1
|
Juric V, Mayes E, Binnewies M, Lee T, Canaday P, Pollack JL, Rudolph J, Du X, Liu VM, Dash S, Palmer R, Jahchan NS, Ramoth ÅJ, Lacayo S, Mankikar S, Norng M, Brassell C, Pal A, Chan C, Lu E, Sriram V, Streuli M, Krummel MF, Baker KP, Liang L. TREM1 activation of myeloid cells promotes antitumor immunity. Sci Transl Med 2023; 15:eadd9990. [PMID: 37647386 DOI: 10.1126/scitranslmed.add9990] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/02/2023] [Indexed: 09/01/2023]
Abstract
Myeloid cells in the tumor microenvironment (TME) can exist in immunosuppressive and immunostimulatory states that impede or promote antitumor immunity, respectively. Blocking suppressive myeloid cells or increasing stimulatory cells to enhance antitumor immune responses is an area of interest for therapeutic intervention. Triggering receptor expressed on myeloid cells-1 (TREM1) is a proinflammatory receptor that amplifies immune responses. TREM1 is expressed on neutrophils, subsets of monocytes and tissue macrophages, and suppressive myeloid populations in the TME, including tumor-associated neutrophils, monocytes, and tumor-associated macrophages. Depletion or inhibition of immunosuppressive myeloid cells, or stimulation by TREM1-mediated inflammatory signaling, could be used to promote an immunostimulatory TME. We developed PY159, an afucosylated humanized anti-TREM1 monoclonal antibody with enhanced FcγR binding. PY159 is a TREM1 agonist that induces signaling, leading to up-regulation of costimulatory molecules on monocytes and macrophages, production of proinflammatory cytokines and chemokines, and enhancement of T cell activation in vitro. An antibody against mouse TREM1, PY159m, promoted antitumor efficacy in syngeneic mouse tumor models. These results suggest that PY159-mediated agonism of TREM1 on tumoral myeloid cells can promote a proinflammatory TME and offer a promising strategy for immunotherapy.
Collapse
Affiliation(s)
- Vladislava Juric
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Erin Mayes
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Mikhail Binnewies
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Tian Lee
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Pamela Canaday
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Joshua L Pollack
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Joshua Rudolph
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Xiaoyan Du
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Victoria M Liu
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Subhadra Dash
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Rachael Palmer
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Nadine S Jahchan
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Åsa Johanna Ramoth
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Sergio Lacayo
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Shilpa Mankikar
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Manith Norng
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Chris Brassell
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Aritra Pal
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Christopher Chan
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Erick Lu
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Venkataraman Sriram
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Michel Streuli
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kevin P Baker
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Linda Liang
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| |
Collapse
|
2
|
Gu Y, Lin S, Wu Y, Xu P, Zhu W, Wang Y, Cheng X, Zhang LW, Stauber RH, Wang Y, Gao M. Targeting STING Activation by Antigen-Inspired MnO 2 Nanovaccines Optimizes Tumor Radiotherapy. Adv Healthc Mater 2023; 12:e2300028. [PMID: 36876892 DOI: 10.1002/adhm.202300028] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/27/2023] [Indexed: 03/07/2023]
Abstract
Immune checkpoint blockers therapy can improve the radiotherapy-induced immunosuppression by enhancing interferon secretion, but still suffer from low clinical response rate and potential adverse effects. Mn2+ -mediated activation of interferon gene stimulator (STING) pathway provides an alternative for combination radioimmunotherapy of tumor. However, it is still a challenge for specific delivery of Mn2+ to innate immune cells and targeting activation of STING pathway. Herein, a novel antigen-inspired MnO2 nanovaccine is fabricated as Mn2+ source and functionalized with mannose, enabling it to target innate immune cells to activate the STING pathway. Meanwhile, the release of Mn2+ in the intracellular lysosomes can also be for magnetic resonance imaging to monitor the dynamic distribution of nanovaccines in vivo. The targeting activation of STING pathway can enhance radiotherapy-induced immune responses for inhibiting local and distant tumors, and resisting tumor metastasis. The study proposes an optimized radiotherapy strategy through targeting STING activation of antigen-inspired nanovaccines.
Collapse
Affiliation(s)
- Yuan Gu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Subin Lin
- Department of Orthopedic, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, P. R. China
| | - Yanxian Wu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Pei Xu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Wen Zhu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Yangyun Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Xiaju Cheng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Leshuai W Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Roland H Stauber
- Nanobiomedicine/ENT Department, University Medical Center Mainz, 55131, Mainz, Germany
| | - Yong Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Mingyuan Gao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
3
|
Wang L, Jia Q, Chu Q, Zhu B. Targeting tumor microenvironment for non-small cell lung cancer immunotherapy. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:18-29. [PMID: 39170874 PMCID: PMC11332857 DOI: 10.1016/j.pccm.2022.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/21/2022] [Accepted: 11/23/2022] [Indexed: 08/23/2024]
Abstract
The tumor microenvironment (TME) is composed of different cellular and non-cellular elements. Constant interactions between tumor cells and the TME are responsible for tumor initiation, tumor progression, and responses to therapies. Immune cells in the TME can be classified into two broad categories, namely adaptive and innate immunity. Targeting these immune cells has attracted substantial research and clinical interest. Current research focuses on identifying key molecular players and developing targeted therapies. These approaches may offer more efficient ways of treating different cancers. In this review, we explore the heterogeneity of the TME in non-small cell lung cancer, summarize progress made in targeting the TME in preclinical and clinical studies, discuss the potential predictive value of the TME in immunotherapy, and highlight the promising effects of bispecific antibodies in the era of immunotherapy.
Collapse
Affiliation(s)
- Lei Wang
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| |
Collapse
|
4
|
Tian H, Cao J, Li B, Nice EC, Mao H, Zhang Y, Huang C. Managing the immune microenvironment of osteosarcoma: the outlook for osteosarcoma treatment. Bone Res 2023; 11:11. [PMID: 36849442 PMCID: PMC9971189 DOI: 10.1038/s41413-023-00246-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/17/2022] [Accepted: 12/29/2022] [Indexed: 03/01/2023] Open
Abstract
Osteosarcoma, with poor survival after metastasis, is considered the most common primary bone cancer in adolescents. Notwithstanding the efforts of researchers, its five-year survival rate has only shown limited improvement, suggesting that existing therapeutic strategies are insufficient to meet clinical needs. Notably, immunotherapy has shown certain advantages over traditional tumor treatments in inhibiting metastasis. Therefore, managing the immune microenvironment in osteosarcoma can provide novel and valuable insight into the multifaceted mechanisms underlying the heterogeneity and progression of the disease. Additionally, given the advances in nanomedicine, there exist many advanced nanoplatforms for enhanced osteosarcoma immunotherapy with satisfactory physiochemical characteristics. Here, we review the classification, characteristics, and functions of the key components of the immune microenvironment in osteosarcoma. This review also emphasizes the application, progress, and prospects of osteosarcoma immunotherapy and discusses several nanomedicine-based options to enhance the efficiency of osteosarcoma treatment. Furthermore, we examine the disadvantages of standard treatments and present future perspectives for osteosarcoma immunotherapy.
Collapse
Affiliation(s)
- Hailong Tian
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Jiangjun Cao
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Bowen Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Edouard C. Nice
- grid.1002.30000 0004 1936 7857Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800 Australia
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, 315020, People's Republic of China.
| | - Yi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
5
|
Cancer Stem Cell Relationship with Pro-Tumoral Inflammatory Microenvironment. Biomedicines 2023; 11:biomedicines11010189. [PMID: 36672697 PMCID: PMC9855358 DOI: 10.3390/biomedicines11010189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Inflammatory processes and cancer stem cells (CSCs) are increasingly recognized as factors in the development of tumors. Emerging evidence indicates that CSCs are associated with cancer properties such as metastasis, treatment resistance, and disease recurrence. However, the precise interaction between CSCs and the immune microenvironment remains unexplored. Although evasion of the immune system by CSCs has been extensively studied, new research demonstrates that CSCs can also control and even profit from the immune response. This review provides an overview of the reciprocal interplay between CSCs and tumor-infiltrating immune cells, collecting pertinent data about how CSCs stimulate leukocyte reprogramming, resulting in pro-tumor immune cells that promote metastasis, chemoresistance, tumorigenicity, and even a rise in the number of CSCs. Tumor-associated macrophages, neutrophils, Th17 and regulatory T cells, mesenchymal stem cells, and cancer-associated fibroblasts, as well as the signaling pathways involved in these pro-tumor activities, are among the immune cells studied. Although cytotoxic leukocytes have the potential to eliminate CSCs, immune evasion mechanisms in CSCs and their clinical implications are also known. We intended to compile experimental findings that provide direct evidence of interactions between CSCs and the immune system and CSCs and the inflammatory milieu. In addition, we aimed to summarize key concepts in order to comprehend the cross-talk between CSCs and the tumor microenvironment as a crucial process for the effective design of anti-CSC therapies.
Collapse
|
6
|
Drouin M, Saenz J, Gauttier V, Evrard B, Teppaz G, Pengam S, Mary C, Desselle A, Thepenier V, Wilhelm E, Merieau E, Ligeron C, Girault I, Lopez MD, Fourgeux C, Sinha D, Baccelli I, Moreau A, Louvet C, Josien R, Poschmann J, Poirier N, Chiffoleau E. CLEC-1 is a death sensor that limits antigen cross-presentation by dendritic cells and represents a target for cancer immunotherapy. SCIENCE ADVANCES 2022; 8:eabo7621. [PMID: 36399563 PMCID: PMC9674301 DOI: 10.1126/sciadv.abo7621] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Tumors exploit numerous immune checkpoints, including those deployed by myeloid cells to curtail antitumor immunity. Here, we show that the C-type lectin receptor CLEC-1 expressed by myeloid cells senses dead cells killed by programmed necrosis. Moreover, we identified Tripartite Motif Containing 21 (TRIM21) as an endogenous ligand overexpressed in various cancers. We observed that the combination of CLEC-1 blockade with chemotherapy prolonged mouse survival in tumor models. Loss of CLEC-1 reduced the accumulation of immunosuppressive myeloid cells in tumors and invigorated the activation state of dendritic cells (DCs), thereby increasing T cell responses. Mechanistically, we found that the absence of CLEC-1 increased the cross-presentation of dead cell-associated antigens by conventional type-1 DCs. We identified antihuman CLEC-1 antagonist antibodies able to enhance antitumor immunity in CLEC-1 humanized mice. Together, our results demonstrate that CLEC-1 acts as an immune checkpoint in myeloid cells and support CLEC-1 as a novel target for cancer immunotherapy.
