79 results on '"Jason M Schenkel"'
Search Results
2. IL-33 Signaling Alters Regulatory T Cell Diversity in Support of Tumor Development
- Author
-
Amy Li, Rebecca H. Herbst, David Canner, Jason M. Schenkel, Olivia C. Smith, Jonathan Y. Kim, Michelle Hillman, Arjun Bhutkar, Michael S. Cuoco, C. Garrett Rappazzo, Patricia Rogers, Celeste Dang, Livnat Jerby-Arnon, Orit Rozenblatt-Rosen, Le Cong, Michael Birnbaum, Aviv Regev, and Tyler Jacks
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Regulatory T cells (Tregs) can impair anti-tumor immune responses and are associated with poor prognosis in multiple cancer types. Tregs in human tumors span diverse transcriptional states distinct from those of peripheral Tregs, but their contribution to tumor development remains unknown. Here, we use single-cell RNA sequencing (RNA-seq) to longitudinally profile dynamic shifts in the distribution of Tregs in a genetically engineered mouse model of lung adenocarcinoma. In this model, interferon-responsive Tregs are more prevalent early in tumor development, whereas a specialized effector phenotype characterized by enhanced expression of the interleukin-33 receptor ST2 is predominant in advanced disease. Treg-specific deletion of ST2 alters the evolution of effector Treg diversity, increases infiltration of CD8+ T cells into tumors, and decreases tumor burden. Our study shows that ST2 plays a critical role in Treg-mediated immunosuppression in cancer, highlighting potential paths for therapeutic intervention. : Li et al. show in a genetic mouse model of lung adenocarcinoma that during tumor development regulatory T cell (Treg) diversity shifts from an interferon-responsive to a ST2-positive, Klrg1+Areg+ effector-like phenotype. Treg-specific deletion of ST2 alters Treg heterogeneity, increases tumor infiltration by CD8+ T cells, and decreases tumor burden. Keywords: regulatory T cell, autochthonous mouse model of cancer, lung adenocarcinoma, tumor immunosuppression, single cell RNA sequencing, Treg heterogeneity, interleukin-33, ST2, Il1rl1
- Published
- 2019
- Full Text
- View/download PDF
3. Non-oncogenic Acute Viral Infections Disrupt Anti-cancer Responses and Lead to Accelerated Cancer-Specific Host Death
- Author
-
Frederick J. Kohlhapp, Erica J. Huelsmann, Andrew T. Lacek, Jason M. Schenkel, Jevgenijs Lusciks, Joseph R. Broucek, Josef W. Goldufsky, Tasha Hughes, Janet P. Zayas, Hubert Dolubizno, Ryan T. Sowell, Regina Kühner, Sarah Burd, John C. Kubasiak, Arman Nabatiyan, Sh’Rae Marshall, Praveen K. Bommareddy, Shengguo Li, Jenna H. Newman, Claude E. Monken, Sasha H. Shafikhani, Amanda L. Marzo, Jose A. Guevara-Patino, Ahmed Lasfar, Paul G. Thomas, Edmund C. Lattime, Howard L. Kaufman, and Andrew Zloza
- Subjects
cancer ,infection ,concomitant ,melanoma ,breast cancer ,influenza ,mouse ,PD-1 ,CD8+ T cells ,viral ,Biology (General) ,QH301-705.5 - Abstract
In light of increased cancer prevalence and cancer-specific deaths in patients with infections, we investigated whether infections alter anti-tumor immune responses. We report that acute influenza infection of the lung promotes distal melanoma growth in the dermis and leads to accelerated cancer-specific host death. Furthermore, we show that during influenza infection, anti-melanoma CD8+ T cells are shunted from the tumor to the infection site, where they express high levels of the inhibitory receptor programmed cell death protein 1 (PD-1). Immunotherapy to block PD-1 reverses this loss of anti-tumor CD8+ T cells from the tumor and decreases infection-induced tumor growth. Our findings show that acute non-oncogenic infection can promote cancer growth, raising concerns regarding acute viral illness sequelae. They also suggest an unexpected role for PD-1 blockade in cancer immunotherapy and provide insight into the immune response when faced with concomitant challenges.
- Published
- 2016
- Full Text
- View/download PDF
4. Low neoantigen expression and poor T-cell priming underlie early immune escape in colorectal cancer
- Author
-
Alex M. Jaeger, JJ Patten, Olivia Smith, Haley Hauck, Ömer H. Yilmaz, Tyler Jacks, George Eng, Peter M. K. Westcott, Jason M. Schenkel, Coralie Backlund, Ryan Elbashir, Darrell J. Irvine, Zackery A. Ely, Daniel Zhang, Nathan J. Sacks, and Mary Clare Beytagh
- Subjects
Cancer Research ,Colorectal cancer ,T-Lymphocytes ,T cell ,medicine.medical_treatment ,Mice ,Immune system ,Antigens, Neoplasm ,Animals ,Humans ,Medicine ,integumentary system ,business.industry ,Cancer ,Microsatellite instability ,Immunotherapy ,medicine.disease ,Transplantation ,medicine.anatomical_structure ,Oncology ,Immunoediting ,Cancer research ,Microsatellite Instability ,Colorectal Neoplasms ,business - Abstract
Immune evasion is a hallmark of cancer and therapies that restore immune surveillance have proven highly effective in cancers with high tumor mutation burden (TMB) (for example, those with microsatellite instability). Whether low TMB cancers, which are largely refractory to immunotherapy, harbor potentially immunogenic neoantigens remains unclear. Here, we show that tumors from all patients with microsatellite stable colorectal cancer express clonal predicted neoantigens despite low TMB. Unexpectedly, these neoantigens are broadly expressed at lower levels compared to those in colorectal cancer with microsatellite instability. Using a versatile platform for modulating neoantigen expression in colorectal cancer organoids and transplantation into the distal colon of mice, we show that low expression precludes productive cross-priming and drives immediate T-cell dysfunction. Notably, experimental or therapeutic rescue of priming rendered T cells capable of controlling tumors with low neoantigen expression. These findings underscore a critical role of neoantigen expression level in immune evasion and therapy response. Jacks and colleagues demonstrate the effects of neoantigen expression level on T-cell priming and immune surveillance during tumor development and progression and explore implications for immunotherapies, using in vivo models of colorectal cancer.
- Published
- 2021
- Full Text
- View/download PDF
5. Oncolytic Viruses—Natural and Genetically Engineered Cancer Immunotherapies
- Author
-
Sachin R. Jhawar, Aditya Thandoni, Praveen K. Bommareddy, Suemair Hassan, Frederick J. Kohlhapp, Sharad Goyal, Jason M. Schenkel, Ann W. Silk, and Andrew Zloza
- Subjects
oncolytic viruses ,cancer immunotherapy ,oncoimmunology ,pathogens ,viruses ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
There has long been interest in innovating an approach by which tumor cells can be selectively and specifically targeted and destroyed. The discovery of viruses that lyse tumor cells, termed oncolytic viruses (OVs), has led to a revolution in the treatment of cancer. The potential of OVs to improve the therapeutic ratio is derived from their ability to preferentially infect and replicate in cancer cells while avoiding destruction of normal cells surrounding the tumor. Two main mechanisms exist through which these viruses are reported to improve outcomes: direct lysis of tumor cells and indirect augmentation of host anti-tumor immunity. With these factors in mind, viruses are chosen or modified to selectively target tumor cells, decrease pathogenicity to normal cells, decrease the antiviral immune response (to prevent viral clearance), and increase the antitumor immune response. While only one OV has been approved for the treatment of cancer in the United States, and only two other OVs have been approved worldwide, a wide spectrum of OVs are in various stages of preclinical development and in clinical trials. These viruses are being studied as alternatives and adjuncts to more traditional cancer therapies including surgical resection, chemotherapy, radiation, hormonal therapies, targeted therapies, and other immunotherapies. Here, we review the natural characteristics and genetically engineered modifications that enhance the effectiveness of OVs for the treatment of cancer.
- Published
- 2017
- Full Text
- View/download PDF
6. Single-nucleus and spatial transcriptome profiling of pancreatic cancer identifies multicellular dynamics associated with neoadjuvant treatment
- Author
-
William L. Hwang, Karthik A. Jagadeesh, Jimmy A. Guo, Hannah I. Hoffman, Payman Yadollahpour, Jason W. Reeves, Rahul Mohan, Eugene Drokhlyansky, Nicholas Van Wittenberghe, Orr Ashenberg, Samouil L. Farhi, Denis Schapiro, Prajan Divakar, Eric Miller, Daniel R. Zollinger, George Eng, Jason M. Schenkel, Jennifer Su, Carina Shiau, Patrick Yu, William A. Freed-Pastor, Domenic Abbondanza, Arnav Mehta, Joshua Gould, Conner Lambden, Caroline B. M. Porter, Alexander Tsankov, Danielle Dionne, Julia Waldman, Michael S. Cuoco, Lan Nguyen, Toni Delorey, Devan Phillips, Jaimie L. Barth, Marina Kem, Clifton Rodrigues, Debora Ciprani, Jorge Roldan, Piotr Zelga, Vjola Jorgji, Jonathan H. Chen, Zackery Ely, Daniel Zhao, Kit Fuhrman, Robin Fropf, Joseph M. Beechem, Jay S. Loeffler, David P. Ryan, Colin D. Weekes, Cristina R. Ferrone, Motaz Qadan, Martin J. Aryee, Rakesh K. Jain, Donna S. Neuberg, Jennifer Y. Wo, Theodore S. Hong, Ramnik Xavier, Andrew J. Aguirre, Orit Rozenblatt-Rosen, Mari Mino-Kenudson, Carlos Fernandez-del Castillo, Andrew S. Liss, David T. Ting, Tyler Jacks, and Aviv Regev
- Subjects
Pancreatic Neoplasms ,Gene Expression Profiling ,Genetics ,Biomarkers, Tumor ,Humans ,Prognosis ,Transcriptome ,Article ,Neoadjuvant Therapy ,Carcinoma, Pancreatic Ductal - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal and treatment-refractory cancer. Molecular stratification in pancreatic cancer remains rudimentary and does not yet inform clinical management or therapeutic development. Here, we construct a high-resolution molecular landscape of the cellular subtypes and spatial communities that compose PDAC using single-nucleus RNA sequencing and whole-transcriptome digital spatial profiling (DSP) of 43 primary PDAC tumor specimens that either received neoadjuvant therapy or were treatment naive. We uncovered recurrent expression programs across malignant cells and fibroblasts, including a newly identified neural-like progenitor malignant cell program that was enriched after chemotherapy and radiotherapy and associated with poor prognosis in independent cohorts. Integrating spatial and cellular profiles revealed three multicellular communities with distinct contributions from malignant, fibroblast and immune subtypes: classical, squamoid-basaloid and treatment enriched. Our refined molecular and cellular taxonomy can provide a framework for stratification in clinical trials and serve as a roadmap for therapeutic targeting of specific cellular phenotypes and multicellular interactions.
- Published
- 2022
7. Deciphering the immunopeptidome in vivo reveals novel tumor antigens
- Author
-
Alex M. Jaeger, Lauren E. Stopfer, Ryuhjin Ahn, Emma A. Sanders, Demi A. Sandel, William A. Freed-Pastor, William M. Rideout, Santiago Naranjo, Tim Fessenden, Kim B. Nguyen, Peter S. Winter, Ryan E. Kohn, Peter M. K. Westcott, Jason M. Schenkel, Sean-Luc Shanahan, Alex K. Shalek, Stefani Spranger, Forest M. White, and Tyler Jacks
- Subjects
Proteomics ,Antigen Presentation ,Multidisciplinary ,Lung Neoplasms ,Histocompatibility Antigens Class I ,CD8-Positive T-Lymphocytes ,Article ,Pancreatic Neoplasms ,Mice ,Antigens, Neoplasm ,Alveolar Epithelial Cells ,Animals ,RNA, Messenger ,Peptides ,Carcinoma, Pancreatic Ductal - Abstract
Immunosurveillance of cancer requires the presentation of peptide antigens on major histocompatibility complex class I (MHC-I)(1,2,3,4,5). Current approaches to profile MHC-I associated peptides, collectively known as the “immunopeptidome”, are limited to in vitro investigation or bulk tumor lysates, limiting our understanding of cancer-specific patterns of antigen presentation in vivo(6). To overcome these limitations, we engineered an inducible affinity tag into the mouse MHC-I gene (H2-K1) and targeted this allele to the Kras(LSL-G12D/+); p53(fl/fl) (KP) mouse model (KP/K(b)Strep)(7). This approach allowed us to precisely isolate MHC-I peptides from autochthonous pancreatic ductal adenocarcinoma (PDAC) and lung adenocarcinoma (LUAD) in vivo. In addition, we profiled the LUAD immunopeptidome from the alveolar type 2 cell-of-origin through late-stage disease. Differential peptide presentation in LUAD was not predictable by mRNA expression or translation efficiency and is likely driven by post-translational mechanisms. Vaccination with peptides presented by LUAD in vivo provoked CD8(+) T cell responses in naïve and tumor-bearing mice. Many peptides unique to LUAD, including immunogenic peptides, exhibited minimal expression of the cognate mRNA, provoking reconsideration of antigen prediction pipelines that triage peptides according to transcript abundance(8). Beyond cancer, the K(b)Strep allele is compatible with other Cre-driver lines to explore antigen presentation in vivo in the pursuit of understanding basic immunology, infectious disease, and autoimmunity.
- Published
- 2022
8. Modeling diverse genetic subtypes of lung adenocarcinoma with a next-generation alveolar type 2 organoid platform
- Author
-
Santiago Naranjo, Christina M. Cabana, Lindsay M. LaFave, Rodrigo Romero, Sean-Luc Shanahan, Arjun Bhutkar, Peter M.K. Westcott, Jason M. Schenkel, Arkopravo Ghosh, Laura Z. Liao, Isabella Del Priore, Dian Yang, and Tyler Jacks
- Subjects
Organoids ,Proto-Oncogene Proteins B-raf ,Proto-Oncogene Proteins p21(ras) ,Mice ,Lung Neoplasms ,Genetics ,Animals ,Humans ,Receptor Protein-Tyrosine Kinases ,Adenocarcinoma of Lung ,Developmental Biology - Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Lung adenocarcinoma (LUAD), the most common histological subtype, accounts for 40% of all cases. While existing genetically engineered mouse models (GEMMs) recapitulate the histological progression and transcriptional evolution of human LUAD, they are time-consuming and technically demanding. In contrast, cell line transplant models are fast and flexible, but these models fail to capture the full spectrum of disease progression. Organoid technologies provide a means to create next-generation cancer models that integrate the most advantageous features of autochthonous and transplant-based systems. However, robust and faithful LUAD organoid platforms are currently lacking. Here, we describe optimized conditions to continuously expand murine alveolar type 2 (AT2) cells, a prominent cell of origin for LUAD, in organoid culture. These organoids display canonical features of AT2 cells, including marker gene expression, the presence of lamellar bodies, and an ability to differentiate into the AT1 lineage. We used this system to develop flexible and versatile immunocompetent organoid-based models of KRAS, BRAF, and ALK mutant LUAD. Notably, organoid-based tumors display extensive burden and complete penetrance and are histopathologically indistinguishable from their autochthonous counterparts. Altogether, this organoid platform is a powerful, versatile new model system to study LUAD.
