1
|
Zi R, Shen K, Zheng P, Su X, Yang Y, Chen Y, Dai H, Mao C, Lu Y, Wang L, Ma H, Wang W, Li Q, Lu W, Li C, Zheng S, Shi H, Liu X, Chen Z, Liang H, Ou J. NPC1L1 on pancreatic adenocarcinoma cell functions as a two-pronged checkpoint against antitumor activity. Innovation (N Y) 2025; 6:100783. [PMID: 40098667 PMCID: PMC11910884 DOI: 10.1016/j.xinn.2024.100783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 12/30/2024] [Indexed: 03/19/2025] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a highly lethal malignancy with an immunosuppressive microenvironment and a limited immunotherapy response. Cholesterol is necessary for rapid growth of cancer cells, and cholesterol metabolism reprogramming is a hallmark of PAAD. How PAAD cells initiate cholesterol reprogramming to sustain their growth demand and suppressive immunomicroenvironment remains elusive. In this study, we for the first time revealed that PAAD cells overcome cholesterol shortage and immune surveillance via ectopically overexpressing NPC1L1, a cholesterol transporter, but function as a two-pronged checkpoint, which not only directly suppresses TCR activation of CD8+T cells but also hijacks the intracellular cholesterol from CD8+T cells. In vivo, we showed that ezetimibe, an NPC1L1 inhibitor usually for hypercholesterolemia, efficiently prevented PAAD cells from depriving cholesterol of CD8+T cells, and improved the anti-tumor immunity of PAAD to synergize with PD-1 blockade, suggesting NPC1L1 as a promising target to rescue the anti-tumor activity in PAAD.
Collapse
Affiliation(s)
- Ruiyang Zi
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kaicheng Shen
- Department of Oncology, Fuling Hospital of Chongqing University, Chongqing 408000, China
| | - Pengfei Zheng
- College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xingxing Su
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yishi Yang
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yanrong Chen
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Haisu Dai
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chengyi Mao
- Department of Pathology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Yongling Lu
- Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Hongbo Ma
- Department of Oncology, Fuling Hospital of Chongqing University, Chongqing 408000, China
| | - Wei Wang
- Department of Oncology, Fuling Hospital of Chongqing University, Chongqing 408000, China
| | - Qingyun Li
- Genecast Biotechnology Co., Wuxi 214104, China
| | - Wei Lu
- Galixir Technologies, Beijing 100086, China
| | | | | | - Hui Shi
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaohong Liu
- National University of Singapore (Chongqing) Research Institute, Chongqing 401123, China
| | - Zhiyu Chen
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Houjie Liang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Juanjuan Ou
- Yu-Yue Pathology Scientific Research Center, Chongqing 401329, China
- Center for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu 610042, China
| |
Collapse
|
2
|
Afeyan AB, Wu CJ, Oliveira G. Rapid parallel reconstruction and specificity screening of hundreds of T cell receptors. Nat Protoc 2025; 20:539-586. [PMID: 39516267 PMCID: PMC11896752 DOI: 10.1038/s41596-024-01061-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/06/2024] [Indexed: 11/16/2024]
Abstract
The ability to screen the reactivity of T cell receptors (TCRs) is essential to understanding how antigen-specific T cells drive productive or dysfunctional immune responses during infections, cancer and autoimmune diseases. Methods to profile large numbers of TCRs are critical for characterizing immune responses sustained by diverse T cell clones. Here we provide a medium-throughput approach to reconstruct dozens to hundreds of TCRs in parallel, which can be simultaneously screened against primary human tissues and broad curated panels of antigenic targets. Using Gibson assembly and miniaturized lentiviral transduction, individual TCRs are rapidly cloned and expressed in T cells; before screening, TCR cell lines undergo combinatorial labeling with dilutions of three fluorescent dyes, which allows retrieval of the identity of individual T cell effectors when they are organized and tested in pools using flow cytometry. Upon incubation with target cells, we measure the upregulation of CD137 on T cells as a readout of TCR activation. This approach is scalable and simultaneously captures the reactivity of pooled TCR cell lines, whose activation can be deconvoluted in real time, thus providing a path for screening the reactivity of dozens of TCRs against broad panels of synthetic antigens or against cellular targets, such as human tumor cells. We applied this pipeline to systematically deconvolute the antitumoral and antiviral reactivity and antigenic specificity of TCRs from human tumor-infiltrating lymphocytes. This protocol takes ~2 months, from experimental design to data analysis, and requires standard expertise in cloning, cell culture and flow cytometry.
Collapse
Affiliation(s)
- Alexander B Afeyan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Lahusen A, Minhöfer N, Lohse KA, Blechner C, Lindenmayer J, Eiseler T, Wellstein A, Kleger A, Seufferlein T, Windhorst S, Lin YN. Pancreatic cancer cell-intrinsic transglutaminase-2 promotes T cell suppression through microtubule-dependent secretion of immunosuppressive cytokines. J Immunother Cancer 2025; 13:e010579. [PMID: 39824529 PMCID: PMC11748943 DOI: 10.1136/jitc-2024-010579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/30/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is mostly refractory to immunotherapy due to immunosuppression in the tumor microenvironment and cancer cell-intrinsic T cell tolerance mechanisms. PDAC is described as a "cold" tumor type with poor infiltration by T cells and factors leading to intratumoral T cell suppression have thus received less attention. Here, we identify a cancer cell-intrinsic mechanism that contributes to a T cell-resistant phenotype and describes potential combinatorial therapy. METHODS We used an unbiased screening approach of T cell resistant and sensitive murine KPC (KrasLSL-G12D/+; Trp53fl/fl; Ptf1aCre/+ ) PDAC cells in a three-dimensional co-culture platform with syngeneic antigen-educated T cells to identify potential cell-intrinsic drivers of T cell suppression in PDAC. Comparative transcriptomic analysis was performed to reveal promising candidates that mediate resistance to T cells. We investigated their contribution by shRNA-mediated knockdown and pharmacological inhibition in murine in vitro and in vivo studies, as well as in patient-derived organoids (PDOs). A combination of transcriptomic analyses, cytometric and immunohistochemistry techniques allowed us to validate the underlying T cell response phenotypes of PDAC cells. The action of TGM2 via interaction with tubulin and the impact of microtubule dynamics and vesicle trafficking were evaluated by protein analyses and live-cell imaging. Correlation analyses via TCGA data complemented the functional studies. RESULTS We identified transglutaminase 2 (TGM2) as a mediator of T cell suppression in PDAC. We report that high levels of TGM2 expression in patients' tumors correlate with immunosuppressive signatures and poor overall survival. We found that TGM2 regulates vesicle trafficking by modulating microtubule network density and dynamics in pancreatic cancer cells, thus facilitating the secretion of immunosuppressive cytokines, which impair effector T cell functionality. In TGM2-expressing PDOs, pharmacological TGM2 inhibition or treatment with nocodazole increased T cell-mediated apoptosis. Also, pretreatment of TGM2high PDOs with sublethal doses of the spindle poisons paclitaxel or vincristine increased CD8+T cell activation and sensitized PDOs toward T cell-mediated cytotoxicity. CONCLUSIONS These findings indicate that targeting microtubular function therapeutically may enhance antitumor T cell responses by impacting activity of immunosuppressive cytokines in the PDAC microenvironment.
Collapse
Affiliation(s)
- Anton Lahusen
- Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Nora Minhöfer
- Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | | | - Christine Blechner
- Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Tim Eiseler
- Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Anton Wellstein
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA
| | - Alexander Kleger
- Core Facility Organoids, Ulm University, Ulm, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
- Division of Interdisciplinary Pancreatology, Internal Medicine I, Ulm, Germany
| | | | - Sabine Windhorst
- Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yuan-Na Lin
- Internal Medicine I, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
4
|
Sun DY, Hu YJ, Li X, Peng J, Dai ZJ, Wang S. Unlocking the full potential of memory T cells in adoptive T cell therapy for hematologic malignancies. Int Immunopharmacol 2025; 144:113392. [PMID: 39608170 DOI: 10.1016/j.intimp.2024.113392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 11/30/2024]
Abstract
In recent years, immune cell therapy, particularly adoptive cell therapy (ACT), has shown superior therapeutic effects on hematologic malignancies. However, a challenge lies in ensuring that genetically engineered specific T cells maintain lasting anti-tumor effects within the host. The enduring success of ACT therapy hinges on the persistence of memory T (TM) cells, a diverse cell subset crucial for tumor immune response and immune memory upkeep. Notably, TM cell subsets at varying differentiation stages exhibit distinct biological traits and anti-tumor capabilities. Poorly differentiated TM cells are pivotal for favorable clinical outcomes in ACT. The differentiation of TM cells is influenced by multiple factors, including metabolism and cytokines. Consequently, current research focuses on investigating the differentiation patterns of TM cells and enhancing the production of poorly differentiated TM cells with potent anti-tumor properties in vitro, which is a prominent area of interest globally. This review delves into the differentiation features of TM cells, outlining their distribution in patients and their impact on ACT treatment. It comprehensively explores cutting-edge strategies to boost ACT efficacy through TM cell differentiation induction, aiming to unlock the full potential of TM cells in treating hematologic malignancies and offering novel insights for tumor immune cell therapy.
Collapse
Affiliation(s)
- Ding-Ya Sun
- Xiangya School of Pharmaceutical Sciences, Department of Pharmacology, Central South University, Changsha, China
| | - Yi-Jie Hu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Xin Li
- International Medicine Institute, Changsha Medical University, Changsha, China
| | - Jun Peng
- Xiangya School of Pharmaceutical Sciences, Department of Pharmacology, Central South University, Changsha, China.
| | - Zhi-Jie Dai
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Shan Wang
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| |
Collapse
|
5
|
Shah A, Ganguly K, Rauth S, Sheree SS, Khan I, Ganti AK, Ponnusamy MP, Kumar S, Jain M, Batra SK. Unveiling the resistance to therapies in pancreatic ductal adenocarcinoma. Drug Resist Updat 2024; 77:101146. [PMID: 39243602 PMCID: PMC11770815 DOI: 10.1016/j.drup.2024.101146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/09/2024]
Abstract
Despite the ongoing advances in interventional strategies (surgery, chemotherapy, radiotherapy, and immunotherapy) for managing pancreatic ductal adenocarcinoma (PDAC), the development of therapy refractory phenotypes remains a significant challenge. Resistance to various therapeutic modalities in PDAC emanates from a combination of inherent and acquired factors and is attributable to cancer cell-intrinsic and -extrinsic mechanisms. The critical determinants of therapy resistance include oncogenic signaling and epigenetic modifications that drive cancer cell stemness and metabolic adaptations, CAF-mediated stromagenesis that results in ECM deposition altered mechanotransduction, and secretome and immune evasion. We reviewed the current understanding of these multifaceted mechanisms operating in the PDAC microenvironment, influencing the response to chemotherapy, radiotherapy, and immunotherapy regimens. We then describe how the lessons learned from these studies can guide us to discover novel therapeutic regimens to prevent, delay, or revert resistance and achieve durable clinical responses.
Collapse
Affiliation(s)
- Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Koelina Ganguly
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Shamema S Sheree
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Imran Khan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Apar K Ganti
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Division of Oncology-hematology, Department of Internal Medicine, VA Nebraska Western Iowa Health Care System and University of Nebraska Medical Center, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha 68198-5870, USA
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha 68198-5870, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha 68198-5870, USA.
| |
Collapse
|
6
|
Zhang Z, Zhang W, Liu X, Yan Y, Fu W. T lymphocyte‑related immune response and immunotherapy in gastric cancer (Review). Oncol Lett 2024; 28:537. [PMID: 39319215 PMCID: PMC11421013 DOI: 10.3892/ol.2024.14670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024] Open
Abstract
Gastric cancer (GC) remains a global healthcare challenge because of its high incidence and poor prognosis. The efficacy of current chemotherapy regimens for advanced GC is limited. T cells, which have been implicated in the progression of GC, have a significant impact in the tumor microenvironment. With a more detailed understanding of the mechanisms underlying the cancer immunoediting process, immunotherapy may become a promising treatment option for patients with GC. Several clinical trials are currently investigating different mechanisms targeting the tumor immune response. The present review summarized T cell-involved immune responses and various immunotherapy strategies for GC.
Collapse
Affiliation(s)
- Zhaoxiong Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Wenxin Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Xin Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yongjia Yan
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Weihua Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
7
|
Salek M, Förster JD, Becker JP, Meyer M, Charoentong P, Lyu Y, Lindner K, Lotsch C, Volkmar M, Momburg F, Poschke I, Fröhling S, Schmitz M, Offringa R, Platten M, Jäger D, Zörnig I, Riemer AB. optiPRM: A Targeted Immunopeptidomics LC-MS Workflow With Ultra-High Sensitivity for the Detection of Mutation-Derived Tumor Neoepitopes From Limited Input Material. Mol Cell Proteomics 2024; 23:100825. [PMID: 39111711 PMCID: PMC11405902 DOI: 10.1016/j.mcpro.2024.100825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 09/08/2024] Open
Abstract
Personalized cancer immunotherapies such as therapeutic vaccines and adoptive transfer of T cell receptor-transgenic T cells rely on the presentation of tumor-specific peptides by human leukocyte antigen class I molecules to cytotoxic T cells. Such neoepitopes can for example arise from somatic mutations and their identification is crucial for the rational design of new therapeutic interventions. Liquid chromatography mass spectrometry (LC-MS)-based immunopeptidomics is the only method to directly prove actual peptide presentation and we have developed a parameter optimization workflow to tune targeted assays for maximum detection sensitivity on a per peptide basis, termed optiPRM. Optimization of collision energy using optiPRM allows for the improved detection of low abundant peptides that are very hard to detect using standard parameters. Applying this to immunopeptidomics, we detected a neoepitope in a patient-derived xenograft from as little as 2.5 × 106 cells input. Application of the workflow on small patient tumor samples allowed for the detection of five mutation-derived neoepitopes in three patients. One neoepitope was confirmed to be recognized by patient T cells. In conclusion, optiPRM, a targeted MS workflow reaching ultra-high sensitivity by per peptide parameter optimization, makes the identification of actionable neoepitopes possible from sample sizes usually available in the clinic.
