1
|
Huang L, Zhang C, Jiang A, Lin A, Zhu L, Mou W, Zeng D, Liu Z, Tang B, Zhang J, Cheng Q, Miao K, Wei T, Luo P. T-cell Senescence in the Tumor Microenvironment. Cancer Immunol Res 2025; 13:618-632. [PMID: 40232041 DOI: 10.1158/2326-6066.cir-24-0894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/07/2024] [Accepted: 01/24/2025] [Indexed: 04/16/2025]
Abstract
T-cell senescence occurs in the tumor microenvironment (TME) and influences cancer outcomes as well as the effectiveness of immunotherapies. The TME triggers this T-cell senescence via multiple pathways, including persistent stimulation with tumor-associated antigens, altered metabolic pathways, and activation of chronic inflammatory responses. Senescent T cells exhibit characteristics such as genomic instability, loss of protein homeostasis, metabolic dysregulation, and epigenetic alterations. Direct cross-talk between senescent T cells and other immune cells further exacerbates the immunosuppressive TME. This immune-tumor cell interaction within the TME contributes to impaired tumor antigen recognition and surveillance by T cells. The presence of senescent T cells is often associated with poor prognosis and reduced efficacy of immunotherapies; thus, targeting the tumor-promoting mechanisms of T-cell senescence may provide novel insights into improving tumor immunotherapy and patient outcomes. This review explores the contributors to tumor-derived T-cell senescence, the link between T-cell senescence and tumor prognosis, and the potential for targeting T-cell senescence to enhance tumor immunotherapy.
Collapse
Affiliation(s)
- Lihaoyun Huang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lingxuan Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weiming Mou
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bufu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| | - Kai Miao
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
2
|
Li L, Jiang N, Jin Y, Yang C, Fu Y. The role of CD57+γδ T cells in the immune pathogenesis of patients with sepsis. J Leukoc Biol 2025; 117:qiaf015. [PMID: 39936498 DOI: 10.1093/jleuko/qiaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/16/2024] [Accepted: 02/11/2025] [Indexed: 02/13/2025] Open
Abstract
The immune responses are dysregulated during sepsis. This study aimed to investigate the role of CD57+γδ T cells in the immune pathogenesis of patients with sepsis. We characterized the transcriptomic profile of peripheral γδ T cells from patients with sepsis using smart RNA sequencing. Additionally, we assessed the ratios and immune functional signatures of CD57+γδ T cells in sepsis through flow cytometry. The cytotoxic capacity of the CD57+γδ T-cell subset was also validated in vitro. Our findings revealed a reduction in the proportion of peripheral CD57+γδ T cells in patients with sepsis, which was correlated with clinical outcomes. Compared with CD57-γδ T cells, CD57+γδ T cells exhibited a markedly altered immunophenotype, including decreased expression of costimulatory molecules and increased expression of proinflammatory factors and cytotoxic granules. Our study proposed a potential role of CD57 in sepsis and its utility as a novel biomarker for patients with sepsis.
Collapse
Affiliation(s)
- Liman Li
- Department of Laboratory Medicine, West China Hospital of Sichuan University, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
- Laboratory Medicine Research Center of West China Hospital Chengdu, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
| | - Nenggang Jiang
- Department of Laboratory Medicine, West China Hospital of Sichuan University, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
- Laboratory Medicine Research Center of West China Hospital Chengdu, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
| | - Yongmei Jin
- Department of Laboratory Medicine, West China Hospital of Sichuan University, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
- Laboratory Medicine Research Center of West China Hospital Chengdu, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
| | - Chuan Yang
- Laboratory of Pulmonary Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
| | - Yang Fu
- Department of Laboratory Medicine, West China Hospital of Sichuan University, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
- Laboratory Medicine Research Center of West China Hospital Chengdu, 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, Chengdu, Sichuan Province 610041, China
| |
Collapse
|
3
|
Madsen CO, Velasco Santiago M, Martinenaite E, Holz Borch T, Donia M, Svane IM, Hansen M. Peripheral immune biomarkers associated with response to adoptive cell therapy with tumor infiltrating lymphocytes. Clin Exp Immunol 2025; 219:uxaf010. [PMID: 39965099 PMCID: PMC12010344 DOI: 10.1093/cei/uxaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/11/2024] [Accepted: 02/17/2025] [Indexed: 02/20/2025] Open
Abstract
Adoptive cell therapy (ACT) with ex-vivo expanded tumor-infiltrating lymphocytes (TILs, TIL-ACT) has shown clinical efficacy in a significant proportion of patients with metastatic melanoma. To further target TIL-ACT toward responsive patients, identifying predictive biomarkers and understanding broader immune dynamics remain critical. This study investigated the peripheral blood immune landscape in 47 patients with metastatic melanoma undergoing TIL-ACT, assessing antitumor reactivity and peripheral immune cell profiles before and after treatment. Responders displayed increased frequency of circulating tumor-reactive cells post-treatment, and higher baseline levels of activated CD57-expressing T cells, serving as potential biomarkers of response. In contrast, persistent high serum levels of interleukin (IL)-6 and IL-8, higher frequencies of CD38-expressing T cells, and regulatory T cells (Tregs) post-treatment, correlated with unfavorable outcomes. These findings contribute to understanding the peripheral immune landscape associated with response to TIL-ACT, offering valuable insights into predictive biomarkers and mechanisms to improve patient selection.