- Published
- 2022
9. Therapeutic avenues for cancer neuroscience: translational frontiers and clinical opportunities
- Author
-
Diana D Shi, Jimmy A Guo, Hannah I Hoffman, Jennifer Su, Mari Mino-Kenudson, Jaimie L Barth, Jason M Schenkel, Jay S Loeffler, Helen A Shih, Theodore S Hong, Jennifer Y Wo, Andrew J Aguirre, Tyler Jacks, Lei Zheng, Patrick Y Wen, Timothy C Wang, and William L Hwang
- Subjects
Clinical Trials as Topic ,Oncology ,Drug Resistance, Neoplasm ,Neoplasms ,Neurosciences ,Tumor Microenvironment ,Humans ,Nervous System Physiological Phenomena ,Cancer Pain ,Immune Checkpoint Inhibitors ,Article - Abstract
With increasing attention on the essential roles of the tumour microenvironment in recent years, the nervous system has emerged as a novel and crucial facilitator of cancer growth. In this Review, we describe the foundational, translational, and clinical advances illustrating how nerves contribute to tumour proliferation, stress adaptation, immunomodulation, metastasis, electrical hyperactivity and seizures, and neuropathic pain. Collectively, this expanding knowledge base reveals multiple therapeutic avenues for cancer neuroscience that warrant further exploration in clinical studies. We discuss the available clinical data, including ongoing trials investigating novel agents targeting the tumour-nerve axis, and the therapeutic potential for repurposing existing neuroactive drugs as an anti-cancer approach, particularly in combination with established treatment regimens. Lastly, we discuss the clinical challenges of these treatment strategies and highlight unanswered questions and future directions in the burgeoning field of cancer neuroscience.
- Published
- 2022
10. Modeling diverse genetic subtypes of lung adenocarcinoma with a next-generation alveolar type 2 organoid platform
- Author
-
Santiago Naranjo, Christina M. Cabana, Lindsay M. LaFave, Peter M.K. Westcott, Rodrigo Romero, Arkopravo Ghosh, Laura Z. Liao, Jason M. Schenkel, Isabella Del Priore, Arjun Bhutkar, Dian Yang, and Tyler Jacks
- Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Lung adenocarcinoma (LUAD), the most common histological subtype, accounts for 40% of all cases. While genetically engineered mouse models (GEMMs) recapitulate the histological progression and transcriptional evolution of human LUAD, they are slow and technically demanding. In contrast, cell line transplant models are fast and flexible, but are often derived from clonal idiosyncratic tumors that fail to capture the full spectrum of clinical disease. Organoid technologies provide a means to create next-generation cancer models that integrate the most relevant features of autochthonous and transplant-based systems, yet robust and faithful LUAD organoid platforms are currently lacking. Here, we describe optimized conditions to continuously expand murine alveolar type 2 cells (AT2), a prominent cell-of-origin for LUAD, in organoid culture. These organoids display canonical features of AT2 cells, including marker gene expression, the presence of lamellar bodies, and an ability to differentiate into the AT1 lineage. We used this system to develop flexible and versatile immunocompetent organoid-based models of KRAS and ALK-mutant LUAD. Notably, the resultant tumors closely resemble their autochthonous murine counterparts and human LUAD. In contrast to comparable organoid platforms, our system supports long-term maintenance of the AT2 cellular identity, providing unprecedented ease and reliability to study AT2 and LUAD biology in vitro and in vivo.
- Published
- 2021
- Full Text
- View/download PDF
11. Antigen Dominance Hierarchies Shape TCF1+ Progenitor CD8 T Cell Phenotypes in Tumors
- Author
-
Jason M. Schenkel, Sara Z. Tavana, Michelle Hillman, Peter M. K. Westcott, Megan L. Burger, Sarah E. Blatt, Tyler Jacks, David Canner, Marta M. Holovatska, Michael Manos, Giorgio Gaglia, Izumi de los Rios Kobara, Aviv Regev, Alex M. Jaeger, Arjun Bhutkar, Sandro Santagata, William L. Hwang, F. Stephen Hodi, Cecily C. Ritch, Amanda M. Cruz, Tamina Kienka, Andrea Garmilla, Alexia L. Barandiaran, Amy Li, and Grace E. Crossland
- Subjects
Receptors, CCR6 ,Subdominant ,Lung Neoplasms ,Programmed Cell Death 1 Receptor ,Receptors, Antigen, T-Cell ,Adenocarcinoma of Lung ,Immunodominance ,Biology ,CD8-Positive T-Lymphocytes ,Major histocompatibility complex ,General Biochemistry, Genetics and Molecular Biology ,Article ,Epitopes ,Mice ,Antigen ,Antigens, Neoplasm ,Cytotoxic T cell ,Animals ,Humans ,CTLA-4 Antigen ,Amino Acid Sequence ,Hepatocyte Nuclear Factor 1-alpha ,RNA-Seq ,Immune Checkpoint Inhibitors ,Progenitor ,Stem Cells ,Vaccination ,Immune checkpoint ,Phenotype ,biology.protein ,Cancer research ,Female ,Single-Cell Analysis ,Peptides ,CD8 - Abstract
Summary CD8 T cell responses against different tumor neoantigens occur simultaneously, yet little is known about the interplay between responses and its impact on T cell function and tumor control. In mouse lung adenocarcinoma, we found that immunodominance is established in tumors, wherein CD8 T cell expansion is predominantly driven by the antigen that most stably binds MHC. T cells responding to subdominant antigens were enriched for a TCF1+ progenitor phenotype correlated with response to immune checkpoint blockade (ICB) therapy. However, the subdominant T cell response did not preferentially benefit from ICB due to a dysfunctional subset of TCF1+ cells marked by CCR6 and Tc17 differentiation. Analysis of human samples and sequencing datasets revealed that CCR6+ TCF1+ cells exist across human cancers and are not correlated with ICB response. Vaccination eliminated CCR6+ TCF1+ cells and dramatically improved the subdominant response, highlighting a strategy to optimally engage concurrent neoantigen responses against tumors.
- Published
- 2021
12. Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1(+) CD8(+) T cells in the tumor draining lymph nodes
- Author
-
William L. Hwang, Megan L. Burger, Orit Rozenblatt-Rosen, Le Cong, Michael S. Cuoco, Tyler Jacks, Jonathan Y. Kim, David Canner, George Eng, Amy Li, Sean-Luc Shanahan, Peter M. K. Westcott, Olivia Smith, Patricia Rogers, Rebecca H. Herbst, Kristen E. Pauken, Grace Gibbons, Aviv Regev, Jason M. Schenkel, Jin K. Park, Michelle Hillman, and William A. Freed-Pastor
- Subjects
Lung Neoplasms ,Immunology ,Adenocarcinoma of Lung ,Dendritic cell ,Dendritic Cells ,Biology ,CD8-Positive T-Lymphocytes ,medicine.disease ,Tumor antigen ,Immune checkpoint ,Article ,Mice ,Infectious Diseases ,Tumor progression ,T-Lymphocyte Subsets ,medicine ,Cancer research ,T Cell Transcription Factor 1 ,Immunology and Allergy ,Adenocarcinoma ,Cytotoxic T cell ,Animals ,Lymph ,Lymph Nodes ,CD8 - Abstract
In tumors, a subset of CD8(+) T cells expressing the transcription factor TCF-1 drives the response to immune checkpoint blockade. We examined the mechanisms that maintain these cells in an autochthonous model of lung adenocarcinoma. Longitudinal sampling and single-cell sequencing of tumor-antigen specific TCF-1(+) CD8(+) T cells revealed that whereas intratumoral TCF-1(+) CD8(+) T cells acquired dysfunctional features and decreased in number as tumors progressed, TCF-1(+) CD8(+) T cell frequency in the tumor draining LN (dLN) remained stable. Two discrete intratumoral TCF-1(+) CD8(+) T cell subsets developed over time – a proliferative SlamF6(+) subset and a non-cycling SlamF6- subset; blocking dLN egress decreased the frequency of intratumoral SlamF6+ TCF-1(+) CD8(+) T cells. Conventional type I dendritic cell (cDC1) in dLN decreased in number with tumor progression, and Flt3L+anti-CD40 treatment recovered SlamF6(+) T cell frequencies and decreased tumor burden. Thus, cDC1s in tumor dLN maintain a reservoir of TCF-1(+) CD8(+) T cells and their decrease contributes to failed anti-tumor immunity.
- Published
- 2021
13. Intratumoral injection of the seasonal flu shot converts immunologically cold tumors to hot and serves as an immunotherapy for cancer
- Author
-
Joseph R. Broucek, Eileena F. Giurini, Nora L. Herzog, Lisa K. Denzin, Gabriel Guevara-Aleman, C. Brent Chesson, Ricardo Estupinian, Marco Rossi, Mary J. Fidler, Raquel E. Redondo, Jai S. Rudra, Joyce Paras, Jochen Reiser, Amanda L. Marzo, Jyoti Malhotra, Shang Jui Wang, Emily S. Ruiz, Stuti Buddhadev, Howard L. Kaufman, Saeed Tarabichi, Sachin R. Jhawar, Frederick J. Kohlhapp, Daniel J. Medina, Mones M. Aboelatta, David Rotter, Shengguo Li, Leonard Y. Lee, Vineet Gupta, John Langenfeld, Ann W. Silk, Russell J. Pepe, Kajal Gupta, Timothy M. Kuzel, Jason M. Schenkel, Derek B. Sant'Angelo, Abdulkareem Abed, Jenna H. Newman, Michael J. Lee, Praveen K. Bommareddy, Salvatore M. Aspromonte, Ahmed Lasfar, Eric A. Singer, Michael P. Kane, Christina Nowicki, Arnold B. Rabson, Andrew Zloza, Paul G. Thomas, and Josef W. Goldufsky
- Subjects
Lung Neoplasms ,Skin Neoplasms ,medicine.medical_treatment ,CD8-Positive T-Lymphocytes ,Injections, Intralesional ,Mice ,Influenza A Virus, H1N1 Subtype ,Immunology and Inflammation ,0302 clinical medicine ,Cancer immunotherapy ,Neoplasms ,vaccine ,Tumor Microenvironment ,Medicine ,Lung ,Skin ,B-Lymphocytes ,Immunity, Cellular ,0303 health sciences ,Multidisciplinary ,Vaccination ,Biological Sciences ,Interleukin-10 ,3. Good health ,Basic-Leucine Zipper Transcription Factors ,PNAS Plus ,Influenza Vaccines ,Immunotherapy ,Seasons ,influenza ,flu shot ,Adjuvant ,Squalene ,Virus ,03 medical and health sciences ,Adjuvants, Immunologic ,Influenza, Human ,Animals ,Humans ,cancer ,030304 developmental biology ,Tumor microenvironment ,business.industry ,Cancer ,medicine.disease ,Blockade ,Mice, Inbred C57BL ,Repressor Proteins ,intratumoral ,Cancer research ,business ,030215 immunology - Abstract
Significance Immunotherapy has revolutionized cancer treatment, yielding unprecedented long-term responses and survival. However, a significant proportion of patients remain refractory, which correlates with the absence of immune-infiltrated (“hot”) tumors. Here, we observed that FDA-approved unadjuvanted seasonal influenza vaccines administered via intratumoral injection not only provide protection against active influenza virus lung infection, but also reduce tumor growth by increasing antitumor CD8+ T cells and decreasing regulatory B cells within the tumor. Ultimately, intratumoral unadjuvanted seasonal influenza vaccine converts immunologically inactive “cold” tumors to “hot,” generates systemic responses, and sensitizes resistant tumors to checkpoint blockade. Repurposing the “flu shot” may increase response rates to immunotherapy, and based on its current FDA approval and safety profile, may be quickly translated for clinical care., Reprogramming the tumor microenvironment to increase immune-mediated responses is currently of intense interest. Patients with immune-infiltrated “hot” tumors demonstrate higher treatment response rates and improved survival. However, only the minority of tumors are hot, and a limited proportion of patients benefit from immunotherapies. Innovative approaches that make tumors hot can have immediate impact particularly if they repurpose drugs with additional cancer-unrelated benefits. The seasonal influenza vaccine is recommended for all persons over 6 mo without prohibitive contraindications, including most cancer patients. Here, we report that unadjuvanted seasonal influenza vaccination via intratumoral, but not intramuscular, injection converts “cold” tumors to hot, generates systemic CD8+ T cell-mediated antitumor immunity, and sensitizes resistant tumors to checkpoint blockade. Importantly, intratumoral vaccination also provides protection against subsequent active influenza virus lung infection. Surprisingly, a squalene-based adjuvanted vaccine maintains intratumoral regulatory B cells and fails to improve antitumor responses, even while protecting against active influenza virus lung infection. Adjuvant removal, B cell depletion, or IL-10 blockade recovers its antitumor effectiveness. Our findings propose that antipathogen vaccines may be utilized for both infection prevention and repurposing as a cancer immunotherapy.
