1
|
Tang Q, Leng S, Tan Y, Cheng H, Liu Q, Wang Z, Xu Y, Zhu L, Wang C. Chitosan/dextran-based organohydrogel delivers EZH2 inhibitor to epigenetically reprogram chemo/immuno-resistance in unresectable metastatic melanoma. Carbohydr Polym 2024; 346:122645. [PMID: 39245506 DOI: 10.1016/j.carbpol.2024.122645] [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: 03/06/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024]
Abstract
Melanoma either intrinsically possesses resistance or rapidly acquires resistance to anti-tumor therapy, which often leads to local recurrence or distant metastasis after resection. In this study, we found histone 3 lysine 27 (H3K27) demethylated by an inhibitor of histone methyltransferase EZH2 could epigenetically reverse the resistance to chemo-drug paclitaxel (PTX), or enhance the efficacy of immune checkpoint inhibitor anti-TIGIT via downregulating TIGIT ligand CD155. Next, to address the complexity in the combination of multiple bioactive molecules with distinct therapeutic properties, we developed a polysaccharides-based organohydrogel (OHG) configured with a heterogenous network. Therein, hydroxypropyl chitosan (HPC)-stabilized emulsions for hydrophobic drug entrapment were crosslinked with oxidized dextran (Odex) to form a hydrophilic gel matrix to facilitate antibody accommodation, which demonstrated a tunable sustained release profile by optimizing emulsion/gel volume ratios. As results, local injection of OHG loaded with EZH2 inhibitor UNC1999, PTX and anti-TIGIT did not only synergistically enhance the cytotoxicity of PTX, but also reprogrammed the immune resistance via bi-directionally blocking TIGIT/CD155 axis, leading to the recruitment of cytotoxic effector cells into tumor and conferring a systemic immune memory to prevent lung metastasis. Hence, this polysaccharides-based OHG represents a potential in-situ epigenetic-, chemo- and immunotherapy platform to treat unresectable metastatic melanoma.
Collapse
Affiliation(s)
- Qi Tang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
| | - Shaolong Leng
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, PR China
| | - Yinqiu Tan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China; Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, PR China
| | - Huan Cheng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Qi Liu
- The First Dongguan Affiliated Hospital Guangdong Medical University No. 42, Jiaoping Road Dongguan, Guangdong 523710, PR China
| | - Zhongjuan Wang
- Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, No.245, People East Road, Kunming 650051, PR China
| | - Yunsheng Xu
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, PR China.
| | - Linyu Zhu
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, PR China.
| | - Cuifeng Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China; Department of neurosurgery, JiuJiang Hospital of Traditional Chinese Medicine, Jiujiang, PR China.
| |
Collapse
|
2
|
Spagnolo CC, Campo I, Campennì A, Cardile D, Cannavò S, Silvestris N, Santarpia M, Ruggeri RM. Challenges and pitfalls in the management of endocrine toxicities from immune checkpoint inhibitors: a case presentation of synchronous thyrotoxicosis and primary adrenal insufficiency in a melanoma patient. Hormones (Athens) 2024; 23:759-764. [PMID: 38421588 DOI: 10.1007/s42000-024-00535-0] [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: 01/16/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors have revolutionized the therapeutic approach to several solid tumors, becoming the standard of care for cancer treatment in different disease settings. Despite the fact that these agents are better tolerated than conventional chemotherapy, their use is associated with a specific toxicity profile, so-called immune-related adverse events (irAEs), that can involve several organs. Endocrine irAEs are among the most frequent toxicities (around 10 to 16%) and include hypophysitis, thyroid disorders, adrenalitis, and diabetes mellitus. Some of them may be life-threatening if not promptly recognized (such as diabetic ketoacidosis and acute adrenal crisis). CASE PRESENTATION A 55-year-old woman with a personal history of euthyroid Hashimoto's thyroiditis was diagnosed with a metastatic melanoma, BRAF wild type. Under treatment with anti-PD-1 pembrolizumab, she developed thyrotoxicosis followed by hypothyroidism due to destructive thyroiditis and concurrent primary adrenal insufficiency due to adrenalitis. CONCLUSIONS The simultaneous occurrence of adrenal and thyroid autoimmune diseases, resembling autoimmune polyendocrine syndrome type 2, may occur as a rare but serious side effect of ICI treatment. It often presents with abrupt onset and rapid evolution towards polyglandular insufficiency. Physicians should be aware of the potential association of two or more endocrine disorders and careful monitoring of endocrine function is needed during ICI therapy.
Collapse
Affiliation(s)
- Calogera Claudia Spagnolo
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, AOU G. Martino, V. Consolare Valeria 1, 98125, Messina, Italy
| | - Irene Campo
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, AOU G. Martino, V. Consolare Valeria 1, 98125, Messina, Italy
| | - Alfredo Campennì
- Unit of Nuclear Medicine, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125, Messina, Italy
| | - Davide Cardile
- Unit of Nuclear Medicine, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125, Messina, Italy
| | - Salvatore Cannavò
- Endocrinology Unit, Department of Human Pathology of Adulthood and Childhood DETEV, University of Messina, 98125, Messina, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, AOU G. Martino, V. Consolare Valeria 1, 98125, Messina, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, AOU G. Martino, V. Consolare Valeria 1, 98125, Messina, Italy.
| | - Rosaria Maddalena Ruggeri
- Endocrinology Unit, Department of Human Pathology of Adulthood and Childhood DETEV, University of Messina, 98125, Messina, Italy
| |
Collapse
|
3
|
Rebaudi F, De Franco F, Goda R, Obino V, Vita G, Baronti C, Iannone E, Pitto F, Massa B, Fenoglio D, Jandus C, Poggio F, Fregatti P, Melaiu O, Bozzo M, Candiani S, Papaccio F, Greppi M, Pesce S, Marcenaro E. The landscape of combining immune checkpoint inhibitors with novel Therapies: Secret alliances against breast cancer. Cancer Treat Rev 2024; 130:102831. [PMID: 39342797 DOI: 10.1016/j.ctrv.2024.102831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/04/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
This review focuses on the immune checkpoint inhibitors (ICIs) in the context of breast cancer (BC) management. These innovative treatments, by targeting proteins expressed on both tumor and immune cells, aim to overcome tumor-induced immune suppression and reactivate the immune system. The potential of this approach is the subject of numerous clinical studies. Here, we explore the key studies and emerging therapies related to ICIs providing a detailed analysis of their specific and combined use in BC treatment.
Collapse
Affiliation(s)
- Federico Rebaudi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Fabiana De Franco
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Rayan Goda
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Valentina Obino
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Giorgio Vita
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy
| | - Camilla Baronti
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Eleonora Iannone
- Breast Surgery Clinic, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesca Pitto
- Department of Pathology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Barbara Massa
- Department of Pathology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniela Fenoglio
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy; Biotherapy Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Camilla Jandus
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland; Geneva Center for Inflammation Research, Geneva, Switzerland
| | - Francesca Poggio
- Department of Medical Oncology, Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Piero Fregatti
- Breast Surgery Clinic, IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Integrated Surgical and Diagnostic Sciences (DISC), University of Genoa, Genoa, Italy
| | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Matteo Bozzo
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| | - Simona Candiani
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Federica Papaccio
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Marco Greppi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy.
| | - Silvia Pesce
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Emanuela Marcenaro
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
4
|
Gang X, Yan J, Li X, Shi S, Xu L, Liu R, Cai L, Li H, Zhao M. Immune checkpoint inhibitors rechallenge in non-small cell lung cancer: Current evidence and future directions. Cancer Lett 2024; 604:217241. [PMID: 39260670 DOI: 10.1016/j.canlet.2024.217241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/23/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Immunotherapy, remarkably immune checkpoint inhibitors (ICIs), has significantly altered the treatment landscape for non-small cell lung cancer (NSCLC). Despite their success, the discontinuation of ICIs therapy may occur due to factors such as prior treatment completion, disease progression during ICIs treatment, or immune-related adverse events (irAEs). As numerous studies highlight the dynamic nature of immune responses and the sustained benefits of ICIs, ICIs rechallenge has become an attractive and feasible option. However, the decision-making process for ICIs rechallenge in clinical settings is complicated by numerous uncertainties. This review systematically analyses existing clinical research evidence, classifying ICIs rechallenge into distinct clinical scenarios, exploring methods to overcome ICIs resistance in rechallenge instances, and identifying biomarkers to select patients likely to benefit from rechallenge. By integrating recent studies and new technologies, we offer crucial recommendations for future clinical trial design and provide a practical guideline to maximize the therapeutic benefits of immunotherapy for NSCLC patients.