Collapse
Affiliation(s)
- Cecilie Oelvang Madsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Marta Velasco Santiago
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Evelina Martinenaite
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Troels Holz Borch
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Marco Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Morten Hansen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
4
|
Lopez J, Powles T, Braiteh F, Siu LL, LoRusso P, Friedman CF, Balmanoukian AS, Gordon M, Yachnin J, Rottey S, Karydis I, Fisher GA, Schmidt M, Schuler M, Sullivan RJ, Burris HA, Galvao V, Henick BS, Dirix L, Jaeger D, Ott PA, Wong KM, Jerusalem G, Schiza A, Fong L, Steeghs N, Leidner RS, Rittmeyer A, Laurie SA, Gort E, Aljumaily R, Melero I, Sabado RL, Rhee I, Mancuso MR, Muller L, Fine GD, Yadav M, Kim L, Leveque VJP, Robert A, Darwish M, Qi T, Zhu J, Zhang J, Twomey P, Rao GK, Low DW, Petry C, Lo AA, Schartner JM, Delamarre L, Mellman I, Löwer M, Müller F, Derhovanessian E, Cortini A, Manning L, Maurus D, Brachtendorf S, Lörks V, Omokoko T, Godehardt E, Becker D, Hawner C, Wallrapp C, Albrecht C, Kröner C, Tadmor AD, Diekmann J, Vormehr M, Jork A, Paruzynski A, Lang M, Blake J, Hennig O, Kuhn AN, Sahin U, Türeci Ö, Camidge DR. Autogene cevumeran with or without atezolizumab in advanced solid tumors: a phase 1 trial. Nat Med 2025; 31:152-164. [PMID: 39762422 PMCID: PMC11750724 DOI: 10.1038/s41591-024-03334-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 10/02/2024] [Indexed: 01/23/2025]
Abstract
Effective targeting of somatic cancer mutations to enhance the efficacy of cancer immunotherapy requires an individualized approach. Autogene cevumeran is a uridine messenger RNA lipoplex-based individualized neoantigen-specific immunotherapy designed from tumor-specific somatic mutation data obtained from tumor tissue of each individual patient to stimulate T cell responses against up to 20 neoantigens. This ongoing phase 1 study evaluated autogene cevumeran as monotherapy (n = 30) and in combination with atezolizumab (n = 183) in pretreated patients with advanced solid tumors. The primary objective was safety and tolerability; exploratory objectives included evaluation of pharmacokinetics, pharmacodynamics, preliminary antitumor activity and immunogenicity. Non-prespecified interim analysis showed that autogene cevumeran was well tolerated and elicited poly-epitopic neoantigen-specific responses, encompassing CD4+ and/or CD8+ T cells, in 71% of patients, most of them undetectable at baseline. Responses were detectable up to 23 months after treatment initiation. CD8+ T cells specific for several neoantigens constituted a median of 7.3% of circulating CD8+ T cells, reaching up to 23% in some patients. Autogene cevumeran-induced T cells were found within tumor lesions constituting up to 7.2% of tumor-infiltrating T cells. Clinical activity was observed, including one objective response in monotherapy dose escalation and in two patients with disease characteristics unfavorable for response to immunotherapy treated in combination with atezolizumab. These findings support the continued development of autogene cevumeran in earlier treatment lines. ClinicalTrials.gov registration: NCT03289962 .
Collapse
Affiliation(s)
- Juanita Lopez
- The Royal Marsden Hospital and the Institute of Cancer Research, Sutton, UK.
| | - Thomas Powles
- Barts Cancer Institute, Centre for Experimental Cancer Medicine, Queen Mary University of London, London, UK
| | - Fadi Braiteh
- Comprehensive Cancer Centers of Nevada, Las Vegas, NV, USA
| | - Lillian L Siu
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | - Claire F Friedman
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine Weill Cornell Medical College, New York, NY, USA
| | - Ani S Balmanoukian
- The Angeles Clinic and Research Institute, a Cedars-Sinai affiliate, Los Angeles, CA, USA
| | | | | | - Sylvie Rottey
- Drug Research Unit Ghent, Ghent University Hospital, Ghent, Belgium
| | - Ioannis Karydis
- University Hospital Southampton NHS Trust and University of Southampton, Southampton, UK
| | - George A Fisher
- Department of Medicine (Oncology), Stanford University, Stanford, CA, USA
| | | | - Martin Schuler
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | | | - Vladimir Galvao
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Brian S Henick
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Luc Dirix
- ZAS Ziekenhuizen, Oncology Center Antwerp (OCA) Campus Sint-Augustinus, Antwerp, Belgium
| | - Dirk Jaeger
- National Center for Tumor Diseases, University Hospital of Heidelberg, Heidelberg, Germany
| | - Patrick A Ott
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Kit Man Wong
- University of Washington School of Medicine, Seattle, WA, USA
- Seagen, Inc., Bothell, WA, USA
| | | | - Aglaia Schiza
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Lawrence Fong
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | | | - Rom S Leidner
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | | | - Scott A Laurie
- The Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada
| | - Eelke Gort
- University Medical Center Utrecht, Utrecht, Netherlands
| | - Raid Aljumaily
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ignacio Melero
- University of Navarra and Instituto de Investigacion Sanitaria de Navarra and CIBERONC, Pamplona, Spain
| | | | - Ina Rhee
- Genentech, Inc., South San Francisco, CA, USA
| | | | - Lars Muller
- Genentech, Inc., South San Francisco, CA, USA
- Gilead Sciences, Foster City, CA, USA
| | | | | | - Leesun Kim
- Bluejay Therapeutics, San Mateo, CA, USA
| | | | | | | | - Ting Qi
- Genentech, Inc., South San Francisco, CA, USA
| | - Jiawen Zhu
- Genentech, Inc., South San Francisco, CA, USA
| | - Jingbin Zhang
- Genentech, Inc., South San Francisco, CA, USA
- Artera, Inc., Los Altos, CA, USA
| | - Patrick Twomey
- Genentech, Inc., South San Francisco, CA, USA
- AbbVie, Inc., Santa Clara, CA, USA
| | | | | | - Chris Petry
- Genentech, Inc., South San Francisco, CA, USA
| | - Amy A Lo
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | - Ira Mellman
- Genentech, Inc., South San Francisco, CA, USA
| | - Martin Löwer
- TRON Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ugur Sahin
- BioNTech, Mainz, Germany
- Helmholtz Institute for Translational Oncology Mainz (HI-TRON Mainz) by DKFZ, Mainz, Germany
| | - Özlem Türeci
- BioNTech, Mainz, Germany
- Helmholtz Institute for Translational Oncology Mainz (HI-TRON Mainz) by DKFZ, Mainz, Germany
| | - D Ross Camidge
- Department of Medicine-Medical Oncology, University of Colorado Cancer Center, Denver, CO, USA
| |
Collapse
|
5
|
King LA, Veth M, Iglesias-Guimarais V, Blijdorp I, Kloosterman J, Vis AN, Roovers RC, Hulsik DL, Riedl T, Adang AE, Parren PW, van Helden PM, de Gruijl TD, van der Vliet HJ. Leveraging Vγ9Vδ2 T cells against prostate cancer through a VHH-based PSMA-Vδ2 bispecific T cell engager. iScience 2024; 27:111289. [PMID: 39628574 PMCID: PMC11612814 DOI: 10.1016/j.isci.2024.111289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/17/2024] [Accepted: 10/28/2024] [Indexed: 12/06/2024] Open
Abstract
Vγ9Vδ2 T cells constitute a homogeneous effector T cell population that lyses tumors of different origin, including the prostate. We generated a bispecific T cell engager (bsTCE) to direct Vγ9Vδ2 T cells to PSMA+ prostate cancer (PCa) cells. The PSMA-Vδ2 bsTCE triggered healthy donor and PCa patient-derived Vγ9Vδ2 T cells to lyse PSMA+ PCa cell lines and patient-derived tumor cells while sparing normal prostate cells and enhanced Vγ9Vδ2 T cell antigen cross-presentation to CD8+ T cells. Vγ9Vδ2 T cell expressed NKG2D and DNAM-1 contributed to Vγ9Vδ2 T cell activation and tumor lysis at low PSMA-Vδ2 bsTCE concentrations. In vivo models confirmed the antitumor efficacy of the bsTCE and demonstrated a half-life of 6-7 days. Tissue-cross reactivity analysis was in line with known tissue distribution of PSMA and Vγ9Vδ2 T cells. Together these data show the PSMA-Vδ2 bsTCE to represent a promising anti-tumor strategy and supports its ongoing evaluation in a phase 1/2a clinical trial in therapy refractory metastatic castration-resistant PCa.