- Published
- 2019
- Full Text
- View/download PDF
14. CRISPR-mediated modeling and functional validation of candidate tumor suppressor genes in small cell lung cancer
- Author
-
Kim L. Mercer, Jason M. Schenkel, Roderick T. Bronson, Elliot H. Akama-Garren, Tyler Jacks, Arjun Bhutkar, Sheng Rong Ng, and William M. Rideout
- Subjects
Candidate gene ,Lung Neoplasms ,Apoptosis ,Retinoblastoma-Like Protein p107 ,Disease ,law.invention ,Mice ,0302 clinical medicine ,Loss of Function Mutation ,law ,CRISPR ,Genes, Tumor Suppressor ,Lung ,Gene Editing ,0303 health sciences ,Multidisciplinary ,Biological Sciences ,humanities ,Tumor Burden ,3. Good health ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Disease Progression ,Mice, Transgenic ,Biology ,Cell Line ,03 medical and health sciences ,p107 ,Genetics ,medicine ,Animals ,Humans ,Lung cancer ,neoplasms ,Gene ,Cell Proliferation ,Neoplasm Staging ,030304 developmental biology ,Retinoblastoma-Like Protein p130 ,medicine.disease ,Small Cell Lung Carcinoma ,respiratory tract diseases ,Disease Models, Animal ,GEMM ,Tumor progression ,Cancer research ,Feasibility Studies ,Suppressor ,small cell lung cancer ,CRISPR-Cas Systems ,Tumor Suppressor Protein p53 - Abstract
Significance SCLC is a deadly disease for which treatment outcomes have not improved significantly for over 30 y due to the lack of effective new therapies. Large-scale sequencing studies have identified many recurrently mutated genes in human SCLC tumors, whose functions remain poorly understood. We have adapted the CRISPR-Cas9 system to rapidly model mutations in target genes in a mouse model of SCLC. Using this system, we show that the gene p107 functions as a tumor suppressor gene in SCLC. Furthermore, loss of p107 confers a distinct tumor phenotype from loss of its close relative p130. Our results demonstrate the utility of our system for better understanding the genetic factors that contribute to SCLC progression., Small cell lung cancer (SCLC) is a highly aggressive subtype of lung cancer that remains among the most lethal of solid tumor malignancies. Recent genomic sequencing studies have identified many recurrently mutated genes in human SCLC tumors. However, the functional roles of most of these genes remain to be validated. Here, we have adapted the CRISPR-Cas9 system to a well-established murine model of SCLC to rapidly model loss-of-function mutations in candidate genes identified from SCLC sequencing studies. We show that loss of the gene p107 significantly accelerates tumor progression. Notably, compared with loss of the closely related gene p130, loss of p107 results in fewer but larger tumors as well as earlier metastatic spread. In addition, we observe differences in proliferation and apoptosis as well as altered distribution of initiated tumors in the lung, resulting from loss of p107 or p130. Collectively, these data demonstrate the feasibility of using the CRISPR-Cas9 system to model loss of candidate tumor suppressor genes in SCLC, and we anticipate that this approach will facilitate efforts to investigate mechanisms driving tumor progression in this deadly disease.
- Published
- 2019
- Full Text
- View/download PDF
15. IL-33 Signaling Alters Regulatory T Cell Diversity in Support of Tumor Development
- Author
-
Jonathan Y. Kim, Michael E. Birnbaum, Patricia Rogers, Olivia Smith, Tyler Jacks, Celeste Dang, Arjun Bhutkar, Aviv Regev, Jason M. Schenkel, Michelle Hillman, Amy Li, David Canner, Livnat Jerby-Arnon, Le Cong, Orit Rozenblatt-Rosen, Rebecca H. Herbst, C. Garrett Rappazzo, and Michael S. Cuoco
- Subjects
0301 basic medicine ,Male ,Regulatory T cell ,IL1RL1 ,chemical and pharmacologic phenomena ,Biology ,CD8-Positive T-Lymphocytes ,T-Lymphocytes, Regulatory ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,immune system diseases ,Neoplasms ,medicine ,Immune Tolerance ,Tumor Microenvironment ,Animals ,lcsh:QH301-705.5 ,Immunosuppression Therapy ,Effector ,hemic and immune systems ,medicine.disease ,Interleukin-33 ,Phenotype ,Interleukin 33 ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,Cancer research ,Adenocarcinoma ,Female ,030217 neurology & neurosurgery ,CD8 ,Signal Transduction - Abstract
Summary: Regulatory T cells (Tregs) can impair anti-tumor immune responses and are associated with poor prognosis in multiple cancer types. Tregs in human tumors span diverse transcriptional states distinct from those of peripheral Tregs, but their contribution to tumor development remains unknown. Here, we use single-cell RNA sequencing (RNA-seq) to longitudinally profile dynamic shifts in the distribution of Tregs in a genetically engineered mouse model of lung adenocarcinoma. In this model, interferon-responsive Tregs are more prevalent early in tumor development, whereas a specialized effector phenotype characterized by enhanced expression of the interleukin-33 receptor ST2 is predominant in advanced disease. Treg-specific deletion of ST2 alters the evolution of effector Treg diversity, increases infiltration of CD8+ T cells into tumors, and decreases tumor burden. Our study shows that ST2 plays a critical role in Treg-mediated immunosuppression in cancer, highlighting potential paths for therapeutic intervention. : Li et al. show in a genetic mouse model of lung adenocarcinoma that during tumor development regulatory T cell (Treg) diversity shifts from an interferon-responsive to a ST2-positive, Klrg1+Areg+ effector-like phenotype. Treg-specific deletion of ST2 alters Treg heterogeneity, increases tumor infiltration by CD8+ T cells, and decreases tumor burden. Keywords: regulatory T cell, autochthonous mouse model of cancer, lung adenocarcinoma, tumor immunosuppression, single cell RNA sequencing, Treg heterogeneity, interleukin-33, ST2, Il1rl1
- Published
- 2019
16. Use of a Blast Dominance–Hematogone Index for the Flow Cytometric Evaluation of Myelodysplastic Syndrome (MDS)
- Author
-
David M. Dorfman, Graham Dudley, Mai Drew, Jason M. Schenkel, Christopher B. Hergott, and Karry Charest
- Subjects
Adult ,Male ,medicine.medical_specialty ,Population ,Chronic myelomonocytic leukemia ,CD13 Antigens ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Immunophenotyping ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,education ,Aged ,Dominance (genetics) ,Aged, 80 and over ,education.field_of_study ,Receiver operating characteristic ,business.industry ,Area under the curve ,HLA-DR Antigens ,General Medicine ,Middle Aged ,Flow Cytometry ,medicine.disease ,medicine.anatomical_structure ,Normal bone ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Female ,Bone marrow ,business ,030215 immunology - Abstract
OBJECTIVES We tested whether combined flow cytometric assessment of loss of blast heterogeneity and decreased hematogones is a diagnostically useful approach for evaluation of myelodysplastic syndrome (MDS). METHODS Bone marrow samples from patients with known MDS were analyzed by 10-color flow cytometric immunophenotyping and compared with normal bone marrow samples. RESULTS There was loss of blast heterogeneity in patients with MDS compared with normal bone marrow samples, based on the relative size of the dominant blast population (83.0% vs 64.8%) and fewer hematogones (0.08% vs 1.39%). The size of the largest blast population divided by the fraction of hematogones (blast dominance-hematogone [BDH] index) was significantly larger in MDS compared with normal cases (27,084 vs 190, P < .0001; receiver operating characteristic area under the curve = 0.96). CONCLUSIONS The BDH index is more sensitive and specific than loss of blast heterogeneity or decrease in hematogones for detecting MDS in bone marrow samples and may be useful in clinical practice.
- Published
- 2019
- Full Text
- View/download PDF
17. CD4+ resident memory T cells dominate immunosurveillance and orchestrate local recall responses
- Author
-
Nancy J. Fares-Frederickson, Kathryn A. Fraser, Lalit K. Beura, Milcah C. Scott, Jason M. Schenkel, Vaiva Vezys, Elizabeth M. Steinert, Emily A. Thompson, and David Masopust
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Male ,Chemokine ,Immunology ,CD8-Positive T-Lymphocytes ,Granzymes ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Movement ,medicine ,Immunology and Allergy ,Animals ,Arenaviridae Infections ,Lymphocytic choriomeningitis virus ,RNA-Seq ,Immunologic Surveillance ,Research Articles ,biology ,Chimera ,Mucous membrane ,Phenotype ,Small intestine ,Cell biology ,Immunosurveillance ,Granzyme B ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Lymphatic system ,biology.protein ,Female ,Transcriptome ,Immunologic Memory ,CD8 ,030215 immunology - Abstract
CD4 T cell localization impacts function and differentiation. Beura et al. show that memory CD4+ T cells are largely resident in both lymphoid and non-lymphoid tissues, organize local recall responses, and share overlapping transcriptional and location-specific features with CD8+ TRM., This study examines the extent to which memory CD4+ T cells share immunosurveillance strategies with CD8+ resident memory T cells (TRM). After acute viral infection, memory CD4+ T cells predominantly used residence to survey nonlymphoid tissues, albeit not as stringently as observed for CD8+ T cells. In contrast, memory CD4+ T cells were more likely to be resident within lymphoid organs than CD8+ T cells. Migration properties of memory-phenotype CD4+ T cells in non-SPF parabionts were similar, generalizing these results to diverse infections and conditions. CD4+ and CD8+ TRM shared overlapping transcriptional signatures and location-specific features, such as granzyme B expression in the small intestine, revealing tissue-specific and migration property–specific, in addition to lineage-specific, differentiation programs. Functionally, mucosal CD4+ TRM reactivation locally triggered both chemokine expression and broad immune cell activation. Thus, residence provides a dominant mechanism for regionalizing CD4+ T cell immunity, and location enforces shared transcriptional, phenotypic, and functional properties with CD8+ T cells., Graphical Abstract
- Published
- 2019
18. The CD155/TIGIT axis promotes and maintains immune evasion in neoantigen-expressing pancreatic cancer
- Author
-
Jason M. Schenkel, Prajan Divakar, Alex M. Jaeger, Ana P. Garcia, Peter M. K. Westcott, Tyler Hether, Zackery A. Ely, Roderick T. Bronson, Laurens J. Lambert, Jason Reeves, William A. Freed-Pastor, Devan Phillips, Vikram Deshpande, Arjun Bhutkar, Kim L. Mercer, Ömer H. Yilmaz, Tyler Jacks, George Eng, Lin Lin, William L. Hwang, Nimisha B. Pattada, Aviv Regev, Toni Delorey, and William M. Rideout
- Subjects
Cancer Research ,endocrine system diseases ,medicine.medical_treatment ,Article ,Mice ,Immune system ,Lymphocytes, Tumor-Infiltrating ,TIGIT ,Pancreatic cancer ,MHC class I ,medicine ,Animals ,Humans ,CD155 ,Immune Evasion ,biology ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,digestive system diseases ,Pancreatic Neoplasms ,Oncology ,Cancer research ,biology.protein ,Receptors, Virus ,business ,CD8 - Abstract
Summary The CD155/TIGIT axis can be co-opted during immune evasion in chronic viral infections and cancer. Pancreatic adenocarcinoma (PDAC) is a highly lethal malignancy, and immune-based strategies to combat this disease have been largely unsuccessful to date. We corroborate prior reports that a substantial portion of PDAC harbors predicted high-affinity MHC class I-restricted neoepitopes and extend these findings to advanced/metastatic disease. Using multiple preclinical models of neoantigen-expressing PDAC, we demonstrate that intratumoral neoantigen-specific CD8+ T cells adopt multiple states of dysfunction, resembling those in tumor-infiltrating lymphocytes of PDAC patients. Mechanistically, genetic and/or pharmacologic modulation of the CD155/TIGIT axis was sufficient to promote immune evasion in autochthonous neoantigen-expressing PDAC. Finally, we demonstrate that the CD155/TIGIT axis is critical in maintaining immune evasion in PDAC and uncover a combination immunotherapy (TIGIT/PD-1 co-blockade plus CD40 agonism) that elicits profound anti-tumor responses in preclinical models, now poised for clinical evaluation.
- Published
- 2021
19. Deciphering the tumor-specific immunopeptidome in vivo with genetically engineered mouse models
- Author
-
Alex K. Shalek, Kohn R, Forest M. White, Santiago Naranjo, William M. Rideout, Alex M. Jaeger, Demi Sandel, Tim B. Fessenden, Shanahan S, Tyler Jacks, Sanders E, Jason M. Schenkel, William A. Freed-Pastor, Winter Ps, Lauren E. Stopfer, and Stefani Spranger
- Subjects
Immunosurveillance ,biology ,Antigen ,In vivo ,Tumor progression ,Genetically Engineered Mouse ,Antigen presentation ,biology.protein ,Cancer research ,Proteomics ,Major histocompatibility complex - Abstract
Effective immunosurveillance of cancer requires the presentation of peptide antigens on major histocompatibility complex Class I (MHC-I). Recent developments in proteomics have improved the identification of peptides that are naturally presented by MHC-I, collectively known as the “immunopeptidome”. Current approaches to profile tumor immunopeptidomes have been limited to in vitro investigation, which fails to capture the in vivo repertoire of MHC-I peptides, or bulk tumor lysates, which are obscured by the lack of tumor-specific MHC-I isolation. To overcome these limitations, we report here the engineering of a Cre recombinase-inducible affinity tag into the endogenous mouse MHC-I gene and targeting of this allele to the KrasLSL-G12D/+; p53fl/fl (KP) mouse model (KP; KbStrep). This novel approach has allowed us to isolate tumor-specific MHC-I peptides from autochthonous pancreatic ductal adenocarcinoma (PDAC) and lung adenocarcinoma (LUAD) in vivo. With this powerful analytical tool, we were able to profile the evolution of the LUAD immunopeptidome through tumor progression and show that in vivo MHC-I presentation is shaped by post-translational mechanisms. We also uncovered novel, putative LUAD tumor associated antigens (TAAs). Many peptides that were recurrently presented in vivo exhibited very low expression of the cognate mRNA, provoking reconsideration of antigen prediction pipelines that triage peptides according to transcript abundance. Beyond cancer, the KbStrep allele is compatible with a broad range of Cre-driver lines to explore antigen presentation in vivo in the pursuit of understanding basic immunology, infectious disease, and autoimmunity.