Collapse
Affiliation(s)
- Marion Drouin
- OSE Immunotherapeutics, Nantes, France
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Javier Saenz
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | | | - Berangere Evrard
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | | | | | | | | | | | | | - Emmanuel Merieau
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Camille Ligeron
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | | | - Maria-Dolores Lopez
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Cynthia Fourgeux
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Debajyoti Sinha
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | | | - Aurelie Moreau
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Cedric Louvet
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Regis Josien
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
- CHU Nantes, Nantes Université, Laboratoire d’Immunologie, CIMNA, Nantes, France
| | - Jeremie Poschmann
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | | | - Elise Chiffoleau
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
- Corresponding author.
| |
Collapse
|
7
|
Bizymi N, Matthaiou AM, Matheakakis A, Voulgari I, Aresti N, Zavitsanou K, Karasachinidis A, Mavroudi I, Pontikoglou C, Papadaki HA. New Perspectives on Myeloid-Derived Suppressor Cells and Their Emerging Role in Haematology. J Clin Med 2022; 11:jcm11185326. [PMID: 36142973 PMCID: PMC9504532 DOI: 10.3390/jcm11185326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 12/03/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature cells of myeloid origin that have gained researchers’ attention, as they constitute promising biomarkers and targets for novel therapeutic strategies (i.e., blockage of development, differentiation, depletion, and deactivation) in several conditions, including neoplastic, autoimmune, infective, and inflammatory diseases, as well as pregnancy, obesity, and graft rejection. They are characterised in humans by the typical immunophenotype of CD11b+CD33+HLA-DR–/low and immune-modulating properties leading to decreased T-cell proliferation, induction of T-regulatory cells (T-regs), hindering of natural killer (NK) cell functionality, and macrophage M2-polarisation. The research in the field is challenging, as there are still difficulties in defining cell-surface markers and gating strategies that uniquely identify the different populations of MDSCs, and the currently available functional assays are highly demanding. There is evidence that MDSCs display altered frequency and/or functionality and could be targeted in immune-mediated and malignant haematologic diseases, although there is a large variability of techniques and results between different laboratories. This review presents the current literature concerning MDSCs in a clinical point of view in an attempt to trigger future investigation by serving as a guide to the clinical haematologist in order to apply them in the context of precision medicine as well as the researcher in the field of experimental haematology.
Collapse
Affiliation(s)
- Nikoleta Bizymi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Laboratory of Molecular and Cellular Pneumonology, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Andreas M. Matthaiou
- Laboratory of Molecular and Cellular Pneumonology, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Respiratory Physiology Laboratory, Medical School, University of Cyprus, 2029 Nicosia, Cyprus
| | - Angelos Matheakakis
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Ioanna Voulgari
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Nikoletta Aresti
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Konstantina Zavitsanou
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Anastasios Karasachinidis
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Irene Mavroudi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Charalampos Pontikoglou
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Helen A. Papadaki
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Correspondence: ; Tel.: +30-2810394637
| |
Collapse
|
8
|
Song D, Wei Y, Hu Y, Sun Y, Liu M, Ren Q, Hu Z, Guo Q, Wang Y, Zhou Y. Identification of immunophenotypes in esophageal squamous cell carcinoma based on immune gene sets. Clin Transl Oncol 2022; 24:1100-1114. [PMID: 35098447 DOI: 10.1007/s12094-021-02749-9] [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: 10/13/2021] [Accepted: 12/06/2021] [Indexed: 02/05/2023]
Abstract
PURPOSE Esophageal squamous cell carcinoma (ESCC) is a malignant tumor with high heterogeneity. Research on molecular mechanisms involved in the process of tumor origination and progression is extremely limited to investigating mechanisms of molecular typing for ESCC. METHODS After comprehensively analyzing the gene expression profiles in The Cancer Genome Atlas and Gene Expression Omnibus databases, we identified four immunotypes of ESCC (referred to as C1-C4) based on the gene sets of 28 immune cell subpopulations. The discrepancies in prognostic value, clinical features, drug sensitivity, and tumor components between the immunotypes were individually analyzed. RESULTS The ranking of immune infiltration is C1 > C4 > C3 > C2. These subtypes are characterized by high and low expression of immune checkpoint proteins, enrichment and insufficiency of immune-related pathways, and differential distribution of immune cell subgroups. Poorer survival was observed in the C1 subtype, which we hypothesized could be caused by an immunosuppressive cell population. Fortunately, C1's susceptibility to anti-PD-1 therapy offers hope for patients with poor prognosis in advanced stages. On the other hand, C4 is sensitive to docetaxel, which may offer novel treatment strategies for ESCC in the future. It is worth noting that immunophenotyping is tightly bound to the abundance of stromal components and stem cells, which could explain the tumor immune escape to some extent. Ultimately, determination of hub genes based on the C1 subtypes provides a reference for the discovery of immunotarget drugs against ESCC. CONCLUSION The identification of immunophenotypes in our study provides new therapeutic strategies for patients with ESCC.
Collapse
Affiliation(s)
- Danlei Song
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Yongjian Wei
- The First Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Yuping Hu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Hospital of Reproductive Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yueting Sun
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Min Liu
- Department of Gastroenterology, Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Qian Ren
- Department of Gastroenterology, Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Zenan Hu
- Department of Gastroenterology, Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Qinghong Guo
- Department of Gastroenterology, Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Yuping Wang
- Department of Gastroenterology, Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Yongning Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China.
- Department of Gastroenterology, Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
9
|
Guan X, Sun L, Shen Y, Jin F, Bo X, Zhu C, Han X, Li X, Chen Y, Xu H, Yue W. Nanoparticle-enhanced radiotherapy synergizes with PD-L1 blockade to limit post-surgical cancer recurrence and metastasis. Nat Commun 2022; 13:2834. [PMID: 35595770 PMCID: PMC9123179 DOI: 10.1038/s41467-022-30543-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 05/05/2022] [Indexed: 01/22/2023] Open
Abstract
Cancer recurrence after surgical resection (SR) is a considerable challenge, and the biological effect of SR on the tumor microenvironment (TME) that is pivotal in determining postsurgical treatment efficacy remains poorly understood. Here, with an experimental model, we demonstrate that the genomic landscape shaped by SR creates an immunosuppressive milieu characterized by hypoxia and high-influx of myeloid cells, fostering cancer progression and hindering PD-L1 blockade therapy. To address this issue, we engineer a radio-immunostimulant nanomedicine (IPI549@HMP) capable of targeting myeloid cells, and catalyzing endogenous H2O2 into O2 to achieve hypoxia-relieved radiotherapy (RT). The enhanced RT-mediated immunogenic effect results in postsurgical TME reprogramming and increased susceptibility to anti-PD-L1 therapy, which can suppress/eradicate locally residual and distant tumors, and elicits strong immune memory effects to resist tumor rechallenge. Our radioimmunotherapy points to a simple and effective therapeutic intervention against postsurgical cancer recurrence and metastasis. Tumor recurrence after surgical resection is associated with a poor clinical outcome. Here the authors design a manganese dioxide-based nanosystem to increase response to radio-immunotherapy by relieving tumor hypoxia and targeting myeloid cells, showing reduced post-surgical cancer recurrence and metastasis.
Collapse
Affiliation(s)
- Xin Guan
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Liping Sun
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Yuting Shen
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Fengshan Jin
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Xiaowan Bo
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Chunyan Zhu
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Xiaoxia Han
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Xiaolong Li
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P.R. China.,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Huixiong Xu
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China. .,Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P.R. China. .,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China. .,Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.
| | - Wenwen Yue
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P.R. China. .,Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P.R. China. .,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China.
| |
Collapse
|
10
|
Li S, Zhu C, Zhou X, Chen L, Bo X, Shen Y, Guan X, Han X, Shan D, Sun L, Chen Y, Xu H, Yue W. Engineering ROS-Responsive Bioscaffolds for Disrupting Myeloid Cell-Driven Immunosuppressive Niche to Enhance PD-L1 Blockade-Based Postablative Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104619. [PMID: 35156339 PMCID: PMC9008797 DOI: 10.1002/advs.202104619] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 01/18/2022] [Indexed: 02/06/2023]
Abstract
The existence of inadequate ablation remains an important cause of treatment failure for loco-regional ablation therapies. Here, using a preclinical model, it is reported that inadequate microwave ablation (iMWA) induces immunosuppressive niche predominated by myeloid cells. The gene signature of ablated tumor presented by transcriptome analyses is highly correlated with immune checkpoint blocking (ICB) resistance. Thus, an in situ scaffold with synergistic delivery of IPI549 and anti-programmed death-ligand 1 blocking antibody (aPDL1) for postablative cancer immunotherapy is designed and engineered, in which IPI549 capable of targeting myeloid cells could disrupt the immunosuppressive niche and subsequently improve ICB-mediated antitumor immune response. Based on five mouse cancer models, it is demonstrated that this biomaterial system (aPDL1&IPI549@Gel) could mimic a "hot" tumor-immunity niche to inhibit tumor progression and metastasis, and protect cured mice against tumor rechallenge. This work enables a new standard-of-care paradigm for the immunotherapy of myeloid cells-mediated "cold" tumors after loco-regional inadequate practices.