Collapse
Affiliation(s)
- Xiaoyu Gang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Jinshan Yan
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xin Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Sha Shi
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Lu Xu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ruotong Liu
- Clinical Medicine, Shenyang Medical College, Shenyang, 110001, China
| | - Lutong Cai
- Psychological Medicine, Shenyang Medical College, Shenyang, 110001, China
| | - Heming Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China; Guangdong Association of Clinical Trials (GACT)/Chinese Thoracic Oncology Group (CTONG) and Guangdong Provincial Key Lab of Translational Medicine in Lung Cancer, Guangzhou, 510000, China.
| | - Mingfang Zhao
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
5
|
Gunawardana J, Law SC, Sabdia MB, Fennell É, Hennessy A, Leahy CI, Murray PG, Bednarska K, Brosda S, Trotman J, Berkahn L, Zaharia A, Birch S, Burgess M, Talaulikar D, Lee JN, Jude E, Hawkes EA, Jain S, Nath K, Snell C, Swain F, Tobin JWD, Keane C, Shanavas M, Blyth E, Steidl C, Savage K, Farinha P, Boyle M, Meissner B, Green MR, Vega F, Gandhi MK. Intra-tumoral and peripheral blood TIGIT and PD-1 as immune biomarkers in nodular lymphocyte predominant Hodgkin lymphoma. Am J Hematol 2024; 99:2096-2107. [PMID: 39152767 PMCID: PMC11469944 DOI: 10.1002/ajh.27459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/28/2024] [Indexed: 08/19/2024]
Abstract
In classical Hodgkin lymphoma (cHL), responsiveness to immune-checkpoint blockade (ICB) is associated with specific tumor microenvironment (TME) and peripheral blood features. The role of ICB in nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) is not established. To gain insights into its potential in NLPHL, we compared TME and peripheral blood signatures between HLs using an integrative multiomic analysis. A discovery/validation approach in 121 NLPHL and 114 cHL patients highlighted >2-fold enrichment in programmed cell death-1 (PD-1) and T-cell Ig and ITIM domain (TIGIT) gene expression for NLPHL versus cHL. Multiplex imaging showed marked increase in intra-tumoral protein expression of PD-1+ (and/or TIGIT+) CD4+ T-cells and PD-1+CD8+ T-cells in NLPHL compared to cHL. This included T-cells that rosetted with lymphocyte predominant (LP) and Hodgkin Reed-Sternberg (HRS) cells. In NLPHL, intra-tumoral PD-1+CD4+ T-cells frequently expressed TCF-1, a marker of heightened T-cell response to ICB. The peripheral blood signatures between HLs were also distinct, with higher levels of PD-1+TIGIT+ in TH1, TH2, and regulatory CD4+ T-cells in NLPHL versus cHL. Circulating PD-1+CD4+ had high levels of TCF-1. Notably, in both lymphomas, highly expanded populations of clonal TIGIT+PD-1+CD4+ and TIGIT+PD-1+CD8+ T-cells in the blood were also present in the TME, indicating that immune-checkpoint expressing T-cells circulated between intra-tumoral and blood compartments. In in vitro assays, ICB was capable of reducing rosette formation around LP and HRS cells, suggesting that disruption of rosetting may be a mechanism of action of ICB in HL. Overall, results indicate that further evaluation of ICB is warranted in NLPHL.
Collapse
Affiliation(s)
- Jay Gunawardana
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Soi C. Law
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Muhammed B. Sabdia
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Éanna Fennell
- School of Medicine, Limerick Digital Cancer Research Centre, Health Research Institute and Bernal Institute, University of Limerick, Limerick, Ireland
| | - Aoife Hennessy
- School of Medicine, Limerick Digital Cancer Research Centre, Health Research Institute and Bernal Institute, University of Limerick, Limerick, Ireland
| | - Ciara I. Leahy
- School of Medicine, Limerick Digital Cancer Research Centre, Health Research Institute and Bernal Institute, University of Limerick, Limerick, Ireland
| | - Paul G. Murray
- School of Medicine, Limerick Digital Cancer Research Centre, Health Research Institute and Bernal Institute, University of Limerick, Limerick, Ireland
- Royal College of Surgeons Ireland, Adliya, Bahrain
| | - Karolina Bednarska
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Sandra Brosda
- Frazer Institute, University of Queensland, Translational Research Institute Brisbane, Australia
| | - Judith Trotman
- Concord Repatriation General Hospital, University of Sydney, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW
| | - Leanne Berkahn
- Department of Haematology, Auckland City Hospital, Auckland, New Zealand
| | - Andreea Zaharia
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Simone Birch
- Princess Alexandra Hospital, Brisbane, Australia
| | - Melinda Burgess
- School of Medicine, Limerick Digital Cancer Research Centre, Health Research Institute and Bernal Institute, University of Limerick, Limerick, Ireland
- Princess Alexandra Hospital, Brisbane, Australia
| | - Dipti Talaulikar
- Haematology Translational Research Unit, ACT Pathology, Canberra Health Services, Canberra, Australia
- College of Health and Medicine, Australian National University, Canberra, Australia
| | - Justina N. Lee
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | | | - Eliza A. Hawkes
- Olivia Newton John Cancer Research and Wellness Centre, Austin Health, Melbourne, Australia
- Transfusion Research Unit, School of Public Health and Preventative Medicine, Monash University, Melbourne Australia
| | - Sanjiv Jain
- Anatomical Pathology Department, The Canberra Hospital, Canberra, Australia
| | - Karthik Nath
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
- Memorial Sloan Kettering Cancer Center, NY, USA
| | - Cameron Snell
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Mater Pathology, Brisbane, Australia
| | - Fiona Swain
- Royal College of Surgeons Ireland, Adliya, Bahrain
- Princess Alexandra Hospital, Brisbane, Australia
| | - Joshua W. D. Tobin
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
- Princess Alexandra Hospital, Brisbane, Australia
| | - Colm Keane
- Frazer Institute, University of Queensland, Translational Research Institute Brisbane, Australia
- Princess Alexandra Hospital, Brisbane, Australia
| | - Mohamed Shanavas
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Emily Blyth
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW
- Department of Haematology, Westmead Hospital, Westmead, NSW, Australia
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW
| | | | - Kerry Savage
- British Columbia Cancer Agency, Vancouver, Canada
| | | | | | | | | | - Francisco Vega
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maher K. Gandhi
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
- Princess Alexandra Hospital, Brisbane, Australia
| |
Collapse
|
6
|
Malhotra J, Huang A, Amini A, Lee P. Novel Immunotherapeutics for the Treatment of Non-Small Cell Lung Cancer (NSCLC) Resistant to PD-1/PD-L1 Inhibitors. Cancers (Basel) 2024; 16:3603. [PMID: 39518043 PMCID: PMC11545025 DOI: 10.3390/cancers16213603] [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: 09/30/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Immunotherapy with PD-1/PD-L1 inhibitors is the standard method of care for the treatment of newly diagnosed advanced or metastatic NSCLC, with or without chemotherapy. Many tumors, however, develop resistance to these immunotherapy agents. There is a need to develop more effective therapies for patients with metastatic NSCLC in the second-line setting and beyond. In this review, we present an overview of novel immunotherapies being investigated regarding the treatment of these patients. We summarize completed, as well as ongoing, trials investigating these therapies as monotherapy or in combination with PD-1/PD-L1 inhibitors. These include immune co-stimulatory antibodies, T-cell agonists, oncolytic viruses, vaccines, TIL therapies, and CAR-T therapies.
Collapse
Affiliation(s)
- Jyoti Malhotra
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Amy Huang
- Department of Medicine, University of Connecticut School of Medicine, Farmington, CT 06030, USA;
| | - Arya Amini
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.A.); (P.L.)
| | - Percy Lee
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.A.); (P.L.)
| |
Collapse
|
7
|
Zhang MG, Gallo RA, Tan CH, Camacho M, Fasih-Ahmad S, Moeyersoms AHM, Sayegh Y, Dubovy SR, Pelaez D, Rong AJ. Single-Cell RNA Profiling of Ocular Adnexal Sebaceous Carcinoma Reveals a Complex Tumor Microenvironment and Identifies New Biomarkers. Am J Ophthalmol 2024; 270:8-18. [PMID: 39393421 DOI: 10.1016/j.ajo.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/23/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
PURPOSE Ocular adnexal sebaceous carcinoma (OaSC) is an aggressive malignancy that often necessitates orbital exenteration. Its tumor composition and transcriptional profile remain largely unknown, which poses a significant barrier to medical advances. Here, we report the first in-depth transcriptomic analysis of OaSC at the single-cell resolution and discern mechanisms underlying cancer progression for the discovery of potential globe-sparing immunotherapies, targeted therapies, and biomarkers to guide clinical management. DESIGN Laboratory investigation with a retrospective observational case series. METHODS Single-cell RNA sequencing was performed on six patient specimens: three primary tumors, two tumors with pagetoid spread, and a normal tarsus sample. Cellular components were identified via gene signatures. Molecular pathways underlying tumorigenesis and pagetoid spread were discerned via gene ontology analysis of the differentially expressed genes between specimens. CALML5 immunohistochemistry was performed on an archival cohort of OaSC, squamous cell carcinoma, ocular surface squamous neoplasia (OSSN), and basal cell carcinoma cases. RESULTS Analysis of 29,219 cells from OaSC specimens revealed tumor, immune, and stromal cells. Tumor-infiltrating immune cells include a diversity of cell types, including exhausted T-cell populations. In primary OaSC tumors, mitotic nuclear division and oxidative phosphorylation pathways are upregulated, while lipid biosynthesis and metabolism pathways are downregulated. Epithelial tissue migration pathways are upregulated in tumor cells undergoing pagetoid spread. Single-cell RNA sequencing analyses also revealed that CALML5 is upregulated in OaSC tumor cells. Diffuse nuclear and cytoplasmic CALML5 staining was present in 28 of 28 (100%) OaSC cases. Diffuse nuclear and membranous CALML5 staining was present in 5 of 25 (20%) squamous cell carcinoma and OSSN cases, while diffuse nuclear staining was present in 1 of 12 (8%) basal cell carcinoma cases. CONCLUSIONS This study reveals a complex OaSC tumor microenvironment and confirms that the CALML5 immunohistochemical stain is a sensitive diagnostic marker.