- Published
- 2021
- Full Text
- View/download PDF
20. Single-cell analyses identify circulating anti-tumor CD8 T cells and markers for their enrichment
- Author
-
Conor Delaney, Kaitlyn A. Lagattuta, Jacob M. Luber, Osmaan Shahid, Kelly P. Burke, Jaclyn M. Long, Kelly M. Mahuron, Margaret M. Lowe, Arlene H. Sharpe, Katy K. Tsai, Melissa Chow, Adil Daud, Megan E. Fung, Jason M. Schenkel, Juhi R. Kuchroo, Samantha Guinn, Kristen E. Pauken, Linglin Huang, Meromit Singer, Kathleen Newcomer, and Michael Rosenblum
- Subjects
0301 basic medicine ,Skin Neoplasms ,Cell ,Programmed Cell Death 1 Receptor ,CD8-Positive T-Lymphocytes ,Inbred C57BL ,Medical and Health Sciences ,Transcriptome ,Mice ,0302 clinical medicine ,Receptors ,Immunology and Allergy ,Cytotoxic T cell ,2.1 Biological and endogenous factors ,Melanoma ,Cancer ,Tumor ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Antigen ,Colonic Neoplasms ,Female ,Single-Cell Analysis ,Tumor Immunology ,T cell ,Immunology ,Receptors, Antigen, T-Cell ,Biology ,Adenocarcinoma ,Article ,Cell Line ,03 medical and health sciences ,Cell Line, Tumor ,Biomarkers, Tumor ,medicine ,Genetics ,Animals ,Humans ,Cluster of differentiation ,T-cell receptor ,NKG2D ,medicine.disease ,T-Cell ,Molecular biology ,Mice, Inbred C57BL ,030104 developmental biology ,CD8 ,Biomarkers - Abstract
Using single-cell RNA sequencing and TCR sequencing, Pauken et al. detect CD8+ T cell clones shared between blood and tumor in mice or melanoma patients, characterize matching clones in blood and tumor, and identify potential biomarkers for their isolation in blood., The ability to monitor anti-tumor CD8+ T cell responses in the blood has tremendous therapeutic potential. Here, we used paired single-cell RNA and TCR sequencing to detect and characterize “tumor-matching” (TM) CD8+ T cells in the blood of mice with MC38 tumors or melanoma patients using the TCR as a molecular barcode. TM cells showed increased activation compared with nonmatching T cells in blood and were less exhausted than matching cells in tumors. Importantly, PD-1, which has been used to identify putative circulating anti-tumor CD8+ T cells, showed poor sensitivity for identifying TM cells. By leveraging the transcriptome, we identified candidate cell surface markers for TM cells in mice and patients and validated NKG2D, CD39, and CX3CR1 in mice. These data show that the TCR can be used to identify tumor-relevant cells for characterization, reveal unique transcriptional properties of TM cells, and develop marker panels for tracking and analysis of these cells., Graphical Abstract
- Published
- 2021
21. Antigen Dominance Hierarchies Shape TCF1+ Progenitor CD8 T Cell Phenotypes in Tumors
- Author
-
Sarah E. Blatt, Izumi de los Rios Kobara, Aviv Regev, Arjun Bhutkar, David Canner, Sandro Santagata, Sara Z. Tavana, Amy Li, Jason M. Schenkel, Tyler Jacks, Grace E. Crossland, Michelle Hillman, Tamina Kienka, Peter M. K. Westcott, Andrea Garmilla, Giorgio Gaglia, William L. Hwang, Megan L. Burger, Cecily C. Ritch, and Amanda M. Cruz
- Subjects
Subdominant ,medicine.anatomical_structure ,Antigen ,T cell ,Cancer research ,medicine ,Cytotoxic T cell ,Immunodominance ,Progenitor cell ,Biology ,Immune checkpoint ,Progenitor - Abstract
CD8 T cell responses against different tumor neoantigens occur simultaneously, yet little is known about their interplay and its impact on T cell function and tumor control. In mouse lung adenocarcinoma, we find that immunodominance is established in tumors, wherein CD8 T cell expansion is predominantly driven by the antigen that most stably binds MHC. T cells responding to subdominant antigens are enriched for a TCF1+ progenitor phenotype that has been correlated with response to immune checkpoint blockade (ICB) therapy. However, the subdominant T cell response does not preferentially benefit from ICB due to a dysfunctional subset of TCF1+ progenitor cells marked by CCR6 and Tc17 differentiation. Analysis of human samples and sequencing datasets indicates CCR6+ TCF1+ cells exist across human cancers and correlate with poor ICB response. Vaccination eliminates CCR6+ TCF1+ cells and dramatically expands the subdominant response, highlighting a strategy to optimally engage concurrent neoantigen responses against tumors.
- Published
- 2021
- Full Text
- View/download PDF
22. Inhibitory signaling sustains a distinct early memory CD8 + T cell precursor that is resistant to DNA damage
- Author
-
Kristen E. Pauken, Zhangying Cai, Bertram Bengsch, Jonathan B. Johnnidis, Shin Foong Ngiow, Dario A. A. Vignali, Michael A. Paley, Yuki Muroyama, Sasikanth Manne, Arlene H. Sharpe, Shufei Song, Christelle Harly, Jason M. Schenkel, John Attanasio, Jesse M. Platt, Zeyu Chen, Allison R. Greenplate, Avinash Bhandoola, Steven L. Reiner, F. Bradley Johnson, Mohamed S. Abdel-Hakeem, Kito Nzingha, Jean Christophe Beltra, Makoto Kurachi, Mohammed Alkhatim A. Ali, Josephine R. Giles, Vesselin T. Tomov, E. John Wherry, University of Pennsylvania [Philadelphia], Washington University in Saint Louis (WUSTL), Massachusetts General Hospital [Boston], Brigham & Women’s Hospital [Boston] (BWH), Harvard Medical School [Boston] (HMS), Massachusetts Institute of Technology (MIT), Immunobiology of Human αβ and γδ T Cells and Immunotherapeutic Applications (CRCINA-ÉQUIPE 1), Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA), National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH), LabEX IGO Immunothérapie Grand Ouest, Freiburg University Medical Center, Centre for Biological Signaling Studies [Freiburg] (BIOSS), University of Freiburg [Freiburg], Kanazawa University (KU), University of Pittsburgh School of Medicine, Pennsylvania Commonwealth System of Higher Education (PCSHE), UPMC Hillman Cancer Center [Pittsburgh, PA, États-Unis], Columbia University [New York], University of Pennsylvania, Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes), Nantes Université (Nantes Univ), Universitäts Klinikum Freiburg = University Medical Center Freiburg (Uniklinik), and Bernardo, Elizabeth
- Subjects
0301 basic medicine ,Effector ,DNA damage ,Chemistry ,T cell ,Lymphocyte ,Immunology ,Eomesodermin ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,General Medicine ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,medicine ,Cytotoxic T cell ,CD5 ,CD8 ,030215 immunology - Abstract
International audience; The developmental origins of memory T cells remain incompletely understood. During the expansion phase of acute viral infection, we identified a distinct subset of virus-specific CD8 + T cells that possessed distinct characteristics including expression of CD62L, T cell factor 1 (TCF-1), and Eomesodermin; relative quiescence; expression of activation markers; and features of limited effector differentiation. These cells were a quantitatively minor subpopulation of the TCF-1 + pool and exhibited self-renewal, heightened DNA damage surveillance activity, and preferential long-term recall capacity. Despite features of memory and somewhat restrained proliferation during the expansion phase, this subset displayed evidence of stronger TCR signaling than other responding CD8 + T cells, coupled with elevated expression of multiple inhibitory receptors including programmed cell death 1 (PD-1), lymphocyte activating gene 3 (LAG-3), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), CD5, and CD160. Genetic ablation of PD-1 and LAG-3 compromised the formation of this CD62L hi TCF-1 + subset and subsequent CD8 + T cell memory. Although central memory phenotype CD8 + T cells were formed in the absence of these cells, subsequent memory CD8 + T cell recall responses were compromised. Together, these results identify an impor tant link between genome integrity maintenance and CD8 + T cell memory. Moreover, the data indicate a role for inhibitory receptors in preserving key memory CD8 + T cell precursors during initial activation and differentiation. Identification of this rare subpopulation within the memory CD8 + T cell precursor pool may help reconcile models of the developmental origin of long-term CD8 + T cell memory.
- Published
- 2021
- Full Text
- View/download PDF
23. Inhibitory signaling sustains a distinct early memory CD8
- Author
-
Jonathan B, Johnnidis, Yuki, Muroyama, Shin Foong, Ngiow, Zeyu, Chen, Sasikanth, Manne, Zhangying, Cai, Shufei, Song, Jesse M, Platt, Jason M, Schenkel, Mohamed, Abdel-Hakeem, Jean-Christophe, Beltra, Allison R, Greenplate, Mohammed-Alkhatim A, Ali, Kito, Nzingha, Josephine R, Giles, Christelle, Harly, John, Attanasio, Kristen E, Pauken, Bertram, Bengsch, Michael A, Paley, Vesselin T, Tomov, Makoto, Kurachi, Dario A A, Vignali, Arlene H, Sharpe, Steven L, Reiner, Avinash, Bhandoola, F Bradley, Johnson, and E John, Wherry
- Subjects
Male ,Mice, Knockout ,Precursor Cells, T-Lymphoid ,Programmed Cell Death 1 Receptor ,Cell Differentiation ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Lymphocyte Activation ,Listeria monocytogenes ,Lymphocyte Activation Gene 3 Protein ,Article ,Disease Models, Animal ,Memory T Cells ,Mice ,Antigens, CD ,Animals ,Humans ,Lymphocytic choriomeningitis virus ,Female ,Listeriosis ,Hepatocyte Nuclear Factor 1-alpha ,Immunologic Memory ,DNA Damage - Abstract
The developmental origins of memory T cells remain incompletely understood. During the expansion phase of acute viral infection, we identified a distinct subset of virus-specific CD8(+) T cells that possessed distinct characteristics including expression of CD62L, TCF-1 and Eomes; relative quiescence; expression of activation markers; and features of limited effector differentiation. These cells were a quantitatively minor subpopulation of the TCF-1(+) pool and exhibited self-renewal, heightened DNA damage surveillance activity, and preferential long-term recall capacity. Despite features of memory and somewhat restrained proliferation during the expansion phase, this subset displayed evidence of stronger TCR signaling than other responding CD8(+) T cells, coupled with elevated expression of multiple inhibitory receptors including PD-1, LAG-3, CTLA-4, CD5, and CD160. Indeed, genetic ablation of PD-1 and LAG-3 compromised the formation of this CD62L(hi) TCF-1(+) subset and subsequent CD8(+) T cell memory. Although central memory phenotype CD8(+) T cells (T(CM)) were formed in the absence of these cells, subsequent memory CD8(+) T cell recall responses were compromised. Together, these results identify an important link between genome integrity maintenance and CD8(+) T cell memory. Moreover, the data indicate a role for inhibitory receptors in preserving key memory CD8(+) T cell precursors during initial activation and differentiation. Identification of this rare subpopulation within the memory CD8(+) T cell precursor pool may help reconcile models of the developmental origin of long-term CD8(+) T cell memory.
- Published
- 2020
24. Single-nucleus and spatial transcriptomics of archival pancreatic cancer reveals multi-compartment reprogramming after neoadjuvant treatment
- Author
-
Cristina R. Ferrone, Eugene Drokhlyansky, Arnav Mehta, Conner Lambden, Alexander M. Tsankov, Joseph M. Beechem, Devan Phillips, Joshua Gould, David T. Ting, Mari Mino-Kenudson, Carlos Fernandez-del Castillo, David P. Ryan, Orr Ashenberg, Nicholas Van Wittenberghe, Caroline B. M. Porter, Lan Nguyen, Jason M. Schenkel, Hannah I. Hoffman, Robin Fropf, Domenic Abbondanza, Jay S. Loeffler, Julia Waldman, Theodore S. Hong, Karthik A. Jagadeesh, William A. Freed-Pastor, Payman Yadollahpour, Kit Fuhrman, Rahul Mohan, Denis Schapiro, Jimmy A. Guo, Toni Delorey, Tyler Jacks, George Eng, Aviv Regev, Daniel R. Zollinger, Andrew J. Aguirre, Marina Kern, Jennifer Y. Wo, William L. Hwang, Colin D. Weekes, Jason Reeves, Michael S. Cuoco, Samouil L. Farhi, Danielle Dionne, Andrew S. Liss, Clifton Rodrigues, Orit Rozenblatt-Rosen, and Debora Ciprani
- Subjects
Cell type ,medicine.medical_treatment ,Cell ,Context (language use) ,Biology ,medicine.disease ,Transcriptome ,Radiation therapy ,medicine.anatomical_structure ,Stroma ,Pancreatic cancer ,Cancer research ,medicine ,Reprogramming - Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a treatment-refractory disease. Characterizing PDAC by mRNA profiling remains particularly challenging. Previously identified bulk expression subtypes were influenced by contaminating stroma and have not yet informed clinical management, whereas single cell RNA-seq (scRNA-seq) of fresh tumors under-represented key cell types. Here, we developed a robust single-nucleus RNA-seq (snRNA-seq) technique for frozen archival PDAC specimens and used it to study both untreated tumors and those that received neoadjuvant chemotherapy and radiotherapy (CRT). Gene expression programs learned across untreated malignant cell and fibroblast profiles uncovered a clinically relevant molecular taxonomy with improved prognostic stratification compared to prior classifications. Moreover, in the increasingly-adopted neoadjuvant treatment context, there was a depletion of classical-like phenotypes in malignant cells in favor of basal-like phenotypes associated with TNF-NFkB and interferon signaling as well as the presence of novel acinar and neuroendocrine classical-like states, which may be more resilient to cytotoxic treatment. Spatially-resolved transcriptomics revealed an association between malignant cells expressing these basal-like programs and higher immune infiltration with increased lymphocytic content, whereas those exhibiting classical-like programs were linked to sparser macrophage-predominant microniches, perhaps pointing to susceptibility to distinct therapeutic strategies. Our refined molecular taxonomy and spatial resolution can help advance precision oncology in PDAC through informative stratification in clinical trials and insights into differential therapeutic targeting leveraging the immune system.
- Published
- 2020
- Full Text
- View/download PDF
25. Plateletpheresis-associated lymphopenia in frequent platelet donors
- Author
-
A. Helena Jonsson, Duane R. Wesemann, Revital Yefidoff-Freedman, Idayat Brimah, Donna Neuberg, Mahboubeh Rahmani, Zhihan J. Li, Dan L. Longo, Martha Ellis, Brooke M. Fortin, Michael B. Brenner, Nicolas C. Issa, Steven R. Sloan, Robin Smeland-Wagman, Lindsey R. Baden, Deepak A. Rao, Nancy Berliner, John M. Gansner, Richard M. Kaufman, and Jason M. Schenkel
- Subjects
Adult ,Blood Platelets ,Male ,Regulatory T cell ,Immunology ,Plateletpheresis ,Blood Donors ,030204 cardiovascular system & hematology ,Biochemistry ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Immunophenotyping ,Receptor repertoire ,Lymphopenia ,medicine ,Humans ,Platelet ,Aged ,Platelet Count ,Transfusion Medicine ,business.industry ,Cell Biology ,Hematology ,Middle Aged ,Prognosis ,Cross-Sectional Studies ,medicine.anatomical_structure ,Apheresis ,Leukoreduction ,Female ,business ,CD8 ,030215 immunology - Abstract
More than 1 million apheresis platelet collections are performed annually in the United States. After 2 healthy plateletpheresis donors were incidentally found to have low CD4+ T-lymphocyte counts, we investigated whether plateletpheresis causes lymphopenia. We conducted a cross-sectional single-center study of platelet donors undergoing plateletpheresis with the Trima Accel, which removes leukocytes continuously with its leukoreduction system chamber. We recruited 3 groups of platelet donors based on the total number of plateletpheresis sessions in the prior 365 days: 1 or 2, 3 to 19, or 20 to 24. CD4+ T-lymphocyte counts were
- Published
- 2019
- Full Text
- View/download PDF
26. Sensing and alarm function of vaccine-elicited SIV-gag specific CD8 TRM in the reproductive mucosa of rhesus macaques
- Author
-
Vineet Joag, Clare Quarnstrom, Andrew Soerens, James Michael Stolley, Jason M Schenkel, Kathryn Fraser, Vaiva Vezys, Pamela J Skinner, Eric Hunter, Benjamin Bimber, Rama Rao Amara, and David Masopust
- Subjects
Immunology ,Immunology and Allergy - Abstract
TRM constitute a recently identified lymphocyte lineage that occupies non-lymphoid tissues (NLT) without recirculating. Experiments in mice demonstrated that TRM reactivation triggers antiviral responses in neighboring cells by promoting local stimulation of innate and adaptive immune cells, and by recruiting immune effectors. Collectively this is referred to as ‘sensing and alarm’ function. We wished to broadly identify TRM functions in non-human primates (NHP). Here we applied a prime-boost vaccine modality in rhesus macaques to generate abundant SIVgag-specific CD8 TRM in the female reproductive tract (FRT) and 14 other NLT. To assess sensing and alarm function, macaques were challenged intravaginally with CM9 peptide from the SIV gag protein and necropsied 24 or 48 hours later. CD8 TRM reactivation increased expression of CD69 and granzyme B in SIV-gag specific CD8 T cells throughout the FRT at 24h, in situ proliferation (Ki67 expression) by 48h, and rapid antiviral and IFN response gene expression in essentially all hematopoietic and non-hematopoietic cells by 24h. Upregulation of effector genes in CD8 T cells, CD4 T cells, NK cells, and ILCs peaked at 24h and persisted at 48h. Mucosal CD4 T cells expressed various HIV restriction factors, and had reduced expression of the HIV-coreceptor CCR5. Increased numbers of vaginal T and B cells coincided with increased expression of chemokines and VCAM-1 on endothelial and stromal cells and a concomitant reduction in circulating T cells, B cells and SIV-env-specific B cells. These data demonstrate that vaccine-elicited CD8 TRM rapidly trigger local activation and the recruitment of innate, cellular, and humoral immune responses to the site of antigen exposure.