Collapse
Affiliation(s)
- Lisa A. King
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands
- Cancer Center Amsterdam, 1081 HV Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Myrthe Veth
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands
- Cancer Center Amsterdam, 1081 HV Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | | | - Iris Blijdorp
- Lava Therapeutics NV, 3584 CM Utrecht, the Netherlands
| | - Jan Kloosterman
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands
- Cancer Center Amsterdam, 1081 HV Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - André N. Vis
- Prostate Cancer Network the Netherlands, Amsterdam, the Netherlands
- Department of Urology, Amsterdam UMC, Vrije Universiteit Amsterdam, HV Amsterdam 1081, the Netherlands
| | | | | | - Thilo Riedl
- Lava Therapeutics NV, 3584 CM Utrecht, the Netherlands
| | | | - Paul W.H.I. Parren
- Lava Therapeutics NV, 3584 CM Utrecht, the Netherlands
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | | | - Tanja D. de Gruijl
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands
- Cancer Center Amsterdam, 1081 HV Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Hans J. van der Vliet
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands
- Cancer Center Amsterdam, 1081 HV Amsterdam, the Netherlands
- Lava Therapeutics NV, 3584 CM Utrecht, the Netherlands
| |
Collapse
|
6
|
Wang S, Qi X, Liu D, Xie D, Jiang B, Wang J, Wang X, Wu G. The implications for urological malignancies of non-coding RNAs in the the tumor microenvironment. Comput Struct Biotechnol J 2024; 23:491-505. [PMID: 38249783 PMCID: PMC10796827 DOI: 10.1016/j.csbj.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/08/2023] [Accepted: 12/16/2023] [Indexed: 01/23/2024] Open
Abstract
Urological malignancies are a major global health issue because of their complexity and the wide range of ways they affect patients. There's a growing need for in-depth research into these cancers, especially at the molecular level. Recent studies have highlighted the importance of non-coding RNAs (ncRNAs) – these don't code for proteins but are crucial in controlling genes – and the tumor microenvironment (TME), which is no longer seen as just a background factor but as an active player in cancer progression. Understanding how ncRNAs and the TME interact is key for finding new ways to diagnose and predict outcomes in urological cancers, and for developing new treatments. This article reviews the basic features of ncRNAs and goes into detail about their various roles in the TME, focusing specifically on how different ncRNAs function and act in urological malignancies.
Collapse
Affiliation(s)
- Shijin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Xiaochen Qi
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Deqian Xie
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Bowen Jiang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Jin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Xiaoxi Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| |
Collapse
|
7
|
Li X, Liu Y, Gui J, Gan L, Xue J. Cell Identity and Spatial Distribution of PD-1/PD-L1 Blockade Responders. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400702. [PMID: 39248327 PMCID: PMC11538707 DOI: 10.1002/advs.202400702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/08/2024] [Indexed: 09/10/2024]
Abstract
The programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) axis inhibits T cell activity, impairing anti-tumor immunity. Blocking this axis with therapeutic antibodies is one of the most promising anti-tumor immunotherapies. It has long been recognized that PD-1/PD-L1 blockade reinvigorates exhausted T (TEX) cells already present in the tumor microenvironment (TME). However, recent advancements in high-throughput gene sequencing and bioinformatic tools have provided researchers with a more granular and dynamic insight into PD-1/PD-L1 blockade-responding cells, extending beyond the TME and TEX populations. This review provides an update on the cell identity, spatial distribution, and treatment-induced spatiotemporal dynamics of PD-1/PD-L1 blockade responders. It also provides a synopsis of preliminary reports of potential PD-1/PD-L1 blockade responders other than T cells to depict a panoramic picture. Important questions to answer in further studies and the translational and clinical potential of the evolving understandings are also discussed.
Collapse
Affiliation(s)
- Xintong Li
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Yuanxin Liu
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Jun Gui
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Lu Gan
- Research Laboratory of Emergency MedicineDepartment of Emergency MedicineNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsLaboratory of Clinical Cell TherapyWest China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
8
|
Wu H, Li J, Zhang Z, Zhang Y. Characteristics and mechanisms of T-cell senescence: A potential target for cancer immunotherapy. Eur J Immunol 2024; 54:e2451093. [PMID: 39107923 DOI: 10.1002/eji.202451093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 11/08/2024]
Abstract
Immunosenescence, the aging of the immune system, leads to functional deficiencies, particularly in T cells, which undergo significant changes. While numerous studies have investigated age-related T-cell phenotypes in healthy aging, senescent T cells have also been observed in younger populations during pathological conditions like cancer. This review summarizes the recent advancements in age-associated alterations and markers of T cells, mechanisms, and the relationship between senescent T cells and the tumor microenvironment. We also discuss potential strategies for targeting senescent T cells to prevent age-related diseases and enhance tumor immunotherapy efficacy.