Collapse
Affiliation(s)
- Shaoyue Li
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteSchool of MedicineTongji UniversityShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
- Department of In‐patient UltrasoundThe Second Affiliated HospitalHarbin Medical UniversityHarbin150001P. R. China
| | - Chunyan Zhu
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteSchool of MedicineTongji UniversityShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
| | - Xianli Zhou
- Department of In‐patient UltrasoundThe Second Affiliated HospitalHarbin Medical UniversityHarbin150001P. R. China
| | - Liang Chen
- Department of GastroenterologyShanghai Tenth People's HospitalTongji University School of MedicineShanghai200072P. R. China
| | - Xiaowan Bo
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteSchool of MedicineTongji UniversityShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
| | - Yuting Shen
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteSchool of MedicineTongji UniversityShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
| | - Xin Guan
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteSchool of MedicineTongji UniversityShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
| | - Xiaoxia Han
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteSchool of MedicineTongji UniversityShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
| | - Dandan Shan
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteSchool of MedicineTongji UniversityShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
| | - Liping Sun
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteSchool of MedicineTongji UniversityShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
| | - Yu Chen
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteSchool of MedicineTongji UniversityShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Huixiong Xu
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteSchool of MedicineTongji UniversityShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
- Department of UltrasoundZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Wenwen Yue
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteSchool of MedicineTongji UniversityShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
| |
Collapse
|
11
|
Zhao W, Xu D, Zhang L, Meng H, Zheng Q, Wang J. Anti-inflammation of torachrysone-8-O-β-ᴅ-glucoside by hurdling over morphological changes of macrophages. Int Immunopharmacol 2022; 105:108548. [DOI: 10.1016/j.intimp.2022.108548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/02/2022] [Accepted: 01/13/2022] [Indexed: 12/20/2022]
|
12
|
Wilczyński JR, Wilczyński M, Paradowska E. Cancer Stem Cells in Ovarian Cancer-A Source of Tumor Success and a Challenging Target for Novel Therapies. Int J Mol Sci 2022; 23:ijms23052496. [PMID: 35269636 PMCID: PMC8910575 DOI: 10.3390/ijms23052496] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
Ovarian cancer is the most lethal neoplasm of the female genital organs. Despite indisputable progress in the treatment of ovarian cancer, the problems of chemo-resistance and recurrent disease are the main obstacles for successful therapy. One of the main reasons for this is the presence of a specific cell population of cancer stem cells. The aim of this review is to show the most contemporary knowledge concerning the biology of ovarian cancer stem cells (OCSCs) and their impact on chemo-resistance and prognosis in ovarian cancer patients, as well as to present the treatment options targeted exclusively on the OCSCs. The review presents data concerning the role of cancer stem cells in general and then concentrates on OCSCs. The surface and intracellular OCSCs markers and their meaning both for cancer biology and clinical prognosis, signaling pathways specifically activated in OCSCs, the genetic and epigenetic regulation of OCSCs function including the recent studies on the non-coding RNA regulation, cooperation between OCSCs and the tumor microenvironment (ovarian cancer niche) including very specific environment such as ascites fluid, the role of shear stress, autophagy and metabolic changes for the function of OCSCs, and finally mechanisms of OCSCs escape from immune surveillance, are described and discussed extensively. The possibilities of anti-OCSCs therapy both in experimental settings and in clinical trials are presented, including the recent II phase clinical trials and immunotherapy. OCSCs are a unique population of cancer cells showing a great plasticity, self-renewal potential and resistance against anti-cancer treatment. They are responsible for the progression and recurrence of the tumor. Several completed and ongoing clinical trials have tested different anti-OCSCs drugs which, however, have shown unsatisfactory efficacy in most cases. We propose a novel approach to ovarian cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Jacek R Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
- Correspondence:
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother’s Health Center—Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland;
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland;
| |
Collapse
|
13
|
Li J, Bolyard C, Xin G, Li Z. Targeting Metabolic Pathways of Myeloid Cells Improves Cancer Immunotherapy. Front Cell Dev Biol 2022; 9:747863. [PMID: 34988072 PMCID: PMC8721007 DOI: 10.3389/fcell.2021.747863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/22/2021] [Indexed: 01/20/2023] Open
Abstract
Tumor-infiltrating myeloid cells are a prominent pro-tumorigenic immune cell population that limit host anti-tumor immunity and present a significant obstacle for many cancer immunotherapies. Targeting the mechanisms regulating myeloid cell function within the tumor microenvironment may overcome immunotherapy resistance in some cancers. Recent discoveries in the emerging field of immunometabolism reveal that the metabolic profiles of intratumoral myeloid cells are rewired to adapt to the nutrition-limited tumor microenvironment, and this shapes their pro-tumor phenotypes. Interestingly, metabolic modulation can shift these myeloid cells toward the immune-stimulating anti-tumor phenotype. In this review, we will highlight the roles of specific metabolic pathways in the activation and function of myeloid cells, and discuss the therapeutic value of metabolically reprogramming myeloid cells to augment and improve outcomes with cancer immunotherapy.
Collapse
Affiliation(s)
- Jianying Li
- Pelotonia Institute of Immuno-Oncology, the Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, United States
| | - Chelsea Bolyard
- Pelotonia Institute of Immuno-Oncology, the Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, United States
| | - Gang Xin
- Pelotonia Institute of Immuno-Oncology, the Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, United States.,Department of Microbial Infection and Immunity, the Ohio State University College of Medicine, Columbus, OH, United States
| | - Zihai Li
- Pelotonia Institute of Immuno-Oncology, the Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, United States.,Department of Medical Oncology, the Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|
14
|
Hanssens H, Meeus F, De Veirman K, Breckpot K, Devoogdt N. The antigen-binding moiety in the driver's seat of CARs. Med Res Rev 2022; 42:306-342. [PMID: 34028069 PMCID: PMC9292017 DOI: 10.1002/med.21818] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 02/17/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022]
Abstract
Immuno-oncology has been at the forefront of cancer treatment in recent decades. In particular immune checkpoint and chimeric antigen receptor (CAR)-T cell therapy have achieved spectacular results. Over the years, CAR-T cell development has followed a steady evolutionary path, focusing on increasing T cell potency and sustainability, which has given rise to different CAR generations. However, there was less focus on the mode of interaction between the CAR-T cell and the cancer cell; more specifically on the targeting moiety used in the CAR and its specific properties. Recently, the importance of optimizing this domain has been recognized and the possibilities have been exploited. Over the last 10 years-in addition to the classical scFv-based CARs-single domain CARs, natural receptor-ligand CARs, universal CARs and CARs targeting more than one antigen have emerged. In addition, the specific parameters of the targeting domain and their influence on T cell activation are being examined. In this review, we concisely present the history of CAR-T cell therapy, and then expand on various developments in the CAR ectodomain. We discuss different formats, each with their own advantages and disadvantages, as well as the developments in affinity tuning, avidity effects, epitope location, and influence of the extracellular spacer.
Collapse
Affiliation(s)
- Heleen Hanssens
- In Vivo Cellular and Molecular Imaging LaboratoryVrije Universiteit BrusselBrusselsBelgium
- Laboratory of Hematology and ImmunologyVrije Universiteit BrusselBrusselsBelgium
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical SciencesVrije Universiteit BrusselBrusselsBelgium
| | - Fien Meeus
- In Vivo Cellular and Molecular Imaging LaboratoryVrije Universiteit BrusselBrusselsBelgium
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical SciencesVrije Universiteit BrusselBrusselsBelgium
| | - Kim De Veirman
- Laboratory of Hematology and ImmunologyVrije Universiteit BrusselBrusselsBelgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical SciencesVrije Universiteit BrusselBrusselsBelgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging LaboratoryVrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
15
|
Lyu N, Yi JZ, Zhao M. Immunotherapy in older patients with hepatocellular carcinoma. Eur J Cancer 2021; 162:76-98. [PMID: 34954439 DOI: 10.1016/j.ejca.2021.11.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/31/2021] [Accepted: 11/21/2021] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common types of cancer globally and is currently the third leading cause of cancer-related deaths. Recently, immunotherapy using immune checkpoint inhibitors (ICIs) has been shown with encouraging anticancer activity and safety in clinical trials. To reverse the phenomenon of tumours evading immune response, ICIs can be used to stimulate the natural antitumour potential of cancer cells by blocking the relevant checkpoints to activate T cells. However, the components and functions of the immune system may undergo a series of changes with ageing, known as 'immunosenescence,' potentially affecting the antitumour effect and safety of immunotherapy. In the current phase III clinical trials of ICIs including nivolumab, pembrolizumab and atezolizumab, the proportion of patients with HCC older than 65 years in CheckMate 459, KEYNOTE-240 and IMbrave150 is 51%, 58% and 50%, respectively, which is less than 70%-73% of epidemiological investigation. Therefore, the elderly population recruited in clinical trials may not accurately represent the real-world elderly patients with HCC, which affects the extrapolation of the efficacy and safety profile obtained in clinical trials to the elderly population in the real world. This review provides the latest advances in ICIs immuno-treatment available for HCC and relevant information about their therapeutic effects and safety on elderly patients. We discuss the benefits of ICIs for older HCC patients, and relevant recommendations about conducting further clinical trials are proposed for more complete answers to this clinical issue.
Collapse
Affiliation(s)
- Ning Lyu
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jun-Zhe Yi
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ming Zhao
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
16
|
Multifaceted Roles of Chemokines and Chemokine Receptors in Tumor Immunity. Cancers (Basel) 2021; 13:cancers13236132. [PMID: 34885241 PMCID: PMC8656932 DOI: 10.3390/cancers13236132] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Various immune cells are involved in host immune responses to cancer. T-helper (Th) 1 cells, cytotoxic CD8+ T cells, and natural killer cells are the major effector cells in anti-tumor immunity, whereas cells such as regulatory T cells and myeloid-derived suppressor cells are negatively involved in anti-tumor immunity. Th2 cells and Th17 cells have been shown to have both pro-tumor and anti-tumor activities. The migratory properties of various immune cells are essential for their function and critically regulated by the chemokine superfamily. In this review, we summarize the roles of various immune cells in tumor immunity and their migratory regulation by the chemokine superfamily. We also assess the therapeutic possibilities of targeting chemokines and chemokine receptors in cancer immunotherapy. Abstract Various immune cells are involved in host tumor immune responses. In particular, there are many T cell subsets with different roles in tumor immunity. T-helper (Th) 1 cells are involved in cellular immunity and thus play the major role in host anti-tumor immunity by inducing and activating cytotoxic T lymphocytes (CTLs). On the other hand, Th2 cells are involved in humoral immunity and suppressive to Th1 responses. Regulatory T (Treg) cells negatively regulate immune responses and contribute to immune evasion of tumor cells. Th17 cells are involved in inflammatory responses and may play a role in tumor progression. However, recent studies have also shown that Th17 cells are capable of directly inducting CTLs and thus may promote anti-tumor immunity. Besides these T cell subsets, there are many other innate immune cells such as dendritic cells (DCs), natural killer (NK) cells, and myeloid-derived suppressor cells (MDSCs) that are involved in host immune responses to cancer. The migratory properties of various immune cells are critical for their functions and largely regulated by the chemokine superfamily. Thus, chemokines and chemokine receptors play vital roles in the orchestration of host immune responses to cancer. In this review, we overview the various immune cells involved in host responses to cancer and their migratory properties regulated by the chemokine superfamily. Understanding the roles of chemokines and chemokine receptors in host immune responses to cancer may provide new therapeutic opportunities for cancer immunotherapy.
Collapse
|
17
|
Wu B, Zhong C, Lang Q, Liang Z, Zhang Y, Zhao X, Yu Y, Zhang H, Xu F, Tian Y. Poliovirus receptor (PVR)-like protein cosignaling network: new opportunities for cancer immunotherapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:267. [PMID: 34433460 PMCID: PMC8390200 DOI: 10.1186/s13046-021-02068-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/10/2021] [Indexed: 12/14/2022]
Abstract
Immune checkpoint molecules, also known as cosignaling molecules, are pivotal cell-surface molecules that control immune cell responses by either promoting (costimulatory molecules) or inhibiting (coinhibitory molecules) a signal. These molecules have been studied for many years. The application of immune checkpoint drugs in the clinic provides hope for cancer patients. Recently, the poliovirus receptor (PVR)-like protein cosignaling network, which involves several immune checkpoint receptors, i.e., DNAM-1 (DNAX accessory molecule-1, CD226), TIGIT (T-cell immunoglobulin (Ig) and immunoreceptor tyrosine-based inhibitory motif (ITIM)), CD96 (T cell activation, increased late expression (TACLILE)), and CD112R (PVRIG), which interact with their ligands CD155 (PVR/Necl-5), CD112 (PVRL2/nectin-2), CD111 (PVRL1/nectin-1), CD113 (PVRL3/nectin-3), and Nectin4, was discovered. As important components of the immune system, natural killer (NK) and T cells play a vital role in eliminating and killing foreign pathogens and abnormal cells in the body. Recently, increasing evidence has suggested that this novel cosignaling network axis costimulates and coinhibits NK and T cell activation to eliminate cancer cells after engaging with ligands, and this activity may be effectively targeted for cancer immunotherapy. In this article, we review recent advances in research on this novel cosignaling network. We also briefly outline the structure of this cosignaling network, the signaling cascades and mechanisms involved after receptors engage with ligands, and how this novel cosignaling network costimulates and coinhibits NK cell and T cell activation for cancer immunotherapy. Additionally, this review comprehensively summarizes the application of this new network in preclinical trials and clinical trials. This review provides a new immunotherapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Baokang Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Chongli Zhong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Qi Lang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Zhiyun Liang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Yizhou Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Xin Zhao
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Yang Yu
- Department of Surgery, Jinzhou Medical University, Jinzhou, 121001, Liaoning Province, China
| | - Heming Zhang
- Department of College of Medical and Biological Information Engineering, Northeastern University, Shenyang, 110819, Liaoning Province, China
| | - Feng Xu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
18
|
Hot or cold: Bioengineering immune contextures into in vitro patient-derived tumor models. Adv Drug Deliv Rev 2021; 175:113791. [PMID: 33965462 DOI: 10.1016/j.addr.2021.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, immune checkpoint inhibitors (ICI) have proven to be tremendously effective for a subset of cancer patients. However, it is difficult to predict the response of individual patients and efforts are now directed at understanding the mechanisms of ICI resistance. Current models of patient tumors poorly recapitulate the immune contexture, which describe immune parameters that are associated with patient survival. In this Review, we discuss parameters that influence the induction of different immune contextures found within tumors and how engineering strategies may be leveraged to recapitulate these contextures to develop the next generation of immune-competent patient-derived in vitro models.