- Published
- 2022
- Full Text
- View/download PDF
27. Integrated stress response and immune cell infiltration in an ibrutinib‐refractory mantle cell lymphoma patient following <scp>ONC</scp> 201 treatment
- Author
-
Maria Badillo, Hun J. Lee, Jorge E. Romaguera, Jason M. Schenkel, Joshua E. Allen, Chi Young Ok, Michael Wang, Evita Sadimin, Rohinton Tarapore, Lee Schalop, Varun V. Prabhu, and Andrew Zloza
- Subjects
0301 basic medicine ,business.industry ,Hematology ,medicine.disease ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,030220 oncology & carcinogenesis ,Ibrutinib ,Cancer research ,Refractory Mantle Cell Lymphoma ,Medicine ,Integrated stress response ,Mantle cell lymphoma ,business ,Immune cell infiltration - Published
- 2018
- Full Text
- View/download PDF
28. A phase II trial of riluzole, an antagonist of metabotropic glutamate receptor 1 (GRM1) signaling, in patients with advanced melanoma
- Author
-
Jason M. Schenkel, James S. Goydos, Michael P. Kane, Janice M. Mehnert, Weichung Joe Shih, Jiadong Li, Evita Sadimin, Suzie Chen, Liesel Dudek, Jonathan H. Lee, Hongxia Lin, Ann W. Silk, Byeong-Seon Jeong, and Andrew Zloza
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,MAP Kinase Signaling System ,Down-Regulation ,Dermatology ,Receptors, Metabotropic Glutamate ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Internal medicine ,Clinical endpoint ,medicine ,Humans ,Adverse effect ,Melanoma ,Riluzole ,business.industry ,Antagonist ,Middle Aged ,medicine.disease ,Clinical trial ,030104 developmental biology ,030220 oncology & carcinogenesis ,Metabotropic glutamate receptor 1 ,Female ,business ,medicine.drug - Abstract
Studies demonstrate that GRM, expressed by >60% of human melanomas, may be a therapeutic target. We performed a phase II trial of 100 mg PO bid of riluzole, an inhibitor of GRM1 signaling, in patients with advanced melanoma with the primary endpoint of response rate. Thirteen patients with GRM1-positive tumors were enrolled. No objective responses were observed, and accrual was stopped. Stable disease was noted in six (46%) patients, with one patient on study for 42 weeks. Riluzole was well tolerated, with fatigue (62%) as the most common adverse event. Downregulation of MAPK and PI3K/AKT was noted in 33% of paired tumor biopsies. Hypothesis-generating correlative studies suggested that downregulation of angiogenic markers and increased leukocytes at the active edge of tumor correlate with clinical benefit. Pharmacokinetic analysis showed interpatient variability consistent with prior riluzole studies. Future investigations should interrogate mechanisms of biologic activity and advance the development of agents with improved bioavailability.
- Published
- 2018
- Full Text
- View/download PDF
29. Anti-Tumor TCF1+ CD8 T Cells are Functionally Diverse and Evolve During Tumorigenesis and Progression
- Author
-
Rebecca Herbst, Olivia Smith, Tyler Jacks, Amy Li, Aviv Regev, Michelle Hillman, Jason M. Schenkel, Orit Rozenblatt-rosen, and David Canner
- Subjects
Antitumor activity ,medicine.diagnostic_test ,medicine.medical_treatment ,Leptocytes ,Cancer ,General Medicine ,Biology ,medicine.disease_cause ,medicine.disease ,Flow cytometry ,Cytokine ,Antigen ,medicine ,Cancer research ,Cytotoxic T cell ,Carcinogenesis - Abstract
CD8+ T cells drive protective responses against infection and cancer. In the context of chronic antigen stimulation, CD8+ T cells become progressively dysfunctional, losing the ability to proliferate, secrete cytokines, and kill target cells. While dysfunctional CD8 T cells have been characterized in infection, studies examining longitudinal responses in tumors have been limited. Here, in an autochthonous model of lung adenocarcinoma using high dimensional flow cytometry and single cell RNA sequencing, we demonstrate that tumor specific CD8 T cell heterogeneity is dynamic, revealing multiple dysfunctional-like CD8 T cell populations. Among these states, we identify TCF-1+ CD8 T cells, a population previously associated with superior functionality. Temporal analysis revealed heterogeneity within the TCF-1+ CD8+ T cell compartment - SlamF6+ CD8 T cells were predominant in early stage disease, while SlamF6- CD8 T cells appeared later and were the majority during progression. Functionally, the SlamF6+ population was proliferative and expressed more inhibitory receptors than SlamF6- cells. However, SlamF6+ CD8 T cells gradually lost the ability to divide and secrete cytokines over the course of tumorigenesis, demonstrating that they become dysfunctional over time. Collectively, our results provide new insights into longitudinal response of TCF-1+ T cells over the course of tumorigenesis and have therapeutic implications for modulating the anti-tumor T cell response.
- Published
- 2020
- Full Text
- View/download PDF
30. Increased Dysfunction of Tumor Specific TCF-1+ CD8+ T Cells Associated with Increase in Non-Cycling TCF-1+ SlamF6- Cells Subsets Revealed by Single Cell Analysis
- Author
-
Jonathan Y. Kim, Jason M. Schenkel, Jin K. Park, Tyler Jacks, Aviv Regev, Kristen E. Pauken, Peter M. K. Westcott, George Eng, Michelle Hillman, David Canner, Orit Rozenblatt-Rosen, William A. Freed-Pastor, Amy Li, Megan L. Burger, Patricia Rogers, Rebecca H. Herbst, Olivia Smith, Le Cong, William L. Hwang, and Mike Cuoco
- Subjects
education.field_of_study ,Chemistry ,T cell ,Cell ,Population ,medicine.disease ,medicine.anatomical_structure ,Single-cell analysis ,medicine ,Cancer research ,Adenocarcinoma ,Cytotoxic T cell ,education ,Lymph node ,CD8 - Abstract
During chronic antigen stimulation, CD8+ T cells lose their effector potential. Because much of our understanding of anti-tumor T cell dysfunction comes from shorter-term transplantable models, it is less clear how dysfunction shifts over time in tumors. Using single-cell RNA-Seq and functional assays in an autochthonous model of lung adenocarcinoma, we demonstrate that anti-tumor CD8+ T cell dysfunction is dynamic and heterogeneous. In particular. we found a change over time in the composition of the TCF-1+ compartment, a population known to retain superior functionality, with proliferative TCF-1+ SlamF6+ cells predominant early, and an increase in non-cycling TCF-1+ SlamF6- cells with progression. Blocking egress from the draining lymph node decreased SlamF6+ CD8+ T cells in tumor-bearing lungs, suggesting that the lymph node is a reservoir of more functional anti-tumor CD8+ T cells. Collectively, our results provide insights into TCF-1+ T cell biology with therapeutic implications for anti-tumor immunity.
- Published
- 2020
- Full Text
- View/download PDF
31. The CD155/TIGIT Axis Promotes and Maintains Immune Evasion in Neoantigen-Expressing Pancreatic Cancer
- Author
-
Aviv Regev, Vikram Deshpande, Nimisha B. Pattada, William L. Hwang, Roderick T. Bronson, Jason M. Schenkel, William M. Rideout, Zackery A. Ely, Kim L. Mercer, William A. Freed-Pastor, Toni Delorey, Ana P. Garcia, Peter M. K. Westcott, Tyler Jacks, Laurens J. Lambert, George Eng, Arjun Bhutkar, Devan Phillips, Ömer H. Yilmaz, and Lin Lin
- Subjects
CD40 ,endocrine system diseases ,biology ,business.industry ,Cancer ,medicine.disease ,digestive system diseases ,Immune system ,TIGIT ,Pancreatic cancer ,MHC class I ,biology.protein ,Cancer research ,Medicine ,CD155 ,business ,CD8 - Abstract
The CD155/TIGIT axis can be co-opted during immune evasion in chronic viral infections and cancer. Pancreatic adenocarcinoma (PDAC) is a highly lethal malignancy, and immune-based strategies to combat this disease have been largely unsuccessful to date. We corroborate prior reports that a substantial portion of PDAC harbors predicted high affinity MHC class I-restricted neoepitopes and extend these findings to advanced/metastatic disease. Using two novel preclinical models of neoantigen-expressing PDAC, we demonstrate that intratumoral neoantigen-specific CD8+ T cells adopt multiple states of dysfunction, which are similar to tumor-infiltrating lymphocytes of human PDAC patients. Mechanistically, genetic and/or pharmacologic modulation of the CD155/TIGIT axis was sufficient to promote immune evasion in autochthonous neoantigen-expressing PDAC. Finally, we demonstrate that the CD155/TIGIT axis is critical to maintain immune evasion in PDAC and uncover a combination immunotherapy (TIGIT/PD-1 co-blockade plus CD40 agonism) that elicits profound anti-tumor responses in preclinical models, now poised for clinical evaluation.
- Published
- 2020
- Full Text
- View/download PDF
32. Abstract 94: Multi-compartment reprogramming and spatially-resolved interactions in frozen pancreatic cancer with and without neoadjuvant chemotherapy and radiotherapy at single-cell resolution
- Author
-
Jason M. Schenkel, Payman Yadollahpour, Cristina R. Ferrone, Denis Schapiro, Mari Mino-Kenudson, William A. Freed-Pastor, Kit Fuhrman, Tyler Jacks, Toni Delorey, Samouil L. Farhi, Marina Kern, Jimmy A. Guo, Jaimie L. Barth, Debora Ciprani, George Eng, Clifton Rodrigues, William L. Hwang, David T. Ting, Joseph M. Beechem, Daniel R. Zollinger, Hannah I. Hoffman, Jorge Roldan, Jason Reeves, Andrew S. Liss, J.Y. Wo, Andrew J. Aguirre, Aviv Regev, Eugene Drokhlyansky, Orit Rozenblatt-Rosen, Orr Ashenberg, Domenic Abbondanza, Colin D. Weekes, Carlos Fernandez-del Castillo, Robin Fropf, Devan Phillips, Nicholas Van Wittenberghe, Theodore S. Hong, and Karthik A. Jagadeesh
- Subjects
Cancer Research ,Chemotherapy ,Chemistry ,medicine.medical_treatment ,Spatially resolved ,Cell ,Resolution (electron density) ,medicine.disease ,Radiation therapy ,medicine.anatomical_structure ,Oncology ,Pancreatic cancer ,medicine ,Cancer research ,Compartment (pharmacokinetics) ,Reprogramming - Abstract
A molecular classification of pancreatic ductal adenocarcinoma (PDAC) that informs clinical management remains elusive. Previously identified bulk expression subtypes in the untreated setting were influenced by contaminating stroma whereas single cell RNA-seq (scRNA-seq) of fresh tumors under-represented key cell types. Two consensus subtypes have arisen from these prior efforts: (1) classical-like, and (2) basal-like. Basal-like tumors were associated with worse survival in the metastatic setting but attempts to refine this binary classification have failed to further stratify patient survival. Here, we developed a robust single-nucleus RNA-seq (snRNA-seq) technique for banked frozen PDAC specimens and studied a cohort of untreated resected primary tumors (n ~ 20). Gene expression programs learned across malignant cell and cancer-associated fibroblast (CAF) profiles uncovered a clinically-relevant molecular taxonomy with improved prognostic stratification compared to prior classifications. Digital spatial profiling revealed an association between malignant cells expressing basal-like programs and greater immune infiltration with relatively fewer macrophages, whereas those exhibiting classical-like programs were linked to inflammatory CAFs and macrophage-predominant microniches. Recent clinical trials have supported the increasing adoption of neoadjuvant therapy to aggressively address the risk of micro-metastatic spread and to circumvent concerns of treatment tolerance in the postoperative setting. There is an urgent need to understand how preoperative treatment impacts residual tumor cells and their interactions with other cell types in the tumor microenvironment to identify additional therapeutic vulnerabilities that can be exploited. Towards this end, we performed snRNA-seq on an unmatched cohort of neoadjuvant-treated resected primary tumors (n ~ 25) with most cases involving FOLFIRINOX chemotherapy followed by chemoradiation. Remarkably, the quality of single-nucleus mRNA profiles was comparable between heavily pre-treated and untreated specimens. We identified differentially expressed genes between treated and untreated samples to infer cell-type specific reprogramming in the residual tumor. This analysis revealed that in the neoadjuvant treatment context, there was lower expression of classical-like phenotypes in malignant cells in favor of basal-like phenotypes associated with TNF-NFkB and interferon signaling as well as the presence of novel acinar and neuroendocrine classical-like states. Our refined molecular taxonomy and spatial resolution may help advance precision oncology in PDAC through informative stratification in clinical trials and insights into compartment-specific therapies. Citation Format: William L. Hwang, Karthik A. Jagadeesh, Jimmy A. Guo, Hannah I. Hoffman, Payman Yadollahpour, Jason Reeves, Eugene Drokhlyansky, Nicholas Van Wittenberghe, Samouil Farhi, Denis Schapiro, George Eng, Jason M. Schenkel, William A. Freed-Pastor, Orr Ashenberg, Clifton Rodrigues, Domenic Abbondanza, Toni Delorey, Devan Phillips, Jorge Roldan, Debora Ciprani, Marina Kern, Jaimie L. Barth, Daniel R. Zollinger, Kit Fuhrman, Robin Fropf, Joseph Beechem, Colin Weekes, Cristina R. Ferrone, Jennifer Y. Wo, Theodore S. Hong, Orit Rozenblatt-Rosen, Andrew J. Aguirre, Mari Mino-Kenudson, Carlos Fernandez-del- Castillo, Andrew S. Liss, David T. Ting, Tyler Jacks, Aviv Regev. Multi-compartment reprogramming and spatially-resolved interactions in frozen pancreatic cancer with and without neoadjuvant chemotherapy and radiotherapy at single-cell resolution [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 94.