Collapse
Affiliation(s)
- Mogjiborahman Salek
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Jonas D Förster
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Jonas P Becker
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Marten Meyer
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Department of Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Pornpimol Charoentong
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Heidelberg, Germany
| | - Yanhong Lyu
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Katharina Lindner
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), NCT Heidelberg, A Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Catharina Lotsch
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Michael Volkmar
- T Cell Discovery Platform, Helmholtz Institute for Translational Oncology (HI-TRON) Mainz - A Helmholtz Institute of the DKFZ, Mainz, Germany
| | - Frank Momburg
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Isabel Poschke
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), NCT Heidelberg, A Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Stefan Fröhling
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Division of Translational Medical Oncology, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, A Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), NCT Dresden, A PARTNership between DKFZ, University Hospital Carl Gustav Carus, Faculty of Medicine Carl Gustav Carus of TU Dresden and Helmholtz Center Dresden-Rossendorf, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, A Partnership Between DKFZ, University Hospital Carl Gustav Carus, Faculty of Medicine Carl Gustav Carus of TU Dresden, Helmholtz Center Dresden-Rossendorf and Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Dresden, Germany
| | - Rienk Offringa
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Platten
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), NCT Heidelberg, A Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience (MCTN), Heidelberg University, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Helmholtz Institute for Translational Oncology, Mainz (HI-TRON Mainz) - A Helmholtz Institute of the DKFZ, Mainz, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Angelika B Riemer
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany.
| |
Collapse
|
8
|
Merali N, Jessel MD, Arbe-Barnes EH, Ruby Lee WY, Gismondi M, Chouari T, O'Brien JW, Patel B, Osei-Bordom D, Rockall TA, Sivakumar S, Annels N, Frampton AE. Impact of tertiary lymphoid structures on prognosis and therapeutic response in pancreatic ductal adenocarcinoma. HPB (Oxford) 2024; 26:873-894. [PMID: 38729813 DOI: 10.1016/j.hpb.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/27/2024] [Accepted: 04/19/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is known to have a heterogeneous desmoplastic tumour microenvironment (TME) with a large number of immunosuppressive cells. Recently, high B-cell infiltration in PDAC has received growing interest as a potential therapeutic target. METHODS Our literature review summarises the characteristics of tumour-associated tertiary lymphoid structures (TLSs) and highlight the key studies exploring the clinical outcomes of TLSs in PDAC patients and the direct effect on the TME. RESULTS The location, density and maturity stages of TLSs within tumours play a key role in determining the prognosis and is a new emerging target in cancer immunotherapy. DISCUSSION TLS development is imperative to improve the prognosis of PDAC patients. In the future, studying the genetics and immune characteristics of tumour infiltrating B cells and TLSs may lead towards enhancing adaptive immunity in PDAC and designing personalised therapies.
Collapse
Affiliation(s)
- Nabeel Merali
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK; Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK; Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - Maria-Danae Jessel
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK
| | - Edward H Arbe-Barnes
- UCL Institute of Immunity and Transplantation, The Pears Building, Pond Street, London, UK
| | - Wing Yu Ruby Lee
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Martha Gismondi
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - Tarak Chouari
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK; Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - James W O'Brien
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - Bhavik Patel
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK; Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK; Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - Daniel Osei-Bordom
- Liver and Digestive Health, University College London, Royal Free Hospital, Pond St, London, UK
| | - Timothy A Rockall
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - Shivan Sivakumar
- Oncology Department and Institute of Immunology and Immunotherapy, Birmingham Medical School, University of Birmingham, Birmingham, UK
| | - Nicola Annels
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK
| | - Adam E Frampton
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK; Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK; Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK.
| |
Collapse
|
9
|
Morotti M, Grimm AJ, Hope HC, Arnaud M, Desbuisson M, Rayroux N, Barras D, Masid M, Murgues B, Chap BS, Ongaro M, Rota IA, Ronet C, Minasyan A, Chiffelle J, Lacher SB, Bobisse S, Murgues C, Ghisoni E, Ouchen K, Bou Mjahed R, Benedetti F, Abdellaoui N, Turrini R, Gannon PO, Zaman K, Mathevet P, Lelievre L, Crespo I, Conrad M, Verdeil G, Kandalaft LE, Dagher J, Corria-Osorio J, Doucey MA, Ho PC, Harari A, Vannini N, Böttcher JP, Dangaj Laniti D, Coukos G. PGE 2 inhibits TIL expansion by disrupting IL-2 signalling and mitochondrial function. Nature 2024; 629:426-434. [PMID: 38658764 PMCID: PMC11078736 DOI: 10.1038/s41586-024-07352-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024]
Abstract
Expansion of antigen-experienced CD8+ T cells is critical for the success of tumour-infiltrating lymphocyte (TIL)-adoptive cell therapy (ACT) in patients with cancer1. Interleukin-2 (IL-2) acts as a key regulator of CD8+ cytotoxic T lymphocyte functions by promoting expansion and cytotoxic capability2,3. Therefore, it is essential to comprehend mechanistic barriers to IL-2 sensing in the tumour microenvironment to implement strategies to reinvigorate IL-2 responsiveness and T cell antitumour responses. Here we report that prostaglandin E2 (PGE2), a known negative regulator of immune response in the tumour microenvironment4,5, is present at high concentrations in tumour tissue from patients and leads to impaired IL-2 sensing in human CD8+ TILs via the PGE2 receptors EP2 and EP4. Mechanistically, PGE2 inhibits IL-2 sensing in TILs by downregulating the IL-2Rγc chain, resulting in defective assembly of IL-2Rβ-IL2Rγc membrane dimers. This results in impaired IL-2-mTOR adaptation and PGC1α transcriptional repression, causing oxidative stress and ferroptotic cell death in tumour-reactive TILs. Inhibition of PGE2 signalling to EP2 and EP4 during TIL expansion for ACT resulted in increased IL-2 sensing, leading to enhanced proliferation of tumour-reactive TILs and enhanced tumour control once the cells were transferred in vivo. Our study reveals fundamental features that underlie impairment of human TILs mediated by PGE2 in the tumour microenvironment. These findings have therapeutic implications for cancer immunotherapy and cell therapy, and enable the development of targeted strategies to enhance IL-2 sensing and amplify the IL-2 response in TILs, thereby promoting the expansion of effector T cells with enhanced therapeutic potential.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Dinoprostone/metabolism
- Down-Regulation
- Ferroptosis
- Interleukin Receptor Common gamma Subunit/biosynthesis
- Interleukin Receptor Common gamma Subunit/deficiency
- Interleukin Receptor Common gamma Subunit/metabolism
- Interleukin-2/antagonists & inhibitors
- Interleukin-2/immunology
- Interleukin-2/metabolism
- Interleukin-2 Receptor beta Subunit/metabolism
- Lymphocytes, Tumor-Infiltrating/cytology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mitochondria/metabolism
- Oxidative Stress
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors
- Signal Transduction
- TOR Serine-Threonine Kinases/metabolism
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Matteo Morotti
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Alizee J Grimm
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Helen Carrasco Hope
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Marion Arnaud
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Mathieu Desbuisson
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Nicolas Rayroux
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - David Barras
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Maria Masid
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Baptiste Murgues
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Bovannak S Chap
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Marco Ongaro
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Ioanna A Rota
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Catherine Ronet
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Aspram Minasyan
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Johanna Chiffelle
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Sebastian B Lacher
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Sara Bobisse
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Clément Murgues
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Eleonora Ghisoni
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Khaoula Ouchen
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Ribal Bou Mjahed
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Fabrizio Benedetti
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Naoill Abdellaoui
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Riccardo Turrini
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Philippe O Gannon
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Khalil Zaman
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Patrice Mathevet
- Department of Gynaecology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Loic Lelievre
- Department of Gynaecology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Isaac Crespo
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Target and Therapeutics Centre, Helmholtz Munich, Neuherberg, Germany
| | - Gregory Verdeil
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Lana E Kandalaft
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Julien Dagher
- Unit of Translational Oncopathology, Institute of Pathology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Jesus Corria-Osorio
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Marie-Agnes Doucey
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Ping-Chih Ho
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Alexandre Harari
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Nicola Vannini
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Denarda Dangaj Laniti
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland.
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
| | - George Coukos
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland.
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
| |
Collapse
|
10
|
Rupp L, Dietsche I, Kießler M, Sommer U, Muckenhuber A, Steiger K, van Eijck CWF, Richter L, Istvanffy R, Jäger C, Friess H, van Eijck CHJ, Demir IE, Reyes CM, Schmitz M. Neoadjuvant chemotherapy is associated with suppression of the B cell-centered immune landscape in pancreatic ductal adenocarcinoma. Front Immunol 2024; 15:1378190. [PMID: 38629072 PMCID: PMC11018975 DOI: 10.3389/fimmu.2024.1378190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is typically diagnosed at advanced stages and associated with early distant metastasis and poor survival. Besides clinical factors, the tumor microenvironment (TME) emerged as a crucial determinant of patient survival and therapy response in many tumors, including PDAC. Thus, the presence of tumor-infiltrating lymphocytes and the formation of tertiary lymphoid structures (TLS) is associated with longer survival in PDAC. Although neoadjuvant therapy (NeoTx) has improved the management of locally advanced tumors, detailed insight into its effect on various TME components is limited. While a remodeling towards a proinflammatory state was reported for PDAC-infiltrating T cells, the effect of NeoTx on B cell subsets, including plasma cells, and TLS formation is widely unclear. We thus investigated the frequency, composition, and spatial distribution of PDAC-infiltrating B cells in primary resected (PR) versus neoadjuvant-treated patients using a novel multiplex immunohistochemistry panel. The NeoTx group displayed significantly lower frequencies of pan B cells, GC B cells, plasmablasts, and plasma cells, accompanied by a reduced abundance of TLS. This finding was supported by bulk RNA-sequencing analysis of an independent fresh frozen tissue cohort, which revealed that major B cell pathways were downregulated in the NeoTx group. We further observed that plasma cells frequently formed aggregates that localized close to TLS and that TLS+ patients displayed significantly higher plasma cell frequencies compared to TLS- patients in the PR group. Additionally, high densities of CD20+ intratumoral B cells were significantly associated with longer overall survival in the PR group. While CD20+ B cells held no prognostic value for NeoTx patients, an increased frequency of proliferating CD20+Ki67+ B cells emerged as an independent prognostic factor for longer survival in the NeoTx group. These results indicate that NeoTx differentially affects PDAC-infiltrating immune cells and may have detrimental effects on the existing B cell landscape and the formation of TLS. Gaining further insight into the underlying molecular mechanisms is crucial to overcome the intrinsic immunotherapy resistance of PDAC and develop novel strategies to improve the long-term outcome of PDAC patients.
Collapse
Affiliation(s)
- Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Ina Dietsche
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Maximilian Kießler
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Ulrich Sommer
- Institute of Pathology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Alexander Muckenhuber
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Casper W. F. van Eijck
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Leonard Richter
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Rouzanna Istvanffy
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Carsten Jäger
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Casper H. J. van Eijck
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of General Surgery, Hepato-Pancreato-Biliary (HPB) Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Türkiye
- Else Kröner Clinician Scientist Professor for Translational Pancreatic Surgery, Technical University of Munich, Munich, Germany
| | - Carmen Mota Reyes
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Pothuri VS, Hogg GD, Conant L, Borcherding N, James CA, Mudd J, Williams G, Seo YD, Hawkins WG, Pillarisetty VG, DeNardo DG, Fields RC. Intratumoral T-cell receptor repertoire composition predicts overall survival in patients with pancreatic ductal adenocarcinoma. Oncoimmunology 2024; 13:2320411. [PMID: 38504847 PMCID: PMC10950267 DOI: 10.1080/2162402x.2024.2320411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy that is refractory to immune checkpoint inhibitor therapy. However, intratumoral T-cell infiltration correlates with improved overall survival (OS). Herein, we characterized the diversity and antigen specificity of the PDAC T-cell receptor (TCR) repertoire to identify novel immune-relevant biomarkers. Demographic, clinical, and TCR-beta sequencing data were collated from 353 patients across three cohorts that underwent surgical resection for PDAC. TCR diversity was calculated using Shannon Wiener index, Inverse Simpson index, and "True entropy." Patients were clustered by shared repertoire specificity. TCRs predictive of OS were identified and their associated transcriptional states were characterized by single-cell RNAseq. In multivariate Cox regression models controlling for relevant covariates, high intratumoral TCR diversity predicted OS across multiple cohorts. Conversely, in peripheral blood, high abundance of T-cells, but not high diversity, predicted OS. Clustering patients based on TCR specificity revealed a subset of TCRs that predicts OS. Interestingly, these TCR sequences were more likely to encode CD8+ effector memory and CD4+ T-regulatory (Tregs) T-cells, all with the capacity to recognize beta islet-derived autoantigens. As opposed to T-cell abundance, intratumoral TCR diversity was predictive of OS in multiple PDAC cohorts, and a subset of TCRs enriched in high-diversity patients independently correlated with OS. These findings emphasize the importance of evaluating peripheral and intratumoral TCR repertoires as distinct and relevant biomarkers in PDAC.
Collapse
Affiliation(s)
- Vikram S. Pothuri
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Graham D. Hogg
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Leah Conant
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas Borcherding
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - C. Alston James
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacqueline Mudd
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Greg Williams
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Yongwoo David Seo
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, USA
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - William G. Hawkins
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MOUSA
| | - Venu G. Pillarisetty
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WAUSA
| | - David G. DeNardo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MOUSA
| | - Ryan C. Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MOUSA
| |
Collapse
|
12
|
Wang S, Wang H, Li C, Liu B, He S, Tu C. Tertiary lymphoid structures in cancer: immune mechanisms and clinical implications. MedComm (Beijing) 2024; 5:e489. [PMID: 38469550 PMCID: PMC10925885 DOI: 10.1002/mco2.489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 03/13/2024] Open
Abstract
Cancer is a major cause of death globally, and traditional treatments often have limited efficacy and adverse effects. Immunotherapy has shown promise in various malignancies but is less effective in tumors with low immunogenicity or immunosuppressive microenvironment, especially sarcomas. Tertiary lymphoid structures (TLSs) have been associated with a favorable response to immunotherapy and improved survival in cancer patients. However, the immunological mechanisms and clinical significance of TLS in malignant tumors are not fully understood. In this review, we elucidate the composition, neogenesis, and immune characteristics of TLS in tumors, as well as the inflammatory response in cancer development. An in-depth discussion of the unique immune characteristics of TLSs in lung cancer, breast cancer, melanoma, and soft tissue sarcomas will be presented. Additionally, the therapeutic implications of TLS, including its role as a marker of therapeutic response and prognosis, and strategies to promote TLS formation and maturation will be explored. Overall, we aim to provide a comprehensive understanding of the role of TLS in the tumor immune microenvironment and suggest potential interventions for cancer treatment.