Collapse
Affiliation(s)
- Han Wu
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Junru Li
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Roozitalab G, Abedi B, Imani S, Farghadani R, Jabbarzadeh Kaboli P. Comprehensive assessment of TECENTRIQ® and OPDIVO®: analyzing immunotherapy indications withdrawn in triple-negative breast cancer and hepatocellular carcinoma. Cancer Metastasis Rev 2024; 43:889-918. [PMID: 38409546 DOI: 10.1007/s10555-024-10174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 02/05/2024] [Indexed: 02/28/2024]
Abstract
Atezolizumab (TECENTRIQ®) and nivolumab (OPDIVO®) are both immunotherapeutic indications targeting programmed cell death 1 ligand 1 (PD-L1) and programmed cell death 1 (PD-1), respectively. These inhibitors hold promise as therapies for triple-negative breast cancer (TNBC) and hepatocellular carcinoma (HCC) and have demonstrated encouraging results in reducing the progression and spread of tumors. However, due to their adverse effects and low response rates, the US Food and Drug Administration (FDA) has withdrawn the approval of atezolizumab in TNBC and nivolumab in HCC treatment. The withdrawals of atezolizumab and nivolumab have raised concerns regarding their effectiveness and the ability to predict treatment responses. Therefore, the current study aims to investigate the immunotherapy withdrawal of PD-1/PD-L1 inhibitors, specifically atezolizumab for TNBC and nivolumab for HCC. This study will examine both the structural and clinical aspects. This review provides detailed insights into the structure of the PD-1 receptor and its ligands, the interactions between PD-1 and PD-L1, and their interactions with the withdrawn antibodies (atezolizumab and nivolumab) as well as PD-1 and PD-L1 modifications. In addition, this review further assesses these antibodies in the context of TNBC and HCC. It seeks to elucidate the factors that contribute to diverse responses to PD-1/PD-L1 therapy in different types of cancer and propose approaches for predicting responses, mitigating the potential risks linked to therapy withdrawals, and optimizing patient outcomes. By better understanding the mechanisms underlying responses to PD-1/PD-L1 therapy and developing strategies to predict these responses, it is possible to create more efficient treatments for TNBC and HCC.
Collapse
Affiliation(s)
- Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Behnaz Abedi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People's Republic of China
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Parham Jabbarzadeh Kaboli
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan.
| |
Collapse
|
10
|
Núñez K, Sandow T, Gimenez J, Hibino M, Cohen A, Thevenot P. Yttrium-90 Induces an Effector Memory Response with Neoantigen Clonotype Expansion: Implications for Immunotherapy. CANCER RESEARCH COMMUNICATIONS 2024; 4:2163-2173. [PMID: 39069671 PMCID: PMC11331567 DOI: 10.1158/2767-9764.crc-24-0228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/14/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Yttrium-90 (90Y) transarterial radioembolization can safely and effectively treat hepatocellular carcinoma (HCC). Clinical trials combining 90Y with immunotherapy are aimed at improving treatment response rates. The impact of transient 90Y-induced lymphopenia on T-cell homeostasis and functional dynamics is unknown. Paired blood specimens were collected prior to first-cycle 90Y and at imaging follow-up in patients with HCC Barcelona Clinic Liver Cancer stages A-B. Flow cytometry and T-cell receptor (TCR) sequencing were used to monitor changes in T-cell subsets and TCR repertoire following 90Y. Objective response (OR) rates were determined using modified RECIST and defined as either OR or nonobjective response. Time-to-progression (TTP) was defined as progression to Barcelona Clinic Liver Cancer stage C within 6 months following 90Y. 90Y induced shifts in both CD4+ (P = 0.049) and CD8+ (P < 0.001) toward an effector memory T-cell response independent of treatment response rate. Nonresponders to 90Y were characterized by a sustained elevation in both naïve CD4+ cells (P = 0.019) and programmed cell death protein 1 expression in CD8+ cells (P = 0.003). Paired analysis of the TCR repertoire revealed a variable induction of neoantigen clonotypes and expansion of existing clonotypes independent of 90Y response. In patients with an OR, changes in TCR clonality did not influence TTP. However, polyclonal profiles in patients without an OR were associated with shorter TTP (P = 0.005; HR, 10.8) and 75% disease progression rates 6 months following treatment. 90Y induces a population shift from central to effector memory accompanied by neoantigen T-cell responses independent of treatment response rate. Monoclonal shifts in the post-90Y T-cell repertoire had superior overall TTP and improved TTP in patients with a first-cycle nonobjective response. SIGNIFICANCE 90Y can safely treat HCC; however, it causes transient lymphopenia. In this article, 90Y stimulates a peripheral effector memory response independent of initial treatment response. TCR sequencing revealed that polyclonal profiles in patients without an OR to treatment were associated with rapid progression rates 6 months after 90Y.
Collapse
Affiliation(s)
- Kelley Núñez
- Institute of Translational Research, Ochsner Health System, New Orleans, Louisiana.
| | - Tyler Sandow
- Interventional Radiology, Ochsner Health System, New Orleans, Louisiana.
| | - Juan Gimenez
- Interventional Radiology, Ochsner Health System, New Orleans, Louisiana.
| | - Mina Hibino
- Institute of Translational Research, Ochsner Health System, New Orleans, Louisiana.
| | - Ari Cohen
- Multi-Organ Transplant Institute, Ochsner Health System, New Orleans, Louisiana.