Collapse
Affiliation(s)
- Michelle G Zhang
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ryan A Gallo
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Charissa H Tan
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Matthew Camacho
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sohaib Fasih-Ahmad
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Acadia H M Moeyersoms
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Yoseph Sayegh
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sander R Dubovy
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Daniel Pelaez
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Andrew J Rong
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA; Division of Oculofacial Plastic, Reconstructive, and Orbital Surgery (A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
8
|
Liu Y, Zhang J, Li S, Chen W, Wu R, Hao Z, Xu J. Prediction of TNFRSF9 expression and molecular pathological features in thyroid cancer using machine learning to construct Pathomics models. Endocrine 2024; 86:324-332. [PMID: 38753243 DOI: 10.1007/s12020-024-03862-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/04/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND The TNFRSF9 molecule is pivotal in thyroid carcinoma (THCA) development. This study utilizes Pathomics techniques to predict TNFRSF9 expression in THCA tissue and explore its molecular mechanisms. METHODS Transcriptome data, pathology images, and clinical information from the cancer genome atlas (TCGA) were analyzed. Image segmentation and feature extraction were performed using the OTSU's algorithm and pyradiomics package. The dataset was split for training and validation. Features were selected using maximum relevance minimum redundancy recursive feature elimination (mRMR_RFE) and modeling conducted with the gradient boosting machine (GBM) algorithm. Model evaluation included receiver operating characteristic curve (ROC) analysis. The Pathomics model output a probabilistic pathomics score (PS) for gene expression prediction, with its prognostic value assessed in TNFRSF9 expression groups. Subsequent analysis involved gene set variation analysis (GSVA), immune gene expression, cell abundance, immunotherapy susceptibility, and gene mutation analysis. RESULTS High TNFRSF9 expression correlated with worsened progression-free interval (PFI) and acted as an independent risk factor [hazard ratio (HR) = 2.178, 95% confidence interval (CI) 1.045-4.538, P = 0.038]. Nine pathohistological features were identified. The GBM Pathomics model demonstrated good prediction efficacy [area under the curve (AUC) 0.819 and 0.769] and clinical benefits. High PS was a PFI risk factor (HR = 2.156, 95% CI 1.047-4.440, P = 0.037). Patients with high PS potentially exhibited enriched pathways, increased TIGIT gene expression, Tregs infiltration (P < 0.0001), and higher rates of gene mutations (BRAF, TTN, TG). CONCLUSIONS The GBM Pathomics model constructed based on the pathohistological features of H&E-stained sections well predicted the expression level of TNFRSF9 molecules in THCA.
Collapse
Affiliation(s)
- Ying Liu
- Department of Endocrine and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, Jiangxi, China
- Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, Jiangxi, China
| | - Junping Zhang
- Department of Endocrine and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shanshan Li
- Department of Endocrine and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wen Chen
- Department of Endocrine and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Rongqian Wu
- Department of Endocrine and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zejin Hao
- Department of Endocrine and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jixiong Xu
- Department of Endocrine and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, Jiangxi, China.
- Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, Jiangxi, China.
| |
Collapse
|
9
|
Li C, Yu X, Han X, Lian C, Wang Z, Shao S, Shao F, Wang H, Ma S, Liu J. Innate immune cells in tumor microenvironment: A new frontier in cancer immunotherapy. iScience 2024; 27:110750. [PMID: 39280627 PMCID: PMC11399700 DOI: 10.1016/j.isci.2024.110750] [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] [Indexed: 09/18/2024] Open
Abstract
Innate immune cells, crucial in resisting infections and initiating adaptive immunity, play diverse and significant roles in tumor development. These cells, including macrophages, granulocytes, dendritic cells (DCs), innate lymphoid cells, and innate-like T cells, are pivotal in the tumor microenvironment (TME). Innate immune cells are crucial components of the TME, based on which various immunotherapy strategies have been explored. Immunotherapy strategies, such as novel immune checkpoint inhibitors, STING/CD40 agonists, macrophage-based surface backpack anchoring, ex vivo polarization approaches, DC-based tumor vaccines, and CAR-engineered innate immune cells, aim to enhance their anti-tumor potential and counteract cancer-induced immunosuppression. The proximity of innate immune cells to tumor cells in the TME also makes them excellent drug carriers. In this review, we will first provide a systematic overview of innate immune cells within the TME and then discuss innate cell-based therapeutic strategies. Furthermore, the research obstacles and perspectives within the field will also be addressed.
Collapse
Affiliation(s)
- Changhui Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xinyu Yu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xinyan Han
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Chen Lian
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Zijin Wang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Fangwei Shao
- National Key Laboratory of Biobased Transportation Fuel Technology, ZJU-UIUC Institute, Zhejiang University, Hangzhou 310027, China
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Shenglin Ma
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou 310002, China
| | - Jian Liu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Biomedical and Heath Translational Research Center of Zhejiang Province, Haining, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
10
|
Farkas AM, Youssef D, Tran MA, Balan S, Newman JH, Audenet F, Anastos H, Velazquez LG, Peros A, Ananthanarayanan A, Daza J, Gonzalez-Gugel E, Sadanala K, Theorell J, Galsky MD, Horowitz A, Sfakianos JP, Bhardwaj N. Natural Killer Cell Dysfunction In Human Bladder Cancer Is Caused By Tissue-Specific Suppression of SLAMF6 Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591366. [PMID: 38746459 PMCID: PMC11092609 DOI: 10.1101/2024.04.30.591366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
NK cells are innate lymphocytes critical for surveillance of viruses and tumors, however the mechanisms underlying NK cell dysfunction in cancer are incompletely understood. We assessed the effector function of NK cells from bladder cancer patients and found severe dysfunction in NK cells derived from tumors versus peripheral blood. While both peripheral and tumor-infiltrating NK cells exhibited conserved patterns of inhibitory receptor over-expression, this did not explain the observed defects in NK surveillance in bladder tumors. Rather, TME-specific TGF-β and metabolic perturbations such as hypoxia directly suppressed NK cell function. Specifically, an oxygen-dependent reduction in signaling through SLAMF6 was mechanistically responsible for poor NK cell function, as tumor-infiltrating NK cells cultured ex vivo under normoxic conditions exhibited complete restoration of function, while deletion of SLAMF6 abrogated NK cell cytolytic function even under normoxic conditions. Collectively, this work highlights the role of tissue-specific factors in dictating NK cell function, and implicates SLAMF6 signaling as a rational target for immuno-modulation to improve NK cell function in bladder cancer.
Collapse
|
11
|
Shapira-Frommer R, Niu J, Perets R, Peters S, Shouse G, Lugowska I, Garassino MC, Sands J, Keenan T, Zhao B, Healy J, Ahn MJ. The KEYVIBE program: vibostolimab and pembrolizumab for the treatment of advanced malignancies. Future Oncol 2024; 20:1983-1991. [PMID: 39230120 PMCID: PMC11497960 DOI: 10.1080/14796694.2024.2343272] [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: 12/12/2023] [Accepted: 04/11/2024] [Indexed: 09/05/2024] Open
Abstract
Vibostolimab, a humanized IgG1 monoclonal antibody, blocks the interaction between TIGIT and its ligands, preventing the immunosuppressive effects of TIGIT. The addition of vibostolimab to the PD-1 inhibitor pembrolizumab has shown promising antitumor activity, warranting further exploration of vibostolimab as a potential therapeutic option. The KEYVIBE program consists of nine trials that will evaluate the safety and efficacy of vibostolimab monotherapy and vibostolimab-based combination therapy in advanced solid tumors and hematological malignancies. These studies will also evaluate coformulated immunotherapy addressing issues that occur with the sequential administration of immunotherapy. The KEYVIBE program will provide further insight into the clinical utility of vibostolimab-based therapy across multiple indications and various stages of disease.
Collapse
Affiliation(s)
- Ronnie Shapira-Frommer
- Division of Oncology and Clinical Research Institute at Rambam, Rambam Medical Center, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, 5265601, Israel
| | - Jiaxin Niu
- Banner MD Anderson Cancer Center, Gilbert, AZ85234, USA
| | - Ruth Perets
- Clinical Research Institute at Rambam, Rambam Medical Center & Technion—Israel Institute of Technology, Haifa, 3109601,Israel
| | - Solange Peters
- Vaud University Hospital Center, University of Lausanne, Lausanne, 1005, Switzerland
| | - Geoffrey Shouse
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA91010, USA
| | - Iwona Lugowska
- Early Phase Clinical Trials Unit, Maria Sklodowska Curie National Research Institute of Oncology, Warsaw, 00-001, Poland
| | | | - Jacob Sands
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA02215, USA
| | | | - Bin Zhao
- Merck & Co., Inc., Rahway, NJ07065, USA
| | | | - Myung-Ju Ahn
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| |
Collapse
|
12
|
Sun J, Tian Y, Yang C. Target therapy of TIGIT; a novel approach of immunotherapy for the treatment of colorectal cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03346-7. [PMID: 39158733 DOI: 10.1007/s00210-024-03346-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024]
Abstract
The T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT), a newly discovered checkpoint, is characterized by its elevated expression on CD4 + T cells, CD8 + T cells, natural killer (NK) cells, regulatory T cells (Tregs), and tumor-infiltrating lymphocytes (TILs). Research to date has been shown that TIGIT has been linked to exhaustion of NK cell both and T cells in numerous cancers. CD155, being the specific ligand of TIGIT in humans, emerges as a key target for immunotherapy owing to its crucial interaction with TIGIT. Furthermore, numerous studies have demonstrated that the combination of TIGIT with other immune checkpoint inhibitors (ICIs) and/or traditional treatments elicits a potent antitumor response in colorectal cancer (CRC). This review provides an overview of the structure, function, and signaling pathways associated with TIGIT across multiple immune system cell types. Additionally, focusing on the role of TIGIT in the progression of CRC, this study reviewed various studies exploring TIGIT-based immunotherapy in CRC.