Collapse
|
19
|
Zhang H, Zhu G. Beyond Promoter: The Role of Macrophage in Invasion and Progression of Renal Cell Carcinoma. Curr Stem Cell Res Ther 2021; 15:588-596. [PMID: 32096752 DOI: 10.2174/1574888x15666200225093210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/28/2019] [Accepted: 12/11/2019] [Indexed: 11/22/2022]
Abstract
Renal cell carcinoma (RCC) is one of the common urologic neoplasms, and its incidence has been increasing over the past several decades; however, its pathogenesis is still unknown up to now. Recent studies have found that in addition to tumor cells, other cells in the tumor microenvironment also affect the biological behavior of the tumor. Among them, macrophages exist in a large amount in tumor microenvironment, and they are generally considered to play a key role in promoting tumorigenesis. Therefore, we summarized the recent researches on macrophage in the invasiveness and progression of RCC in latest years, and we also introduced and discussed many studies about macrophage in RCC to promote angiogenesis by changing tumor microenvironment and inhibit immune response in order to activate tumor progression. Moreover, macrophage interactes with various cytokines to promote tumor proliferation, invasion and metastasis, and it also promotes tumor stem cell formation and induces drug resistance in the progression of RCC. The highlight of this review is to make a summary of the roles of macrophage in the invasion and progression of RCC; at the same time to raise some potential and possible targets for future RCC therapy.
Collapse
Affiliation(s)
- Haibao Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Guodong Zhu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
20
|
Rafael TS, de Vries HM, Ottenhof SR, Hofland I, Broeks A, de Jong J, Bekers E, Horenblas S, de Menezes RX, Jordanova ES, Brouwer OR. Distinct Patterns of Myeloid Cell Infiltration in Patients With hrHPV-Positive and hrHPV-Negative Penile Squamous Cell Carcinoma: The Importance of Assessing Myeloid Cell Densities Within the Spatial Context of the Tumor. Front Immunol 2021; 12:682030. [PMID: 34194435 PMCID: PMC8236714 DOI: 10.3389/fimmu.2021.682030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/24/2021] [Indexed: 12/13/2022] Open
Abstract
Comprehensive analysis of tumor infiltrating myeloid cells in the tumor microenvironment of penile squamous cell carcinoma (PSCC) is lacking. In this retrospective study, for the first time, PSCC resection specimens (N = 103) were annotated into the following compartments: intratumoral tumor (IT Tumor), intratumoral stroma (IT Stroma), peritumoral tumor (PT Tumor) and peritumoral stroma (PT Stroma) compartments. We then quantified CD14+, CD68+ and CD163+ myeloid cells within these compartments using an image analysis software and assessed their association with various clinical parameters, including high-risk human papillomavirus (hrHPV) status. In the total cohort, hrHPV status, grade of differentiation, age and tumor size were associated with myeloid cell densities. hrHPV+ tumors had higher infiltration rates of CD14+, CD68+ and CD163+ myeloid cells in the IT tumor compartment (p < 0.001, for all) compared to hrHPV- tumors. Furthermore, when examining the association between compartment-specific infiltration and differentiation grade, increased myeloid cell densities in the IT tumor compartment were associated with a more advanced histological grade (p < 0.001, for all). This association remained significant when the hrHPV- cohort (N = 60) was analyzed (CD14+ p = 0.001; CD68+ p < 0.001; CD163+ p = 0.004). Subgroup analysis in the hrHPV+ group (N = 43) showed that high infiltration rates of CD68+ and CD163+ cells in the PT tumor compartment were associated with lymph node (LN) metastasis (p = 0.031 and p = 0.026, respectively). Regarding the association between myeloid cell densities and disease-specific survival, the risk of death was found to decrease slightly as the number of myeloid cells in the IT tumor compartment increased (CD14+ p = 0.04; CD68+ p = 0.05; CD163+ p = 0.02). However, after adjusting for hrHPV, no independent association between myeloid densities and disease-specific survival were found. Altogether, these findings demonstrate the importance of assessing myeloid cell densities within the spatial context of the tumor. Further studies are needed to unravel the specific phenotype of myeloid cells residing in the different compartments, their effect on clinical parameters and the impact of hrHPV on the recruitment of myeloid cell populations in PSCC.
Collapse
Affiliation(s)
- Tynisha S Rafael
- Department of Urology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Hielke M de Vries
- Department of Urology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Sarah R Ottenhof
- Department of Urology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ingrid Hofland
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jeroen de Jong
- Department of Pathology, Reinier Haga Medisch Diagnostisch Centrum (MDC), The Hague, Netherlands
| | - Elise Bekers
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Simon Horenblas
- Department of Urology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Renée X de Menezes
- Biostatistics Center, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ekaterina S Jordanova
- Department of Urology, Netherlands Cancer Institute, Amsterdam, Netherlands.,Center for Gynecologic Oncology Amsterdam (CGOA), Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Oscar R Brouwer
- Department of Urology, Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
21
|
Maes K, Mondino A, Lasarte JJ, Agirre X, Vanderkerken K, Prosper F, Breckpot K. Epigenetic Modifiers: Anti-Neoplastic Drugs With Immunomodulating Potential. Front Immunol 2021; 12:652160. [PMID: 33859645 PMCID: PMC8042276 DOI: 10.3389/fimmu.2021.652160] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/09/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer cells are under the surveillance of the host immune system. Nevertheless, a number of immunosuppressive mechanisms allow tumors to escape protective responses and impose immune tolerance. Epigenetic alterations are central to cancer cell biology and cancer immune evasion. Accordingly, epigenetic modulating agents (EMAs) are being exploited as anti-neoplastic and immunomodulatory agents to restore immunological fitness. By simultaneously acting on cancer cells, e.g. by changing expression of tumor antigens, immune checkpoints, chemokines or innate defense pathways, and on immune cells, e.g. by remodeling the tumor stroma or enhancing effector cell functionality, EMAs can indeed overcome peripheral tolerance to transformed cells. Therefore, combinations of EMAs with chemo- or immunotherapy have become interesting strategies to fight cancer. Here we review several examples of epigenetic changes critical for immune cell functions and tumor-immune evasion and of the use of EMAs in promoting anti-tumor immunity. Finally, we provide our perspective on how EMAs could represent a game changer for combinatorial therapies and the clinical management of cancer.
Collapse
Affiliation(s)
- Ken Maes
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, Vrije Universiteit Brussel (VUB), Universiteit Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Anna Mondino
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Juan José Lasarte
- Immunology and Immunotherapy Program, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | - Xabier Agirre
- Laboratory of Cancer Epigenetics, Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Pamplona, Spain.,Hemato-oncology Program, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | - Karin Vanderkerken
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Felipe Prosper
- Laboratory of Cancer Epigenetics, Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Pamplona, Spain.,Hemato-oncology Program, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain.,Hematology and Cell Therapy Department, Clínica Universidad de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
22
|
Van Hoecke L, Verbeke R, Dewitte H, Lentacker I, Vermaelen K, Breckpot K, Van Lint S. mRNA in cancer immunotherapy: beyond a source of antigen. Mol Cancer 2021; 20:48. [PMID: 33658037 PMCID: PMC7926200 DOI: 10.1186/s12943-021-01329-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 02/01/2021] [Indexed: 02/08/2023] Open
Abstract
mRNA therapeutics have become the focus of molecular medicine research. Various mRNA applications have reached major milestones at high speed in the immuno-oncology field. This can be attributed to the knowledge that mRNA is one of nature's core building blocks carrying important information and can be considered as a powerful vector for delivery of therapeutic proteins to the patient.For a long time, the major focus in the use of in vitro transcribed mRNA was on development of cancer vaccines, using mRNA encoding tumor antigens to modify dendritic cells ex vivo. However, the versatility of mRNA and its many advantages have paved the path beyond this application. In addition, due to smart design of both the structural properties of the mRNA molecule as well as pharmaceutical formulations that improve its in vivo stability and selective targeting, the therapeutic potential of mRNA can be considered as endless.As a consequence, many novel immunotherapeutic strategies focus on the use of mRNA beyond its use as the source of tumor antigens. This review aims to summarize the state-of-the-art on these applications and to provide a rationale for their clinical application.
Collapse
Affiliation(s)
- Lien Van Hoecke
- VIB-UGent Center for Inflammation Research, Technologiepark 71, 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, 9052 Ghent, Belgium
| | - Rein Verbeke
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Karim Vermaelen
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Tumor Immunology Laboratory, Department of Respiratory Medicine and Immuno-Oncology Network Ghent, Ghent University Hospital, Corneel Heymanslaan 10 MRB2, 9000 Ghent, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103 Building E, 1090 Brussels, Belgium
| | - Sandra Van Lint
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Tumor Immunology Laboratory, Department of Respiratory Medicine and Immuno-Oncology Network Ghent, Ghent University Hospital, Corneel Heymanslaan 10 MRB2, 9000 Ghent, Belgium
| |
Collapse
|
23
|
Campesato LF, Weng CH, Merghoub T. Innate immune checkpoints for cancer immunotherapy: expanding the scope of non T cell targets. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1031. [PMID: 32953831 PMCID: PMC7475486 DOI: 10.21037/atm-20-1816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Luis F Campesato
- Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chien-Huan Weng
- Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
24
|
Faramand R, Jain M, Staedtke V, Kotani H, Bai R, Reid K, Lee SB, Spitler K, Wang X, Cao B, Pinilla J, Lazaryan A, Khimani F, Shah B, Chavez JC, Nishihori T, Mishra A, Mullinax J, Gonzalez R, Hussaini M, Dam M, Brandjes BD, Bachmeier CA, Anasetti C, Locke FL, Davila ML. Tumor Microenvironment Composition and Severe Cytokine Release Syndrome (CRS) Influence Toxicity in Patients with Large B-Cell Lymphoma Treated with Axicabtagene Ciloleucel. Clin Cancer Res 2020; 26:4823-4831. [PMID: 32669372 DOI: 10.1158/1078-0432.ccr-20-1434] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/14/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE One of the challenges of adoptive T-cell therapy is the development of immune-mediated toxicities including cytokine release syndrome (CRS) and neurotoxicity (NT). We aimed to identify factors that place patients at high risk of severe toxicity or treatment-related death in a cohort of 75 patients with large B-cell lymphoma treated with a standard of care CD19 targeted CAR T-cell product (axicabtagene ciloleucel). EXPERIMENTAL DESIGN Serum cytokine and catecholamine levels were measured prior to lymphodepleting chemotherapy, on the day of CAR T infusion and daily thereafter while patients remained hospitalized. Tumor biopsies were taken within 1 month prior to CAR T infusion for evaluation of gene expression. RESULTS We identified an association between pretreatment levels of IL6 and life-threatening CRS and NT. Because the risk of toxicity was related to pretreatment factors, we hypothesized that the tumor microenvironment (TME) may influence CAR T-cell toxicity. In pretreatment patient tumor biopsies, gene expression of myeloid markers was associated with higher toxicity. CONCLUSIONS These results suggest that a proinflammatory state and an unfavorable TME preemptively put patients at risk for toxicity after CAR T-cell therapy. Tailoring toxicity management strategies to patient risk may reduce morbidity and mortality.
