, Alejandro; Leinwand, Joshua; Kurz, Emma; Rossi, Juan A Kochen; Mishra, Ankita; Liria, Miguel; Sanchez, Gustavo; Panta, Jyoti; Loke, P'ng; Aykut, Berk; Miller, George
The drivers and the specification of CD4+ T cell differentiation in the tumor microenvironment and their contributions to tumor immunity or tolerance are incompletely understood. Using models of pancreatic ductal adenocarcinoma (PDA), we show that a distinct subset of tumor-infiltrating dendritic cells (DC) promotes PDA growth by directing a unique TH-program. Specifically, CD11b+CD103- DC predominate in PDA, express high IL-23 and TGF-β, and induce FoxP3neg tumor-promoting IL-10+IL-17+IFNγ+ regulatory CD4+ T cells. The balance between this distinctive TH program and canonical FoxP3+ TREGS is unaffected by pattern recognition receptor ligation and is modulated by DC expression of retinoic acid. This TH-signature is mimicked in human PDA where it is associated with immune-tolerance and diminished patient survival. Our data suggest that CD11b+CD103- DC promote CD4+ T cell tolerance in PDA which may underscore its resistance to immunotherapy.
PMID: 30926808
ISSN: 2041-1723
CID: 3779022

RIP1 Kinase Promotes Macrophage Mediated Adaptive Immune Tolerance in Pancreatic Adenocarcinoma [Meeting Abstract]

Hundeyin, M.; Wang, W.; Miller, G.
ISI:000459144900363
ISSN: 1068-9265
CID: 3705472

STAT3 inhibition induces Bax-dependent apoptosis in liver tumor myeloid-derived suppressor cells

Guha, Prajna; Gardell, Jillian; Darpolor, Josephine; Cunetta, Marissa; Lima, Matthew; Miller, George; Espat, N Joseph; Junghans, Richard P; Katz, Steven C
Immunosuppressive myeloid-derived suppressor cells (MDSC) subvert antitumor immunity and limit the efficacy of chimeric antigen receptor T cells (CAR-T). Previously, we reported that the GM-CSF/JAK2/STAT3 axis drives liver-associated MDSC (L-MDSC) proliferation and blockade of this axis rescued antitumor immunity. We extended these findings in our murine liver metastasis (LM) model, by treating tumor-bearing mice with STAT3 inhibitors (STATTIC or BBI608) to further our understanding of how STAT3 drives L-MDSC suppressive function. STAT3 inhibition caused significant reduction of tumor burden as well as L-MDSC frequencies due to decrease in pSTAT3 levels. L-MDSC isolated from STATTIC or BBI608-treated mice had significantly reduced suppressive function. STAT3 inhibition of L-MDSC was associated with enhanced antitumor activity of CAR-T. Further investigation demonstrated activation of apoptotic signaling pathways in L-MDSC following STAT3 inhibition as evidenced by an upregulation of the pro-apoptotic proteins Bax, cleaved caspase-3, and downregulation of the anti-apoptotic protein Bcl-2. Accordingly, there was also a decrease of pro-survival markers, pErk and pAkt, and an increase in pro-death marker, Fas, with activation of downstream JNK and p38 MAPK. These findings represent a previously unrecognized link between STAT3 inhibition and Fas-induced apoptosis of MDSCs. Our findings suggest that inhibiting STAT3 has potential clinical application for enhancing the efficacy of CAR-T cells in LM through modulation of L-MDSC.
PMID: 30158673
ISSN: 1476-5594
CID: 3609752

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