Collapse
Affiliation(s)
- Jing Sun
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Yan Tian
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Changqing Yang
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China.
| |
Collapse
|
13
|
Ye QN, Zhu L, Liang J, Zhao DK, Tian TY, Fan YN, Ye SY, Liu H, Huang XY, Cao ZT, Shen S, Wang J. Orchestrating NK and T cells via tri-specific nano-antibodies for synergistic antitumor immunity. Nat Commun 2024; 15:6211. [PMID: 39043643 PMCID: PMC11266419 DOI: 10.1038/s41467-024-50474-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 07/08/2024] [Indexed: 07/25/2024] Open
Abstract
The functions of natural killer (NK) and T cells in innate and adaptive immunity, as well as their functions in tumor eradication, are complementary and intertwined. Here we show that utilization of multi-specific antibodies or nano-antibodies capable of simultaneously targeting both NK and T cells could be a valuable approach in cancer immunotherapy. Here, we introduce a tri-specific Nano-Antibody (Tri-NAb), generated by immobilizing three types of monoclonal antibodies (mAbs), using an optimized albumin/polyester composite nanoparticle conjugated with anti-Fc antibody. This Tri-NAb, targeting PDL1, 4-1BB, and NKG2A (or TIGIT) simultaneously, effectively binds to NK and CD8+ T cells, triggering their activation and proliferation, while facilitating their interaction with tumor cells, thereby inducing efficient tumor killing. Importantly, the antitumor efficacy of Tri-NAb is validated in multiple models, including patient-derived tumor organoids and humanized mice, highlighting the translational potential of NK and T cell co-targeting.
Collapse
Affiliation(s)
- Qian-Ni Ye
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, P. R. China
| | - Long Zhu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, P. R. China
| | - Jie Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, P. R. China
| | - Dong-Kun Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, P. R. China
| | - Tai-Yu Tian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, P. R. China
| | - Ya-Nan Fan
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, P. R. China
| | - Si-Yi Ye
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, P. R. China
| | - Hua Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, P. R. China
| | - Xiao-Yi Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, P. R. China
| | - Zhi-Ting Cao
- School of Biopharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Song Shen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, P. R. China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, P. R. China.
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, P. R. China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, P. R. China.
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, P. R. China.
| |
Collapse
|
14
|
Vanmeerbeek I, Naulaerts S, Sprooten J, Laureano RS, Govaerts J, Trotta R, Pretto S, Zhao S, Cafarello ST, Verelst J, Jacquemyn M, Pociupany M, Boon L, Schlenner SM, Tejpar S, Daelemans D, Mazzone M, Garg AD. Targeting conserved TIM3 +VISTA + tumor-associated macrophages overcomes resistance to cancer immunotherapy. SCIENCE ADVANCES 2024; 10:eadm8660. [PMID: 39028818 PMCID: PMC11259173 DOI: 10.1126/sciadv.adm8660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 06/14/2024] [Indexed: 07/21/2024]
Abstract
Despite the success of immunotherapy, overcoming immunoresistance in cancer remains challenging. We identified a unique niche of tumor-associated macrophages (TAMs), coexpressing T cell immunoglobulin and mucin domain-containing 3 (TIM3) and V-domain immunoglobulin suppressor of T cell activation (VISTA), that dominated human and mouse tumors resistant to most of the currently used immunotherapies. TIM3+VISTA+ TAMs were sustained by IL-4-enriching tumors with low (neo)antigenic and T cell-depleted features. TIM3+VISTA+ TAMs showed an anti-inflammatory and protumorigenic phenotype coupled with inability to sense type I interferon (IFN). This was established with cancer cells succumbing to immunogenic cell death (ICD). Dying cancer cells not only triggered autocrine type I IFNs but also exposed HMGB1/VISTA that engaged TIM3/VISTA on TAMs to suppress paracrine IFN-responses. Accordingly, TIM3/VISTA blockade synergized with paclitaxel, an ICD-inducing chemotherapy, to repolarize TIM3+VISTA+ TAMs to proinflammatory TAMs that killed cancer cells via tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) signaling. We propose targeting TIM3+VISTA+ TAMs to overcome immunoresistant tumors.
Collapse
Affiliation(s)
- Isaure Vanmeerbeek
- Laboratory of Cell Stress and Immunity, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Naulaerts
- Laboratory of Cell Stress and Immunity, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jenny Sprooten
- Laboratory of Cell Stress and Immunity, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Raquel S. Laureano
- Laboratory of Cell Stress and Immunity, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jannes Govaerts
- Laboratory of Cell Stress and Immunity, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Rosa Trotta
- Laboratory of Tumour Inflammation and Angiogenesis, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Tumour Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Samantha Pretto
- Laboratory of Tumour Inflammation and Angiogenesis, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Tumour Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Shikang Zhao
- Laboratory of Tumour Inflammation and Angiogenesis, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Tumour Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sarah Trusso Cafarello
- Laboratory of Tumour Inflammation and Angiogenesis, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Tumour Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Joren Verelst
- Laboratory of Tumour Inflammation and Angiogenesis, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Tumour Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Maarten Jacquemyn
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, KU Leuven, Rega Institute, Leuven, Belgium
| | - Martyna Pociupany
- Laboratory of Cell Stress and Immunity, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | | | - Susan M. Schlenner
- Laboratory of Adaptive Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Sabine Tejpar
- Laboratory for Molecular Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Dirk Daelemans
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, KU Leuven, Rega Institute, Leuven, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumour Inflammation and Angiogenesis, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Tumour Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Abhishek D. Garg
- Laboratory of Cell Stress and Immunity, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Tong LW, Hu YS, Yu SJ, Li CL, Shao JW. Current application and future perspective of CRISPR/cas9 gene editing system mediated immune checkpoint for liver cancer treatment. NANOTECHNOLOGY 2024; 35:402002. [PMID: 38964289 DOI: 10.1088/1361-6528/ad5f33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 07/04/2024] [Indexed: 07/06/2024]
Abstract
Liver cancer, which is well-known to us as one of human most prevalent malignancies across the globe, poses a significant risk to live condition and life safety of individuals in every region of the planet. It has been shown that immune checkpoint treatment may enhance survival benefits and make a significant contribution to patient prognosis, which makes it a promising and popular therapeutic option for treating liver cancer at the current time. However, there are only a very few numbers of patients who can benefit from the treatment and there also exist adverse events such as toxic effects and so on, which is still required further research and discussion. Fortunately, the clustered regularly interspaced short palindromic repeat/CRISPR-associated nuclease 9 (CRISPR/Cas9) provides a potential strategy for immunotherapy and immune checkpoint therapy of liver cancer. In this review, we focus on elucidating the fundamentals of the recently developed CRISPR/Cas9 technology as well as the present-day landscape of immune checkpoint treatment which pertains to liver cancer. What's more, we aim to explore the molecular mechanism of immune checkpoint treatment in liver cancer based on CRISPR/Cas9 technology. At last, its encouraging and powerful potential in the future application of the clinic is discussed, along with the issues that already exist and the difficulties that must be overcome. To sum up, our ultimate goal is to create a fresh knowledge that we can utilize this new CRISPR/Cas9 technology for the current popular immune checkpoint therapy to overcome the treatment issues of liver cancer.
Collapse
Affiliation(s)
- Ling-Wu Tong
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Yong-Shan Hu
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Shi-Jing Yu
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Cheng-Lei Li
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Jing-Wei Shao
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| |
Collapse
|
16
|
Shemesh CS, Wang Y, An A, Ding H, Chan P, Liu Q, Chen YW, Wu B, Wu Q, Wang X. Phase I pharmacokinetic, safety, and preliminary efficacy study of tiragolumab in combination with atezolizumab in Chinese patients with advanced solid tumors. Cancer Chemother Pharmacol 2024; 94:45-55. [PMID: 38451273 PMCID: PMC11258083 DOI: 10.1007/s00280-024-04650-y] [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: 12/21/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE Tiragolumab is an immunoglobulin G1 monoclonal antibody targeting the immune checkpoint T cell immunoreceptor with immunoglobulin and immunoreceptor ITIM domains. Targeting multiple immune pathways may improve anti-tumor responses. The phase I YP42514 study assessed the pharmacokinetics (PK), safety, and preliminary efficacy of tiragolumab plus atezolizumab in Chinese patients with advanced solid tumors. METHODS Adult patients from mainland China with Eastern Cooperative Oncology Group performance score 0/1, life expectancy of ≥ 12 weeks, and adequate hematologic/end organ function were eligible. Patients received tiragolumab 600 mg and atezolizumab 1200 mg intravenous every 3 weeks. Key endpoints were PK (serum concentrations of tiragolumab and atezolizumab) and safety. Results from this study were compared with the global phase I study, GO30103 (NCT02794571). RESULTS In this study, 20 patients received a median of five doses of tiragolumab plus atezolizumab. Median age was 57.5 years, 85.0% of patients were male and the most common tumor type was non-small cell lung cancer. Exposures in Chinese patients were comparable to the global GO30103 population: geometric mean ratio was 1.07 for Cycle 1 tiragolumab area under the concentration-time curve0-21 and 0.92 and 0.93 for Cycle 1 peak and trough atezolizumab exposure, respectively. Treatment-related adverse events were consistent across the Chinese and global populations. Two patients (10.0%) in this study achieved a partial response. CONCLUSION In this study, tiragolumab plus atezolizumab was tolerable and demonstrated preliminary anti-tumor activity. There were no meaningful differences in the PK or safety of tiragolumab plus atezolizumab between the Chinese and global populations. CLINICAL TRIAL REGISTRATION NUMBER China Clinical Trial Registry Identifier CTR20210219/YP42514. Date of registration 16 March 2021.