Collapse
Affiliation(s)
- Siyu Wang
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Xiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Hua Wang
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Chenbei Li
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Binfeng Liu
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Shasha He
- Department of OncologyThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Chao Tu
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Shenzhen Research Institute of Central South UniversityGuangdongChina
- Changsha Medical UniversityChangshaChina
| |
Collapse
|
13
|
Zhang H, Xu W, Zhu H, Chen X, Tsai HI. Overcoming the limitations of immunotherapy in pancreatic ductal adenocarcinoma: Combining radiotherapy and metabolic targeting therapy. J Cancer 2024; 15:2003-2023. [PMID: 38434964 PMCID: PMC10905401 DOI: 10.7150/jca.92502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/20/2024] [Indexed: 03/05/2024] Open
Abstract
As a novel anticancer therapy, immunotherapy has demonstrated robust efficacy against a few solid tumors but poor efficacy against pancreatic ductal adenocarcinoma (PDAC). This poor outcome is primarily attributable to the intrinsic cancer cell resistance and T-cell exhaustion, which is also the reason for the failure of conventional therapy. The present review summarizes the current PDAC immunotherapy avenues and the underlying resistance mechanisms. Then, the review discusses synergistic combination therapies, such as radiotherapy (RT) and metabolic targeting. Research suggests that RT boosts the antigen of PDAC, which facilitates the anti-tumor immune cell infiltration and exerts function. Metabolic reprogramming contributes to restoring the exhausted T cell function. The current review will help in tailoring combination regimens to enhance the efficacy of immunotherapy. In addition, it will help provide new approaches to address the limitations of the immunosuppressive tumor microenvironment (TME) by examining the relationship among immunotherapy, RT, and metabolism targeting therapy in PDAC.
Collapse
Affiliation(s)
- Han Zhang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
| | - Wenjin Xu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xuelian Chen
- Department of Radiology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| | - Hsiang-I Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
14
|
Gao Z, Azar J, Zhu H, Williams-Perez S, Kang SW, Marginean C, Rubinstein MP, Makawita S, Lee HS, Camp ER. Translational and oncologic significance of tertiary lymphoid structures in pancreatic adenocarcinoma. Front Immunol 2024; 15:1324093. [PMID: 38361928 PMCID: PMC10867206 DOI: 10.3389/fimmu.2024.1324093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/11/2024] [Indexed: 02/17/2024] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is an aggressive tumor with poor survival and limited treatment options. PDAC resistance to immunotherapeutic strategies is multifactorial, but partially owed to an immunosuppressive tumor immune microenvironment (TiME). However, the PDAC TiME is heterogeneous and harbors favorable tumor-infiltrating lymphocyte (TIL) populations. Tertiary lymphoid structures (TLS) are organized aggregates of immune cells that develop within non-lymphoid tissue under chronic inflammation in multiple contexts, including cancers. Our current understanding of their role within the PDAC TiME remains limited; TLS are complex structures with multiple anatomic features such as location, density, and maturity that may impact clinical outcomes such as survival and therapy response in PDAC. Similarly, our understanding of methods to manipulate TLS is an actively developing field of research. TLS may function as anti-tumoral immune niches that can be leveraged as a therapeutic strategy to potentiate both existing chemotherapeutic regimens and potentiate future immune-based therapeutic strategies to improve patient outcomes. This review seeks to cover anatomy, relevant features, immune effects, translational significance, and future directions of understanding TLS within the context of PDAC.
Collapse
Affiliation(s)
- Zachary Gao
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Joseph Azar
- The Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Huili Zhu
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Sophia Williams-Perez
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Sung Wook Kang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Celia Marginean
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Mark P. Rubinstein
- The Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Shalini Makawita
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Hyun-Sung Lee
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - E. Ramsay Camp
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX, United States
| |
Collapse
|
15
|
Luo B, Wang P, Tian J, Chu X, Lu X, Yang Y, Zhou Y, Li Y, Que Z. Jinfukang inhibits lung cancer metastasis by regulating T cell receptors. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116885. [PMID: 37422099 DOI: 10.1016/j.jep.2023.116885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/27/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Metastasis is the leading cause of death in lung cancer worldwide, and immune escape plays a vital role in the process of metastasis. Clinical studies have proven that Jinfukang (JFK) can effectively treat lung cancer metastasis by regulating T lymphocytes. However, it is still unknown whether JFK plays a role in treating lung cancer metastasis by regulating T-cell receptors (TCRs). AIM OF THE STUDY To explore the effect of JFK in inhibiting lung cancer metastasis by regulating TCR. MATERIALS AND METHODS A lung metastasis model was established in C57BL/6J and BALB/c-nude mice by tail vein injection of Lewis lung cancer cells. JFK was given by continuous intragastric administration. Anatomical observation combined with hematoxylin-eosin staining was used to evaluate lung metastasis. T cells, MDSCs, and macrophages in the peripheral blood were detected by flow cytometry, and the proliferation and immune cell infiltration of lung metastases were observed by immunohistochemistry and immunofluorescence. The diversity and gene expression of TCR in peripheral blood and lung tissues were detected by immune repertoire sequencing, and bioinformatics analysis was carried out. RESULTS Compared with the control group, the number of pulmonary metastatic nodules in JFK-treated mice showed a decreasing trend, and it significantly reduced the burden of lung tumor metastasis in mice. We found that the expression level of Ki-67 protein in lung metastatic tumor tissues of mice treated with JFK was significantly reduced, while the infiltration level of CD8+ T lymphocytes and NK cells was significantly increased. In addition, we also found that JFK could significantly increase the proportion of CD4+ T, CD8+ T and NKT cells in the peripheral blood of mice. Moreover, JFK reduced the ratio of M-MDSCs and increased the ratio of PMN-MDSCs in the peripheral blood of mice. JFK increased the ratio of M1 macrophages in the peripheral blood of Lewis tumor-bearing mice. The sequencing of TCR in the peripheral blood and lung tissue of mice indicated that there was no notable difference in TCR diversity as the tumor progressed and JFK treatment was administered. However, the downregulation of TRBV16, TRBV17, TRBV1 and the upregulation of the TRBV12-2 gene in the TCR caused by tumor progression can be reversed by JFK. CONCLUSION These results suggest that JFK may upregulate the proportion of CD4+ T, CD8+ T and NKT cells in peripheral blood, reverse the TCR changes caused by tumor metastasis, and promote the infiltration of CD8+ T and NK cells in tumor tissues, thereby inhibiting the growth of tumors and ultimately reducing the burden of lung cancer metastasis. This will provide new strategies for developing Chinese herbal medicine to treat metastasis by regulating TCR.
Collapse
Affiliation(s)
- Bin Luo
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, China; Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
| | - Panpan Wang
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, China.
| | - Jianhui Tian
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, China.
| | - Xiaoge Chu
- Department of Hematological Oncology, Shanghai Jing'an District Beizhan Hospital, 200070, Shanghai, China.
| | - Xinyi Lu
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, China.
| | - Yun Yang
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, China.
| | - Yiyang Zhou
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, China.
| | - Yan Li
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, China.
| | - Zujun Que
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, China.
| |
Collapse
|
16
|
Meng Z, Rodriguez Ehrenfried A, Tan CL, Steffens LK, Kehm H, Zens S, Lauenstein C, Paul A, Schwab M, Förster JD, Salek M, Riemer AB, Wu H, Eckert C, Leonhardt CS, Strobel O, Volkmar M, Poschke I, Offringa R. Transcriptome-based identification of tumor-reactive and bystander CD8 + T cell receptor clonotypes in human pancreatic cancer. Sci Transl Med 2023; 15:eadh9562. [PMID: 37967201 DOI: 10.1126/scitranslmed.adh9562] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/16/2023] [Indexed: 11/17/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is generally refractory to immune checkpoint blockade, although patients with genetically unstable tumors can show modest therapeutic benefit. We previously demonstrated the presence of tumor-reactive CD8+ T cells in PDAC samples. Here, we charted the tumor-infiltrating T cell repertoire in PDAC by combining single-cell transcriptomics with functional testing of T cell receptors (TCRs) for reactivity against autologous tumor cells. On the basis of a comprehensive dataset including 93 tumor-reactive and 65 bystander TCR clonotypes, we delineated a gene signature that effectively distinguishes between these T cell subsets in PDAC, as well as in other tumor indications. This revealed a high frequency of tumor-reactive TCR clonotypes in three genetically unstable samples. In contrast, the T cell repertoire in six genetically stable PDAC tumors was largely dominated by bystander T cells. Nevertheless, multiple tumor-reactive TCRs were successfully identified in each of these samples, thereby providing a perspective for personalized immunotherapy in this treatment-resistant indication.
Collapse
Affiliation(s)
- Zibo Meng
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Aaron Rodriguez Ehrenfried
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Helmholtz-Institute for Translational Oncology by DKFZ (HI-TRON), 55131 Mainz, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Chin Leng Tan
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Laura K Steffens
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Hannes Kehm
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Stefan Zens
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Claudia Lauenstein
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Alina Paul
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Marius Schwab
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Jonas D Förster
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Division of Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), partner site Heidelberg, 69120 Heidelberg, Germany
| | - Mogjiborahman Salek
- Division of Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), partner site Heidelberg, 69120 Heidelberg, Germany
| | - Angelika B Riemer
- Division of Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), partner site Heidelberg, 69120 Heidelberg, Germany
| | - Heshui Wu
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Christoph Eckert
- Pathology Institute, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Carl-Stephan Leonhardt
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Oliver Strobel
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Michael Volkmar
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Helmholtz-Institute for Translational Oncology by DKFZ (HI-TRON), 55131 Mainz, Germany
| | - Isabel Poschke
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Rienk Offringa
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| |
Collapse
|
17
|
James CA, Baer JM, Zou C, Panni UY, Knolhoff BL, Hogg GD, Kingston NL, Kang LI, Lander VE, Luo J, Tao Y, Watson MA, Aft R, Fields RC, Hawkins WG, DeNardo DG. Systemic Alterations in Type-2 Conventional Dendritic Cells Lead to Impaired Tumor Immunity in Pancreatic Cancer. Cancer Immunol Res 2023; 11:1055-1067. [PMID: 37229629 PMCID: PMC10524961 DOI: 10.1158/2326-6066.cir-21-0946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 10/04/2022] [Accepted: 05/23/2023] [Indexed: 05/27/2023]
Abstract
Intratumoral T-cell dysfunction is a hallmark of pancreatic tumors, and efforts to improve dendritic cell (DC)-mediated T-cell activation may be critical in treating these immune therapy unresponsive tumors. Recent evidence indicates that mechanisms that induce dysfunction of type 1 conventional DCs (cDC1) in pancreatic adenocarcinomas (PDAC) are drivers of the lack of responsiveness to checkpoint immunotherapy. However, the impact of PDAC on systemic type 2 cDC2 development and function has not been well studied. Herein, we report the analysis of 3 cohorts, totaling 106 samples, of human blood and bone marrow (BM) from patients with PDAC for changes in cDCs. We found that circulating cDC2s and their progenitors were significantly decreased in the blood of patients with PDAC, and repressed numbers of cDC2s were associated with poor prognosis. Serum cytokine analyses identified IL6 as significantly elevated in patients with PDAC and negatively correlated with cDC numbers. In vitro, IL6 impaired the differentiation of cDC1s and cDC2s from BM progenitors. Single-cell RNA sequencing analysis of human cDC progenitors in the BM and blood of patients with PDAC showed an upregulation of the IL6/STAT3 pathway and a corresponding impairment of antigen processing and presentation. These results suggested that cDC2s were systemically suppressed by inflammatory cytokines, which was linked to impaired antitumor immunity.
Collapse
Affiliation(s)
- C. Alston James
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John M. Baer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chong Zou
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Usman Y. Panni
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brett L. Knolhoff
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Graham D. Hogg
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Natalie L Kingston
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Liang-I Kang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Varintra E. Lander
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jingqin Luo
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yu Tao
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mark A. Watson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rebecca Aft
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ryan C. Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - William G. Hawkins
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David G. DeNardo
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
18
|
Zou X, Guan C, Gao J, Shi W, Cui Y, Zhong X. Tertiary lymphoid structures in pancreatic cancer: a new target for immunotherapy. Front Immunol 2023; 14:1222719. [PMID: 37529035 PMCID: PMC10388371 DOI: 10.3389/fimmu.2023.1222719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/26/2023] [Indexed: 08/03/2023] Open
Abstract
Pancreatic cancer (PC) is extremely malignant and shows limited response to available immunotherapies due to the hypoxic and immunosuppressive nature of its tumor microenvironment (TME). The aggregation of immune cells (B cells, T cells, dendritic cells, etc.), which is induced in various chronic inflammatory settings such as infection, inflammation, and tumors, is known as the tertiary lymphoid structure (TLS). Several studies have shown that TLSs can be found in both intra- and peritumor tissues of PC. The role of TLSs in peritumor tissues in tumors remains unclear, though intratumoral TLSs are known to play an active role in a variety of tumors, including PC. The formation of intratumoral TLSs in PC is associated with a good prognosis. In addition, TLSs can be used as an indicator to assess the effectiveness of treatment. Targeted induction of TLS formation may become a new avenue of immunotherapy for PC. This review summarizes the formation, characteristics, relevant clinical outcomes, and clinical applications of TLSs in the pancreatic TME. We aim to provide new ideas for future immunotherapy of PC.