Collapse
Affiliation(s)
- Rawan Faramand
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Michael Jain
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Verena Staedtke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hiroshi Kotani
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Renyuan Bai
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kayla Reid
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Sae Bom Lee
- Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Kristen Spitler
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Biwei Cao
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Javier Pinilla
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aleksander Lazaryan
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Farhad Khimani
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Bijal Shah
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Julio C Chavez
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Asmita Mishra
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - John Mullinax
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Sarcoma, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Ricardo Gonzalez
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Sarcoma, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mohammad Hussaini
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Marian Dam
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Brigett D Brandjes
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Christina A Bachmeier
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Claudio Anasetti
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Frederick L Locke
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Marco L Davila
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida. .,Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
25
|
Qu J, Jiang M, Wang L, Zhao D, Qin K, Wang Y, Tao J, Zhang X. Mechanism and potential predictive biomarkers of immune checkpoint inhibitors in NSCLC. Biomed Pharmacother 2020; 127:109996. [DOI: 10.1016/j.biopha.2020.109996] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/31/2020] [Accepted: 02/05/2020] [Indexed: 12/20/2022] Open
|
26
|
Neophytou CM, Pierides C, Christodoulou MI, Costeas P, Kyriakou TC, Papageorgis P. The Role of Tumor-Associated Myeloid Cells in Modulating Cancer Therapy. Front Oncol 2020; 10:899. [PMID: 32656079 PMCID: PMC7325995 DOI: 10.3389/fonc.2020.00899] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022] Open
Abstract
Myeloid cells include various cellular subtypes that are distinguished into mononuclear and polymorphonuclear cells, derived from either common myeloid progenitor cells (CMPs) or myeloid stem cells. They play pivotal roles in innate immunity since, following invasion by pathogens, myeloid cells are recruited and initiate phagocytosis and secretion of inflammatory cytokines into local tissues. Moreover, mounting evidence suggests that myeloid cells may also regulate cancer development by infiltrating the tumor to directly interact with cancer cells or by affecting the tumor microenvironment. Importantly, mononuclear phagocytes, including macrophages and dendritic cells (DCs), can have either a positive or negative impact on the efficacy of chemotherapy, radiotherapy as well as targeted anti-cancer therapies. Tumor-associated macrophages (TAMs), profusely found in the tumor stroma, can promote resistance to chemotherapeutic drugs, such as Taxol and Paclitaxel, whereas the suppression of TAMs can lead to an improved radiotherapy outcome. On the contrary, the presence of TAMs may be beneficial for targeted therapies as they can facilitate the accumulation of large quantities of nanoparticles carrying therapeutic compounds. Tumor infiltrating DCs, however, are generally thought to enhance cytotoxic therapies, including those using anthracyclines. This review focuses on the role of tumor-infiltrating and stroma myeloid cells in modulating tumor responses to various treatments. We herein report the impact of myeloid cells in a number of therapeutic approaches across a wide range of malignancies, as well as the efforts toward the elimination of myeloid cells or the exploitation of their presence for the enhancement of therapeutic efficacy against cancer.
Collapse
Affiliation(s)
- Christiana M Neophytou
- European University Research Centre, Nicosia, Cyprus.,Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Chryso Pierides
- The Center for the Study of Haematological Malignancies, Nicosia, Cyprus
| | | | - Paul Costeas
- The Center for the Study of Haematological Malignancies, Nicosia, Cyprus.,The Cyprus Cancer Research Institute, Nicosia, Cyprus
| | | | - Panagiotis Papageorgis
- European University Research Centre, Nicosia, Cyprus.,Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| |
Collapse
|
27
|
Chaib M, Chauhan SC, Makowski L. Friend or Foe? Recent Strategies to Target Myeloid Cells in Cancer. Front Cell Dev Biol 2020; 8:351. [PMID: 32509781 PMCID: PMC7249856 DOI: 10.3389/fcell.2020.00351] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) is a complex network of epithelial and stromal cells, wherein stromal components provide support to tumor cells during all stages of tumorigenesis. Among these stromal cell populations are myeloid cells, which are comprised mainly of tumor-associated macrophages (TAM), dendritic cells (DC), myeloid-derived suppressor cells (MDSC), and tumor-associated neutrophils (TAN). Myeloid cells play a major role in tumor growth through nurturing cancer stem cells by providing growth factors and metabolites, increasing angiogenesis, as well as promoting immune evasion through the creation of an immune-suppressive microenvironment. Immunosuppression in the TME is achieved by preventing critical anti-tumor immune responses by natural killer and T cells within the primary tumor and in metastatic niches. Therapeutic success in targeting myeloid cells in malignancies may prove to be an effective strategy to overcome chemotherapy and immunotherapy limitations. Current therapeutic approaches to target myeloid cells in various cancers include inhibition of their recruitment, alteration of function, or functional re-education to an antitumor phenotype to overcome immunosuppression. In this review, we describe strategies to target TAMs and MDSCs, consisting of single agent therapies, nanoparticle-targeted approaches and combination therapies including chemotherapy and immunotherapy. We also summarize recent molecular targets that are specific to myeloid cell populations in the TME, while providing a critical review of the limitations of current strategies aimed at targeting a single subtype of the myeloid cell compartment. The goal of this review is to provide the reader with an understanding of the critical role of myeloid cells in the TME and current therapeutic approaches including ongoing or recently completed clinical trials.
Collapse
Affiliation(s)
- Mehdi Chaib
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Subhash C Chauhan
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Liza Makowski
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Hematology Oncology, Department of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States.,Center for Cancer Research, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
28
|
Pastò A, Consonni FM, Sica A. Influence of Innate Immunity on Cancer Cell Stemness. Int J Mol Sci 2020; 21:ijms21093352. [PMID: 32397392 PMCID: PMC7247585 DOI: 10.3390/ijms21093352] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/02/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Even if cancer stem cells (CSCs) represent only a small proportion of the tumor mass, they significantly account for tumor maintenance, resistance to therapies, relapse and metastatic spread, due to their increased capacity of self-renewal, multipotency, tumorigenicity and quiescence. Emerging evidence suggests that the immune contexture within the tumor microenvironment (TME) determines both the response to therapy and the clinical outcome. In this context, CSCs acquire immune evasion skills by editing immune cell functions and sculpting the immunosuppressive landscape of TME. Reciprocally, infiltrating immune cells influence CSCs self-renewal, tumorigenicity and metastasis. In this review, we summarize the immunomodulatory properties of CSCs, as well as the impact of innate immune cells on cancer cells stemness in the different phases of cancer immunoediting process and neoplastic progression.
Collapse
Affiliation(s)
- Anna Pastò
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center–IRCCS–, via Manzoni 56, 20089 Rozzano (MI), Italy;
| | - Francesca Maria Consonni
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A. Avogadro, via Bovio 6, 28100 Novara, Italy;
| | - Antonio Sica
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center–IRCCS–, via Manzoni 56, 20089 Rozzano (MI), Italy;
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A. Avogadro, via Bovio 6, 28100 Novara, Italy;
- Correspondence: ; Tel.: +39-0321-375-881; Fax: +39-0321-375-621
| |
Collapse
|
29
|
Wang L, Simons DL, Lu X, Tu TY, Avalos C, Chang AY, Dirbas FM, Yim JH, Waisman J, Lee PP. Breast cancer induces systemic immune changes on cytokine signaling in peripheral blood monocytes and lymphocytes. EBioMedicine 2020; 52:102631. [PMID: 31981982 PMCID: PMC6992943 DOI: 10.1016/j.ebiom.2020.102631] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/12/2019] [Accepted: 01/06/2020] [Indexed: 12/20/2022] Open
Abstract
Background It is increasingly recognized that cancer progression induces systemic immune changes in the host. Alterations in number and function of immune cells have been identified in cancer patients’ peripheral blood and lymphoid organs. Recently, we found dysregulated cytokine signaling in peripheral blood T cells from breast cancer (BC) patients, even those with localized disease. Methods We used phosphoflow cytometry to determine the clinical significance of cytokine signaling responsiveness in peripheral blood monocytes from non-metastatic BC patients at diagnosis. We also examined the correlation between cytokine signaling in peripheral monocytes and the number of tumor-infiltrating macrophages in paired breast tumors. Findings Our results show that cytokine (IFNγ) signaling may also be dysregulated in peripheral blood monocytes at diagnosis, specifically in BC patients who later relapsed. Some patients exhibited concurrent cytokine signaling defects in monocytes and lymphocytes at diagnosis, which predict the risk of future relapse in two independent cohorts of BC patients. Moreover, IFNγ signaling negatively correlates with expression of CSF1R on monocytes, thus modulating their ability to infiltrate into tumors. Interpretation Our results demonstrate that tumor-induced systemic immune changes are evident in peripheral blood immune cells for both myeloid and lymphoid lineages, and point to cytokine signaling responsiveness as important biomarkers to evaluate the overall immune status of BC patients. Funding This study was supported by the Department of Defense Breast Cancer Research Program (BCRP), The V Foundation, Stand Up to Cancer (SU2C), and Breast Cancer Research Foundation (BCRF).