Collapse
Affiliation(s)
- Colby S Shemesh
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA.
| | - Yongsheng Wang
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Andrew An
- Safety Science, F. Hoffmann-La Roche Ltd, Beijing, China
| | - Hao Ding
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Phyllis Chan
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Qi Liu
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Yih-Wen Chen
- Bioanalytical Science, Genentech Inc., South San Francisco, CA, USA
| | - Benjamin Wu
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Qiong Wu
- Product Development Oncology, F. Hoffmann-La Roche Ltd, Shanghai, China
| | - Xian Wang
- Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Becker W, Olkhanud PB, Seishima N, Moreno PA, Goldfarbmuren KC, Maeng HM, Berzofsky JA. Second-generation checkpoint inhibitors and Treg depletion synergize with a mouse cancer vaccine in accordance with tumor microenvironment characterization. J Immunother Cancer 2024; 12:e008970. [PMID: 38955422 PMCID: PMC11218019 DOI: 10.1136/jitc-2024-008970] [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: 05/28/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Despite advances in checkpoint inhibitor (CPI) therapy for cancer treatment, many cancers remain resistant. Tumors deemed "cold" based on lack of T cell infiltration show reduced potential for CPI therapy. Cancer vaccines may overcome the inadequacy of existing T cells by inducing the needed antitumor T cell response to synergize with CPIs and overcome resistance. METHODS CT26 and TC1 tumor cells were injected subcutaneously into mice. Mice were treated with combinations of CPIs alone or a cancer vaccine specific to the tumor antigen E7 present in TC1 cells. CPIs for the TC1 model were selected because of immunophenotyping TC1 tumors. Antitumor and protumor immunity, tumor size and survival, sequence and timing of vaccine and CPI administration, and efficacy of treatment in young and aged mice were probed. RESULTS While "hot" CT26 tumors are treatable with combinations of second-generation CPIs alone or with anti-TGFβ, "cold" TC1 tumor reduction requires the synergy of a tumor-antigen-specific vaccine in combination with two CPIs, anti-TIGIT and anti-PD-L1, predicted by tumor microenvironment (TME) characterization. The synergistic triple combination delays tumor growth better than any pairwise combination and improves survival in a CD8+T cell-dependent manner. Depletion of CD4+T cells improved the treatment response, and depleting regulatory T cells (Treg) revealed Tregs to be inhibiting the response as also predicted from TME analysis. We found the sequence of CPI and vaccine administration dictates the success of the treatment, and the triple combination administered concurrently induces the highest E7-specific T cell response. Contrary to young mice, in aged mice, the cancer vaccine alone is ineffective, requiring the CPIs to delay tumor growth. CONCLUSIONS These findings show how pre-existing or vaccine-mediated de novo T cell responses can both be amplified by and facilitate synergistic CPIs and Treg depletion that together lead to greater survival, and how analysis of the TME can help rationally design combination therapies and precision medicine to enhance clinical response to CPI and cancer vaccine therapy.
Collapse
Affiliation(s)
- William Becker
- Vaccine Branch, CCR, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Purevdorj B Olkhanud
- Vaccine Branch, CCR, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Noriko Seishima
- Vaccine Branch, CCR, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Paloma A Moreno
- Vaccine Branch, CCR, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine C Goldfarbmuren
- Vaccine Branch, CCR, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- Advanced Biomedical Computational Science, Leidos Biomedical Research Inc, Frederick, Maryland, USA
| | - Hoyoung M Maeng
- Vaccine Branch, CCR, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jay A Berzofsky
- Vaccine Branch, CCR, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
18
|
Yamamoto N, Koyama T, Sato J, Yoshida T, Sudo K, Iwasa S, Kondo S, Yonemori K, Kawasaki A, Satake K, Shibata S, Shimizu T. Phase I study of the anti-TIGIT antibody tiragolumab in combination with atezolizumab in Japanese patients with advanced or metastatic solid tumors. Cancer Chemother Pharmacol 2024; 94:109-115. [PMID: 38206370 PMCID: PMC11258096 DOI: 10.1007/s00280-023-04627-3] [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: 08/24/2023] [Accepted: 11/24/2023] [Indexed: 01/12/2024]
Abstract
PURPOSE Tiragolumab is a monoclonal antibody that binds to the inhibitory immune checkpoint TIGIT (T-cell immunoreceptor with Ig and ITIM domains). In early phase clinical trials, tiragolumab in combination with the programmed death-ligand 1-inhibitor atezolizumab was well tolerated and has demonstrated preliminary anti-tumor activity in patients with advanced/metastatic solid tumors. We report the results of a phase I study of tiragolumab plus atezolizumab in Japanese patients (jRCT2080224926). METHODS Japanese patients ≥ 20 years old received tiragolumab (600 mg) and atezolizumab (1200 mg) intravenously every 21 days until unacceptable toxicity or disease progression. Primary endpoints were safety and pharmacokinetic (PK) parameters of tiragolumab plus atezolizumab. Secondary endpoints were anti-tumor activity. RESULTS Three patients were enrolled with diagnoses of non-small cell lung cancer, pancreatic cancer, and cholangiocarcinoma. No dose-limiting toxicities were observed. Two patients experienced treatment-related adverse events (AEs) of any grade. There were no grade ≥ 3 AEs, serious AEs, AEs leading to discontinuation, modification or withdrawal of any study drug, or AEs leading to death. At cycle 1, mean PK parameters of tiragolumab were as follows: Cmax 217 μg/mL; Cmin 54.9 μg/mL; area under the concentration-time curve from 0 to the last measurable concentration, 2000 μg·day/mL; t1/2, 17.6 days. Best overall response was stable disease in two patients. CONCLUSION Tiragolumab plus atezolizumab was well tolerated in Japanese patients with advanced/metastatic solid tumors, and no differences in tiragolumab PK characteristics were noted between Japanese patients enrolled in this study, and non-Japanese patients enrolled in a global phase Ia/Ib study. These results may support the inclusion of Japanese patients in ongoing global phase III clinical trials. TRIAL REGISTRATION NUMBER jRCT2080224926.
Collapse
Affiliation(s)
- Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-Ku, Tokyo, 104-0045, Japan.
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Jun Sato
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Tatsuya Yoshida
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Kazuki Sudo
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Satoru Iwasa
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shunsuke Kondo
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Kan Yonemori
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Atsuko Kawasaki
- Chugai Pharmaceutical Co., Ltd, 1-1 Nihonbashi-Muromachi 2-Chome Chuo-Ku, Tokyo, 103-8324, Japan
| | - Kyoko Satake
- Chugai Pharmaceutical Co., Ltd, 1-1 Nihonbashi-Muromachi 2-Chome Chuo-Ku, Tokyo, 103-8324, Japan
| | - Shoyo Shibata
- Chugai Pharmaceutical Co., Ltd, 1-1 Nihonbashi-Muromachi 2-Chome Chuo-Ku, Tokyo, 103-8324, Japan
| | - Toshio Shimizu
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-Ku, Tokyo, 104-0045, Japan
| |
Collapse
|
19
|
Rosellini M, Marchetti A, Tassinari E, Mollica V, Massari F, Santoni M. Do we need alternative PD-1 inhibitors for the treatment of renal cell carcinoma? Expert Opin Biol Ther 2024; 24:411-414. [PMID: 38898658 DOI: 10.1080/14712598.2024.2369190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Affiliation(s)
- Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Elisa Tassinari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | | |
Collapse
|
20
|
Lang Y, Huang H, Jiang H, Wu S, Chen Y, Xu B, Liu Y, Zhu D, Zheng X, Chen L, Jiang J. TIGIT Blockade Reshapes the Tumor Microenvironment Based on the Single-cell RNA-Sequencing Analysis. J Immunother 2024; 47:172-181. [PMID: 38545758 DOI: 10.1097/cji.0000000000000511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/26/2024] [Indexed: 05/09/2024]
Abstract
SUMMARY Immune checkpoint blockade therapy is a pivotal approach in treating malignant tumors. TIGIT has emerged as a focal point of interest among the diverse targets for tumor immunotherapy. Nonetheless, there is still a lack of comprehensive understanding regarding the immune microenvironment alterations following TIGIT blockade treatment. To bridge this knowledge gap, we performed single-cell sequencing on mice both before and after the administration of anti-TIGIT therapy. Our analysis revealed that TIGIT was predominantly expressed on T cells and natural killer (NK) cells. The blockade of TIGIT exhibited inhibitory effects on Treg cells by downregulating the expression of Foxp3 and reducing the secretion of immunosuppressive cytokines. In addition, TIGIT blockade facilitated the activation of NK cells, leading to an increase in cell numbers, and promoted cDC1 maturation through the secretion of XCL1 and Flt3L. This activation, in turn, stimulated the TCR signaling of CD8 + T cells, thereby enhancing their antitumor effect. Consequently, anti-TIGIT therapy demonstrated substantial potential for cancer immunotherapy. Our research provided novel insights into future therapeutic strategies targeting TIGIT for patients with cancer.
Collapse
Affiliation(s)
- Yanyan Lang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Hao Huang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Hongwei Jiang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Yaping Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Bin Xu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Yingting Liu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Dawei Zhu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
21
|
Shalata W, Attal ZG, Solomon A, Shalata S, Abu Saleh O, Tourkey L, Abu Salamah F, Alatawneh I, Yakobson A. Melanoma Management: Exploring Staging, Prognosis, and Treatment Innovations. Int J Mol Sci 2024; 25:5794. [PMID: 38891988 PMCID: PMC11171767 DOI: 10.3390/ijms25115794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Melanoma, a malignant neoplasm originating from melanocytes, stands as one of the most prevalent cancers globally, ranking fifth in terms of estimated new cases in recent years. Its aggressive nature and propensity for metastasis pose significant challenges in oncology. Recent advancements have led to a notable shift towards targeted therapies, driven by a deeper understanding of cutaneous tumor pathogenesis. Immunotherapy and tyrosine kinase inhibitors have emerged as promising strategies, demonstrating the potential to improve clinical outcomes across all disease stages, including neoadjuvant, adjuvant, and metastatic settings. Notably, there has been a groundbreaking development in the treatment of brain metastasis, historically associated with poor prognosis in oncology but showcasing impressive results in melanoma patients. This review article provides a comprehensive synthesis of the most recent knowledge on staging and prognostic factors while highlighting emerging therapeutic modalities, with a particular focus on neoadjuvant and adjuvant strategies, notably immunotherapy and targeted therapies, including the ongoing trials.