Collapse
Affiliation(s)
- Xinlei Zou
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Canghai Guan
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianjun Gao
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wujiang Shi
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunfu Cui
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangyu Zhong
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
Sato Y, Silina K, van den Broek M, Hirahara K, Yanagita M. The roles of tertiary lymphoid structures in chronic diseases. Nat Rev Nephrol 2023:10.1038/s41581-023-00706-z. [PMID: 37046081 PMCID: PMC10092939 DOI: 10.1038/s41581-023-00706-z] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 04/14/2023]
Abstract
Tertiary lymphoid structures (TLSs) are ectopic lymphoid tissues that drive antigen-specific immune responses at sites of chronic inflammation. Unlike secondary lymphoid organs such as lymph nodes, TLSs lack capsules and have their own unique characteristics and functions. The presumed influence of TLSs on the disease course has led to widespread interest in obtaining a better understanding of their biology and function. Studies using single-cell analyses have suggested heterogeneity in TLS composition and phenotype, and consequently, functional correlates with disease progression are sometimes conflicting. The presence of TLSs correlates with a favourable disease course in cancer and infection. Conversely, in autoimmune diseases and chronic age-related inflammatory diseases including chronic kidney disease, the presence of TLSs is associated with a more severe disease course. However, the detailed mechanisms that underlie these clinical associations are not fully understood. To what extent the mechanisms of TLS development and maturation are shared across organs and diseases is also still obscure. Improved understanding of TLS development and function at the cellular and molecular levels may enable the exploitation of these structures to improve therapies for chronic diseases, including chronic kidney disease.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Karina Silina
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | | | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.
| |
Collapse
|
20
|
Pearce H, Croft W, Nicol SM, Margielewska-Davies S, Powell R, Cornall R, Davis SJ, Marcon F, Pugh MR, Fennell É, Powell-Brett S, Mahon BS, Brown RM, Middleton G, Roberts K, Moss P. Tissue-Resident Memory T Cells in Pancreatic Ductal Adenocarcinoma Coexpress PD-1 and TIGIT and Functional Inhibition Is Reversible by Dual Antibody Blockade. Cancer Immunol Res 2023; 11:435-449. [PMID: 36689623 PMCID: PMC10068448 DOI: 10.1158/2326-6066.cir-22-0121] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/02/2022] [Accepted: 01/19/2023] [Indexed: 01/24/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor clinical outlook. Responses to immune checkpoint blockade are suboptimal and a much more detailed understanding of the tumor immune microenvironment is needed if this situation is to be improved. Here, we characterized tumor-infiltrating T-cell populations in patients with PDAC using cytometry by time of flight (CyTOF) and single-cell RNA sequencing. T cells were the predominant immune cell subset observed within tumors. Over 30% of CD4+ T cells expressed a CCR6+CD161+ Th17 phenotype and 17% displayed an activated regulatory T-cell profile. Large populations of CD8+ tissue-resident memory (TRM) T cells were also present and expressed high levels of programmed cell death protein 1 (PD-1) and TIGIT. A population of putative tumor-reactive CD103+CD39+ T cells was also observed within the CD8+ tumor-infiltrating lymphocytes population. The expression of PD-1 ligands was limited largely to hemopoietic cells whilst TIGIT ligands were expressed widely within the tumor microenvironment. Programmed death-ligand 1 and CD155 were expressed within the T-cell area of ectopic lymphoid structures and colocalized with PD-1+TIGIT+ CD8+ T cells. Combinatorial anti-PD-1 and TIGIT blockade enhanced IFNγ secretion and proliferation of T cells in the presence of PD-1 and TIGIT ligands. As such, we showed that the PDAC microenvironment is characterized by the presence of substantial populations of TRM cells with an exhausted PD-1+TIGIT+ phenotype where dual checkpoint receptor blockade represents a promising avenue for future immunotherapy.
Collapse
Affiliation(s)
- Hayden Pearce
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Wayne Croft
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre for Computational Biology, University of Birmingham, Birmingham, United Kingdom
| | - Samantha M. Nicol
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sandra Margielewska-Davies
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Richard Powell
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Richard Cornall
- Nuffield Department of Medicine and Medical Research Council Human Immunology Unit, University of Oxford, Oxford, United Kingdom
| | - Simon J. Davis
- Radcliffe Department of Medicine and Medical Research Council Human Immunology Unit, University of Oxford, Oxford, United Kingdom
| | - Francesca Marcon
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Matthew R. Pugh
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Éanna Fennell
- Health Research Institute, Bernal Institute and School of Medicine, University of Limerick, Limerick, Ireland
| | - Sarah Powell-Brett
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Brinder S. Mahon
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Rachel M. Brown
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Gary Middleton
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Keith Roberts
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Paul Moss
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| |
Collapse
|
21
|
Golan T, Raitses-Gurevich M, Beller T, Carroll J, Brody JR. Strategies for the Management of Patients with Pancreatic Cancer with PARP Inhibitors. Cancer Treat Res 2023; 186:125-142. [PMID: 37978134 DOI: 10.1007/978-3-031-30065-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
A subset of patients with pancreatic adenocarcinomas (PDAC) harbor mutations that are exploitable in the context of DNA-damage response and repair (DDR) inhibitory strategies. Between 8-18% of PDACs harbor specific mutations in the DDR pathway such as BRCA1/2 mutations, and a higher prevalence exists in high-risk populations (e.g., Ashkenazi Jews). Herein, we will review the current trials and data on the treatment of PDAC patients who harbor such mutations and who appear sensitive to platinum and/or poly ADP ribose polymerase inhibitor (PARPi) based therapies due to a concept known as synthetic lethality. Although this current best-in-class precision treatment shows clinical promise, the specter of resistance limits the extent of therapeutic responses. We therefore also evaluate promising pre-clinical and clinical approaches in the pipeline that may either work with existing therapies to break resistance or work separately with combination therapies against this subset of PDACs.
Collapse
Affiliation(s)
- Talia Golan
- Cancer Center, Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Maria Raitses-Gurevich
- Cancer Center, Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Beller
- Cancer Center, Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - James Carroll
- Department of Surgery, Brenden Colson Center for Pancreatic Care, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Jonathan R Brody
- Department of Surgery, Brenden Colson Center for Pancreatic Care, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
22
|
Schalck A, Sakellariou-Thompson D, Forget MA, Sei E, Hughes TG, Reuben A, Bai S, Hu M, Kumar T, Hurd MW, Katz MH, Tzeng CWD, Pant S, Javle M, Fogelman DR, Maitra A, Haymaker CL, Kim MP, Navin NE, Bernatchez C. Single-Cell Sequencing Reveals Trajectory of Tumor-Infiltrating Lymphocyte States in Pancreatic Cancer. Cancer Discov 2022; 12:2330-2349. [PMID: 35849783 PMCID: PMC9547957 DOI: 10.1158/2159-8290.cd-21-1248] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 05/09/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has few effective treatments. Immunotherapy, an attractive alternative strategy, remains challenging with the lack of knowledge on the tumor-infiltrating lymphocyte (TIL) landscape in PDAC. To generate a reference of T-cell subpopulations, we profiled 80,000 T cells from 57 PDAC samples, 22 uninvolved/normal samples, and cultured TIL using single-cell transcriptomic and T-cell receptor analysis. These data revealed 20 cell states and heterogeneous distributions of TIL populations. The CD8+ TIL contained a putative transitional GZMK+ population based on T-cell receptor clonotype sharing, and cell-state trajectory analysis showed similarity to a GZMB+PRF1+ cytotoxic and a CXCL13+ dysfunctional population. Statistical analysis suggested that certain TIL states, such as dysfunctional and inhibitory populations, often occurred together. Finally, analysis of cultured TIL revealed that high-frequency clones from effector populations were preferentially expanded. These data provide a framework for understanding the PDAC TIL landscape for future TIL use in immunotherapy for PDAC. SIGNIFICANCE To improve the efficacy of immunotherapy in PDAC, there is a great need to understand the PDAC TIL landscape. This study represents a reference of PDAC TIL subpopulations and their relationships and provides a foundation upon which to base future immunotherapeutic efforts. This article is highlighted in the In This Issue feature, p. 2221.
Collapse
Affiliation(s)
- Aislyn Schalck
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Donastas Sakellariou-Thompson
- Department of Biologics Development, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marie-Andrée Forget
- Department of Biologics Development, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emi Sei
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tara G. Hughes
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shanshan Bai
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Min Hu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapsi Kumar
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Mark W. Hurd
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew H.G. Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ching-Wei D. Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Milind Javle
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David R. Fogelman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- The MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cara L. Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael P. Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas E. Navin
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chantale Bernatchez
- Department of Biologics Development, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
23
|
Targeting the Metabolic Rewiring in Pancreatic Cancer and Its Tumor Microenvironment. Cancers (Basel) 2022; 14:cancers14184351. [PMID: 36139512 PMCID: PMC9497173 DOI: 10.3390/cancers14184351] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/20/2022] [Accepted: 09/03/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with only a few effective therapeutic options. A characteristic feature of PDAC is its unique tumor microenvironment (TME), termed desmoplasia, which shows extensive fibrosis and extracellular matrix deposition, generating highly hypoxic and nutrient-deprived conditions within the tumor. To thrive in this harsh TME, PDAC undergoes extensive metabolic rewiring that includes the altered use of glucose and glutamine, constitutive activation of autophagy-lysosomal pathways, and nutrient acquisition from host cells in the TME. Notably, these properties support PDAC metabolism and mediate therapeutic resistance, including immune suppression. A deeper understanding of the unique metabolic properties of PDAC and its TME may aid in the development of novel therapeutic strategies against this deadly disease.
Collapse
|
24
|
Yamamoto K, Iwadate D, Kato H, Nakai Y, Tateishi K, Fujishiro M. Targeting autophagy as a therapeutic strategy against pancreatic cancer. J Gastroenterol 2022; 57:603-618. [PMID: 35727403 PMCID: PMC9392712 DOI: 10.1007/s00535-022-01889-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/28/2022] [Indexed: 02/07/2023]
Abstract
Macroautophagy (hereafter autophagy) is a catabolic process through which cytosolic components are captured in the autophagosome and degraded in the lysosome. Autophagy plays two major roles: nutrient recycling under starvation or stress conditions and maintenance of cellular homeostasis by removing the damaged organelles or protein aggregates. In established cancer cells, autophagy-mediated nutrient recycling promotes tumor progression, whereas in normal/premalignant cells, autophagy suppresses tumor initiation by eliminating the oncogenic/harmful molecules. Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease that is refractory to most currently available treatment modalities, including immune checkpoint blockade and molecular-targeted therapy. One prominent feature of PDAC is its constitutively active and elevated autophagy-lysosome function, which enables PDAC to thrive in its nutrient-scarce tumor microenvironment. In addition to metabolic support, autophagy promotes PDAC progression in a metabolism-independent manner by conferring resistance to therapeutic treatment or facilitating immune evasion. Besides to cell-autonomous autophagy in cancer cells, host autophagy (autophagy in non-cancer cells) supports PDAC progression, further highlighting autophagy as a promising therapeutic target in PDAC. Based on a growing list of compelling preclinical evidence, there are numerous ongoing clinical trials targeting the autophagy-lysosome pathway in PDAC. Given the multifaceted and context-dependent roles of autophagy in both cancer cells and normal host cells, a deeper understanding of the mechanisms underlying the tumor-promoting roles of autophagy as well as of the consequences of autophagy inhibition is necessary for the development of autophagy inhibition-based therapies against PDAC.
Collapse
Affiliation(s)
- Keisuke Yamamoto
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Dosuke Iwadate
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroyuki Kato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yousuke Nakai
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Keisuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
25
|
Skorupan N, Palestino Dominguez M, Ricci SL, Alewine C. Clinical Strategies Targeting the Tumor Microenvironment of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:4209. [PMID: 36077755 PMCID: PMC9454553 DOI: 10.3390/cancers14174209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 12/04/2022] Open
Abstract
Pancreatic cancer has a complex tumor microenvironment which engages in extensive crosstalk between cancer cells, cancer-associated fibroblasts, and immune cells. Many of these interactions contribute to tumor resistance to anti-cancer therapies. Here, new therapeutic strategies designed to modulate the cancer-associated fibroblast and immune compartments of pancreatic ductal adenocarcinomas are described and clinical trials of novel therapeutics are discussed. Continued advances in our understanding of the pancreatic cancer tumor microenvironment are generating stromal and immune-modulating therapeutics that may improve patient responses to anti-tumor treatment.
Collapse
Affiliation(s)
- Nebojsa Skorupan
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Medical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mayrel Palestino Dominguez
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samuel L. Ricci
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christine Alewine
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
26
|
Ullman NA, Burchard PR, Dunne RF, Linehan DC. Immunologic Strategies in Pancreatic Cancer: Making Cold Tumors Hot. J Clin Oncol 2022; 40:2789-2805. [PMID: 35839445 PMCID: PMC9390820 DOI: 10.1200/jco.21.02616] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/08/2022] [Accepted: 05/18/2022] [Indexed: 12/12/2022] Open
Abstract
The rising incidence and persistent dismal 5-year overall survival of pancreatic ductal adenocarcinoma (PDAC) highlight the need for new effective systemic therapies. Immunotherapy has shown significant benefits in solid organ tumors, but has thus far been disappointing in the treatment of PDAC. There have been several promising preclinical studies, but translation into the clinic has proved to be challenging. This is likely a result of PDAC's complex immunosuppressive tumor microenvironment that acts to insulate the tumor against an effective cytotoxic immune response. Here, we summarize the mechanisms of immunosuppression within the PDAC tumor microenvironment and provide an up-to-date review of completed and ongoing clinical trials using various immunotherapy strategies.