- Published
- 2021
- Full Text
- View/download PDF
33. Single-cell analyses characterize circulating anti-tumor CD8+ T cells in mice and humans and identify markers for their enrichment
- Author
-
Kristen E Pauken, Osmaan Shahid, Kaitlyn A Lagattuta, Kelly M Mahuron, Jacob M Luber, Margaret M Lowe, Linglin Huang, Conor Delaney, Jaclyn Long, Megan E Fung, Kathleen Newcomer, Katy K Tsai, Melissa Chow, Samantha Guinn, Juhi R Kuchroo, Kelly P Burke, Jason M Schenkel, Michael D Rosenblum, Adil I Daud, Arlene H Sharpe, and Meromit Singer
- Subjects
Immunology ,Immunology and Allergy - Abstract
The ability to monitor anti-tumor CD8+ T cell responses in the blood has tremendous therapeutic potential, but is currently challenging due to the limited number of reagents to track antigen-specific T cells. Here, we used paired single-cell RNA sequencing and T cell receptor (TCR) sequencing to detect and characterize “tumor matching” (TM) CD8+ T cells in the blood of mice with MC38 tumors or melanoma patients using the TCR as a molecular barcode. TM cells showed increased activation compared to non-matching T cells in blood, and appeared less exhausted than matching counterparts in tumor. Importantly, PD-1, which has been used to identify putative circulating anti-tumor CD8+ T cells, showed poor sensitivity for identifying TM cells in both mice and humans. By leveraging the transcriptome, we identified candidate cell surface marker panels for TM cells in mice and melanoma patients, and validated CX3CR1, NKG2D, and CD39 proteins in mice. Combinations of markers performed better than single markers in identifying TM cells from non-TM cells in the blood, providing a platform to potentially track TM cells based on surface markers instead of the TCR. These data demonstrate that the TCR can be used to identify tumor-relevant populations for comprehensive characterization, reveal unique transcriptional properties of TM cells, and develop marker panels for tracking and analysis of these cells.
- Published
- 2021
- Full Text
- View/download PDF
34. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade
- Author
-
Jernej Godec, Omar Khan, Alexander C. Huang, Jason M. Schenkel, Zeyu Chen, W. Nicholas Haining, E. John Wherry, Makoto Kurachi, Caroline Bartman, R. Anthony Barnitz, Shelley L. Berger, Morgan A. Sammons, Sasikanth Manne, Pamela M. Odorizzi, Bertram Bengsch, Golnaz Vahedi, Adam M. Drake, Kristen E. Pauken, and Debattama R. Sen
- Subjects
0301 basic medicine ,Transcription, Genetic ,CD8-Positive T-Lymphocytes ,Biology ,B7-H1 Antigen ,Article ,Epigenesis, Genetic ,Mice ,03 medical and health sciences ,Antigen ,Epigenetic Profile ,Animals ,Cytotoxic T cell ,Cell Lineage ,Gene Regulatory Networks ,Epigenetics ,Multidisciplinary ,Effector ,Interleukin-7 ,Cellular Reprogramming ,Cell biology ,Blockade ,Mice, Inbred C57BL ,030104 developmental biology ,Pd 1 blockade ,Female ,Immunotherapy ,Immunologic Memory - Abstract
Blocking Programmed Death–1 (PD-1) can reinvigorate exhausted CD8 T cells (T EX ) and improve control of chronic infections and cancer. However, whether blocking PD-1 can reprogram T EX into durable memory T cells (T MEM ) is unclear. We found that reinvigoration of T EX in mice by PD-L1 blockade caused minimal memory development. After blockade, reinvigorated T EX became reexhausted if antigen concentration remained high and failed to become T MEM upon antigen clearance. T EX acquired an epigenetic profile distinct from that of effector T cells (T EFF ) and T MEM cells that was minimally remodeled after PD-L1 blockade. This finding suggests that T EX are a distinct lineage of CD8 T cells. Nevertheless, PD-1 pathway blockade resulted in transcriptional rewiring and reengagement of effector circuitry in the T EX epigenetic landscape. These data indicate that epigenetic fate inflexibility may limit current immunotherapies.
- Published
- 2016
- Full Text
- View/download PDF
35. Normalizing the environment recapitulates adult human immune traits in laboratory mice
- Author
-
Kathryn A. Fraser, Sara E. Hamilton, Marc K. Jenkins, Pamela C. Rosato, Emily A. Thompson, Oludare A. Odumade, Kerry A. Casey, Vaiva Vezys, Kevin Bi, W. Nicholas Haining, Jason M. Schenkel, Ali Filali-Mouhim, Rafick Pierre Sekaly, David Masopust, Lalit K. Beura, and Stephen C. Jameson
- Subjects
0301 basic medicine ,Multidisciplinary ,Cellular differentiation ,Laboratory mouse ,Environmental exposure ,Biology ,Blood cell ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Immune system ,Immunity ,Immunology ,medicine ,Specific Pathogen Free Organism ,Specific-pathogen-free - Abstract
Our current understanding of immunology was largely defined in laboratory mice, partly because they are inbred and genetically homogeneous, can be genetically manipulated, allow kinetic tissue analyses to be carried out from the onset of disease, and permit the use of tractable disease models. Comparably reductionist experiments are neither technically nor ethically possible in humans. However, there is growing concern that laboratory mice do not reflect relevant aspects of the human immune system, which may account for failures to translate disease treatments from bench to bedside. Laboratory mice live in abnormally hygienic specific pathogen free (SPF) barrier facilities. Here we show that standard laboratory mouse husbandry has profound effects on the immune system and that environmental changes produce mice with immune systems closer to those of adult humans. Laboratory mice--like newborn, but not adult, humans--lack effector-differentiated and mucosally distributed memory T cells. These cell populations were present in free-living barn populations of feral mice and pet store mice with diverse microbial experience, and were induced in laboratory mice after co-housing with pet store mice, suggesting that the environment is involved in the induction of these cells. Altering the living conditions of mice profoundly affected the cellular composition of the innate and adaptive immune systems, resulted in global changes in blood cell gene expression to patterns that more closely reflected the immune signatures of adult humans rather than neonates, altered resistance to infection, and influenced T-cell differentiation in response to a de novo viral infection. These data highlight the effects of environment on the basal immune state and response to infection and suggest that restoring physiological microbial exposure in laboratory mice could provide a relevant tool for modelling immunological events in free-living organisms, including humans.
- Published
- 2016
- Full Text
- View/download PDF
36. Abstract PO016: Single-cell analyses characterize circulating anti-tumor CD8 T cells and identify markers for their isolation
- Author
-
Kelly M. Mahuron, Katy K. Tsai, Jacob M. Luber, Melissa Chow, Linglin Huang, Michael Rosenblum, Jason M. Schenkel, Megan E. Fung, Meromit Singer, Margaret M. Lowe, Osmaan Shahid, Arlene H. Sharpe, Kathleen Newcomer, Kelly P. Burke, Juhi R. Kuchroo, Kaitlyn A. Lagattuta, Adil Daud, Conor Delaney, Kristen E. Pauken, Jaclyn M. Long, and Samantha Guinn
- Subjects
Cancer Research ,medicine.medical_treatment ,Melanoma ,T cell ,Immunology ,T-cell receptor ,Cell ,Immunotherapy ,Biology ,medicine.disease ,Molecular biology ,Transcriptome ,medicine.anatomical_structure ,medicine ,Cytotoxic T cell ,CD8 - Abstract
The ability to monitor anti-tumor CD8+ T cell responses in the blood has tremendous therapeutic potential. Here, we used paired single-cell RNA sequencing and T cell receptor (TCR) sequencing to detect and characterize “tumor matching” (TM) CD8+ T cells in the blood of mice with MC38 tumors and melanoma patients using the TCR as a molecular barcode. TM cells showed increased activation compared to non-matching T cells in blood, and appeared less exhausted than matching counterparts in tumor. Importantly, PD-1, which has been used to identify putative circulating anti-tumor CD8+ T cells, showed poor sensitivity for identifying TM cells. By leveraging the transcriptome we identified candidate cell surface marker panels for TM cells in mice and melanoma patients, and validated markers in mice. Here, using combinations of markers provided better performance than single markers in identifying TM cells from non-TM cells in the blood. These data demonstrate that the TCR can be used to identify tumor-relevant populations for comprehensive characterization, reveal unique transcriptional properties of TM cells, and develop marker panels for tracking and analysis of these cells. Citation Format: Kristen E. Pauken, Osmaan Shahid, Kaitlyn A. Lagattuta, Kelly M. Mahuron, Jacob M. Luber, Margaret M. Lowe, Linglin Huang, Conor Delaney, Jaclyn M. Long, Megan E. Fung, Kathleen Newcomer, Katy K. Tsai, Melissa Chow, Samantha Guinn, Juhi R. Kuchroo, Kelly P. Burke, Jason M. Schenkel, Michael D. Rosenblum, Adil I. Daud, Arlene H. Sharpe, Meromit Singer. Single-cell analyses characterize circulating anti-tumor CD8 T cells and identify markers for their isolation [abstract]. In: Abstracts: AACR Virtual Special Conference: Tumor Immunology and Immunotherapy; 2020 Oct 19-20. Philadelphia (PA): AACR; Cancer Immunol Res 2021;9(2 Suppl):Abstract nr PO016.
- Published
- 2021
- Full Text
- View/download PDF
37. Abstract PR-007: Single-nucleus and spatial transcriptomics of archival pancreatic ductal adenocarcinoma reveals multi-compartment reprogramming after neoadjuvant treatment
- Author
-
Jason Reeves, Mari Mino-Kenudson, Debora Ciprani, Carlos Fernandez-del Castillo, Jason M. Schenkel, Hannah I. Hoffman, Domenic Abbondanza, Jimmy A. Guo, Tyler Jacks, Samouil L. Farhi, Aviv Regev, George Eng, William A. Freed-Pastor, Denis Schapiro, Daniel R. Zollinger, Eugene Drokhlyansky, Clifton Rodrigues, William L. Hwang, Jennifer Y. Wo, Orit Rozenblatt-Rosen, Nicholas Van Wittenberghe, Theodore S. Hong, Karthik A. Jagadeesh, Andrew J. Aguirre, and Andrew S. Liss
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Cell type ,business.industry ,medicine.medical_treatment ,Cancer ,Context (language use) ,medicine.disease ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Stroma ,030220 oncology & carcinogenesis ,Internal medicine ,Pancreatic cancer ,medicine ,business ,Reprogramming ,Neoadjuvant therapy - Abstract
Molecular subtyping of pancreatic ductal adenocarcinoma (PDAC) remains in its nascent stages and does not currently inform clinical management or therapeutic development. Previously identified bulk expression subtypes in the untreated setting were influenced by contaminating stroma whereas single cell RNA-seq (scRNA-seq) of fresh tumors under-represented key cell types. Two consensus subtypes have arisen from these prior efforts: (1) classical-pancreatic, encompassing a spectrum of pancreatic lineage precursors, and (2) basal-like/squamous/quasi-mesenchymal, characterized by loss of endodermal identity and aberrations in chromatin modifiers. Basal-like tumors were associated with poorer responses to chemotherapy and worse survival in the metastatic setting but attempts to refine this binary classification have failed to further stratify patient survival. Recent clinical trials have supported the increasing adoption of neoadjuvant therapy to aggressively address the risk of micro-metastatic spread and to circumvent concerns of treatment tolerance in the postoperative setting. There is an urgent need to understand how preoperative treatment reprograms residual tumor cells to identify additional therapeutic vulnerabilities that can be exploited in combination with neoadjuvant CRT. Here, we developed a robust single-nucleus RNA-seq (snRNA-seq) technique for frozen archival PDAC specimens and used it to study both untreated tumors (n = 15) and those that received neoadjuvant CRT (n = 11). Gene expression programs learned across malignant cell and fibroblast profiles uncovered a clinically relevant molecular taxonomy with improved prognostic stratification (median survival: 11.2 months in highest risk group to 44.7 months in lowest risk group) compared to prior classifications. Moreover, in the neoadjuvant treatment context, there was lower expression of classical-like phenotypes in malignant cells in favor of basal-like phenotypes associated with TNF-NFkB and interferon signaling as well as the presence of novel acinar and neuroendocrine classical-like states, which may be more resilient to cytotoxic treatment. These results suggest that differentiated endodermal phenotypes are only prevalent enough to be detected under treatment selection pressure and when observed in treatment-naïve bulk studies, may reflect normal cell contamination. Spatially-resolved transcriptomics revealed an association between malignant cells expressing basal-like programs and higher immune infiltration with increased lymphocytic content, whereas those exhibiting classical-like programs were linked to sparser macrophage-predominant microniches, perhaps pointing to distinct therapeutic susceptibilities. Our refined molecular taxonomy and spatial resolution may help advance precision oncology in PDAC through informative stratification in clinical trials and insights into differential therapeutic targeting leveraging the immune system. Citation Format: William L. Hwang, Karthik A. Jagadeesh, Jimmy A. Guo, Hannah I. Hoffman, Eugene Drokhlyansky, Nicholas Van Wittenberghe, Samouil Farhi, Denis Schapiro, Jason Reeves, Daniel R. Zollinger, George Eng, Jason M. Schenkel, William A. Freed-Pastor, Clifton Rodrigues, Domenic Abbondanza, Debora Ciprani, Jennifer Y. Wo, Theodore S. Hong, Andrew J. Aguirre, Orit Rozenblatt-Rosen, Mari Mino-Kenudson, Carlos Fernandez-del Castillo, Andrew S. Liss, Tyler E. Jacks, Aviv Regev. Single-nucleus and spatial transcriptomics of archival pancreatic ductal adenocarcinoma reveals multi-compartment reprogramming after neoadjuvant treatment [abstract]. In: Proceedings of the AACR Virtual Special Conference on Pancreatic Cancer; 2020 Sep 29-30. Philadelphia (PA): AACR; Cancer Res 2020;80(22 Suppl):Abstract nr PR-007.