Collapse
Affiliation(s)
- Walid Shalata
- The Legacy Heritage Cancer Center and Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Zoe Gabrielle Attal
- Medical School for International Health, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Adam Solomon
- Medical School for International Health, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Sondos Shalata
- Nutrition Unit, Galilee Medical Center, Nahariya 22000, Israel
| | - Omar Abu Saleh
- Department of Dermatology and Venereology, The Emek Medical Centre, Afula 18341, Israel
| | - Lena Tourkey
- The Legacy Heritage Cancer Center and Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Fahed Abu Salamah
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Department of Dermatology, Soroka Medical Center, Beer Sheva 84105, Israel
| | - Ibrahim Alatawneh
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Department of Dermatology, Soroka Medical Center, Beer Sheva 84105, Israel
| | - Alexander Yakobson
- The Legacy Heritage Cancer Center and Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
22
|
Ke J, Huang S, He Z, Lei S, Lin S, Duan M. TIGIT Regulates T Cell Inflammation in Airway Inflammatory Diseases. Inflammation 2024:10.1007/s10753-024-02045-y. [PMID: 38780694 DOI: 10.1007/s10753-024-02045-y] [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: 03/16/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
TIGIT, a co-inhibitory receptor found on T cells and NK cells, transmits inhibitory signals upon binding to its ligand. This interaction suppresses the activation of various signaling pathways, leading to functional exhaustion of cells, ultimately dampening excessive inflammatory responses or facilitating immune evasion in tumors. Dysregulated TIGIT expression has been noted in T cells across different inflammatory conditions, exhibiting varying effects based on T cell subsets. TIGIT predominantly restrains the effector function of pro-inflammatory T cells, upholds the suppressive function of regulatory T cells, and influences Tfh maturation. Mechanistically, the IL27-induced transcription factors c-Maf and Blimp-1 are believed to be key regulators of TIGIT expression in T cells. Notably, TIGIT expression in T cells is implicated in lung diseases, particularly airway inflammatory conditions such as lung cancer, obstructive pulmonary disease, interstitial lung disease, sarcoidosis, and COVID-19. This review emphasizes the significance of TIGIT in the context of T cell immunity and airway inflammatory diseases.
Collapse
Affiliation(s)
- Junyi Ke
- Guangxi Medical University, Nanning, China
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shu Huang
- Wuming Hospital of Guangxi Medical University, Nanning, China
| | | | - Siyu Lei
- Wuming Hospital of Guangxi Medical University, Nanning, China
| | - Shiya Lin
- Guangxi Medical University, Nanning, China
| | - Minchao Duan
- Wuming Hospital of Guangxi Medical University, Nanning, China.
- Department of Respiratory Medicine, Wuming Hospital of Guangxi Medical University, No.26 Yongning Road, Wuming District, Nanning, 530100, China.
| |
Collapse
|
23
|
Yin N, Li X, Zhang X, Xue S, Cao Y, Niedermann G, Lu Y, Xue J. Development of pharmacological immunoregulatory anti-cancer therapeutics: current mechanistic studies and clinical opportunities. Signal Transduct Target Ther 2024; 9:126. [PMID: 38773064 PMCID: PMC11109181 DOI: 10.1038/s41392-024-01826-z] [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: 10/11/2023] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 05/23/2024] Open
Abstract
Immunotherapy represented by anti-PD-(L)1 and anti-CTLA-4 inhibitors has revolutionized cancer treatment, but challenges related to resistance and toxicity still remain. Due to the advancement of immuno-oncology, an increasing number of novel immunoregulatory targets and mechanisms are being revealed, with relevant therapies promising to improve clinical immunotherapy in the foreseeable future. Therefore, comprehending the larger picture is important. In this review, we analyze and summarize the current landscape of preclinical and translational mechanistic research, drug development, and clinical trials that brought about next-generation pharmacological immunoregulatory anti-cancer agents and drug candidates beyond classical immune checkpoint inhibitors. Along with further clarification of cancer immunobiology and advances in antibody engineering, agents targeting additional inhibitory immune checkpoints, including LAG-3, TIM-3, TIGIT, CD47, and B7 family members are becoming an important part of cancer immunotherapy research and discovery, as are structurally and functionally optimized novel anti-PD-(L)1 and anti-CTLA-4 agents and agonists of co-stimulatory molecules of T cells. Exemplified by bispecific T cell engagers, newly emerging bi-specific and multi-specific antibodies targeting immunoregulatory molecules can provide considerable clinical benefits. Next-generation agents also include immune epigenetic drugs and cytokine-based therapeutics. Cell therapies, cancer vaccines, and oncolytic viruses are not covered in this review. This comprehensive review might aid in further development and the fastest possible clinical adoption of effective immuno-oncology modalities for the benefit of patients.
Collapse
Affiliation(s)
- Nanhao Yin
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xintong Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xuanwei Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Shaolong Xue
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, No. 20, Section 3, South Renmin Road, Chengdu, 610041, Sichuan, PR China
| | - Yu Cao
- Department of Emergency Medicine, Laboratory of Emergency Medicine, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
- Institute of Disaster Medicine & Institute of Emergency Medicine, Sichuan University, No. 17, Gaopeng Avenue, Chengdu, 610041, Sichuan, PR China
| | - Gabriele Niedermann
- Department of Radiation Oncology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site DKTK-Freiburg, Robert-Koch-Strasse 3, 79106, Freiburg, Germany.
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| |
Collapse
|
24
|
Wu LY, Park SH, Jakobsson H, Shackleton M, Möller A. Immune Regulation and Immune Therapy in Melanoma: Review with Emphasis on CD155 Signalling. Cancers (Basel) 2024; 16:1950. [PMID: 38893071 PMCID: PMC11171058 DOI: 10.3390/cancers16111950] [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: 03/28/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Melanoma is commonly diagnosed in a younger population than most other solid malignancies and, in Australia and most of the world, is the leading cause of skin-cancer-related death. Melanoma is a cancer type with high immunogenicity; thus, immunotherapies are used as first-line treatment for advanced melanoma patients. Although immunotherapies are working well, not all the patients are benefitting from them. A lack of a comprehensive understanding of immune regulation in the melanoma tumour microenvironment is a major challenge of patient stratification. Overexpression of CD155 has been reported as a key factor in melanoma immune regulation for the development of therapy resistance. A more thorough understanding of the actions of current immunotherapy strategies, their effects on immune cell subsets, and the roles that CD155 plays are essential for a rational design of novel targets of anti-cancer immunotherapies. In this review, we comprehensively discuss current anti-melanoma immunotherapy strategies and the immune response contribution of different cell lineages, including tumour endothelial cells, myeloid-derived suppressor cells, cytotoxic T cells, cancer-associated fibroblast, and nature killer cells. Finally, we explore the impact of CD155 and its receptors DNAM-1, TIGIT, and CD96 on immune cells, especially in the context of the melanoma tumour microenvironment and anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Li-Ying Wu
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Su-Ho Park
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haakan Jakobsson
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
| | - Mark Shackleton
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
- School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Andreas Möller
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
25
|
Dai T, Sun H, Liban T, Vicente-Suarez I, Zhang B, Song Y, Jiang Z, Yu J, Sheng J, Lv B. A novel anti-LAG-3/TIGIT bispecific antibody exhibits potent anti-tumor efficacy in mouse models as monotherapy or in combination with PD-1 antibody. Sci Rep 2024; 14:10661. [PMID: 38724599 PMCID: PMC11082181 DOI: 10.1038/s41598-024-61477-6] [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: 02/01/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
We report the generation of a novel anti-LAG-3/TIGIT bispecific IgG4 antibody, ZGGS15, and evaluated its anti-tumor efficacy in mouse models as monotherapy or in combination with a PD-1 antibody. ZGGS15 exhibited strong affinities for human LAG-3 and TIGIT, with KDs of 3.05 nM and 2.65 nM, respectively. ZGGS15 has EC50s of 0.69 nM and 1.87 nM for binding to human LAG-3 and TIGIT on CHO-K1 cells, respectively. ZGGS15 competitively inhibited the binding of LAG-3 to MHC-II (IC50 = 0.77 nM) and the binding of TIGIT to CD155 (IC50 = 0.24 nM). ZGGS15 does not induce ADCC, CDC, or obvious cytokine production. In vivo results showed that ZGGS15 had better anti-tumor inhibition than single anti-LAG-3 or anti-TIGIT agents and demonstrated a synergistic effect when combined with nivolumab, with a significantly higher tumor growth inhibition of 95.80% (p = 0.001). The tumor volume inhibition rate for ZGGS15 at 2 mg/kg was 69.70%, and for ZGGS15 at 5 mg/kg plus nivolumab at 1 mg/kg, it was 94.03% (p < 0.001). Our data reveal that ZGGS15 exhibits potent anti-tumor efficacy without eliciting ADCC or CDC or causing cytokine production, therefore having a safe profile.