Collapse
|
27
|
Gorchs L, Oosthoek M, Yucel-Lindberg T, Moro CF, Kaipe H. Chemokine Receptor Expression on T Cells Is Modulated by CAFs and Chemokines Affect the Spatial Distribution of T Cells in Pancreatic Tumors. Cancers (Basel) 2022; 14:cancers14153826. [PMID: 35954489 PMCID: PMC9367555 DOI: 10.3390/cancers14153826] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/21/2022] [Accepted: 08/03/2022] [Indexed: 01/18/2023] Open
Abstract
Simple Summary The infiltration of T cells in pancreatic tumors has been correlated with better overall survival. However, the dense desmoplastic stroma, mainly composed by cancer-associated fibroblasts (CAFs), can sequester the T cells in the stroma preventing them from reaching the tumor nests. Chemokines are small molecules capable of directing T cell migration. Here, we explored whether CAFs could modulate the expression of chemokine receptors on T cells and examined if the spatial distribution of T cells within tumors was correlated to chemokine secretion patterns. Overall, we found that CXCR3 ligands was associated with an increased number of T cells in tumor rich areas and that CAFs downregulated the expression of CXCR3 on T cells. Understanding the mechanisms by which T cells are prevented from reaching the tumor nests is of great importance for the development of novel targeting therapies. Abstract The accumulation of T cells is associated with a better prognosis in pancreatic cancer. However, the immunosuppressive tumor microenvironment, largely composed by cancer-associated fibroblasts (CAFs), can prevent T cells from reaching the tumor nests. We examined how human CAFs modulated chemokine receptors known to be associated with T cell trafficking, CXCR3 and CCR5, and T cell exclusion, CXCR4. CAFs decreased the expression of CXCR3 and CCR5 but increased CXCR4 expression in both 2D and 3D cultures, affecting the migratory capacity of T cells towards CXCL10. An immunohistochemistry analysis showed that very few T cells were found in the tumor nests. Within the stroma, CD8+ T cells were localized more distantly from the malignant cells whereas CD4+ T cells were more equally distributed. Tumor tissues with a high production of chemokines were associated with less T cell infiltration when the whole tissue was analyzed. However, when the spatial localization of CD8+ T cells within the tissue was taken into account, levels of CXCR3 ligands and the CCR5 ligand CCL8 showed a positive association with a high relative T cell infiltration in tumor-rich areas. Thus, CXCR3 ligands could mediate T cell trafficking but CAFs could prevent T cells from reaching the malignant cells.
Collapse
Affiliation(s)
- Laia Gorchs
- Department of Laboratory Medicine, Karolinska Institutet, 141 52 Stockholm, Sweden
- Correspondence: (L.G.); (H.K.)
| | - Marlies Oosthoek
- Department of Laboratory Medicine, Karolinska Institutet, 141 52 Stockholm, Sweden
| | | | - Carlos Fernández Moro
- Department of Laboratory Medicine, Karolinska Institutet, 141 52 Stockholm, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, 141 57 Stockholm, Sweden
| | - Helen Kaipe
- Department of Laboratory Medicine, Karolinska Institutet, 141 52 Stockholm, Sweden
- Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, 141 52 Stockholm, Sweden
- Correspondence: (L.G.); (H.K.)
| |
Collapse
|
28
|
Lin YN, Schmidt MO, Sharif GM, Vietsch EE, Kiliti AJ, Barefoot ME, Riegel AT, Wellstein A. Impaired CXCL12 signaling contributes to resistance of pancreatic cancer subpopulations to T cell-mediated cytotoxicity. Oncoimmunology 2022; 11:2027136. [PMID: 35127250 PMCID: PMC8816404 DOI: 10.1080/2162402x.2022.2027136] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 11/18/2022] Open
Abstract
Pancreatic cancer remains largely unresponsive to immune modulatory therapy attributable in part to an immunosuppressive, desmoplastic tumor microenvironment. Here, we analyze mechanisms of cancer cell-autonomous resistance to T cells. We used a 3D co-culture model of cancer cell spheroids from the KPC (LSL-KrasG12D/+ /LSL-Trp53R172H/+ /p48-Cre) pancreatic ductal adenocarcinoma (PDAC) model, to examine interactions with tumor-educated T cells isolated from draining lymph nodes of PDAC-bearing mice. Subpopulations of cancer cells resistant to these tumor-educated T cells were isolated from the in vitro co-culture and their properties compared with sensitive cancer cells. In co-culture with resistant cancer cell subpopulations, tumor-educated T cells showed reduced effector T cell functionality, reduced infiltration into tumor cell spheroids and decreased induction of apoptosis. A combination of comparative transcriptomic analyses, cytometric and immunohistochemistry techniques allowed us to dissect the role of differential gene expression and signaling pathways between sensitive and resistant cells. A decreased expression of the chemokine CXCL12 (SDF-1) was revealed as a common feature in the resistant cell subpopulations. Adding back CXCL12 reversed the resistant phenotype and was inhibited by the CXCR4 inhibitor AMD3100 (plerixafor). We conclude that reduced CXCL12 signaling contributes to PDAC subpopulation resistance to T cell-mediated attack.
Collapse
Affiliation(s)
- Yuan-Na Lin
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Marcel O. Schmidt
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Ghada M. Sharif
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Eveline E. Vietsch
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
- Department of Surgery, Erasmus Mc, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Amber J. Kiliti
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Megan E. Barefoot
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Anna T. Riegel
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Anton Wellstein
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| |
Collapse
|
29
|
Pinkert J, Boehm HH, Trautwein M, Doecke WD, Wessel F, Ge Y, Gutierrez EM, Carretero R, Freiberg C, Gritzan U, Luetke-Eversloh M, Golfier S, Von Ahsen O, Volpin V, Sorrentino A, Rathinasamy A, Xydia M, Lohmayer R, Sax J, Nur-Menevse A, Hussein A, Stamova S, Beckmann G, Glueck JM, Schoenfeld D, Weiske J, Zopf D, Offringa R, Kreft B, Beckhove P, Willuda J. T cell-mediated elimination of cancer cells by blocking CEACAM6–CEACAM1 interaction. Oncoimmunology 2021; 11:2008110. [PMID: 35141051 PMCID: PMC8820806 DOI: 10.1080/2162402x.2021.2008110] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6), a cell surface receptor, is expressed on normal epithelial tissue and highly expressed in cancers of high unmet medical need, such as non-small cell lung, pancreatic, and colorectal cancer. CEACAM receptors undergo homo- and heterophilic interactions thereby regulating normal tissue homeostasis and angiogenesis, and in cancer, tumor invasion and metastasis. CEACAM6 expression on malignant plasma cells inhibits antitumor activity of T cells, and we hypothesize a similar function on epithelial cancer cells. The interactions between CEACAM6 and its suggested partner CEACAM1 on T cells were studied. A humanized CEACAM6-blocking antibody, BAY 1834942, was developed and characterized for its immunomodulating effects in co-culture experiments with T cells and solid cancer cells and in comparison to antibodies targeting the immune checkpoints programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), and T cell immunoglobulin mucin-3 (TIM-3). The immunosuppressive activity of CEACAM6 was mediated by binding to CEACAM1 expressed by activated tumor-specific T cells. BAY 1834942 increased cytokine secretion by T cells and T cell-mediated killing of cancer cells. The in vitro efficacy of BAY 1834942 correlated with the degree of CEACAM6 expression on cancer cells, suggesting potential in guiding patient selection. BAY 1834942 was equally or more efficacious compared to blockade of PD-L1, and at least an additive efficacy was observed in combination with anti-PD-1 or anti-TIM-3 antibodies, suggesting an efficacy independent of the PD-1/PD-L1 axis. In summary, CEACAM6 blockade by BAY 1834942 reactivates the antitumor response of T cells. This warrants clinical evaluation.
Collapse
Affiliation(s)
- Jessica Pinkert
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans-Henning Boehm
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | - Florian Wessel
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yingzi Ge
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eva Maria Gutierrez
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rafael Carretero
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Uwe Gritzan
- Pharmaceutical Division, Bayer AG, Cologne, Germany
| | | | - Sven Golfier
- Pharmaceutical Division, Bayer AG, Berlin, Germany
| | | | - Valentina Volpin
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Antonio Sorrentino
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Anchana Rathinasamy
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Maria Xydia
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Robert Lohmayer
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
- Institute of Theoretical Physics, University of Regensburg, Regensburg, Germany
| | - Julian Sax
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Ayse Nur-Menevse
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Abir Hussein
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Slava Stamova
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | | | | | | | - Joerg Weiske
- Pharmaceutical Division, Bayer AG, Berlin, Germany
| | - Dieter Zopf
- Pharmaceutical Division, Bayer AG, Berlin, Germany
| | - Rienk Offringa
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Philipp Beckhove
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
- Hematology and Oncology Department, University Hospital Regensburg, Regensburg, Germany
| | | |
Collapse
|
30
|
Goulart MR, Stasinos K, Fincham REA, Delvecchio FR, Kocher HM. T cells in pancreatic cancer stroma. World J Gastroenterol 2021; 27:7956-7968. [PMID: 35046623 PMCID: PMC8678814 DOI: 10.3748/wjg.v27.i46.7956] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/18/2021] [Accepted: 11/28/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly devastating disease with a dismal 5-year survival rate. PDAC has a complex tumour microenvironment; characterised by a robust desmoplastic stroma, extensive infiltration of immunesuppressive cells such as immature myeloid cells, tumour-associated macrophages, neutrophils and regulatory T cells, and the presence of exhausted and senescent T cells. The cross-talk between cells in this fibrotic tumour establishes an immune-privileged microenvironment that supports tumour cell escape from immune-surveillance, disease progression and spread to distant organs. PDAC tumours, considered to be non-immunogenic or cold, express low mutation burden, low infiltration of CD8+ cytotoxic lymphocytes that are localised along the invasive margin of the tumour border in the surrounding fibrotic tissue, and often display an exhausted phenotype. Here, we review the role of T cells in pancreatic cancer, examine the complex interactions of these crucial effector units within pancreatic cancer stroma and shed light on the increasingly attractive use of T cells as therapy.
Collapse
Affiliation(s)
- Michelle R Goulart
- Centre for Tumour Biology Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Konstantinos Stasinos
- Centre for Tumour Biology Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, United Kingdom
- Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London E1 1BB, United Kingdom
| | - Rachel Elizabeth Ann Fincham
- Centre for Tumour Biology Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Francesca R Delvecchio
- Centre for Tumour Biology Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, United Kingdom
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Hemant M Kocher
- Centre for Tumour Biology Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, United Kingdom
- Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London E1 1BB, United Kingdom
| |
Collapse
|
31
|
Blobner J, Kilian M, Tan CL, Aslan K, Sanghvi K, Meyer J, Fischer M, Jähne K, Breckwoldt MO, Sahm F, von Deimling A, Bendszus M, Wick W, Platten M, Green E, Bunse L. Comparative evaluation of T-cell receptors in experimental glioma-draining lymph nodes. Neurooncol Adv 2021; 3:vdab147. [PMID: 34738084 PMCID: PMC8562732 DOI: 10.1093/noajnl/vdab147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Glioblastomas, the most common primary malignant brain tumors, are considered immunologically cold malignancies due to growth in an immune sanctuary site. While peptide vaccines have shown to generate intra-tumoral antigen-specific T cells, the identification of these tumor-specific T cells is challenging and requires detailed analyses of tumor tissue. Several studies have shown that CNS antigens may be transported via lymphatic drainage to cervical lymph nodes, where antigen-specific T-cell responses can be generated. Therefore, we investigated whether glioma-draining lymph nodes (TDLN) may constitute a reservoir of tumor-reactive T cells. Methods We addressed our hypothesis by flow cytometric analyses of chicken ovalbumin (OVA)-specific CD8+ T cells as well as T-cell receptor beta (TCRβ) next-generation-sequencing (TCRβ-NGS) of T cells from tumor tissue, TDLN, spleen, and inguinal lymph nodes harvested from experimental mouse GL261 glioma models. Results Longitudinal dextramer-based assessment of specific CD8+ T cells from TDLN did not show tumor model antigen reactivity. Unbiased immunogenomic analysis revealed a low overlap of TCRβ sequences from glioma-infiltrating CD8+ T cells between mice. Enrichment scores, calculated by the ratio of productive frequencies of the different TCRβ-CDR3 amino-acid (aa) rearrangements of CD8+ T cells derived from tumor, TDLN, inguinal lymph nodes, and spleen demonstrated a higher proportion of tumor-associated TCR in the spleen compared to TDLN. Conclusions In experimental glioblastoma, our data did not provide evidence that glioma-draining cervical lymph nodes are a robust reservoir for spontaneous glioma-specific T cells highlighting the requirement for detailed analyses of glioma-infiltrating T cells for the discovery of tumor-specific TCR.