- Faculty of Medicine, University of Queensland, Brisbane, Australia.
| | - Paul Thevenot
- Institute of Translational Research, Ochsner Health System, New Orleans, Louisiana.
| |
Collapse
|
11
|
Grau Bejar JF, Yaniz Galende E, Zeng Q, Genestie C, Rouleau E, de Bruyn M, Klein C, Le Formal A, Edmond E, Moreau M, Plat A, Gouy S, Maulard A, Pautier P, Michels J, Oaknin A, Colomba-Blameble E, Leary A. Immune predictors of response to immune checkpoint inhibitors in mismatch repair-deficient endometrial cancer. J Immunother Cancer 2024; 12:e009143. [PMID: 38955419 PMCID: PMC11218029 DOI: 10.1136/jitc-2024-009143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Patients with mismatch repair-deficient (MMRd) endometrial cancer (EC) can derive great benefit from immune checkpoint inhibitors (ICI). However not all responses and predictors of primary resistance are lacking. METHODS We compared the immune tumor microenvironment of MMRd EC ICI-responders (Rs) and ICI non-responders (NRs), using spatial multiplexed immune profiling and unsupervised hierarchical clustering analysis. RESULTS Overall, NRs exhibited drastically lower CD8+, absent terminally differentiated T cells, lack of mature tertiary lymphoid structures and dendritic cells, as well as loss of human leukocyte antigen class I. However, no single marker could predict R versus NR with confidence. Clustering analysis identified a combination of four immune features that demonstrated that accurately predicted ICI response, with a discriminative power of 92%. Finally, 80% of NRs lacked programmed death-ligand 1, however, 60% exhibited another actionable immune checkpoint (T-cell immunoglobulin and mucin containing protein-3, indoleamine 2,3-dioxygenase 1, or lymphocyte activation gene 3). CONCLUSIONS These findings underscore the potential of immune tumor microenvironment features for identifying patients with MMRd EC and primary resistance to ICI who should be oriented towards trials testing novel immunotherapeutic combinations.
Collapse
Affiliation(s)
- Juan Francisco Grau Bejar
- Gynecological Oncology Programme, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Gynecological Cancer Translational Research Laboratory, INSERM U981, Gustave Roussy Institute, Villejuif, France
| | - Elisa Yaniz Galende
- Gynecological Cancer Translational Research Laboratory, INSERM U981, Gustave Roussy Institute, Villejuif, France
| | - Qinghe Zeng
- Laboratoire d'Informatique Paris Descartes (LIPADE), Université Paris Cité, Paris, France
- Centre d'Histologie, Imagerie cellulaire et Cytométrie (CHIC), Centre de Recherche des Cordeliers, Centre de Recherche des Cordeliers, Paris, France
| | | | - Etienne Rouleau
- Department of Medical Biology and Pathology, Cancer Genetics Laboratory, Gustave Roussy Institute, Villejuif, France
| | - Marco de Bruyn
- Obstetrics & Gynecology, University of Groningen Faculty of Medical Sciences, Groningen, The Netherlands
| | - Christophe Klein
- Centre d'Histologie, Imagerie cellulaire et Cytométrie (CHIC), Centre de Recherche des Cordeliers, Centre de Recherche des Cordeliers, Paris, France
| | - Audrey Le Formal
- Gynecological Cancer Translational Research Laboratory, INSERM U981, Gustave Roussy Institute, Villejuif, France
| | - Elodie Edmond
- Experimental and Translational Pathology Platform (PETRA), AMMICa Inserm US23/UAR CNRS 3655, Gustave Roussy Institute, Villejuif, France
| | - Maëva Moreau
- Department of Medical Biology and Pathology, Cancer Genetics Laboratory, Gustave Roussy Institute, Villejuif, France
| | - Annechien Plat
- Obstetrics & Gynecology, University of Groningen Faculty of Medical Sciences, Groningen, The Netherlands
| | - Sebastien Gouy
- Department of Gynecologic Surgery, Department of Surgery, Gustave Roussy Institute, Villejuif, France
| | - Amandine Maulard
- Department of Gynecologic Surgery, Department of Surgery, Gustave Roussy Institute, Villejuif, France
| | - Patricia Pautier
- Gynecological Cancer Unit, Department of Medical Oncology, Gustave Roussy Institute, Villejuif, France
| | - Judith Michels
- Gynecological Cancer Unit, Department of Medical Oncology, Gustave Roussy Institute, Villejuif, France
| | - Ana Oaknin
- Gynecological Oncology Programme, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Emeline Colomba-Blameble
- Gynecological Cancer Unit, Department of Medical Oncology, Gustave Roussy Institute, Villejuif, France
| | - Alexandra Leary
- Gynecological Cancer Translational Research Laboratory, INSERM U981, Gustave Roussy Institute, Villejuif, France
- Gynecological Cancer Unit, Department of Medical Oncology, Gustave Roussy Institute, Villejuif, France
| |
Collapse
|
12
|
Kinney BL, Brammer B, Kansal V, Parrish CJ, Kissick HT, Liu Y, Saba NF, Buchwald ZS, El-Deiry MW, Patel MR, Boyce BJ, Kaka AS, Gross JH, Baddour HM, Chen AY, Schmitt NC. CD28-CD57+ T cells from head and neck cancer patients produce high levels of cytotoxic granules and type II interferon but are not senescent. Oncoimmunology 2024; 13:2367777. [PMID: 38887372 PMCID: PMC11181932 DOI: 10.1080/2162402x.2024.2367777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
T lymphocytes expressing CD57 and lacking costimulatory receptors CD27/CD28 have been reported to accumulate with aging, chronic infection, and cancer. These cells are described as senescent, with inability to proliferate but enhanced cytolytic and cytokine-producing capacity. However, robust functional studies on these cells taken directly from cancer patients are lacking. We isolated these T cells and their CD27/28+ counterparts from blood and tumor samples of 50 patients with previously untreated head and neck cancer. Functional studies confirmed that these cells have enhanced ability to degranulate and produce IFN-γ. They also retain the ability to proliferate, thus are not senescent. These data suggest that CD27/28-CD57+ CD8+ T cells are a subset of highly differentiated, CD45RA+ effector memory (TEMRA) cells with retained proliferative capacity. Patients with > 34% of these cells among CD8+ T cells in the blood had a higher rate of locoregional disease relapse, suggesting these cells may have prognostic significance.