Collapse
Affiliation(s)
- Tongcheng Dai
- Suzhou Zelgen Biopharmaceuticals Co., Ltd, Kunshan, China
| | - Hao Sun
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tyler Liban
- Gensun Biopharma Inc., Thousand Oaks, CA, USA
| | | | - Bin Zhang
- Suzhou Zelgen Biopharmaceuticals Co., Ltd, Kunshan, China
| | - Yongping Song
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhongxing Jiang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jifeng Yu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | | | - Binhua Lv
- Suzhou Zelgen Biopharmaceuticals Co., Ltd, Kunshan, China.
| |
Collapse
|
26
|
Li Y, Li B, Wang Q, Zhang X, Zhang Q, Zhou X, Shi R, Wu Y, Zhai W, Chen Z, Zhou X, Zhao W. Dual targeting of TIGIT and PD-1 with a novel small molecule for cancer immunotherapy. Biochem Pharmacol 2024; 223:116162. [PMID: 38527557 DOI: 10.1016/j.bcp.2024.116162] [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: 01/21/2024] [Revised: 03/13/2024] [Accepted: 03/21/2024] [Indexed: 03/27/2024]
Abstract
Immune checkpoint inhibitors have unveiled promising clinical prospects in cancer treatment. Nonetheless, their effectiveness remains restricted, marked by consistently low response rates and affecting only a subset of patients. The co-blockade of TIGIT with PD-1 has exhibited substantial anti-tumor effects. Notably, there is a dearth of reports on small-molecule inhibitors concurrently targeting both TIGIT and PD-1. In this study, we employed Microscale Thermophoresis (MST) to screen our laboratory's existing repository of small molecules. Our findings illuminated Gln(TrT) 's affinity for both TIGIT and PD-1, affirming its potential to effectively inhibit TIGIT/PVR and PD-1/PD-L1 pathways. In vitro co-culture experiments substantiated Gln(TrT)'s proficiency in restoring Jurkat T-cell functionality by blocking both TIGIT/PVR and PD-1/PD-L1 interactions. In the MC38 murine tumor model, Gln(TrT) emerges as a pivotal modulator, promoting the intratumoral infiltration and functional competence of CD8+ T cells. Furthermore, whether used as a monotherapy or in conjunction with radiotherapy, Gln(TrT) substantially impedes MC38 tumor progression, significantly extending the survival of murine subjects.
Collapse
Affiliation(s)
- Yang Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Beibei Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Qingchao Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiangrui Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Qiongqiong Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiuman Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Ranran Shi
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; International Joint Laboratory for Protein and Peptide Drugs of Henan Province, Zhengzhou University, Zhengzhou 450001, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; International Joint Laboratory for Protein and Peptide Drugs of Henan Province, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhen Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaowen Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Wenshan Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
27
|
Spagnolo CC, Pepe F, Ciappina G, Nucera F, Ruggeri P, Squeri A, Speranza D, Silvestris N, Malapelle U, Santarpia M. Circulating biomarkers as predictors of response to immune checkpoint inhibitors in NSCLC: Are we on the right path? Crit Rev Oncol Hematol 2024; 197:104332. [PMID: 38580184 DOI: 10.1016/j.critrevonc.2024.104332] [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: 02/14/2024] [Accepted: 03/25/2024] [Indexed: 04/07/2024] Open
Abstract
Immune checkpoints inhibitors (ICIs) have markedly improved the therapeutic management of advanced NSCLC and, more recently, they have demonstrated efficacy also in the early-stage disease. Despite better survival outcomes with ICIs compared to standard chemotherapy, a large proportion of patients can derive limited clinical benefit from these agents. So far, few predictive biomarkers, including the programmed death-ligand 1 (PD-L1), have been introduced in clinical practice. Therefore, there is an urgent need to identify novel biomarkers to select patients for immunotherapy, to improve efficacy and avoid unnecessary toxicity. A deeper understanding of the mechanisms involved in antitumor immunity and advances in the field of liquid biopsy have led to the identification of a wide range of circulating biomarkers that could potentially predict response to immunotherapy. Herein, we provide an updated overview of these circulating biomarkers, focusing on emerging data from clinical studies and describing modern technologies used for their detection.
Collapse
Affiliation(s)
- Calogera Claudia Spagnolo
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Via S. Pansini, Naples 80131, Italy
| | - Giuliana Ciappina
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Francesco Nucera
- Respiratory Medicine Unit, Department of Biomedical Sciences, Dentistry and Morphological and Functional Imaging (BIOMORF), University of Messina, Messina 98122, Italy
| | - Paolo Ruggeri
- Respiratory Medicine Unit, Department of Biomedical Sciences, Dentistry and Morphological and Functional Imaging (BIOMORF), University of Messina, Messina 98122, Italy
| | - Andrea Squeri
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Desirèe Speranza
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Via S. Pansini, Naples 80131, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy.
| |
Collapse
|
28
|
Zhong D, Shi Y, Ma W, Liang Y, Liu H, Qin Y, Zhang L, Yang Q, Huang X, Lu Y, Shang J. Single-cell profiling reveals the metastasis-associated immune signature of hepatocellular carcinoma. Immun Inflamm Dis 2024; 12:e1264. [PMID: 38780041 PMCID: PMC11112628 DOI: 10.1002/iid3.1264] [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: 08/03/2023] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
AIM Metastasis is the leading cause of mortality in hepatocellular carcinoma (HCC). The metastasis-associated immune signature in HCC is worth exploring. METHODS Bioinformatic analysis was conducted based on the single-cell transcriptome data derived from HCC patients in different stages. Cellular composition, pseudotime state transition, and cell-cell interaction were further analyzed and verified. RESULTS Generally, HCC with metastasis exhibited suppressive immune microenvironment, while HCC without metastasis exhibited active immune microenvironment. Concretely, effector regulatory T cells (eTregs) were found to be enriched in HCC with metastasis. PHLDA1 was identified as one of exhaustion-specific genes and verified to be associated with worse prognosis in HCC patients. Moreover, A novel cluster of CCR7+ dendritic cells (DCs) was identified with high expression of maturation and migration marker genes. Pseudotime analysis showed that inhibition of differentiation occurred in CCR7+ DCs rather than cDC1 in HCC with metastasis. Furthermore, interaction analysis showed that the reduction of CCR7+ DCs lead to impaired CCR7/CCL19 interaction in HCC with metastasis. CONCLUSIONS HCC with metastasis exhibited upregulation of exhaustion-specific genes of eTregs and inhibition of CCL signal of a novel DC cluster, which added new dimensions to the immune landscape and provided new immune therapeutic targets in advanced HCC.
Collapse
Affiliation(s)
- Deyuan Zhong
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Ying Shi
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacau SARChina
| | - Yuxin Liang
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Hanjie Liu
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| | - Yingying Qin
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| | - Lu Zhang
- Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| | - Qinyan Yang
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| | - Xiaolun Huang
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Yuanjun Lu
- Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| | - Jin Shang
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
- Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer CenterAffiliated Cancer Hospital of University of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
29
|
Zhao X, Cheng Y, Guo C, Nie L, Zhang Q, Zhang M, Sun K, Wang G. Efficacy of cross-line anti-programmed death 1/programmed cell death-ligand 1 antibody in the treatment of advanced nonsmall cell lung cancer: A retrospective study. Health Sci Rep 2024; 7:e2114. [PMID: 38736477 PMCID: PMC11082083 DOI: 10.1002/hsr2.2114] [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: 10/27/2023] [Revised: 04/11/2024] [Accepted: 04/20/2024] [Indexed: 05/14/2024] Open
Abstract
Background and Aims Immune checkpoint inhibitors (ICIs) across multiple treatment lines have not yet been evaluated comprehensively. The purpose of this research was to investigate whether or not continuous cross-line ICIs therapy is effective in treating non-small cell lung cancer (NSCLC). Methods We conducted a retrospective investigation into the medical histories of 47 patients diagnosed with advanced NSCLC and treated with ICIs at the Peking University First Hospital between January 2018 and June 2022. Results Due to the progression of their disease, 14 patients were given the same ICIs, 5 patients were given different ICIs, and 6 patients discontinued taking ICIs altogether. The objective response rates were 7.140% in the ICIs cross-line treatment group, 0% in the replacement of ICIs treatment group, and 0% in the discontinuation of ICIs treatment group. The disease control rates were 64.260% in the ICIs cross-line treatment group, 60% in the replacement of ICIs treatment group, and 0% in the discontinuation of ICIs treatment group. The average overall survival durations of the three groups were 24.020 (95% confidence interval [CI]: 17.061-30.979), 31.643 (95% CI: 23.513-39.774), and 7.997 (95% CI: 3.746-12.247) months, respectively (p = 0.003). The median second progression-free survival (PFS2) durations of the three groups were 4.570 (95% CI: 3.276-5.864), 3.530 (95% CI: 0.674-6.386), and 1.570 (95% CI: 0-4.091) months, respectively (p = 0.091). Conclusions Cross-line ICIs cannot improve the prognosis and PFS2 of patients with NSCLC, but compared to discontinuing ICIs, OS may be prolonged. A few patients may benefit from prolonged ICIs therapy.