Collapse
Affiliation(s)
- Jens Blobner
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences (MCTN), Heidelberg University, Heidelberg, Germany
| | - Michael Kilian
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences (MCTN), Heidelberg University, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Chin Leng Tan
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences (MCTN), Heidelberg University, Heidelberg, Germany
| | - Katrin Aslan
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences (MCTN), Heidelberg University, Heidelberg, Germany
| | - Khwab Sanghvi
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences (MCTN), Heidelberg University, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Jochen Meyer
- DKTK Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neuropathology, Heidelberg University Medical Center, Heidelberg, Germany
| | - Manuel Fischer
- Department of Neuroradiology, Heidelberg University Medical Center, Heidelberg, Germany
| | - Kristine Jähne
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences (MCTN), Heidelberg University, Heidelberg, Germany
| | - Michael O Breckwoldt
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neuroradiology, Heidelberg University Medical Center, Heidelberg, Germany
| | - Felix Sahm
- DKTK Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neuropathology, Heidelberg University Medical Center, Heidelberg, Germany
| | - Andreas von Deimling
- DKTK Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neuropathology, Heidelberg University Medical Center, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Medical Center, Heidelberg, Germany
| | - Wolfgang Wick
- DKTK Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Platten
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences (MCTN), Heidelberg University, Heidelberg, Germany.,Helmholtz Center for Translational Oncology (HI-TRON), Mainz, Germany
| | - Edward Green
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences (MCTN), Heidelberg University, Heidelberg, Germany
| | - Lukas Bunse
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences (MCTN), Heidelberg University, Heidelberg, Germany
| |
Collapse
|
32
|
Suarez-Carmona M, Williams A, Schreiber J, Hohmann N, Pruefer U, Krauss J, Jäger D, Frömming A, Beyer D, Eulberg D, Jungelius JU, Baumann M, Mangasarian A, Halama N. Combined inhibition of CXCL12 and PD-1 in MSS colorectal and pancreatic cancer: modulation of the microenvironment and clinical effects. J Immunother Cancer 2021; 9:e002505. [PMID: 34607895 PMCID: PMC8491418 DOI: 10.1136/jitc-2021-002505] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Immunotherapy in microsatellite stable colorectal or pancreatic cancer has not shown promising results. It has been hypothesized that targeting immunosuppressive molecules like SDF1-alpha/CXCL12 could contribute to immunotherapy and animal models showed promising results on T cell activation and migration in combination with immune checkpoint inhibition. METHODS Here, we describe the successful application of anti-CXCL12 (NOX-A12) in patients with advanced stage pretreated metastatic colorectal and pancreatic cancer (OPERA trial). The treatment consisted of 2 weeks of anti-CXCL12 monotherapy with NOX-A12 followed by combination therapy with pembrolizumab (n=20 patients) until progression or intolerable toxicity had occurred. RESULTS The treatment was safe and well tolerated with 83.8% grade I/II, 15.5% grade III and 0.7% grade V adverse events. Of note, for a majority of patients, time on trial treatment was prolonged compared with their last standard treatment preceding trial participation. Systematic serial biopsies revealed distinct patterns of modulation. Tissue and clinical responses were associated with Th1-like tissue reactivity upon CXCL12 inhibition. A downregulation of a cytokine cassette of interleukin (IL)-2/IL-16/CXCL-10 was associated with tumor resistance and furthermore linked to a rare, CXCL12-associated CD14+CD15+promonocytic population. T cells showed aggregation and directed movement towards the tumor cells in responding tissues. Serum analyses detected homogeneous immunomodulatory patterns in all patients, regardless of tissue responses. CONCLUSIONS We demonstrate that the combination of CXCL12 inhibition and checkpoint inhibition is safe and grants further exploration of synergistic combinatorial strategies.
Collapse
Affiliation(s)
- Meggy Suarez-Carmona
- Department of Translational Immunotherapy, German Cancer Research Centre, Heidelberg, Germany
| | - Anja Williams
- Department of Medical Oncology, National Center of Tumor Diseases, Heidelberg, Baden-Württemberg, Germany
| | - Jutta Schreiber
- Department of Medical Oncology, National Center of Tumor Diseases, Heidelberg, Baden-Württemberg, Germany
| | - Nicolas Hohmann
- Department of Medical Oncology, National Center of Tumor Diseases, Heidelberg, Baden-Württemberg, Germany
| | - Ulrike Pruefer
- Department of Medical Oncology, National Center of Tumor Diseases, Heidelberg, Baden-Württemberg, Germany
| | - Jürgen Krauss
- Department of Medical Oncology, National Center of Tumor Diseases, Heidelberg, Baden-Württemberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center of Tumor Diseases, Heidelberg, Baden-Württemberg, Germany
| | | | | | | | | | | | | | - Niels Halama
- Department of Translational Immunotherapy, German Cancer Research Centre, Heidelberg, Germany
| |
Collapse
|
33
|
Kang W, Feng Z, Luo J, He Z, Liu J, Wu J, Rong P. Tertiary Lymphoid Structures in Cancer: The Double-Edged Sword Role in Antitumor Immunity and Potential Therapeutic Induction Strategies. Front Immunol 2021; 12:689270. [PMID: 34394083 PMCID: PMC8358404 DOI: 10.3389/fimmu.2021.689270] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/05/2021] [Indexed: 12/14/2022] Open
Abstract
The complex tumor microenvironment (TME) plays a vital role in cancer development and dramatically determines the efficacy of immunotherapy. Tertiary lymphoid structures (TLSs) within the TME are well recognized and consist of T cell-rich areas containing dendritic cells (DCs) and B cell-rich areas containing germinal centers (GCs). Accumulating research has indicated that there is a close association between tumor-associated TLSs and favorable clinical outcomes in most types of cancers, though a minority of studies have reported an association between TLSs and a poor prognosis. Overall, the double-edged sword role of TLSs in the TME and potential mechanisms need to be further investigated, which will provide novel therapeutic perspectives for antitumor immunoregulation. In this review, we focus on discussing the main functions of TLSs in the TME and recent advances in the therapeutic manipulation of TLSs through multiple strategies to enhance local antitumor immunity.
Collapse
Affiliation(s)
- Wendi Kang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhichao Feng
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China
| | - Jianwei Luo
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhenhu He
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jun Liu
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jianzhen Wu
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China
| |
Collapse
|
34
|
Baumann D, Drebant J, Hägele T, Burger L, Serger C, Lauenstein C, Dudys P, Erdmann G, Offringa R. p38 MAPK signaling in M1 macrophages results in selective elimination of M2 macrophages by MEK inhibition. J Immunother Cancer 2021; 9:jitc-2020-002319. [PMID: 34285105 PMCID: PMC8292803 DOI: 10.1136/jitc-2020-002319] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2021] [Indexed: 12/17/2022] Open
Abstract
M2 macrophages promote tumor progression and therapy resistance, whereas proimmunogenic M1 macrophages can contribute to the efficacy of cytostatic and immunotherapeutic strategies. The abundance of M2 macrophages in the immune infiltrate of many cancer types has prompted the search for strategies to target and eliminate this subset. From our prior experiments in syngeneic mouse tumor models, we learned that pharmacological inhibition of mitogen-activated protein kinase kinase (MEK) did not merely result in tumor cell death, but also in the modulation of the tumor immune infiltrate. This included a prominent decrease in the numbers of macrophages as well as an increase in the M1/M2 macrophage ratio. Investigation of the mechanism underlying this finding in primary murine macrophage cultures revealed that M2 macrophages are significantly more sensitive to MEK inhibition-induced cell death than their M1 counterparts. Further analyses showed that the p38 MAPK pathway, which is activated in M1 macrophages only, renders these cells resistant to death by MEK inhibition. In conclusion, the dependency of M2 macrophages on the MEK/extracellular-signal regulated kinase (ERK) pathway empowers MEK inhibitors to selectively eliminate this subset from the tumor microenvironment.
Collapse
Affiliation(s)
- Daniel Baumann
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Baden-Württemberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Baden-Württemberg, Germany
| | - Jennifer Drebant
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Baden-Württemberg, Germany
| | - Tanja Hägele
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Baden-Württemberg, Germany
| | - Luisa Burger
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Baden-Württemberg, Germany
| | - Clara Serger
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Baden-Württemberg, Germany
| | - Claudia Lauenstein
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Baden-Württemberg, Germany
| | | | | | - Rienk Offringa
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Baden-Württemberg, Germany .,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Baden-Württemberg, Germany
| |
Collapse
|
35
|
Myxoma Virus Expressing LIGHT (TNFSF14) Pre-Loaded into Adipose-Derived Mesenchymal Stem Cells Is Effective Treatment for Murine Pancreatic Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13061394. [PMID: 33808692 PMCID: PMC8003548 DOI: 10.3390/cancers13061394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 11/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a weakly immunogenic fatal neoplasm. Oncolytic viruses with dual anti-cancer properties-oncolytic and immune response-boosting effects-have great potential for PDAC management. Adipose-derived stem cells (ADSCs) of mesenchymal origin were infected ex vivo with recombinant myxoma virus (MYXV), which encodes murine LIGHT, also called tumor necrosis factor ligand superfamily member 14 (TNFSF14). The viability and proliferation of ADSCs were not remarkably decreased (1-2 days) following MYXV infection, in sharp contrast to cells of pancreatic carcinoma lines studied, which were rapidly killed by the infection. Comparison of the intraperitoneal (IP) vs. the intravenous (IV) route of ADSC/MYXV administration revealed more pancreas-targeted distribution of the virus when ADSCs were delivered IP to mice bearing orthotopically injected PDAC. The biodistribution, tumor burden reduction and anti-tumor adaptive immune response were examined. Bioluminescence data, used to assess the presence of the luciferase-tagged virus after IP injection, indicated enhanced trafficking into the pancreata of mice bearing orthotopically-induced PDAC, as compared to tumor-free animals, resulting in extended survival of the treated PDAC-seeded animals and in the boosted expression of key adaptive immune response markers. We conclude that ADSCs pre-loaded with transgene-armed MYXV and administered IP allow for the effective ferrying of the oncolytic virus to sites of PDAC and mediate improved tumor regression.
Collapse
|
36
|
LAG-3-Expressing Tumor-Infiltrating T Cells Are Associated with Reduced Disease-Free Survival in Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13061297. [PMID: 33803936 PMCID: PMC7998134 DOI: 10.3390/cancers13061297] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary In light of the majority of pancreatic cancer patients not responding to current immune checkpoint blockade, alternative immunotherapeutic targets need to be identified. In this study, we employed multiplex immunofluorescence to investigate the expression of co-stimulatory and inhibitory receptors by tumor-infiltrating T cells in human pancreatic cancer. A comprehensive analysis of the receptor pattern on tumor-infiltrating T cells is essential for the development of new therapeutic strategies, as well as personalized immunotherapy, to identify patients who are likely to benefit from targeting specific immune receptors. Abstract T cells are the predominant immune cell population in the pancreatic tumor microenvironment. High CD8+ and Th1-polarized CD4+ T cell infiltration is associated with prolonged survival in human pancreatic ductal adenocarcinoma (PDAC). However, the expression pattern of co-stimulatory and inhibitory receptors by PDAC-infiltrating T cells and their prognostic significance are not well defined. In this study, we employed multiplex immunofluorescence to investigate the intratumoral expression of the co-stimulatory receptor inducible T-cell co-stimulator (ICOS), the inhibitory receptors lymphocyte-activation gene 3 (LAG-3), programmed death 1 (PD-1), and V-domain immunoglobulin suppressor of T cell activation (VISTA) by tumor-infiltrating T cells (CD3) in a cohort of 69 patients with resected PDAC. T cells were enriched particularly within the stromal area and were highly heterogeneous across tumors. Further, T cells were associated with prolonged disease-free survival (DFS). However, LAG-3 expression by PDAC-infiltrating T cells was correlated with reduced DFS. Our study highlights the biological importance of LAG-3 expression by tumor-infiltrating T cells. LAG-3+ T cells may represent a novel prognostic marker and a particularly attractive target for immunotherapeutic strategies in PDAC.
Collapse
|
37
|
Hofschröer V, Najder K, Rugi M, Bouazzi R, Cozzolino M, Arcangeli A, Panyi G, Schwab A. Ion Channels Orchestrate Pancreatic Ductal Adenocarcinoma Progression and Therapy. Front Pharmacol 2021; 11:586599. [PMID: 33841132 PMCID: PMC8025202 DOI: 10.3389/fphar.2020.586599] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is a devastating disease with a dismal prognosis. Therapeutic interventions are largely ineffective. A better understanding of the pathophysiology is required. Ion channels contribute substantially to the "hallmarks of cancer." Their expression is dysregulated in cancer, and they are "misused" to drive cancer progression, but the underlying mechanisms are unclear. Ion channels are located in the cell membrane at the interface between the intracellular and extracellular space. They sense and modify the tumor microenvironment which in itself is a driver of PDAC aggressiveness. Ion channels detect, for example, locally altered proton and electrolyte concentrations or mechanical stimuli and transduce signals triggered by these microenvironmental cues through association with intracellular signaling cascades. While these concepts have been firmly established for other cancers, evidence has emerged only recently that ion channels are drivers of PDAC aggressiveness. Particularly, they appear to contribute to two of the characteristic PDAC features: the massive fibrosis of the tumor stroma (desmoplasia) and the efficient immune evasion. Our critical review of the literature clearly shows that there is still a remarkable lack of knowledge with respect to the contribution of ion channels to these two typical PDAC properties. Yet, we can draw parallels from ion channel research in other fibrotic and inflammatory diseases. Evidence is accumulating that pancreatic stellate cells express the same "profibrotic" ion channels. Similarly, it is at least in part known which major ion channels are expressed in those innate and adaptive immune cells that populate the PDAC microenvironment. We explore potential therapeutic avenues derived thereof. Since drugs targeting PDAC-relevant ion channels are already in clinical use, we propose to repurpose those in PDAC. The quest for ion channel targets is both motivated and complicated by the fact that some of the relevant channels, for example, KCa3.1, are functionally expressed in the cancer, stroma, and immune cells. Only in vivo studies will reveal which arm of the balance we should put our weights on when developing channel-targeting PDAC therapies. The time is up to explore the efficacy of ion channel targeting in (transgenic) murine PDAC models before launching clinical trials with repurposed drugs.
Collapse
Affiliation(s)
| | - Karolina Najder
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Micol Rugi
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Rayhana Bouazzi
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Marco Cozzolino
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Münster, Germany
| |
Collapse
|
38
|
Bear AS, Vonderheide RH, O'Hara MH. Challenges and Opportunities for Pancreatic Cancer Immunotherapy. Cancer Cell 2020; 38:788-802. [PMID: 32946773 PMCID: PMC7738380 DOI: 10.1016/j.ccell.2020.08.004] [Citation(s) in RCA: 372] [Impact Index Per Article: 74.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is among the most immune-resistant tumor types. Its unique genomic landscape shaped by oncogenic drivers promotes immune suppression from the earliest stages of tumor inception to subvert adaptive T cell immunity. Single-agent immune modulators have thus far proven clinically ineffective, and multi-modal therapies targeting mechanisms of immunotherapy resistance are likely needed. Here, we review novel immunotherapy strategies currently under investigation to (1) confer antigen specificity, (2) enhance T cell effector function, and (3) neutralize immunosuppressive elements within the tumor microenvironment that may be rationally combined to untangle the web of immune resistance in PDA and other tumors.