Collapse
Affiliation(s)
- Brendan L.C. Kinney
- Department of Otolaryngology – Head and Neck Surgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Brianna Brammer
- Department of Otolaryngology – Head and Neck Surgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Vikash Kansal
- Department of Otolaryngology – Head and Neck Surgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Connor J. Parrish
- School of Medicine, St. Louis University School of Medicine, St. Louis, MO, USA
| | - Haydn T. Kissick
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Urology, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Yuan Liu
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Nabil F. Saba
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | | | - Mark W. El-Deiry
- Department of Otolaryngology – Head and Neck Surgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Mihir R. Patel
- Department of Otolaryngology – Head and Neck Surgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Brian J. Boyce
- Department of Otolaryngology – Head and Neck Surgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Azeem S. Kaka
- Department of Otolaryngology – Head and Neck Surgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jennifer H. Gross
- Department of Otolaryngology – Head and Neck Surgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - H. Michael Baddour
- Department of Otolaryngology – Head and Neck Surgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Amy Y. Chen
- Department of Otolaryngology – Head and Neck Surgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Nicole C. Schmitt
- Department of Otolaryngology – Head and Neck Surgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
13
|
Yanagihara T, Hata K, Matsubara K, Kunimura K, Suzuki K, Tsubouchi K, Ikegame S, Baba Y, Fukui Y, Okamoto I. Exploratory mass cytometry analysis reveals immunophenotypes of cancer treatment-related pneumonitis. eLife 2024; 12:RP87288. [PMID: 38607373 PMCID: PMC11014725 DOI: 10.7554/elife.87288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
Anticancer treatments can result in various adverse effects, including infections due to immune suppression/dysregulation and drug-induced toxicity in the lung. One of the major opportunistic infections is Pneumocystis jirovecii pneumonia (PCP), which can cause severe respiratory complications and high mortality rates. Cytotoxic drugs and immune-checkpoint inhibitors (ICIs) can induce interstitial lung diseases (ILDs). Nonetheless, the differentiation of these diseases can be difficult, and the pathogenic mechanisms of such diseases are not yet fully understood. To better comprehend the immunophenotypes, we conducted an exploratory mass cytometry analysis of immune cell subsets in bronchoalveolar lavage fluid from patients with PCP, cytotoxic drug-induced ILD (DI-ILD), and ICI-associated ILD (ICI-ILD) using two panels containing 64 markers. In PCP, we observed an expansion of the CD16+ T cell population, with the highest CD16+ T proportion in a fatal case. In ICI-ILD, we found an increase in CD57+ CD8+ T cells expressing immune checkpoints (TIGIT+ LAG3+ TIM-3+ PD-1+), FCRL5+ B cells, and CCR2+ CCR5+ CD14+ monocytes. These findings uncover the diverse immunophenotypes and possible pathomechanisms of cancer treatment-related pneumonitis.
Collapse
Affiliation(s)
- Toyoshi Yanagihara
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu UniversityFukuokaJapan
- Department of Respiratory Medicine, NHO Fukuoka National HospitalFukuokaJapan
| | - Kentaro Hata
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Keisuke Matsubara
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu UniversityFukuokaJapan
| | - Kazufumi Kunimura
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu UniversityFukuokaJapan
| | - Kunihiro Suzuki
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Kazuya Tsubouchi
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Satoshi Ikegame
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Yoshihiro Baba
- Division of Immunology and Genome Biology, Department of Molecular Genetics, Medical Institute of Bioregulation, Kyushu UniversityFukuokaJapan
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu UniversityFukuokaJapan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu UniversityFukuokaJapan
| |
Collapse
|
14
|
Hou J, Yang X, Xie S, Zhu B, Zha H. Circulating T cells: a promising biomarker of anti-PD-(L)1 therapy. Front Immunol 2024; 15:1371559. [PMID: 38576625 PMCID: PMC10991692 DOI: 10.3389/fimmu.2024.1371559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
Anti-PD-(L)1 therapy has shown great efficacy in some patients with cancer. However, a significant proportion of patients with cancer do not respond to it. Another unmet clinical need for anti-PD-(L)1 therapy is the dynamic monitoring of treatment effects. Therefore, identifying biomarkers that can stratify potential responders before PD-(L)1 treatment and timely monitoring of the efficacy of PD-(L)1 treatment are crucial in the clinical setting. The identification of biomarkers by liquid biopsy has attracted considerable attention. Among the identified biomarkers, circulating T cells are one of the most promising because of their indispensable contribution to anti-PD-(L)1 therapy. The present review aimed to thoroughly explore the potential of circulating T cells as biomarkers of anti-PD-(L)1 therapy and its advantages and limitations.
Collapse
Affiliation(s)
- Junlei Hou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xuezhi Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shuanglong Xie
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Haoran Zha
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| |
Collapse
|
15
|
YADOLLAHVANDMIANDOAB REZA, JALALIZADEH MEHRSA, DIONATO FRANCIELEAPARECIDAVECHIA, BUOSI KEINI, LEME PATRÍCIAAF, COL LUCIANASBDAL, GIACOMELLI CRISTIANEF, ASSIS ALEXDIAS, BASHIRICHELKASARI NASIM, REIS LEONARDOOLIVEIRA. Clinical implications of single cell sequencing for bladder cancer. Oncol Res 2024; 32:597-605. [PMID: 38560564 PMCID: PMC10972735 DOI: 10.32604/or.2024.045442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 01/08/2024] [Indexed: 04/04/2024] Open
Abstract
Bladder cancer (BC) is the 10th most common cancer worldwide, with about 0.5 million reported new cases and about 0.2 million deaths per year. In this scoping review, we summarize the current evidence regarding the clinical implications of single-cell sequencing for bladder cancer based on PRISMA guidelines. We searched PubMed, CENTRAL, Embase, and supplemented with manual searches through the Scopus, and Web of Science for published studies until February 2023. We included original studies that used at least one single-cell technology to study bladder cancer. Forty-one publications were included in the review. Twenty-nine studies showed that this technology can identify cell subtypes in the tumor microenvironment that may predict prognosis or response to immune checkpoint inhibition therapy. Two studies were able to diagnose BC by identifying neoplastic cells through single-cell sequencing urine samples. The remaining studies were mainly a preclinical exploration of tumor microenvironment at single cell level. Single-cell sequencing technology can discriminate heterogeneity in bladder tumor cells and determine the key molecular properties that can lead to the discovery of novel perspectives on cancer management. This nascent tool can advance the early diagnosis, prognosis judgment, and targeted therapy of bladder cancer.