Collapse
Affiliation(s)
- Lei Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Diana L Simons
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Xuyang Lu
- Department of Biostatistics, UCLA, Los Angeles, CA 90095, USA
| | - Travis Y Tu
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Christian Avalos
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Andrew Y Chang
- Department of Medicine, Stanford University Medical Center, Stanford, CA 94305, USA
| | | | - John H Yim
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - James Waisman
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA.
| |
Collapse
|
30
|
Day GL, Bryan ML, Northrup SA, Lyles DS, Westcott MM, Stewart JH. Immune Effects of M51R Vesicular Stomatitis Virus Treatment of Carcinomatosis From Colon Cancer. J Surg Res 2019; 245:127-135. [PMID: 31415934 DOI: 10.1016/j.jss.2019.07.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 06/14/2019] [Accepted: 07/11/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The purpose of this study was to analyze the oncolytic and immunomodulatory functions of an M protein mutant of vesicular stomatitis virus (M51R VSV) in a murine model of peritoneal surface dissemination from colon cancer (PSD from CRC). METHODS Luciferase-expressing CT26 peritoneal tumors were established in Balb/c mice to evaluate the impact of M51R VSV treatment on intraperitoneal tumor growth and overall survival. The mice were treated with either intraperitoneal phosphate buffered saline (n = 10) or 5 × 106 PFU M51R VSV (n = 10) at 5 d after tumor implantation. Tumor bioluminescence was measured every 3 d during the 60-day study period. The immunomodulatory effect of M51R VSV treatment was evaluated in mice treated with either intraperitoneal phosphate buffered saline (n = 21) or M51R VSV (n = 21). Peritoneal lavages were collected at days 1, 3, and 7 after M51R VSV treatment for flow cytometry and multiplex cytokine bead analysis. RESULTS A single, intraperitoneal treatment with M51R VSV inhibited the growth of PSD from CRC as evidenced by decreased bioluminescence and improved survival. This treatment approach also resulted in significantly higher frequencies of peritoneal CD4+ T (10.95 ± 1.17 versus 6.19 ± 0.44, P = 0.004) and B1b cells (5.01 ± 0.97 versus 2.20 ± 0.2, P = 0.024). On the other hand, treatment with M51R VSV resulted in fewer myeloid-derived suppressor cells relative to controls (10.66 ± 1.48 versus 14.47 ± 1.06, P = 0.035). M51R-treated peritoneal cavities also contained lower concentrations of immunosuppressive monocyte chemoattractant protein-1 and interleukin 6 cytokines relative to controls. CONCLUSIONS Our findings suggest that M51R VSV alters the innate and adaptive immune responses in PSD from CRC. Future studies will delineate specific components of antitumor immunity that result in its therapeutic effect.
Collapse
Affiliation(s)
- Gwenyth L Day
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Michelle L Bryan
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Scott A Northrup
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Douglas S Lyles
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Marlena M Westcott
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - John H Stewart
- Department of Surgery, The University of Illinois, Chicago School of Medicine, Chicago, Illinois.
| |
Collapse
|
31
|
Acute Lymphoblastic Leukaemia Cells Impair Dendritic Cell and Macrophage Differentiation: Role of BMP4. Cells 2019; 8:cells8070722. [PMID: 31337120 PMCID: PMC6679123 DOI: 10.3390/cells8070722] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/06/2019] [Accepted: 07/13/2019] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells and macrophages are common components of the tumour immune microenvironment and can contribute to immune suppression in both solid and haematological cancers. The Bone Morphogenetic Protein (BMP) pathway has been reported to be involved in cancer, and more recently in leukaemia development and progression. In the present study, we analyse whether acute lymphoblastic leukaemia (ALL) cells can affect the differentiation of dendritic cells and macrophages and the involvement of BMP pathway in the process. We show that ALL cells produce BMP4 and that conditioned media from ALL cells promote the generation of dendritic cells with immunosuppressive features and skew M1-like macrophage polarization towards a less pro-inflammatory phenotype. Likewise, BMP4 overexpression in ALL cells potentiates their ability to induce immunosuppressive dendritic cells and favours the generation of M2-like macrophages with pro-tumoral features. These results suggest that BMP4 is in part responsible for the alterations in dendritic cell and macrophage differentiation produced by ALL cells.
Collapse
|
32
|
Evaluating a Single Domain Antibody Targeting Human PD-L1 as a Nuclear Imaging and Therapeutic Agent. Cancers (Basel) 2019; 11:cancers11060872. [PMID: 31234464 PMCID: PMC6628009 DOI: 10.3390/cancers11060872] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/13/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022] Open
Abstract
The PD-1:PD-L1 immune checkpoint axis is central in the escape of cancer cells from anticancer immune responses. Monoclonal antibodies (mAbs) specific for PD-L1 have been approved for treatment of various cancer types. Although PD-L1 blockade has proven its merit, there are still several aspects that require further attention to fully capitalize on its potential. One of these is the development of antigen-binding moieties that enable PD-L1 diagnosis and therapy. We generated human PD-L1 binding single domain antibodies (sdAbs) and selected sdAb K2, a sdAb with a high affinity for PD-L1, as a lead compound. SPECT/CT imaging in mice following intravenous injection of Technetium-99m (99mTc)-labeled sdAb K2 revealed high signal-to-noise ratios, strong ability to specifically detect PD-L1 in melanoma and breast tumors, and relatively low kidney retention, which is a unique property for radiolabeled sdAbs. We further showed using surface plasmon resonance that sdAb K2 binds to the same epitope on PD-L1 as the mAb avelumab, and antagonizes PD-1:PD-L1 interactions. Different human cell-based assays corroborated the PD-1:PD-L1 blocking activity, showing enhanced T-cell receptor signaling and tumor cell killing when PD-1POS T cells interacted with PD-L1POS tumor cells. Taken together, we present sdAb K2, which specifically binds to human PD-L1, as a new diagnostic and therapeutic agent in cancer management.
Collapse
|
33
|
Höpken UE, Rehm A. Targeting the Tumor Microenvironment of Leukemia and Lymphoma. Trends Cancer 2019; 5:351-364. [DOI: 10.1016/j.trecan.2019.05.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/13/2022]
|
34
|
Cheng Y, Ma XL, Wei YQ, Wei XW. Potential roles and targeted therapy of the CXCLs/CXCR2 axis in cancer and inflammatory diseases. Biochim Biophys Acta Rev Cancer 2019; 1871:289-312. [DOI: 10.1016/j.bbcan.2019.01.005] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/19/2018] [Accepted: 01/09/2019] [Indexed: 12/16/2022]
|
35
|
The prognosis of head and neck squamous cell carcinoma related to immunosuppressive tumor microenvironment regulated by IL-6 signaling. Oral Oncol 2019; 91:47-55. [PMID: 30926062 DOI: 10.1016/j.oraloncology.2019.02.027] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/26/2019] [Accepted: 02/23/2019] [Indexed: 12/17/2022]
Abstract
Evasion of immune surveillance is a significant factor in head and neck squamous cell carcinoma (HNSCC) carcinogenesis. IL-6 signaling is a critical mechanism for the induction of dysfunctional immune responses. In the present study, we examined the role of IL-6 in the prognosis of HNSCC regarding the immunosuppressive tumor microenvironment. We retrospectively analyzed the clinical outcomes of HNSCC patients and examined its correlation with the levels of IL-6 in tumors and circulating myeloid-derived suppressor cells (MDSCs) in peripheral blood. Furthermore, the relationships between IL and 6, programmed death ligand (PD-L1) expression, and immune response were examined in vitro and in vivo. Our data revealed that IL-6 overexpression was associated with the increased risk of developing disease failure and poor prognosis for HNSCC. The immunoreactivity of IL-6 in HNSCC specimens was positively linked to the staining of PD-L1 and the level of circulating MDSCs. By cellular and animal experiments, there were augmented radiation-induced increases in the expression of PD-L1 and the activation of MDSCs noted in IL-6-positive tumors. When IL-6 signaling was inhibited, the levels of PD-L1 and MDSC recruitment were significantly down-regulated. Furthermore, the neutrophil-to-lymphocyte ratio (NLR) was positively correlated with the levels of IL-6 and PD-L1 in tumor, and circulating MDSCs. In conclusion, IL-6 is a significant predictor of treatment outcome in HNSCC patients, and plays an important role in the induction of immunosuppressive tumor microenvironment mediated by increased MDSCs and PD-L1 expression. Furthermore, IL-6 combined with NLR can assist the clinician to make an informed decision regarding treatment options.
Collapse
|
36
|
Saylor J, Ma Z, Goodridge HS, Huang F, Cress AE, Pandol SJ, Shiao SL, Vidal AC, Wu L, Nickols NG, Gertych A, Knudsen BS. Spatial Mapping of Myeloid Cells and Macrophages by Multiplexed Tissue Staining. Front Immunol 2018; 9:2925. [PMID: 30619287 PMCID: PMC6302234 DOI: 10.3389/fimmu.2018.02925] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 11/29/2018] [Indexed: 12/15/2022] Open
Abstract
An array of phenotypically diverse myeloid cells and macrophages (MC&M) resides in the tumor microenvironment, requiring multiplexed detection systems for visualization. Here we report an automated, multiplexed staining approach, named PLEXODY, that consists of five MC&M-related fluorescently-tagged antibodies (anti - CD68, - CD163, - CD206, - CD11b, and - CD11c), and three chromogenic antibodies, reactive with high- and low-molecular weight cytokeratins and CD3, highlighting tumor regions, benign glands and T cells. The staining prototype and image analysis methods which include a pixel/area-based quantification were developed using tissues from inflamed colon and tonsil and revealed a unique tissue-specific composition of 14 MC&M-associated pixel classes. As a proof-of-principle, PLEXODY was applied to three cases of pancreatic, prostate and renal cancers. Across digital images from these cancer types we observed 10 MC&M-associated pixel classes at frequencies greater than 3%. Cases revealed higher frequencies of single positive compared to multi-color pixels and a high abundance of CD68+/CD163+ and CD68+/CD163+/CD206+ pixels. Significantly more CD68+ and CD163+ vs. CD11b+ and CD11c+ pixels were in direct contact with tumor cells and T cells. While the greatest percentage (~70%) of CD68+ and CD163+ pixels was 0–20 microns away from tumor and T cell borders, CD11b+ and CD11c+ pixels were detected up to 240 microns away from tumor/T cell masks. Together, these data demonstrate significant differences in densities and spatial organization of MC&M-associated pixel classes, but surprising similarities between the three cancer types.