Collapse
Affiliation(s)
- Adham S Bear
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert H Vonderheide
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| | - Mark H O'Hara
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA. mark.o'
| |
Collapse
|
39
|
Seifert AM, Eymer A, Heiduk M, Wehner R, Tunger A, von Renesse J, Decker R, Aust DE, Welsch T, Reissfelder C, Weitz J, Schmitz M, Seifert L. PD-1 Expression by Lymph Node and Intratumoral Regulatory T Cells Is Associated with Lymph Node Metastasis in Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12102756. [PMID: 32987956 PMCID: PMC7599971 DOI: 10.3390/cancers12102756] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Pancreatic cancer is a devastating disease and among the most immune-resistant tumor types. Single-agent immunotherapy has not demonstrated clinical benefits in pancreatic cancer patients, and combinational therapies targeting multiple mechanisms of immunosuppression are likely needed. T cell activation in lymph nodes is required for the efficacy of immunotherapy. Here, we phenotypically and functionally analyze T cells from tumor-draining lymph nodes, blood and tumors from patients with pancreatic cancer to decipher unknown immunosuppressive mechanisms and to identify potential immunotherapeutic targets. Abstract Pancreatic ductal adenocarcinoma (PDAC) is characterized by a mostly immunosuppressive microenvironment. Tumor-draining lymph nodes (TDLN) are a major site for priming of tumor-reactive T cells and also tumor metastasis. However, the phenotype and function of T cells in TDLNs from PDAC patients is unknown. In this study, lymph nodes from the pancreatic head (PH), the hepatoduodenal ligament (HDL) and the interaortocaval (IAC) region were obtained from 25 patients with adenocarcinoma of the pancreatic head. Additionally, tumors and matched blood were analyzed from 16 PDAC patients. Using multicolor flow cytometry, we performed a comprehensive analysis of T cells. CD4+ T cells were the predominant T cell subset in PDAC-draining lymph nodes. Overall, lymph node CD4+ and CD8+ T cells had a similar degree of activation, as measured by CD69, inducible T cell co-stimulator (ICOS) and CD137 (4-1BB) expression and interferon-γ (IFNγ) secretion. Expression of the inhibitory receptor programmed death 1 (PD-1) by lymph node and tumor-infiltrating regulatory T cells (Tregs) correlated with lymph node metastasis. Collectively, Treg cells and PD-1 are two relevant components of the immunosuppressive network in PDAC-draining lymph nodes and may be particularly attractive targets for combinatorial immunotherapeutic strategies in selected patients with node-positive PDAC.
Collapse
Affiliation(s)
- Adrian M. Seifert
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Correspondence:
| | - Annabel Eymer
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (A.E.); (R.W.); (A.T.)
| | - Max Heiduk
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Rebekka Wehner
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (A.E.); (R.W.); (A.T.)
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Antje Tunger
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (A.E.); (R.W.); (A.T.)
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Janusz von Renesse
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
| | - Rahel Decker
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
| | - Daniela E. Aust
- Department of Pathology, University Hospital Carl Gustav Carus, Medical Faculty, University of Dresden, 01307 Dresden, Germany;
- NCT Biobank Dresden, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Thilo Welsch
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Christoph Reissfelder
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Marc Schmitz
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (A.E.); (R.W.); (A.T.)
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Lena Seifert
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| |
Collapse
|
40
|
Halama N, Haberkorn U. The Unmet Needs of the Diagnosis, Staging, and Treatment of Gastrointestinal Tumors. Semin Nucl Med 2020; 50:389-398. [PMID: 32768003 DOI: 10.1053/j.semnuclmed.2020.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
New scientific insights in cancer biology and immunobiology have changed the clinical practice of medical oncology in recent years. The molecular stratification of solid tumors has led to improved clinical outcomes and is a key part in the diagnostic workup. Beyond mutational spectra (like Rat sarcoma [RAS] mutations or tumor mutational burden), the investigation of the immunological microenvironment has attracted more efforts. Especially as immunotherapies have changed the standard treatment for some solid tumors dramatically and have become an important part of routine oncology, also for gastrointestinal tumors. Still only a subgroup of patients benefits from immunotherapy in gastrointestinal tumors with prominent examples from colorectal, pancreatic or gastric cancer. Not only microsatellite instability as a marker for response to immunotherapy has shown its utility, there plenty of other approaches currently being investigated to better stratify and understand the microenvironment. But these insights have not translated into clinical utility. Reasons for this are limited technical capabilities for stratification and for coping with heterogeneity of tumor cells and the microenvironment as such. So the situation for gastrointestinal tumors has shown mainly progress for a subgroup of immunotherapy-receptive tumors (eg, microsatellite instability), but advances for the remaining majority have been in the area of stratification and combinatorial therapies, including approaches without chemotherapy. Molecular stratification (eg, B-Rapidly Accelerated Fibrosarcoma [BRAF] V600E mutation in colorectal cancer or NRG1 fusions in Kirsten-rat sarcoma (KRAS) Wild-Type Pancreatic Cancer) has clearly improved the possibilities for directed therapies, but there is a plethora of clinical situations where further developments are needed to improve patient care. Finding these areas and identifying the technical approach to unravel the complexities is the next decisive step. Here the recent advances are summarized and an outlook on possible diagnostic and treatment options in areas of unmet need is given with the context of new molecular imaging possibilities and cutting edge advances in nuclear medicine.
Collapse
Affiliation(s)
- Niels Halama
- German Cancer Research Center (DKFZ), Department of Translational Immunotherapy, German Cancer Research Center (DKFZ), Germany; Helmholtz-Institute for Translational Oncology Mainz (HI-TRON Mainz), Germany; Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases (NCT), Heidelberg, Germany; Institute for Immunology, University Hospital Heidelberg, University Heidelberg.
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Germany; Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
41
|
Hegde S. Pancreatic Cancer Immuno-oncology in the Era of Precision Medicine. Indian J Surg Oncol 2020; 12:118-127. [PMID: 33994737 DOI: 10.1007/s13193-020-01192-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/19/2020] [Indexed: 10/23/2022] Open
Abstract
Pancreatic malignancies carry a dismal prognosis globally, with pancreatic adenocarcinomas (PDAC) being particularly aggressive and stubborn. Unfortunately, several therapeutic strategies that show promise in other cancers have failed to make sizeable impact on pancreatic tumor outcomes. Responses to immunotherapies are especially rare in pancreatic cancer, and patients are in need of innovative approaches that can result in more durable responses. Current research in preclinical models and humans has suggested this resistance is due to a uniquely inflammatory and dysfunctional tumor microenvironment; these findings lay the groundwork for targeting these barriers and improving outcomes. Clinical analyses have also revealed unprecedented heterogeneity in tumor and stromal biology of PDAC, underscoring the need for more personalized approaches and combinatorial therapies. This review will highlight the current state of translational research focusing on PDAC immunity, summarize ongoing clinical efforts to tackle PDAC vulnerabilities, and underscore some unresolved challenges in implementing therapies more broadly. A better understanding of immune contexture and tumor heterogeneity in this disease will greatly accelerate drug discovery and implementation of precision medicine for PDAC.
Collapse
Affiliation(s)
- Samarth Hegde
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| |
Collapse
|
42
|
Wu QY, Yang CK, Rong LJ, Li JC, Lei LM. Investigation of the association between C-X-C motif chemokine receptor subunits and tumor infiltration levels and prognosis in patients with early-stage pancreatic ductal adenocarcinoma. Oncol Lett 2020; 20:16. [PMID: 32774489 PMCID: PMC7406880 DOI: 10.3892/ol.2020.11877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 04/01/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the malignancies with the highest morality rate due to postoperative local invasion and distant metastasis. Although C-X-C motif chemokine receptor (CXCR) subunits have been reported as prognostic indicators in gastric cancer, the prognostic value of CXCR subunits in PDAC remains poorly understood. In the present study, the expression levels and biological functions of CXCR subunits were investigated using multiple publicly accessible bioinformatic platforms and databases. Survival analysis was used to evaluate the prognostic value of CXCR subunits in 112 early-stage PDAC cases by setting the median expression levels as the cut-off values. A nomogram was constructed to combine CXCR subunit expression levels and clinical data for prognosis prediction. Moreover, the association between CXCR subunit expression levels and tumor infiltration levels were detected in PDAC. The expression levels of CXCR subunits were elevated in PDAC tumor tissues. In the multivariate Cox proportional risk regression model, high CXCR2, CXCR4 and CXCR6 expression levels in early-stage PDAC were associated with a more favorable prognosis. Further, it was demonstrated that the differential expression levels of CXCR subunits in PDAC for combined survival analysis could contribute to risk stratification. The nomogram model demonstrated the contribution of CXCR subunits and clinical features in the prognosis of PDAC. Gene Set Enrichment Analysis suggested that CXCR subunits serve a role in immunomodulatory functions. The expression levels and somatic copy number alterations of CXCR subunits were associated with tumor infiltration levels in PDAC. CXCR subunits were associated with prognosis in patients with early-stage PDAC and may be potential drug targets for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Qiong-Yuan Wu
- Department of Tuina, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, P.R.China
| | - Cheng-Kun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530023, P.R.China
| | - Liang-Jun Rong
- Department of Tuina, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, P.R.China
| | - Jun-Chan Li
- Department of Tuina, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, P.R.China
| | - Long-Ming Lei
- Department of Tuina, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi 530023, P.R.China
| |
Collapse
|
43
|
Baumann D, Hägele T, Mochayedi J, Drebant J, Vent C, Blobner S, Noll JH, Nickel I, Schumacher C, Boos SL, Daniel AS, Wendler S, Volkmar M, Strobel O, Offringa R. Proimmunogenic impact of MEK inhibition synergizes with agonist anti-CD40 immunostimulatory antibodies in tumor therapy. Nat Commun 2020; 11:2176. [PMID: 32358491 PMCID: PMC7195409 DOI: 10.1038/s41467-020-15979-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 04/03/2020] [Indexed: 12/21/2022] Open
Abstract
Cancer types with lower mutational load and a non-permissive tumor microenvironment are intrinsically resistant to immune checkpoint blockade. While the combination of cytostatic drugs and immunostimulatory antibodies constitutes an attractive concept for overcoming this refractoriness, suppression of immune cell function by cytostatic drugs may limit therapeutic efficacy. Here we show that targeted inhibition of mitogen-activated protein kinase (MAPK) kinase (MEK) does not impair dendritic cell-mediated T cell priming and activation. Accordingly, combining MEK inhibitors (MEKi) with agonist antibodies (Abs) targeting the immunostimulatory CD40 receptor results in potent synergistic antitumor efficacy. Detailed analysis of the mechanism of action of MEKi shows that this drug exerts multiple pro-immunogenic effects, including the suppression of M2-type macrophages, myeloid derived suppressor cells and T-regulatory cells. The combination of MEK inhibition with agonist anti-CD40 Ab is therefore a promising therapeutic concept, especially for the treatment of mutant Kras-driven tumors such as pancreatic ductal adenocarcinoma. Immune checkpoint inhibitors have limited efficacy in tumors with lower mutational burden and non-permissive microenvironment. Here, the authors show that combining MEK inhibition with an agonist anti-CD40 immunostimulatory antibody improves antitumor treatment by inducing immunogenic changes in the tumor microenvironment.
Collapse
Affiliation(s)
- Daniel Baumann
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany.,Department of Surgery, Heidelberg University Hospital, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Tanja Hägele
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Julian Mochayedi
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Jennifer Drebant
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Caroline Vent
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany.,Department of Surgery, Heidelberg University Hospital, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Sven Blobner
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Julia Han Noll
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Irena Nickel
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Corinna Schumacher
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Sophie Luise Boos
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany.,Department of Oncogenic signaling pathways of colorectal/pancreatic cancer, Ludwig-Maximilians-Universitaet, Munich, Bavaria, 80539, Germany
| | - Aline Sophie Daniel
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Susann Wendler
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany.,Department of Surgery, Heidelberg University Hospital, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Michael Volkmar
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany.,Department of Surgery, Heidelberg University Hospital, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Oliver Strobel
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Baden-Wuerttemberg, 69120, Germany
| | - Rienk Offringa
- Department of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center Heidelberg, Heidelberg, Baden-Wuerttemberg, 69120, Germany. .,Department of Surgery, Heidelberg University Hospital, Heidelberg, Baden-Wuerttemberg, 69120, Germany.
| |
Collapse
|
44
|
Poschke IC, Hassel JC, Rodriguez-Ehrenfried A, Lindner KAM, Heras-Murillo I, Appel LM, Lehmann J, Lövgren T, Wickström SL, Lauenstein C, Roth J, König AK, Haanen JBAG, van den Berg J, Kiessling R, Bergmann F, Flossdorf M, Strobel O, Offringa R. The Outcome of Ex Vivo TIL Expansion Is Highly Influenced by Spatial Heterogeneity of the Tumor T-Cell Repertoire and Differences in Intrinsic In Vitro Growth Capacity between T-Cell Clones. Clin Cancer Res 2020; 26:4289-4301. [PMID: 32303540 DOI: 10.1158/1078-0432.ccr-19-3845] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 03/15/2020] [Accepted: 04/14/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE During our efforts to develop tumor-infiltrating lymphocyte (TIL) therapy to counter the devastating recurrence rate in patients with primary resectable pancreatic ductal adenocarcinoma (PDA), we found that PDA TILs can readily be expanded in vitro and that the majority of resulting TIL cultures show reactivity against the autologous tumor. However, the fraction of tumor-reactive T cells is low. We investigated to which extent this was related to the in vitro expansion. EXPERIMENTAL DESIGN We compared the clonal composition of TIL preparations before and after in vitro expansion using T-cell receptor (TCR) deep sequencing. Our findings for PDA were benchmarked to experiments with melanoma TILs. RESULTS We found that the TIL TCR repertoire changes dramatically during in vitro expansion, leading to loss of tumor- dominant T-cell clones and overgrowth by newly emerging T-cell clones that are barely detectable in the tumor. These changes are primarily driven by differences in the intrinsic in vitro expansion capacity of T-cell clones. Single-cell experiments showed an association between poor proliferative capacity and expression of markers related to antigen experience and dysfunction. Furthermore, we found that spatial heterogeneity of the TIL repertoire resulted in TCR repertoires that are greatly divergent between TIL cultures derived from distant tumor samples of the same patient. CONCLUSIONS Culture-induced changes in clonal composition are likely to affect tumor reactivity of TIL preparations. TCR deep sequencing provides important insights into the factors that govern the outcome of in vitro TIL expansion and thereby a path toward optimization of the production of TIL preparations with high therapeutic efficacy.See related commentary by Lozano-Rabella and Gros, p. 4177.