Collapse
Affiliation(s)
- REZA YADOLLAHVANDMIANDOAB
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
| | - MEHRSA JALALIZADEH
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
| | | | - KEINI BUOSI
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
| | - PATRÍCIA A. F. LEME
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
| | - LUCIANA S. B. DAL COL
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
| | - CRISTIANE F. GIACOMELLI
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
| | - ALEX DIAS ASSIS
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
| | - NASIM BASHIRICHELKASARI
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
| | - LEONARDO OLIVEIRA REIS
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo, 13083-872, Brazil
- ImmunOncology, Pontifical Catholic University of Campinas, PUC-Campinas, Campinas, Sao Paulo, 13087-571, Brazil
| |
Collapse
|
16
|
Perez-Penco M, Lara de la Torre L, Lecoq I, Martinenaite E, Andersen MH. TGFβ-specific T cells induced by a TGFβ-derived immune modulatory vaccine both directly and indirectly modulate the phenotype of tumor-associated macrophages and fibroblasts. J Immunother Cancer 2024; 12:e008405. [PMID: 38417917 PMCID: PMC10900378 DOI: 10.1136/jitc-2023-008405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2024] [Indexed: 03/01/2024] Open
Abstract
The tumor microenvironment (TME) of pancreatic cancer is highly immunosuppressive. We recently developed a transforming growth factor (TGF)β-based immune modulatory vaccine that controlled tumor growth in a murine model of pancreatic cancer by targeting immunosuppression and desmoplasia in the TME. We found that treatment with the TGFβ vaccine not only reduced the percentage of M2-like tumor-associated macrophages (TAMs) and cancer-associated fibroblasts (CAFs) in the tumor but polarized CAFs away from the myofibroblast-like phenotype. However, whether the immune modulatory properties of the TGFβ vaccine on TAM and CAF phenotypes are a direct consequence of the recognition and subsequent targeting of these subsets by TGFβ-specific T cells or an indirect consequence of the overall modulation induced within the TME remains unknown. Recognition of M2 macrophages and fibroblast by TGFβ-specific T cells was assessed by ELISpot and flow cytometry. The indirect and direct effects of the TGFβ vaccine on these cell subsets were evaluated by culturing M2 macrophages or fibroblasts with tumor-conditioned media or with T cells isolated from the spleen of mice treated with the TGFβ vaccine or a control vaccine, respectively. Changes in phenotype were assessed by flow cytometry and Bio-Plex multiplex system (Luminex). We found that TGFβ-specific T cells induced by the TGFβ vaccine can recognize M2 macrophages and fibroblasts. Furthermore, we demonstrated that the phenotype of M2 macrophages and CAFs can be directly modulated by TGFβ-specific T cells induced by the TGFβ vaccine, as well as indirectly modulated as a result of the immune-modulatory effects of the vaccine within the TME. TAMs tend to have tumor-promoting functions, harbor an immunosuppressive phenotype and are linked to decreased overall survival in pancreatic cancer when they harbor an M2-like phenotype. In addition, myofibroblast-like CAFs create a stiff extracellular matrix that restricts T cell infiltration, impeding the effectiveness of immune therapies in desmoplastic tumors, such as pancreatic ductal adenocarcinoma. Reducing immunosuppression and immune exclusion in pancreatic tumors by targeting TAMs and CAFs with the TGFβ-based immune modulatory vaccine emerges as an innovative strategy for the generation of a more favorable environment for immune-based therapies, such as immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Maria Perez-Penco
- Department of Oncology, Copenhagen University Hospital, National Center for Cancer Immune Therapy (CCIT- DK), Herlev, Denmark
| | - Lucia Lara de la Torre
- Department of Oncology, Copenhagen University Hospital, National Center for Cancer Immune Therapy (CCIT- DK), Herlev, Denmark
| | - Inés Lecoq
- Department of Oncology, Copenhagen University Hospital, National Center for Cancer Immune Therapy (CCIT- DK), Herlev, Denmark
| | - Evelina Martinenaite
- Department of Oncology, Copenhagen University Hospital, National Center for Cancer Immune Therapy (CCIT- DK), Herlev, Denmark
| | - Mads Hald Andersen
- Department of Oncology, Copenhagen University Hospital, National Center for Cancer Immune Therapy (CCIT- DK), Herlev, Denmark
- Department of Immunology, University of Copenhagen, Kobenhavn, Denmark
| |
Collapse
|
17
|
Sun W, Qiu F, Zheng J, Fang L, Qu J, Zhang S, Jiang N, Zhou J, Zeng X, Zhou J. CD57-positive CD8 + T cells define the response to anti-programmed cell death protein-1 immunotherapy in patients with advanced non-small cell lung cancer. NPJ Precis Oncol 2024; 8:25. [PMID: 38297019 PMCID: PMC10830454 DOI: 10.1038/s41698-024-00513-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/08/2023] [Indexed: 02/02/2024] Open
Abstract
Immune checkpoint inhibitors have transformed the treatment landscape of non-small cell lung cancer (NSCLC). However, accurately identifying patients who will benefit from immunotherapy remains a challenge. This study aimed to discover potential biomarkers for predicting immunotherapy response in NSCLC patients. Single-cell mass cytometry (CyTOF) was utilized to analyze immune cell subsets in peripheral blood mononuclear cells (PBMCs) obtained from NSCLC patients before and 12 weeks after single-agent immunotherapy. The CyTOF findings were subsequently validated using flow cytometry and multiplex immunohistochemistry/immunofluorescence in PBMCs and tumor tissues, respectively. RNA sequencing (RNA-seq) was performed to elucidate the underlying mechanisms. In the CyTOF cohort (n = 20), a high frequency of CD57+CD8+ T cells in PBMCs was associated with durable clinical benefit from immunotherapy in NSCLC patients (p = 0.034). This association was further confirmed in an independent cohort using flow cytometry (n = 27; p < 0.001), with a determined cutoff value of 12.85%. The cutoff value was subsequently validated in another independent cohort (AUC = 0.733). We also confirmed the CyTOF findings in pre-treatment formalin-fixed and paraffin-embedded tissues (n = 90; p < 0.001). RNA-seq analysis revealed 475 differentially expressed genes (DEGs) between CD57+CD8+ T cells and CD57-CD8+ T cells, with functional analysis identifying DEGs significantly enriched in immune-related signaling pathways. This study highlights CD57+CD8+ T cells as a promising biomarker for predicting immunotherapy success in NSCLC patients.