Collapse
Affiliation(s)
- Joshua Saylor
- Departments of Biomedical Sciences, Pathology, Surgery and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Zhaoxuan Ma
- Departments of Biomedical Sciences, Pathology, Surgery and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Helen S Goodridge
- Departments of Biomedical Sciences, Pathology, Surgery and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Fangjin Huang
- Departments of Biomedical Sciences, Pathology, Surgery and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Anne E Cress
- Molecular and Cellular Biology, University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| | - Stephen J Pandol
- Departments of Biomedical Sciences, Pathology, Surgery and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Stephen L Shiao
- Departments of Biomedical Sciences, Pathology, Surgery and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Adriana C Vidal
- Departments of Biomedical Sciences, Pathology, Surgery and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Lily Wu
- Department of Molecular and Medical Pharmacology and Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nicholas G Nickols
- Department of Molecular and Medical Pharmacology and Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Arkadiusz Gertych
- Departments of Biomedical Sciences, Pathology, Surgery and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Beatrice S Knudsen
- Departments of Biomedical Sciences, Pathology, Surgery and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
37
|
Tian Y, Matsui S, Touma M, Wu Q, Sugimoto K. MicroRNA-342 inhibits tumor growth via targeting chemokine CXCL12 involved in macrophages recruitment/activation. Genes Cells 2018; 23:1009-1022. [PMID: 30347114 DOI: 10.1111/gtc.12650] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/19/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022]
Abstract
MicroRNAs (miRNAs) play important roles in initiation, development, progression and metastasis of tumors. MiR-342 has been reported as a tumor suppressor or an onco-miRNA based on functions or expression changes in various types of cancers. However, the biological roles and underlying molecular mechanisms of miR-342 in tumorigenesis remain largely unknown. Here, we found that miR-342 was expressed significantly less in a murine MS-K tumor cell line that showed riched blood vessels. Over-expression of miR-342 in MS-K cells inhibited cell proliferation, colony formation, reduced frequency of S phase population in vitro and suppressed tumor growth in vivo. Moreover, increasing miR-342 impeded blood vessels formation and accumulation of macrophages (CD11b+ ) in tumors. By bioinformatic analysis and dual-luciferase reporter assays, chemokine CXCL12 was identified as a direct target of miR-342. Restored Cxcl12 expression in MS-K-miR-342 cells could rescue cell proliferation in vitro. In MS-K-miR-342 tumor-infiltrated macrophages, expression of proangiogenic genes (Vegf-A and Thbs1) and M2-subtype macrophage markers (Cd163, Dectin1 and Ym1) was significantly down-regulated compared with controls. Moreover, lower level of Cxcl12 and its receptor Cxcr4 was observed in the macrophages of MS-K-miR-342 tumors, and MS-K-miR-342 derived miR-342, but not endogenous miR-342, might contribute to Cxcl12 suppression in TAM. These results suggest that miR-342 is involved in MS-K tumor growth as a tumor suppressor by targeting chemokine CXCL12.
Collapse
Affiliation(s)
- Yijun Tian
- Department of Cell Science, Faculty of Graduate School of Science and Technology, Niigata University, Nishi-ku, Niigata, Japan
| | - Sayaka Matsui
- Department of Cell Science, Faculty of Graduate School of Science and Technology, Niigata University, Nishi-ku, Niigata, Japan
| | - Maki Touma
- Department of Cell Science, Faculty of Graduate School of Science and Technology, Niigata University, Nishi-ku, Niigata, Japan
| | - Qiong Wu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Kenkichi Sugimoto
- Department of Cell Science, Faculty of Graduate School of Science and Technology, Niigata University, Nishi-ku, Niigata, Japan
| |
Collapse
|
38
|
Elias R, Hartshorn K, Rahma O, Lin N, Snyder-Cappione JE. Aging, immune senescence, and immunotherapy: A comprehensive review. Semin Oncol 2018; 45:187-200. [PMID: 30539714 DOI: 10.1053/j.seminoncol.2018.08.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 07/07/2018] [Accepted: 08/01/2018] [Indexed: 12/22/2022]
Abstract
The advent of immune checkpoint inhibitors (ICIs) has changed the landscape of cancer treatment. Older adults represent the majority of cancer patients; however, direct data evaluating ICIs in this patient population is lacking. Aging is associated with changes in the immune system known as "immunosenescence" that could impact the efficacy and safety profile of ICIs. In this paper, we review aging-associated changes in the immune system as they may relate to cancer and immunotherapy, with mention of the effect of chronic viral infections and frailty. Furthermore, we summarize the current clinical evidence of ICI effectiveness and toxicity among older adults with cancer.
Collapse
Affiliation(s)
- Rawad Elias
- Hartford HealthCare Cancer Institute, Hartford Hospital, Hartford, CT, USA.
| | - Kevan Hartshorn
- Section of Hematology Oncology, Boston University School of Medicine, Boston, MA, USA
| | - Osama Rahma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nina Lin
- Department of Medicine, Boston Medical Center, Boston University School of Medicine, MA, USA
| | - Jennifer E Snyder-Cappione
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
39
|
Zhou G, Peng K, Song Y, Yang W, Shu W, Yu T, Yu L, Lin M, Wei Q, Chen C, Yin L, Cong Y, Liu Z. CD177+ neutrophils suppress epithelial cell tumourigenesis in colitis-associated cancer and predict good prognosis in colorectal cancer. Carcinogenesis 2018; 39:272-282. [PMID: 29228136 DOI: 10.1093/carcin/bgx142] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/01/2017] [Indexed: 12/25/2022] Open
Abstract
Neutrophils are found to be infiltrated in tumour tissues of patients with colitis-associated cancer (CAC) and colorectal cancer (CRC), and CD177 is mainly expressed in neutrophils. In our study, expression of CD177 in tumour tissues from patients with CAC or CRC was analysed byquantitative real-time polymerase chain reaction, flow cytometry and immunohistochemistry. We recruited 378 patients with CRC, determined CD177 expression in tumours and examined its correlation with clinicopathological features. Moreover, CAC model was induced in wild-type and CD177-/- mice by azoxymethane/dextran sodium sulphate. CD177+ neutrophils were significantly increased in colon tumour tissues from patients with CRC or CAC compared with controls. Expression of CD177 mRNA and percentages of CD177+ neutrophils were also markedly increased in tumour tissues from CRC patients compared with controls. Patients with high density of CD177+ neutrophils had better overall survival and disease-free survival compared with controls. Multivariate analyses revealed that the density of CD177+ neutrophils was an independent factor in predicting overall survival and disease-free survival. Consistently, CD177 depletion aggravated azoxymethane/dextran sodium sulphate-induced CAC in mice. Expression of Ki67 and proliferating cell nuclear antigen was increased in tumour tissues from CD177-/- mice compared with wild-type counterparts. Moreover, CD177-/- neutrophils failed to migrate in response to fMLP[AU: Please expand fMLP, DN, TNM and HIF-1α.] stimulation compared with wild-type controls. Our data indicate that CD177+ neutrophils suppress epithelial cell tumourigenesis and act as an independent factor in predicting the prognosis in patients with CRC. CD177+ neutrophils may serve as a novel therapeutic target in the treatment and predict the prognosis of CAC and CRC.
Collapse
Affiliation(s)
- Guangxi Zhou
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Kangsheng Peng
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yang Song
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Wenjing Yang
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Weigang Shu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Tianming Yu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Lin Yu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Moubin Lin
- Department of General Surgery, Yangpu Hospital, Tongji University, Shanghai, China
| | - Qing Wei
- Department of Pathology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Chunqiu Chen
- Department of General Surgery, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Lu Yin
- Department of General Surgery, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Zhanju Liu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
40
|
Clappaert EJ, Murgaski A, Van Damme H, Kiss M, Laoui D. Diamonds in the Rough: Harnessing Tumor-Associated Myeloid Cells for Cancer Therapy. Front Immunol 2018; 9:2250. [PMID: 30349530 PMCID: PMC6186813 DOI: 10.3389/fimmu.2018.02250] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022] Open
Abstract
Therapeutic approaches that engage immune cells to treat cancer are becoming increasingly utilized in the clinics and demonstrated durable clinical benefit in several solid tumor types. Most of the current immunotherapies focus on manipulating T cells, however, the tumor microenvironment (TME) is abundantly infiltrated by a heterogeneous population of tumor-associated myeloid cells, including tumor-associated macrophages (TAMs), tumor-associated dendritic cells (TADCs), tumor-associated neutrophils (TANs), and myeloid-derived suppressor cells (MDSCs). Educated by signals perceived in the TME, these cells often acquire tumor-promoting properties ultimately favoring disease progression. Upon appropriate stimuli, myeloid cells can exhibit cytoxic, phagocytic, and antigen-presenting activities thereby bolstering antitumor immune responses. Thus, depletion, reprogramming or reactivation of myeloid cells to either directly eradicate malignant cells or promote antitumor T-cell responses is an emerging field of interest. In this review, we briefly discuss the tumor-promoting and tumor-suppressive roles of myeloid cells in the TME, and describe potential therapeutic strategies in preclinical and clinical development that aim to target them to further expand the range of current treatment options.
Collapse
Affiliation(s)
- Emile J. Clappaert
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Aleksandar Murgaski
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Helena Van Damme
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mate Kiss
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Damya Laoui
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
41
|
Chen MF, Tsai MS, Chen WC, Chen PT. Predictive Value of the Pretreatment Neutrophil-to-Lymphocyte Ratio in Head and Neck Squamous Cell Carcinoma. J Clin Med 2018; 7:jcm7100294. [PMID: 30241364 PMCID: PMC6210359 DOI: 10.3390/jcm7100294] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/16/2018] [Accepted: 09/18/2018] [Indexed: 12/18/2022] Open
Abstract
This study assessed the significance of the neutrophil-to-lymphocyte ratio (NLR) in head and neck squamous cell carcinoma (HNSCC), and the relationships of the NLR with the aldehyde dehydrogenase 1 (ALDH1) level in tumors and the proportion of myeloid-derived suppressor cells (MDSCs) in the peripheral circulation. In total, 227 HNSCC patients who had received curative treatment at our hospital were enrolled into the present study. The NLR of each HNSCC patient before treatment was calculated. The associations of NLR with various clinicopathological parameters and prognoses were then examined. In addition, correlations between the proportion of MDSCs and level of ALDH1 with the NLR were assessed. Our data revealed that an elevated NLR was significantly correlated with the risk of developing locoregional recurrence and with a reduced overall survival in HNSCC patients. Multivariate analyses revealed that the NLR pretreatment and surgical resection were significantly correlated with the rate of treatment failure and the overall survival rate in HNSCC patients. Furthermore, the levels of ALDH1 in tumors and MDSCs in the peripheral circulation were significantly correlated with the prognosis of HNSCC, and the NLR was positively correlated with MDSC levels in the circulation and ALDH1 staining intensity in tumor specimens. In conclusion, the NLR has power in predicting the expression of ALDH1 in tumors, the circulating level of MDSCs, and the prognosis in HNSCC. We suggest that the NLR is an important biomarker that can assist the clinician and patient in making informed decisions regarding treatment options for HNSCC patients.
Collapse
Affiliation(s)
- Miao-Fen Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
- Chang Gung University College of Medicine, Taoyuan 33302, Taiwan.
| | - Ming-Shao Tsai
- Chang Gung University College of Medicine, Taoyuan 33302, Taiwan.
- Department of Otolaryngology & Head and Neck Surgery, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Wen-Cheng Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
- Chang Gung University College of Medicine, Taoyuan 33302, Taiwan.
| | - Ping-Tsung Chen
- Chang Gung University College of Medicine, Taoyuan 33302, Taiwan.