Collapse
Affiliation(s)
- Isabel C Poschke
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany. .,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Jessica C Hassel
- Department of Dermatology and National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Aaron Rodriguez-Ehrenfried
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Katharina A M Lindner
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Ignacio Heras-Murillo
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Lena M Appel
- Division of Theoretical Systems Biology, German Cancer Research Center and BioQuant Center, University of Heidelberg, Heidelberg, Germany.,Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Johanna Lehmann
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Tanja Lövgren
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Stina L Wickström
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Claudia Lauenstein
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Jasmin Roth
- Department of Dermatology and National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Anna-Katharina König
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - John B A G Haanen
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Joost van den Berg
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Frank Bergmann
- Department of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Flossdorf
- Division of Theoretical Systems Biology, German Cancer Research Center and BioQuant Center, University of Heidelberg, Heidelberg, Germany.,Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Oliver Strobel
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Rienk Offringa
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
45
|
Hegde S, Krisnawan VE, Herzog BH, Zuo C, Breden MA, Knolhoff BL, Hogg GD, Tang JP, Baer JM, Mpoy C, Lee KB, Alexander KA, Rogers BE, Murphy KM, Hawkins WG, Fields RC, DeSelm CJ, Schwarz JK, DeNardo DG. Dendritic Cell Paucity Leads to Dysfunctional Immune Surveillance in Pancreatic Cancer. Cancer Cell 2020; 37:289-307.e9. [PMID: 32183949 PMCID: PMC7181337 DOI: 10.1016/j.ccell.2020.02.008] [Citation(s) in RCA: 296] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/04/2019] [Accepted: 02/14/2020] [Indexed: 12/26/2022]
Abstract
Here, we utilized spontaneous models of pancreatic and lung cancer to examine how neoantigenicity shapes tumor immunity and progression. As expected, neoantigen expression during lung adenocarcinoma development leads to T cell-mediated immunity and disease restraint. By contrast, neoantigen expression in pancreatic ductal adenocarcinoma (PDAC) results in exacerbation of a fibro-inflammatory microenvironment that drives disease progression and metastasis. Pathogenic TH17 responses are responsible for this neoantigen-induced tumor progression in PDAC. Underlying these divergent T cell responses in pancreas and lung cancer are differences in infiltrating conventional dendritic cells (cDCs). Overcoming cDC deficiency in early-stage PDAC leads to disease restraint, while restoration of cDC function in advanced PDAC restores tumor-restraining immunity and enhances responsiveness to radiation therapy.
Collapse
Affiliation(s)
- Samarth Hegde
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Varintra E Krisnawan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brett H Herzog
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chong Zuo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marcus A Breden
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brett L Knolhoff
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Graham D Hogg
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jack P Tang
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John M Baer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cedric Mpoy
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kyung Bae Lee
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Katherine A Alexander
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Buck E Rogers
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - William G Hawkins
- Department of Surgery, Barnes-Jewish Hospital, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Ryan C Fields
- Department of Surgery, Barnes-Jewish Hospital, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Carl J DeSelm
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Julie K Schwarz
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - David G DeNardo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA.
| |
Collapse
|
46
|
Fan JQ, Wang MF, Chen HL, Shang D, Das JK, Song J. Current advances and outlooks in immunotherapy for pancreatic ductal adenocarcinoma. Mol Cancer 2020; 19:32. [PMID: 32061257 PMCID: PMC7023714 DOI: 10.1186/s12943-020-01151-3] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/06/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an incurable cancer resistant to traditional treatments, although a limited number of early-stage patients can undergo radical resection. Immunotherapies for the treatment of haematological malignancies as well as solid tumours have been substantially improved over the past decades, and impressive results have been obtained in recent preclinical and clinical trials. However, PDAC is likely the exception because of its unique tumour microenvironment (TME). In this review, we summarize the characteristics of the PDAC TME and focus on the network of various tumour-infiltrating immune cells, outlining the current advances in PDAC immunotherapy and addressing the effect of the PDAC TME on immunotherapy. This review further explores the combinations of different therapies used to enhance antitumour efficacy or reverse immunodeficiencies and describes optimizable immunotherapeutic strategies for PDAC. The concordant combination of various treatments, such as targeting cancer cells and the stroma, reversing suppressive immune reactions and enhancing antitumour reactivity, may be the most promising approach for the treatment of PDAC. Traditional treatments, especially chemotherapy, may also be optimized for individual patients to remodel the immunosuppressive microenvironment for enhanced therapy.
Collapse
Affiliation(s)
- Jia-qiao Fan
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Meng-Fei Wang
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hai-Long Chen
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jugal K. Das
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, TX USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, TX USA
| |
Collapse
|
47
|
Nevala-Plagemann C, Hidalgo M, Garrido-Laguna I. From state-of-the-art treatments to novel therapies for advanced-stage pancreatic cancer. Nat Rev Clin Oncol 2020; 17:108-123. [PMID: 31705130 DOI: 10.1038/s41571-019-0281-6] [Citation(s) in RCA: 252] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2019] [Indexed: 12/13/2022]
Abstract
Improvements in the outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) have lagged behind advances made in the treatment of many other malignancies over the past few decades. For most patients with PDAC, cytotoxic chemotherapy remains the mainstay of treatment. For patients with resectable disease, modified 5-fluorouracil, leucovorin, irinotecan and oxaliplatin (mFOLFIRINOX) is the standard-of-care adjuvant therapy, although data from several randomized trials have shown improved outcomes with neoadjuvant treatment strategies. For patients with advanced-stage or metastatic disease, comprehensive genomic profiling has revealed several potentially actionable alterations in small subsets of patients and the feasibility of implementing such strategies is beginning to be confirmed. Novel therapies targeting certain aberrations, most notably BRCA1/2 mutations, mismatch repair (MMR) deficiencies or NTRK1-3 fusions, have shown considerable activity in clinical trials, and larotrectinib, entrectinib and pembrolizumab have received FDA approval for the treatment of patients with tumours harbouring NTRK fusions and MMR deficiencies, respectively, regardless of primary tumour histology. In this Review, we describe the available data on the activity of these and other agents in patients with PDAC. Our discussion is structured according to the acronym 'PRIME' to organize the various treatment strategies currently undergoing evaluation in clinical trials: Pathway inhibition, alteration of DNA Repair pathways, Immunotherapy, cancer Metabolism and targeting the Extracellular tumour microenvironment.
Collapse
Affiliation(s)
| | - Manuel Hidalgo
- Division of Hematology and Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Ignacio Garrido-Laguna
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA.
- Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
48
|
Han J, Lotze MT. The Adaptome as Biomarker for Assessing Cancer Immunity and Immunotherapy. Methods Mol Biol 2020; 2055:369-397. [PMID: 31502161 DOI: 10.1007/978-1-4939-9773-2_17] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In terms of diagnosing and treating diseases, our adaptive immune system is the "best doctor." It carries out these tasks with unmatched precision, with the help of both T and B cell receptors, our most diverse set of genes, distinguishing one individual from another. It does this by generating autologous extraordinary diversity in the receptors, ranging from 1015 to 1025 for each chain of the rearranged receptors. By combining multiplex PCR and next-generation sequencing (NGS), we have developed high throughput methods to study adaptive immunity. The adaptome is the sum-total of expressed T and B cell receptor genes in a sample, composed of seven chains, including the alpha/beta and gamma/delta chains for T cells, and heavy/lambda or kappa chains for B cells. Immune repertoire is the sum-total of the individual clonotypes within one chain, including individual complementarity-determining regions (CDR) 3 sequences. In order to reflect the breadth and depth of the true adaptome, the following criteria assessing any method needs to be ascertained: (1) Methods need to be inclusive and quantitative; (2) Analysis should consider what questions need to be addressed and whether bulk or single cell sequencing provide the best tools for assessing the underlying biology and addressing important questions; (3) Measures of clonal diversity are key to understand the underlying structure and providence of the repertoire; and (4) Convergent evolution may allow a surprising degree of homologous or identical CDR3s associated with individual disease entities, creating hope for novel diagnostics and/or disease burden assessments. Integrating studies of the peripheral blood, lymph nodes, and tumor allows dynamic interrogation of the alterations occurring with age, treatment, and response to emergent and established therapies.
Collapse
Affiliation(s)
- Jian Han
- iRepertoire, Inc., Huntsville, AL, USA.,Hudson Alpha Institute, Huntsville, AL, USA
| | - Michael T Lotze
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
49
|
Scott EM, Jacobus EJ, Lyons B, Frost S, Freedman JD, Dyer A, Khalique H, Taverner WK, Carr A, Champion BR, Fisher KD, Seymour LW, Duffy MR. Bi- and tri-valent T cell engagers deplete tumour-associated macrophages in cancer patient samples. J Immunother Cancer 2019; 7:320. [PMID: 31753017 PMCID: PMC6873687 DOI: 10.1186/s40425-019-0807-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/06/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Tumour-associated macrophages (TAMs) are often implicated in cancer progression but can also exert anti-tumour activities. Selective eradication of cancer-promoting (M2-like) TAM subsets is a highly sought-after goal. Here, we have devised a novel strategy to achieve selective TAM depletion, involving the use of T cell engagers to direct endogenous T cell cytotoxicity towards specific M2-like TAMs. To avoid "on-target off-tumour" toxicities, we have explored localising expression of the T cell engagers to the tumour with enadenotucirev (EnAd), an oncolytic adenovirus in Phase I/II clinical trials. METHOD A panel of bi- and tri-valent T cell engagers (BiTEs/TriTEs) was constructed, recognising CD3ε on T cells and CD206 or folate receptor β (FRβ) on M2-like macrophages. Initial characterisation of BiTE/TriTE activity and specificity was performed with M1- and M2-polarised monocyte-derived macrophages and autologous lymphocytes from healthy human peripheral blood donors. T cell engagers were inserted into the genome of EnAd, and oncolytic activity and BiTE secretion assessed with DLD-1 tumour cells. Clinically-relevant ex vivo models (whole malignant ascites from cancer patients) were employed to assess the efficacies of the free- and virally-encoded T cell engagers. RESULTS T cells activated by the CD206- and FRβ-targeting BiTEs/TriTEs preferentially killed M2- over M1-polarised autologous macrophages, with EC50 values in the nanomolar range. A TriTE with bivalent CD3ε binding - the first of its kind - demonstrated enhanced potency whilst retaining target cell selectivity, whereas a CD28-containing TriTE elicited non-specific T cell activation. In immunosuppressive malignant ascites, both free and EnAd-encoded T cell engagers triggered endogenous T cell activation and IFN-γ production, leading to increased T cell numbers and depletion of CD11b+CD64+ ascites macrophages. Strikingly, surviving macrophages exhibited a general increase in M1 marker expression, suggesting microenvironmental repolarisation towards a pro-inflammatory state. CONCLUSIONS This study is the first to achieve selective depletion of specific M2-like macrophage subsets, opening the possibility of eradicating cancer-supporting TAMs whilst sparing those with anti-tumour potential. Targeted TAM depletion with T cell engager-armed EnAd offers a powerful therapeutic approach combining direct cancer cell cytotoxicity with reversal of immune suppression.
Collapse
Affiliation(s)
- Eleanor M. Scott
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Egon J. Jacobus
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Brian Lyons
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Sally Frost
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | | | - Arthur Dyer
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Hena Khalique
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | | | - Alison Carr
- Churchill Hospital, Oxford University Hospital NHS Trust, Oxford, OX3 7LE UK
| | | | - Kerry D. Fisher
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Len W. Seymour
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | | |
Collapse
|
50
|
Characterization of spatial distribution of tumor-infiltrating CD8 + T cells refines their prognostic utility for pancreatic cancer survival. Mod Pathol 2019; 32:1495-1507. [PMID: 31186528 DOI: 10.1038/s41379-019-0291-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 12/13/2022]
Abstract
The abundance of cytotoxic T-cell infiltrates has important implications for patient outcome and therapeutic design for pancreatic ductal adenocarcinoma. However, intratumoral heterogeneity remains a challenge to understanding the complex immune microenvironment. We hypothesized that characterizing CD8+ cell distribution within pancreatic adenocarcinoma tissues might refine the prognostic value of tumor-infiltrating CD8+ lymphocytes. Using multiplex immunohistochemistry-based image analysis on whole-tissue sections of 214 pancreatic ductal adenocarcinomas, we measured CD8+ cell densities in the tumor center, the tumor margin, and the whole tumor, along with the proximity of CD8+ cells to carcinoma cells. Multivariable Cox regression analysis was performed to assess the associations of CD8+ cell densities with pancreatic cancer-specific survival, adjusting for clinicopathologic and immune-related features, including tumor expressions of TP53, SMAD4, and the programmed cell death 1 ligand 1 (CD274, PD-L1) and the extent of tertiary lymphoid structures. There was substantial heterogeneity in CD8+ cell density, with the mean density in the tumor center less than half that in the tumor margin. Tumor CD274 expression and extensive tertiary lymphoid structures were appeared to be associated with higher CD8+ cell density in the tumor margin (P = 0.037 and P = 0.005, respectively), but not with that in the tumor center (P > 0.50). The association of higher CD8+ cell density with prolonged survival was significant for the whole tumor (Ptrend = 0.009); however, the association was stronger for the tumor center (Ptrend = 0.002) and insignificant for the tumor margin (Ptrend = 0.07). Tumor cell-CD8+ cell distance correlated strongly with CD8+ cell density, whereas the density of CD8+ cells proximate to cancer cells exhibited no prognostic association. In conclusion, spatial computational analysis on pancreatic ductal adenocarcinoma reveals the prognostic validity of CD8+ cell density in the tumor center, where CD8+ cell infiltration is ununiformly restricted, likely suggesting pro-tumorigenic roles of the immunosuppressive tumor microenvironment of pancreatic cancer.
Collapse
|