- Published
- 2020
- Full Text
- View/download PDF
38. Abstract 3447: T cell antigen expression levels govern progression and therapy response in a novel model of colon cancer
- Author
-
Mary Clare Beytagh, William L. Hwang, Olivia Smith, Peter M. K. Westcott, Omer H Yilmaz, Tyler Jacks, Nathan J. Sacks, Jatin Roper, Jason M. Schenkel, Zackery A. Ely, George Eng, and Daniel Zhang
- Subjects
Cancer Research ,Therapy response ,Oncology ,Colorectal cancer ,business.industry ,medicine ,Cancer research ,T-cell Antigen ,medicine.disease ,business - Abstract
Genetically-engineered mouse models have greatly advanced our understanding of cancer, yet do not recapitulate the mutational complexity of human cancer, and likely lack neo-antigens capable of eliciting potent anti-tumor T cell responses. We developed a novel orthotopic organoid transplant model of colorectal cancer (CRC) harboring the strong T cell antigen SIINFEKL, and demonstrate the importance of antigen expression level in the anti-tumor T cell response. Although SIINFEKL low-expressing organoids (SIINLow) elicit an endogenous antigen-specific T cell response, the magnitude is substantially lower and kinetics delayed relative to SIINFEKL high-expressing organoids (SIINHi). Consistently, transplant of SIINHi results in rejection and T cell memory, while SIINLow results in tumor progression, terminally-exhausted T cells, and metastasis to the liver. We have shown that suboptimal T cell priming is the major factor underlying SIINLow tumor escape. Importantly, co-transplant of SIINHi and SIINLow organoids at distinct sites in the colon of the same animal results in complete rejection of both lines. In addition, co-transplant rescues the SIINLow tumor-infiltrating antigen-specific T cell response to a magnitude and quality comparable to that of SIINHi tumors. Single-cell RNA-sequencing of antigen-specific T cells from SIINHi and SIINLow tumors 8 days post-transplant revealed distinct clusters dominated by SIINLow-primed T cells, including a cluster enriched for immediate early response genes, Tox, and a number of immune checkpoints, indicative of early dysfunction. Collectively, our results establish the existence of a neo-antigen expression threshold at which T cell priming is limiting, resulting in attenuated magnitude and functionality of the T cell response, and tumor escape. To assess the therapeutic relevance of a poorly-expressed neo-antigen in CRC, we performed preclinical trials with immune checkpoint blockade and agonistic-CD40 (aCD40), which has been shown to potentiate T cell priming and response in poorly-immunogenic mouse and human pancreatic adenocarcinoma. While monotherapies showed only modest effects, the combination of checkpoint blockade and aCD40 resulted in an 80% response rate with a number of complete responses. In conclusion, antigen expression level is a critical determinant of T cell dysfunction, resulting from poor priming. Our results argue that targeting T cell priming may be a promising therapeutic strategy to invigorate anti-tumor immunity in human CRC, the majority of which remains refractory to immunotherapy. Citation Format: Peter Maxwell Kienitz Westcott, Nathan J. Sacks, Olivia Smith, Jason Schenkel, Zackery Ely, Daniel Zhang, Mary Clare Beytagh, William Hwang, George Eng, Jatin Roper, Omer Yilmaz, Tyler Jacks. T cell antigen expression levels govern progression and therapy response in a novel model of colon cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3447.
- Published
- 2020
- Full Text
- View/download PDF
39. Longitudinal single cell profiling of regulatory T cells identifies IL-33 as a driver of tumor immunosuppression
- Author
-
Jonathan Y. Kim, Jason M. Schenkel, Celeste Dang, Rebecca H. Herbst, Michelle Hillman, Mike Cuoco, Orit Rozenblatt-Rosen, Aviv Regev, Rappazzo Cg, Le Cong, Aming Li, Michael E. Birnbaum, Tyler Jacks, Olivia Smith, Patricia Rogers, David Canner, and Arjun Bhutkar
- Subjects
Effector ,medicine.medical_treatment ,Cell ,Immunosuppression ,hemic and immune systems ,chemical and pharmacologic phenomena ,Biology ,medicine.disease_cause ,Interleukin 33 ,Immune system ,medicine.anatomical_structure ,immune system diseases ,hemic and lymphatic diseases ,Cancer research ,medicine ,Receptor ,Carcinogenesis ,CD8 - Abstract
Regulatory T cells (Tregs) can impair anti-tumor immune responses and are associated with poor prognosis in multiple cancer types. Tregs in human tumors span diverse transcriptional states distinct from those of peripheral Tregs, but their contribution to tumor development remains unknown. Here, we used single cell RNA-Seq to longitudinally profile conventional CD4+ T cells (Tconv) and Tregs in a genetic mouse model of lung adenocarcinoma. Tissue-infiltrating and peripheral CD4+ T cells differed, highlighting divergent pathways of activation during tumorigenesis. Longitudinal shifts in Treg heterogeneity suggested increased terminal differentiation and stabilization of an effector phenotype over time. In particular, effector Tregs had enhanced expression of the interleukin 33 receptor ST2. Treg-specific deletion of ST2 reduced effector Tregs, increased infiltration of CD8+ T cells into tumors, and decreased tumor burden. Our study shows that ST2 plays a critical role in Treg-mediated immunosuppression in cancer, highlighting new potential paths for therapeutic intervention.
- Published
- 2019
- Full Text
- View/download PDF
40. Longitudinal Single Cell Profiling of Regulatory T Cells Identifies IL-33 as a Driver of Tumor Immunosuppression
- Author
-
Orit Rozenblatt-Rosen, Jason M. Schenkel, Jonathan Y. Kim, Michael S. Cuoco, Tyler Jacks, Amy Li, C. Garrett Rappazzo, Michelle Hillman, David Canner, Rebecca H. Herbst, Arjun Bhutkar, Olivia Smith, Aviv Regev, Livnat Jerby-Arnon, Michael E. Birnbaum, Patricia Rogers, Celeste Dang, and Le Cong
- Subjects
Effector ,Cell ,Cancer ,hemic and immune systems ,chemical and pharmacologic phenomena ,Biology ,medicine.disease ,medicine.disease_cause ,Interleukin 33 ,medicine.anatomical_structure ,Immune system ,medicine ,Cancer research ,Adenocarcinoma ,Carcinogenesis ,CD8 - Abstract
Regulatory T cells (Tregs) can impair anti-tumor immune responses and are associated with poor prognosis in multiple cancer types. Tregs in human tumors span diverse transcriptional states distinct from those of peripheral Tregs, but their contribution to tumor development remains unknown. Here, we used single cell RNA-Seq to longitudinally profile conventional CD4+ T cells (Tconv) and Tregs in a genetic mouse model of lung adenocarcinoma. Tissue-infiltrating and peripheral CD4+ T cells differed, highlighting divergent pathways of activation during tumorigenesis. Longitudinal shifts in Tregs heterogeneity suggested stabilization of a specialized effector phenotype over time that had enhanced expression of the interleukin 33 receptor ST2. Tregs-specific deletion of ST2 altered lung effector Tregs diversity, increased infiltration of CD8+ T cells into tumors, and decreased tumor burden. Our study shows that ST2 plays a critical role in Tregs-mediated immunosuppression in cancer, highlighting new potential paths for therapeutic intervention.
- Published
- 2019
- Full Text
- View/download PDF
41. Abstract PR11: Antigen dominance hierarchies shape tumor T-cell phenotypes and immunotherapy responses
- Author
-
Jason M. Schenkel, Grace E. Crossland, David Canner, Megan L. Burger, and Tyler Jacks
- Subjects
Cancer Research ,Subdominant ,medicine.medical_treatment ,Immunogenicity ,T cell ,Immunology ,Context (language use) ,Immunotherapy ,Biology ,Epitope ,medicine.anatomical_structure ,Dominance (ethology) ,Antigen ,medicine - Abstract
In order to drive a productive T-cell response to checkpoint immunotherapy, tumors must be sufficiently antigenic. However, antigens vary in their degree of immunogenicity and consequently drive T-cell responses of different magnitudes and quality. In a genetically engineered mouse model of lung adenocarcinoma expressing model T-cell antigens, we find that antigen dominance hierarchies are established in tumors, where the T-cell response is largely focused on a dominant epitope as observed in viral infections. Interestingly, while T cells responding to a subdominant antigen are fewer in number, we find that they are less exhausted and express more markers of memory cells. T cells responding to the dominant antigen rapidly contract, while those responding to the subdominant antigen persist better over time. Despite their seemingly greater functional potential, T cells responding to the subdominant antigen fail to respond significantly to checkpoint immunotherapy in the context of this dominance hierarchy. Remarkably, removal of the dominant antigen unleashes the subdominant response and these cells expand to fill the effector niche previously occupied by the dominant response. Current experiments are testing the ability of vaccination to “break dominance” and improve the contribution of the subdominant response to tumor control. These findings could have great relevance to the design of personalized cancer vaccines, where targeting subdominant antigens over dominant antigens may lead to surprising therapeutic benefit. This abstract is also being presented as Poster B36. Citation Format: Megan L. Burger, Grace E. Crossland, Jason M. Schenkel, David A. Canner, Tyler Jacks. Antigen dominance hierarchies shape tumor T-cell phenotypes and immunotherapy responses [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2019 Nov 17-20; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(3 Suppl):Abstract nr PR11.
- Published
- 2020
- Full Text
- View/download PDF
42. Cutting Edge: Identification of Autoreactive CD4+ and CD8+ T Cell Subsets Resistant to PD-1 Pathway Blockade
- Author
-
Clare F. Quarnstrom, Jason M. Schenkel, Marc K. Jenkins, Nathanael L. Sahli, Kevin C. Osum, Kristen E. Pauken, Tijana Martinov, Vaiva Vezys, Bruce R. Blazar, James Heffernan, Justin A. Spanier, Brian T. Fife, and Christine E. Nelson
- Subjects
CD4-Positive T-Lymphocytes ,T cell ,Programmed Cell Death 1 Receptor ,Immunology ,CD8-Positive T-Lymphocytes ,Biology ,B7-H1 Antigen ,Article ,Autoimmune Diseases ,Mice ,Interleukin 21 ,Mice, Inbred NOD ,Immune Tolerance ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Clonal Anergy ,Mice, Knockout ,ZAP70 ,CD28 ,Cell Differentiation ,Natural killer T cell ,Disease Models, Animal ,medicine.anatomical_structure ,Cancer research ,Female ,Disease Susceptibility ,Signal Transduction - Abstract
Programmed death-1 (PD-1) promotes T cell tolerance. Despite therapeutically targeting this pathway for chronic infections and tumors, little is known about how different T cell subsets are affected during blockade. We examined PD-1/PD ligand 1 (PD-L1) regulation of self-antigen–specific CD4 and CD8 T cells in autoimmune-susceptible models. PD-L1 blockade increased insulin-specific effector CD4 T cells in type 1 diabetes. However, anergic islet-specific CD4 T cells were resistant to PD-L1 blockade. Additionally, PD-L1 was critical for induction, but not maintenance, of CD8 T cell intestinal tolerance. PD-L1 blockade enhanced functionality of effector T cells, whereas established tolerant or anergic T cells were not dependent on PD-1/PD-L1 signaling to remain unresponsive. This highlights the existence of Ag-experienced T cell subsets that do not rely on PD-1/PD-L1 regulation. These findings illustrate how positive treatment outcomes and autoimmunity development during PD-1/PD-L1 inhibition are linked to the differentiation state of a T cell.
- Published
- 2015
- Full Text
- View/download PDF
43. Intravital mucosal imaging of CD8+ resident memory T cells shows tissue-autonomous recall responses that amplify secondary memory
- Author
-
Raissa Fonseca, David Masopust, Lalit K. Beura, Emily A. Thompson, Vaiva Vezys, Javed Mohammed, Heather D. Hickman, Jason S. Mitchell, Brandon J. Burbach, Sathi Wijeyesinghe, Brian T. Fife, and Jason M. Schenkel
- Subjects
0301 basic medicine ,Intravital Microscopy ,Cellular differentiation ,T cell ,Immunology ,Population ,Biology ,CD8-Positive T-Lymphocytes ,Article ,03 medical and health sciences ,Mice ,Antigen ,medicine ,Immunology and Allergy ,Animals ,education ,Immunity, Mucosal ,Immunologic Surveillance ,Skin ,education.field_of_study ,Mucous Membrane ,Cell biology ,Immunosurveillance ,030104 developmental biology ,Lymphatic system ,medicine.anatomical_structure ,Female ,Immunologic Memory ,Intravital microscopy ,CD8 - Abstract
CD8+ T cell immunosurveillance dynamics influence the outcome of intracellular infections and cancer. Here we used two-photon intravital microscopy to visualize the responses of CD8+ resident memory T cells (TRM cells) within the reproductive tracts of live female mice. We found that mucosal TRM cells were highly motile, but paused and underwent in situ division after local antigen challenge. TRM cell reactivation triggered the recruitment of recirculating memory T cells that underwent antigen-independent TRM cell differentiation in situ. However, the proliferation of pre-existing TRM cells dominated the local mucosal recall response and contributed most substantially to the boosted secondary TRM cell population. We observed similar results in skin. Thus, TRM cells can autonomously regulate the expansion of local immunosurveillance independently of central memory or proliferation in lymphoid tissue. Masopust and colleagues show that mucosal tissue-resident memory T cells proliferate in situ in response to local antigen and dominate the local recall response.
- Published
- 2018
44. Tissue-Resident Memory T Cells
- Author
-
David Masopust and Jason M. Schenkel
- Subjects
Lineage (genetic) ,T-Lymphocytes ,Cellular differentiation ,Lymphocyte ,Immunology ,Population ,Cell Ontogeny ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,T-Lymphocyte Subsets ,Immunity ,medicine ,Animals ,Humans ,Immunology and Allergy ,education ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,Effector ,Cell Differentiation ,Cell biology ,Infectious Diseases ,medicine.anatomical_structure ,Immunologic Memory ,Function (biology) ,030215 immunology - Abstract
Tissue-resident memory T (Trm) cells constitute a recently identified lymphocyte lineage that occupies tissues without recirculating. They provide a first response against infections reencountered at body surfaces, where they accelerate pathogen clearance. Because Trm cells are not present within peripheral blood, they have not yet been well characterized, but are transcriptionally, phenotypically, and functionally distinct from recirculating central and effector memory T cells. In this review, we will summarize current knowledge of Trm cell ontogeny, regulation, maintenance, and function and will highlight technical considerations for studying this population.