- Department of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| |
Collapse
|
42
|
Awad RM, De Vlaeminck Y, Maebe J, Goyvaerts C, Breckpot K. Turn Back the TIMe: Targeting Tumor Infiltrating Myeloid Cells to Revert Cancer Progression. Front Immunol 2018; 9:1977. [PMID: 30233579 PMCID: PMC6127274 DOI: 10.3389/fimmu.2018.01977] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022] Open
Abstract
Tumor cells frequently produce soluble factors that favor myelopoiesis and recruitment of myeloid cells to the tumor microenvironment (TME). Consequently, the TME of many cancer types is characterized by high infiltration of monocytes, macrophages, dendritic cells and granulocytes. Experimental and clinical studies show that most myeloid cells are kept in an immature state in the TME. These studies further show that tumor-derived factors mold these myeloid cells into cells that support cancer initiation and progression, amongst others by enabling immune evasion, tumor cell survival, proliferation, migration and metastasis. The key role of myeloid cells in cancer is further evidenced by the fact that they negatively impact on virtually all types of cancer therapy. Therefore, tumor-associated myeloid cells have been designated as the culprits in cancer. We review myeloid cells in the TME with a focus on the mechanisms they exploit to support cancer cells. In addition, we provide an overview of approaches that are under investigation to deplete myeloid cells or redirect their function, as these hold promise to overcome resistance to current cancer therapies.
Collapse
|
43
|
Broos K, Lecocq Q, Raes G, Devoogdt N, Keyaerts M, Breckpot K. Noninvasive imaging of the PD-1:PD-L1 immune checkpoint: Embracing nuclear medicine for the benefit of personalized immunotherapy. Am J Cancer Res 2018; 8:3559-3570. [PMID: 30026866 PMCID: PMC6037030 DOI: 10.7150/thno.24762] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/23/2018] [Indexed: 12/19/2022] Open
Abstract
Molecular imaging of the immune checkpoint receptor PD-1 and its ligand PD-L1 is increasingly investigated as a strategy to guide and monitor PD-1:PD-L1-targeted immune checkpoint therapy. We provide an overview of the current state-of-the-art on PD-1- and PD-L1-specific imaging agents for quantitative, real-time assessment of PD-1:PD-L1 expression in the tumor environment and discuss their potential for clinical translation.
Collapse
|
44
|
Son B, Lee S, Youn H, Kim E, Kim W, Youn B. The role of tumor microenvironment in therapeutic resistance. Oncotarget 2018; 8:3933-3945. [PMID: 27965469 PMCID: PMC5354804 DOI: 10.18632/oncotarget.13907] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/01/2016] [Indexed: 12/20/2022] Open
Abstract
Cancer cells undergo unlimited progression and survival owing to activation of oncogenes. However, support of the tumor microenvironment is essential to the formation of clinically relevant tumors. Recent evidence indicates that the tumor microenvironment is a critical regulator of immune escape, progression, and distant metastasis of cancer. Moreover, the tumor microenvironment is known to be involved in acquired resistance of tumors to various therapies. Despite significant advances in chemotherapy and radiotherapy, occurrence of therapeutic resistance leads to reduced efficacy. This review highlights myeloid cells, cancer-associated fibroblasts, and mesenchymal stem cells consisting of the tumor microenvironment, as well as the relevant signaling pathways that eventually render cancer cells to be therapeutically resistant.
Collapse
Affiliation(s)
- Beomseok Son
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - EunGi Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Wanyeon Kim
- Integrative Graduate Program of Ship and Offshore Plant Technology for Ocean Energy Resource, Pusan National University, Busan 46241, Republic of Korea.,Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea.,Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
45
|
Pyter LM, McKim DB, Husain Y, Calero H, Godbout JP, Sheridan JF, Marucha PT, Engeland CG. Effects of dermal wounding on distal primary tumor immunobiology in mice. J Surg Res 2018; 221:328-335. [PMID: 29229147 PMCID: PMC5788460 DOI: 10.1016/j.jss.2017.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/04/2017] [Accepted: 09/15/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Before primary oral tumors are treated, various prophylactic procedures that require tissue repair are often necessary (e.g. biopsies, tooth extractions, radiation, and tracheotomies). Wound healing and tumor growth harness similar immune/inflammatory mechanisms. Our previous work indicates that tumors impair wound healing, although the extent to which tissue repair conversely influences tumor growth is poorly understood. Here, we test the hypothesis that dermal wound healing exacerbates primary tumor growth and influences tumor immunobiology. MATERIALS AND METHODS Female, immunocompetent mice were inoculated subcutaneously with murine oral cancer cells (AT-84) to induce flank tumors. Half of the mice received dermal excisional wounds (4 × 3.5 mm diameter) on their dorsum 16 days later, whereas the skin of controls remained intact. Tumor and blood tissues were harvested 1 and 5 days post wounding, and tumor myeloid cell populations and inflammatory gene expression were measured. Circulating myeloid cells, cytokines, and corticosterone were also quantified. RESULTS Wounding increased tumor mass, early tumor infiltration of macrophages, and tumor inflammatory gene expression. While wounding attenuated tumor growth-induced increases in circulating myeloid cells, no effects of wounding on circulating cytokine/endocrine measures were observed. CONCLUSIONS These results indicate that modest skin immune/inflammatory processes can enhance distal tumor growth and alter innate tumor immunity. The implication for this work is that, in the presence of a tumor, the benefits of tissue-damaging procedures that occur clinically must be weighed against the potential consequences for tumor biology.
Collapse
Affiliation(s)
- Leah M Pyter
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, Ohio; Departments of Psychiatry and Behavioral Health, Ohio State University, Columbus, Ohio; Department of Neuroscience, Ohio State University, Columbus, Ohio; Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois.
| | - Daniel B McKim
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Yasmin Husain
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| | - Humberto Calero
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| | - Jonathan P Godbout
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Neuroscience, Ohio State University, Columbus, Ohio
| | - John F Sheridan
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Biosciences, College of Dentistry, Ohio State University, Columbus, Ohio
| | - Phillip T Marucha
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| | - Christopher G Engeland
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
46
|
Myeloid-Derived Suppressor Cells in the Tumor Microenvironment: Current Knowledge and Future Perspectives. Arch Immunol Ther Exp (Warsz) 2017; 66:113-123. [PMID: 29032490 DOI: 10.1007/s00005-017-0492-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/05/2017] [Indexed: 01/08/2023]
Abstract
The current knowledge on tumor-infiltrating myeloid-derived suppressor cells (MDSCs) is based mainly on the extensive work performed in murine models. Data obtained for human counterparts are generated on the basis of tumor analysis from patient samples. Both sources of information led to determination of the main suppressive mechanisms used by these cell subsets in tumor-bearing hosts. As a result of the identification of protein targets responsible for MDSCs suppressive activity, different therapeutics agents have been used to eliminate/reduce their adverse effect. In the present work, we review the current knowledge on suppressive mechanisms of MDSCs and therapeutic treatments that interfere with their differentiation, expansion or activity. Based on the accumulation of new evidences supporting their importance for tumor progression and metastasis, the interest in these cell types is increasing. We revise the methods of MDSC generation/differentiation ex vivo that may help in overcoming problems associated with limited numbers of cells available from animals and patients for their study.
Collapse
|
47
|
Abstract
Macrophages play essential roles in the response to injury and infection and contribute to the development and/or homeostasis of the various tissues they reside in. Conversely, macrophages also influence the pathogenesis of metabolic, neurodegenerative, and neoplastic diseases. Mechanisms that contribute to the phenotypic diversity of macrophages in health and disease remain poorly understood. Here we review the recent application of genome-wide approaches to characterize the transcriptomes and epigenetic landscapes of tissue-resident macrophages. These studies are beginning to provide insights into how distinct tissue environments are interpreted by transcriptional regulatory elements to drive specialized programs of gene expression.
Collapse
|
48
|
Dougall WC, Kurtulus S, Smyth MJ, Anderson AC. TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy. Immunol Rev 2017; 276:112-120. [PMID: 28258695 DOI: 10.1111/imr.12518] [Citation(s) in RCA: 329] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/18/2016] [Indexed: 12/13/2022]
Abstract
While therapies targeting the co-inhibitory or immune checkpoint receptors PD-1 and CTLA-4 have shown remarkable success in many cancers, not all patients benefit from these therapies. This has catalyzed enormous interest in the targeting of other immune checkpoint receptors. In this regard, TIGIT and CD96 have recently entered the limelight as novel immune checkpoint receptor targets. TIGIT and CD96 together with the co-stimulatory receptor CD226 form a pathway that is analogous to the CD28/CTLA-4 pathway, in which shared ligands and differential receptor:ligand affinities fine-tune the immune response. Although the roles of TIGIT and CD96 as immune checkpoint receptors in T cell and natural killer cell biology are just beginning to be uncovered, accumulating data support the targeting of these receptors for improving anti-tumor immune responses. A clear understanding of the immune cell populations regulated by TIGIT and CD96 is key to the design of immunotherapies that target these receptors in combination with other existing immune checkpoint blockade therapies.
Collapse
Affiliation(s)
| | - Sema Kurtulus
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.,School of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Ana C Anderson
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
49
|
Watanabe R, Shirai T, Namkoong H, Zhang H, Berry GJ, Wallis BB, Schaefgen B, Harrison DG, Tremmel JA, Giacomini JC, Goronzy JJ, Weyand CM. Pyruvate controls the checkpoint inhibitor PD-L1 and suppresses T cell immunity. J Clin Invest 2017; 127:2725-2738. [PMID: 28604383 PMCID: PMC5490755 DOI: 10.1172/jci92167] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/27/2017] [Indexed: 01/12/2023] Open
Abstract
Patients with coronary artery disease (CAD) are at high risk for reactivation of the varicella zoster virus (VZV) and development of herpes zoster (HZ). Here, we found that macrophages from patients with CAD actively suppress T cell activation and expansion, leading to defective VZV-specific T cell immunity. Monocyte-derived and plaque-infiltrating macrophages from patients with CAD spontaneously expressed high surface density of the immunoinhibitory ligand programmed death ligand-1 (PD-L1), thereby providing negative signals to programmed death-1+ (PD-1+) T cells. We determined that aberrant PD-L1 expression in patient-derived macrophages was metabolically controlled. Oversupply of the glycolytic intermediate pyruvate in mitochondria from CAD macrophages promoted expression of PD-L1 via induction of the bone morphogenetic protein 4/phosphorylated SMAD1/5/IFN regulatory factor 1 (BMP4/p-SMAD1/5/IRF1) signaling pathway. Thus, CAD macrophages respond to nutrient excess by activating the immunoinhibitory PD-1/PD-L1 checkpoint, leading to impaired T cell immunity. This finding indicates that metabolite-based immunotherapy may be a potential strategy for restoring adaptive immunity in CAD.
Collapse
Affiliation(s)
- Ryu Watanabe
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Tsuyoshi Shirai
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hong Namkoong
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Hui Zhang
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Gerald J. Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Barbara B. Wallis
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Benedikt Schaefgen
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jennifer A. Tremmel
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - John C. Giacomini
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Jörg J. Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Cornelia M. Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
50
|
Matsumoto Y, Mabuchi S, Kozasa K, Kuroda H, Sasano T, Yokoi E, Komura N, Sawada K, Kimura T. The significance of tumor-associated neutrophil density in uterine cervical cancer treated with definitive radiotherapy. Gynecol Oncol 2017; 145:469-475. [DOI: 10.1016/j.ygyno.2017.02.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/02/2017] [Accepted: 02/04/2017] [Indexed: 12/11/2022]
|