Collapse
Affiliation(s)
- Wenjia Sun
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Fengqi Qiu
- Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Jing Zheng
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liangjie Fang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shumeng Zhang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Nan Jiang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xun Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jianya Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
18
|
Hou C, Wang Z, Lu X. Impact of immunosenescence and inflammaging on the effects of immune checkpoint inhibitors. CANCER PATHOGENESIS AND THERAPY 2024; 2:24-30. [PMID: 38328711 PMCID: PMC10846300 DOI: 10.1016/j.cpt.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 02/09/2024]
Abstract
Immune checkpoint inhibitors (ICIs) are employed in immunotherapeutic applications for patients with weakened immune systems and can improve the ability of T cells to kill cancer cells. Although ICIs can potentially treat different types of cancers in various groups of patients, their effectiveness may differ among older individuals. The reason ICIs are less effective in older adults is not yet clearly understood, but age-related changes in the immune system, such as immunosenescence and inflammation, may play a role. Therefore, this review focuses on recent advances in understanding the effects of immunosenescence and inflammation on the efficacy of ICIs.
Collapse
Affiliation(s)
- Chuandong Hou
- Medical School of Chinese PLA, Beijing 100853, China
- Department of Hematology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Zining Wang
- Medical School of Chinese PLA, Beijing 100853, China
- Department of Hematology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Xuechun Lu
- Department of Hematology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
19
|
Elliot TAE, Lecky DAJ, Bending D. T-cell response to checkpoint blockade immunotherapies: from fundamental mechanisms to treatment signatures. Essays Biochem 2023; 67:967-977. [PMID: 37386922 PMCID: PMC10539945 DOI: 10.1042/ebc20220247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/01/2023]
Abstract
Immune checkpoint immunotherapies act to block inhibitory receptors on the surface of T cells and other cells of the immune system. This can increase activation of immune cells and promote tumour clearance. Whilst this is very effective in some types of cancer, significant proportions of patients do not respond to single-agent immunotherapy. To improve patient outcomes, we must first mechanistically understand what drives therapy resistance. Many studies have utilised genetic, transcriptional, and histological signatures to find correlates of effective responses to treatment. It is key that we understand pretreatment predictors of response, but also to understand how the immune system becomes treatment resistant during therapy. Here, we review our understanding of the T-cell signatures that are critical for response, how these immune signatures change during treatment, and how this information can be used to rationally design therapeutic strategies. We highlight how chronic antigen recognition drives heterogeneous T-cell exhaustion and the role of T-cell receptor (TCR) signal strength in exhausted T-cell differentiation and molecular response to therapy. We explore how dynamic changes in negative feedback pathways can promote resistance to single-agent therapy. We speculate that this resistance may be circumvented in the future through identifying the most effective combinations of immunotherapies to promote sustained and durable antitumour responses.
Collapse
Affiliation(s)
- Thomas A E Elliot
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| | - David A J Lecky
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| | - David Bending
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| |
Collapse
|
20
|
Sun Q, Hong Z, Zhang C, Wang L, Han Z, Ma D. Immune checkpoint therapy for solid tumours: clinical dilemmas and future trends. Signal Transduct Target Ther 2023; 8:320. [PMID: 37635168 PMCID: PMC10460796 DOI: 10.1038/s41392-023-01522-4] [Citation(s) in RCA: 211] [Impact Index Per Article: 105.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/11/2023] [Accepted: 05/28/2023] [Indexed: 08/29/2023] Open
Abstract
Immune-checkpoint inhibitors (ICBs), in addition to targeting CTLA-4, PD-1, and PD-L1, novel targeting LAG-3 drugs have also been approved in clinical application. With the widespread use of the drug, we must deeply analyze the dilemma of the agents and seek a breakthrough in the treatment prospect. Over the past decades, these agents have demonstrated dramatic efficacy, especially in patients with melanoma and non-small cell lung cancer (NSCLC). Nonetheless, in the field of a broad concept of solid tumours, non-specific indications, inseparable immune response and side effects, unconfirmed progressive disease, and complex regulatory networks of immune resistance are four barriers that limit its widespread application. Fortunately, the successful clinical trials of novel ICB agents and combination therapies, the advent of the era of oncolytic virus gene editing, and the breakthrough of the technical barriers of mRNA vaccines and nano-delivery systems have made remarkable breakthroughs currently. In this review, we enumerate the mechanisms of each immune checkpoint targets, associations between ICB with tumour mutation burden, key immune regulatory or resistance signalling pathways, the specific clinical evidence of the efficacy of classical targets and new targets among different tumour types and put forward dialectical thoughts on drug safety. Finally, we discuss the importance of accurate triage of ICB based on recent advances in predictive biomarkers and diagnostic testing techniques.
Collapse
Affiliation(s)
- Qian Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhenya Hong
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Cong Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Liangliang Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhiqiang Han
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Ding Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
21
|
Multiparameter single-cell proteomic technologies give new insights into the biology of ovarian tumors. Semin Immunopathol 2023; 45:43-59. [PMID: 36635516 PMCID: PMC9974728 DOI: 10.1007/s00281-022-00979-9] [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: 10/31/2022] [Accepted: 12/11/2022] [Indexed: 01/13/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most lethal gynecological malignancy. Its diagnosis at advanced stage compounded with its excessive genomic and cellular heterogeneity make curative treatment challenging. Two critical therapeutic challenges to overcome are carboplatin resistance and lack of response to immunotherapy. Carboplatin resistance results from diverse cell autonomous mechanisms which operate in different combinations within and across tumors. The lack of response to immunotherapy is highly likely to be related to an immunosuppressive HGSOC tumor microenvironment which overrides any clinical benefit. Results from a number of studies, mainly using transcriptomics, indicate that the immune tumor microenvironment (iTME) plays a role in carboplatin response. However, in patients receiving treatment, the exact mechanistic details are unclear. During the past decade, multiplex single-cell proteomic technologies have come to the forefront of biomedical research. Mass cytometry or cytometry by time-of-flight, measures up to 60 parameters in single cells that are in suspension. Multiplex cellular imaging technologies allow simultaneous measurement of up to 60 proteins in single cells with spatial resolution and interrogation of cell-cell interactions. This review suggests that functional interplay between cell autonomous responses to carboplatin and the HGSOC immune tumor microenvironment could be clarified through the application of multiplex single-cell proteomic technologies. We conclude that for better clinical care, multiplex single-cell proteomic technologies could be an integral component of multimodal biomarker development that also includes genomics and radiomics. Collection of matched samples from patients before and on treatment will be critical to the success of these efforts.
Collapse
|