- Published
- 2014
- Full Text
- View/download PDF
45. Cutting Edge: Control of Mycobacterium tuberculosis Infection by a Subset of Lung Parenchyma–Homing CD4 T Cells
- Author
-
Jason M. Schenkel, David Masopust, Keith D. Kauffman, Daniel L. Barber, Shunsuke Sakai, Cortez McBerry, and Katrin D. Mayer-Barber
- Subjects
Adoptive cell transfer ,Pathology ,medicine.medical_specialty ,Lung ,Tuberculosis ,biology ,Immunology ,CXCR3 ,biology.organism_classification ,medicine.disease ,Mycobacterium tuberculosis ,medicine.anatomical_structure ,Parenchyma ,medicine ,Immunology and Allergy ,Interferon gamma ,medicine.drug ,Homing (hematopoietic) - Abstract
Th1 cells are critical for containment of Mycobacterium tuberculosis infection, but little else is known about the properties of protective CD4 T cell responses. In this study, we show that the pulmonary Th1 response against M. tuberculosis is composed of two populations that are either CXCR3hi and localize to lung parenchyma or are CX3CR1hiKLRG1hi and are retained within lung blood vasculature. M. tuberculosis–specific parenchymal CD4 T cells migrate rapidly back into the lung parenchyma upon adoptive transfer, whereas the intravascular effectors produce the highest levels of IFN-γ in vivo. Importantly, parenchymal T cells displayed greater control of infection compared with the intravascular counterparts upon transfer into susceptible T cell–deficient hosts. Thus, we identified a subset of naturally generated M. tuberculosis–specific CD4 T cells with enhanced protective capacity and showed that control of M. tuberculosis correlates with the ability of CD4 T cells to efficiently enter the lung parenchyma rather than produce high levels of IFN-γ.
- Published
- 2014
- Full Text
- View/download PDF
46. Costimulation via the tumor-necrosis factor receptor superfamily couples TCR signal strength to the thymic differentiation of regulatory T cells
- Author
-
Luke S. Manlove, Hideo Yagita, Jason M. Schenkel, Yan Xing, David L. Owen, Kristin A. Hogquist, Michael A. Farrar, Hongbo Chi, Shawn A. Mahmud, Jonathan S. Boomer, Heather Schmitz, Yanyan Wang, and Jonathan M. Green
- Subjects
Interleukin 2 ,Recombinant Fusion Proteins ,Cellular differentiation ,Immunology ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Thymus Gland ,Biology ,T-Lymphocytes, Regulatory ,Article ,Mice ,Glucocorticoid-Induced TNFR-Related Protein ,CD28 Antigens ,STAT5 Transcription Factor ,medicine ,Animals ,Receptors, Tumor Necrosis Factor, Type II ,Immunology and Allergy ,Progenitor cell ,Receptor ,Cells, Cultured ,Mice, Knockout ,Mice, Inbred BALB C ,T-cell receptor ,CD28 ,Cell Differentiation ,hemic and immune systems ,Receptor Cross-Talk ,Receptors, OX40 ,MAP Kinase Kinase Kinases ,Tumor Necrosis Factor Receptor-Associated Peptides and Proteins ,Cell biology ,Mice, Inbred C57BL ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
Regulatory T cells (Treg cells) express members of the tumor-necrosis factor (TNF) receptor superfamily (TNFRSF), but the role of those receptors in the thymic development of Treg cells is undefined. We found here that Treg cell progenitors had high expression of the TNFRSF members GITR, OX40 and TNFR2. Expression of those receptors correlated directly with the signal strength of the T cell antigen receptor (TCR) and required the coreceptor CD28 and the kinase TAK1. The neutralization of ligands that are members of the TNF superfamily (TNFSF) diminished the development of Treg cells. Conversely, TNFRSF agonists enhanced the differentiation of Treg cell progenitors by augmenting responsiveness of the interleukin 2 receptor (IL-2R) and transcription factor STAT5. Costimulation with the ligand of GITR elicited dose-dependent enrichment for cells of lower TCR affinity in the Treg cell repertoire. In vivo, combined inhibition of GITR, OX40 and TNFR2 abrogated the development of Treg cells. Thus, expression of members of the TNFRSF on Treg cell progenitors translated strong TCR signals into molecular parameters that specifically promoted the development of Treg cells and shaped the Treg cell repertoire.
- Published
- 2014
- Full Text
- View/download PDF
47. Abstract A22: Molecular subtypes and resistance programs in pancreatic ductal adenocarcinoma elucidated with single-nucleus RNA-seq
- Author
-
Eugene Drokhlyansky, Orr Ashenberg, Debora Ciprani, Nicholas Van Wittenberghe, Theodore S. Hong, Carlos Fernandez-del Castillo, Michael S. Cuoco, Karthik A. Jagadeesh, Danielle Dionne, Tyler Jacks, Clifton Rodriguez, George Eng, Andrew S. Liss, Andrew J. Aguirre, William L. Hwang, Alexander M. Tsankov, Mari Mino-Kenudson, Orit Rozenblatt-Rosen, Aviv Regev, Caroline B. M. Porter, Julia Waldman, Jason M. Schenkel, William A. Freed-Pastor, Laurens J. Lambert, and Connor Lambden
- Subjects
Cancer Research ,Stromal cell ,RNA integrity number ,Biology ,medicine.disease ,Transcriptome ,Oncology ,Pancreatic cancer ,Gene expression ,Cancer research ,medicine ,RNA extraction ,Copy-number variation ,Gene - Abstract
Objective: Pancreatic ductal adenocarcinoma (PDAC) remains a treatment-refractory disease as existing molecular subtypes are insufficient and do not currently inform clinical decisions. Rare cell types, including those responsible for resistance, are difficult to detect with bulk transcriptomic profiling. Indeed, several previously identified transcriptomic subtypes of PDAC are unintentionally driven by “contaminating” stromal components. Single-cell transcriptomics provides an unprecedented degree of resolution into the properties of individual cells. However, RNA extraction from RNase- and stroma-rich pancreatic tissue is difficult and prior single-cell efforts have been limited by suboptimal dissociation/RNA quality. We developed a robust single-nucleus RNA-seq (sNuc-seq) technique compatible with frozen archival PDAC specimens and computational techniques to identify the transcriptomic programs driving tumor subtypes and therapeutic resistance. Methods: Patients with localized PDAC undergoing surgical resection with or without neoadjuvant chemoradiotherapy were consented for this IRB-approved study. Specimens were screened for RNA Integrity Number >6. Single nuclei suspensions were extracted from flash-frozen primary PDAC specimens and organoids. Approximately 8,000 nuclei were loaded on the 10x Genomics Chromium platform per sample to generate and sequence 3’ gene expression libraries (Illumina HiSeq 2500, 125 bp paired-end reads). sNuc-seq derived reads were processed using the 10X CellRanger v3.0.2 pipeline. Unsupervised clustering was utilized to identify different cell populations and known marker genes from literature were used to label cell types. Results: Both treatment-naïve (n=12) and treatment-resistant (n=11) specimens yielded high-quality sNuc-seq data (>1,000 nuclei per sample, >1,000 median genes per nucleus). In each tumor, distinct clusters with gene expression profiles consistent with ductal, fibroblast, endothelial, endocrine, lymphocyte, and myeloid cell populations were identified. Malignant cells were confirmed by inferred copy number variation analysis (InferCNV v3.9) and segregated into several distinct clusters for each individual patient highlighting intratumoral heterogeneity. While some malignant clusters corresponded to previously identified basal-squamous and classical-progenitor bulk subtypes, others featured expression profiles distinct from known subtypes, including cells with upregulation of hypoxia-associated or cytoskeletal genes. Conclusions: Applying sNuc-seq to treatment-naïve and pretreated PDAC specimens, we uncovered significant intratumoral heterogeneity in the malignant and stromal compartments and identified malignant cells featuring transcriptomic programs that do not fit previously identified bulk subtypes. Characterization of therapeutic resistance programs, spatial relationships among cell types, and association with clinical outcomes is ongoing. Citation Format: William L. Hwang, Karthik A. Jagadeesh, Orr Ashenberg, Eugene Drokhlyansky, George Eng, Nicholas Van Wittenberghe, William Freed-Pastor, Clifton Rodriguez, Danielle Dionne, Julia Waldman, Michael Cuoco, Alexander Tsankov, Connor Lambden, Caroline Porter, Jason Schenkel, Laurens Lambert, Debora Ciprani, Andrew J. Aguirre, Mari Mino-Kenudson, Theodore S. Hong, Orit Rozenblatt-Rosen, Carlos Fernandez-del Castillo, Andrew S. Liss, Aviv Regev, Tyler E. Jacks. Molecular subtypes and resistance programs in pancreatic ductal adenocarcinoma elucidated with single-nucleus RNA-seq [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2019 Sept 6-9; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2019;79(24 Suppl):Abstract nr A22.
- Published
- 2019
- Full Text
- View/download PDF
48. The integration of T cell migration, differentiation and function
- Author
-
David Masopust and Jason M. Schenkel
- Subjects
History ,Lymphoid Tissue ,T-Lymphocytes ,T cell ,Classification scheme ,Biology ,Lymphocyte Activation ,medicine.disease_cause ,Education ,Autoimmunity ,Antigen ,Cell Movement ,Terminology as Topic ,medicine ,Animals ,Humans ,Extramural ,Cell Differentiation ,Computer Science Applications ,medicine.anatomical_structure ,T cell differentiation ,Immunology ,T cell migration ,Immunologic Memory ,Neuroscience ,Function (biology) - Abstract
T cells function locally. Accordingly, T cells' recognition of antigen, their subsequent activation and differentiation, and their role in the processes of infection control, tumour eradication, autoimmunity, allergy and alloreactivity are intrinsically coupled with migration. Recent discoveries revise our understanding of the regulation and patterns of T cell trafficking and reveal limitations in current paradigms. Here, we review classic and emerging concepts, highlight the challenge of integrating new observations with existing T cell classification schemes and summarize the heuristic framework provided by viewing T cell differentiation and function first through the prism of migration.
- Published
- 2013
- Full Text
- View/download PDF
49. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response
- Author
-
Ramin S. Herati, Bradley Wubbenhorst, Jason M. Schenkel, Robert H. Vonderheide, Kurt D'Andrea, Shannon Harmon, Shawn Kothari, Felix Quagliarello, Michael A. Postow, Brandon Wenz, Katherine L. Nathanson, Ravi K. Amaravadi, Lynn M. Schuchter, Robert J. Orlowski, E. John Wherry, Matthew Adamow, Sasikanth Manne, Phillip Wong, Alexander C. Huang, Josephine R. Giles, Wei Xu, Suzanne McGettigan, Deborah Kuk, Giorgos C. Karakousis, Katherine S. Panageas, Cristina Carrera, Bertram Bengsch, Sangeeth M. George, Tara C. Gangadhar, Rosemarie Mick, Xiaowei Xu, Jedd D. Wolchok, Kristen E. Pauken, and Ryan P. Staupe
- Subjects
Male ,medicine.medical_treatment ,T cell ,Programmed Cell Death 1 Receptor ,Pembrolizumab ,CD8-Positive T-Lymphocytes ,Antibodies, Monoclonal, Humanized ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Medicine ,Cytotoxic T cell ,Humans ,Melanoma ,Neoplasm Staging ,Multidisciplinary ,business.industry ,Immunotherapy ,medicine.disease ,Blockade ,Tumor Burden ,medicine.anatomical_structure ,Ki-67 Antigen ,Phenotype ,Treatment Outcome ,030220 oncology & carcinogenesis ,Monoclonal ,Immunology ,Female ,business ,030215 immunology - Abstract
Despite the success of monotherapies based on blockade of programmed cell death 1 (PD-1) in human melanoma, most patients do not experience durable clinical benefit. Pre-existing T-cell infiltration and/or the presence of PD-L1 in tumours may be used as indicators of clinical response; however, blood-based profiling to understand the mechanisms of PD-1 blockade has not been widely explored. Here we use immune profiling of peripheral blood from patients with stage IV melanoma before and after treatment with the PD-1-targeting antibody pembrolizumab and identify pharmacodynamic changes in circulating exhausted-phenotype CD8 T cells (Tex cells). Most of the patients demonstrated an immunological response to pembrolizumab. Clinical failure in many patients was not solely due to an inability to induce immune reinvigoration, but rather resulted from an imbalance between T-cell reinvigoration and tumour burden. The magnitude of reinvigoration of circulating Tex cells determined in relation to pretreatment tumour burden correlated with clinical response. By focused profiling of a mechanistically relevant circulating T-cell subpopulation calibrated to pretreatment disease burden, we identify a clinically accessible potential on-treatment predictor of response to PD-1 blockade.
- Published
- 2016
50. IL-15 independent maintenance of tissue resident and boosted effector memory CD8 T cells
- Author
-
Vaiva Vezys, Kathryn A. Fraser, David Masopust, Kristen E. Pauken, Lalit K. Beura, Kerry A. Casey, and Jason M. Schenkel
- Subjects
0301 basic medicine ,Cytotoxicity, Immunologic ,T cell ,Immunology ,Immunization, Secondary ,Biology ,CD8-Positive T-Lymphocytes ,Article ,03 medical and health sciences ,Mice ,Immunity ,T-Lymphocyte Subsets ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Homeostasis ,Cells, Cultured ,Cell Proliferation ,Interleukin-15 ,Mucous Membrane ,Effector ,Cell growth ,Viral Vaccines ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Interleukin 15 ,Memory T cell ,Immunologic Memory - Abstract
IL-15 regulates central and effector memory CD8 T cell (TCM and TEM, respectively) homeostatic proliferation, maintenance, and longevity. Consequently, IL-15 availability hypothetically defines the carrying capacity for total memory CD8 T cells within the host. In conflict with this hypothesis, previous observations demonstrated that boosting generates preternaturally abundant TEM that increases the total quantity of memory CD8 T cells in mice. In this article, we provide a potential mechanistic explanation by reporting that boosted circulating TEM do not require IL-15 for maintenance. We also investigated tissue-resident memory CD8 T cells (TRM), which protect nonlymphoid tissues from reinfection. We observed up to a 50-fold increase in the total magnitude of TRM in mouse mucosal tissues after boosting, suggesting that the memory T cell capacity in tissues is flexible and that TRM may not be under the same homeostatic regulation as primary central memory CD8 T cells and TEM. Further analysis identified distinct TRM populations that depended on IL-15 for homeostatic proliferation and survival, depended on IL-15 for homeostatic proliferation but not for survival, or did not depend on IL-15 for either process. These observations on the numerical regulation of T cell memory indicate that there may be significant heterogeneity among distinct TRM populations and also argue against the common perception that developing vaccines that confer protection by establishing abundant TEM and TRM will necessarily erode immunity to previously encountered pathogens as the result of competition for IL-15.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.