Collapse
Affiliation(s)
- Xiang Zhao
- Department of Respiratory and Critical Care MedicinePeking University First HospitalBeijingChina
| | - Yuan Cheng
- Department of Respiratory and Critical Care MedicinePeking University First HospitalBeijingChina
| | - Cuiyan Guo
- Department of Respiratory and Critical Care MedicinePeking University First HospitalBeijingChina
| | - Ligong Nie
- Department of Respiratory and Critical Care MedicinePeking University First HospitalBeijingChina
| | - Qi Zhang
- Department of Respiratory and Critical Care MedicinePeking University First HospitalBeijingChina
| | - Meng Zhang
- Department of Respiratory and Critical Care MedicinePeking University First HospitalBeijingChina
| | - Kunyan Sun
- Department of Respiratory and Critical Care MedicinePeking University First HospitalBeijingChina
| | - Guangfa Wang
- Department of Respiratory and Critical Care MedicinePeking University First HospitalBeijingChina
| |
Collapse
|
30
|
Garralda E, Oh DY, Italiano A, Bedard PL, Delord JP, Calvo E, LoRusso P, Wainberg Z, Cervantes A, Rodriguez-Vida A, Shemesh CS, Sane R, Mendus D, Ding H, Hendricks R, Meng R, Cho BC, Kim TW, Wu B. Pharmacokinetics (PK) of Tiragolumab in First-in-Human Study in Patients with Mixed Solid Tumors (GO30103). J Clin Pharmacol 2024; 64:544-554. [PMID: 38105505 DOI: 10.1002/jcph.2397] [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: 09/21/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Tiragolumab is a first-in-class, fully human IgG1/kappa anti-TIGIT monoclonal antibody that blocks the binding of TIGIT to CD155 (the poliovirus receptor). We summarize the pharmacokinetics (PK) data from the phase 1a/1b GO30103 study of Q3W (every 3 weeks) sequential dosing of tiragolumab (2, 8, 30, 100, 400, 600, or 1200 mg) followed by atezolizumab (1200 mg), Q4W (every 4 weeks) sequential dosing (tiragolumab 840 mg followed by atezolizumab 1680 mg), and Q4W co-infusion (tiragolumab 840 mg plus atezolizumab 1680 mg). Serum samples were collected at multiple time points following tiragolumab and atezolizumab intravenous infusion in patients with solid tumors for PK and immunogenicity assessment. The serum PK profile of tiragolumab appeared to be biphasic, with a rapid distribution phase followed by a slower elimination phase when administered alone or in combination with atezolizumab. In phase 1a, across doses of tiragolumab ranging from 2 to 1200 mg (cycle 1), the geometric mean (GM), coefficient of variation (CV%), serum tiragolumab Cmax ranged from 0.682 to 270 µg/mL (18.6% to 36.5%) and Cmin ranged from 0.0125 to 75.3 µg/mL (0.0% to 24.2%). The GM systemic exposure (area under the plasma drug concentration-time curve, AUC0-21) ranged from 310 to 2670 µg day/mL (20.5% to 27.0%); interindividual variability in AUC0-21 ranged from 20.5% to 43.9%. Tiragolumab exposure increased in an approximately dose-proportional manner when administered alone or with atezolizumab at doses ≥100 mg. Postbaseline, 4/207 patients (1.9%) were positive for treatment-emergent antidrug antibodies (ADA) against tiragolumab, each at a single time point. Tiragolumab combined with atezolizumab demonstrated desirable PK properties, with no drug-drug interactions or immunogenicity liability. There were no meaningful differences in tiragolumab or atezolizumab exposure between the Q4W co-infusion and sequential dosing cohorts. ClinicalTrials.gov: NCT02794571 (date of registration June 6, 2016).
Collapse
Affiliation(s)
- Elena Garralda
- Early Drug Development Unit, Vall d'Hebron, Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Do Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Antoine Italiano
- Early Phase Trials Unit, Institut Bergonié, Bordeaux, and Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - Philippe L Bedard
- Princess Margaret Cancer Centre, University Health Network, Division of Medical Oncology & Hematology, University of Toronto, Toronto, Ontario, Canada
| | - Jean-Pierre Delord
- Medical Oncology Department, IUCT Oncopole, Institut Claudius Regaud, Toulouse, France
| | - Emiliano Calvo
- Centro Integral Oncológico Clara Campal, START Madrid - Clara Campal Comprehensive Cancer Center (CIOCC), Madrid, Spain
| | | | - Zev Wainberg
- University of California Los Angeles, Los Angeles, CA, USA
| | - Andres Cervantes
- Department of Medical Oncology, Hospital Clínico, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Colby S Shemesh
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| | - Rucha Sane
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| | - Diana Mendus
- Product Development Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Hao Ding
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| | - Robert Hendricks
- Department of BioAnalytical Sciences, Genentech, Inc., South San Francisco, CA, USA
| | - Ray Meng
- Product Development Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Byoung Chul Cho
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae Won Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Benjamin Wu
- Early Drug Development Unit, Vall d'Hebron, Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
31
|
Bauer M, Schöbel CM, Wickenhauser C, Seliger B, Jasinski-Bergner S. Deciphering the role of alternative splicing in neoplastic diseases for immune-oncological therapies. Front Immunol 2024; 15:1386993. [PMID: 38736877 PMCID: PMC11082354 DOI: 10.3389/fimmu.2024.1386993] [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: 02/16/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024] Open
Abstract
Alternative splicing (AS) is an important molecular biological mechanism regulated by complex mechanisms involving a plethora of cis and trans-acting elements. Furthermore, AS is tissue specific and altered in various pathologies, including infectious, inflammatory, and neoplastic diseases. Recently developed immuno-oncological therapies include monoclonal antibodies (mAbs) and chimeric antigen receptor (CAR) T cells targeting, among others, immune checkpoint (ICP) molecules. Despite therapeutic successes have been demonstrated, only a limited number of patients showed long-term benefit from these therapies with tumor entity-related differential response rates were observed. Interestingly, splice variants of common immunotherapeutic targets generated by AS are able to completely escape and/or reduce the efficacy of mAb- and/or CAR-based tumor immunotherapies. Therefore, the analyses of splicing patterns of targeted molecules in tumor specimens prior to therapy might help correct stratification, thereby increasing therapy success by antibody panel selection and antibody dosages. In addition, the expression of certain splicing factors has been linked with the patients' outcome, thereby highlighting their putative prognostic potential. Outstanding questions are addressed to translate the findings into clinical application. This review article provides an overview of the role of AS in (tumor) diseases, its molecular mechanisms, clinical relevance, and therapy response.
Collapse
Affiliation(s)
- Marcus Bauer
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Chiara-Maria Schöbel
- Institute for Translational Immunology, Brandenburg Medical School (MHB), Theodor Fontane, Brandenburg an der Havel, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Barbara Seliger
- Institute for Translational Immunology, Brandenburg Medical School (MHB), Theodor Fontane, Brandenburg an der Havel, Germany
- Department of Good Manufacturing Practice (GMP) Development & Advanced Therapy Medicinal Products (ATMP) Design, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Institute for Medical Immunology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Simon Jasinski-Bergner
- Institute for Translational Immunology, Brandenburg Medical School (MHB), Theodor Fontane, Brandenburg an der Havel, Germany
| |
Collapse
|
32
|
Wu JW, Liu Y, Dai XJ, Liu HM, Zheng YC, Liu HM. CD155 as an emerging target in tumor immunotherapy. Int Immunopharmacol 2024; 131:111896. [PMID: 38518596 DOI: 10.1016/j.intimp.2024.111896] [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: 02/19/2024] [Revised: 03/08/2024] [Accepted: 03/16/2024] [Indexed: 03/24/2024]
Abstract
CD155 is an immunoglobulin-like protein overexpressed in almost all the tumor cells, which not only promotes proliferation, adhesion, invasion, and migration of tumor cells, but also regulates immune responses by interacting with TIGIT, CD226 or CD96 receptors expressed on several immune cells, thereby modulating the functionality of these cellular subsets. As a novel immune checkpoint, the inhibition of CD155/TIGIT, either as a standalone treatment or in conjunction with other immune checkpoint inhibitors, has demonstrated efficacy in managing advanced solid malignancies. In this review, we summarize the intricate relationship between on tumor surface CD155 and its receptors, with further discussion on how they regulate the occurrence of tumor immune escape. In addition, novel therapeutic strategies and clinical trials targeting CD155 and its receptors are summarized, providing a strong rationale and way forward for the development of next-generation immunotherapies.
Collapse
Affiliation(s)
- Jiang-Wan Wu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ying Liu
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Xing-Jie Dai
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Hui-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
33
|
Firestone RS, McAvoy D, Shekarkhand T, Serrano E, Hamadeh I, Wang A, Zhu M, Qin WG, Patel D, Tan CR, Hultcrantz M, Mailankody S, Hassoun H, Shah US, Korde N, Maclachlan KH, Landau HJ, Scordo M, Shah GL, Lahoud OB, Giralt S, Murata K, Hosszu KK, Chung DJ, Lesokhin AM, Usmani SZ. CD8 effector T cells enhance teclistamab response in BCMA-exposed and -naïve multiple myeloma. Blood Adv 2024; 8:1600-1611. [PMID: 37878808 PMCID: PMC10987849 DOI: 10.1182/bloodadvances.2023011225] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/27/2023] Open
Abstract
ABSTRACT Teclistamab, a B-cell maturation antigen (BCMA)- and CD3-targeting bispecific antibody, is an effective novel treatment for relapsed/refractory multiple myeloma (R/RMM), but efficacy in patients exposed to BCMA-directed therapies and mechanisms of resistance have yet to be fully delineated. We conducted a real-world retrospective study of commercial teclistamab, capturing both clinical outcomes and immune correlates of treatment response in a cohort of patients (n = 52) with advanced R/RMM. Teclistamab was highly effective with an overall response rate (ORR) of 64%, including an ORR of 50% for patients with prior anti-BCMA therapy. Pretreatment plasma cell BCMA expression levels had no bearing on response. However, comprehensive pretreatment immune profiling identified that effector CD8+ T-cell populations were associated with response to therapy and a regulatory T-cell population associated with nonresponse, indicating a contribution of immune status in outcomes with potential utility as a biomarker signature to guide patient management.
Collapse
Affiliation(s)
- Ross S. Firestone
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Devin McAvoy
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Tala Shekarkhand
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Edith Serrano
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Issam Hamadeh
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alice Wang
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Menglei Zhu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wei Ge Qin
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Dhwani Patel
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Carlyn R. Tan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Hani Hassoun
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Urvi S. Shah
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Neha Korde
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kylee H. Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Heather J. Landau
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael Scordo
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gunjan L. Shah
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Oscar B. Lahoud
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sergio Giralt
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kazunori Murata
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kinga K. Hosszu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David J. Chung
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander M. Lesokhin
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Saad Z. Usmani
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
34
|
Jia F, Sun S, Li J, Wang W, Huang H, Hu X, Pan S, Chen W, Shen L, Yao Y, Zheng S, Chen H, Xia W, Yuan H, Zhou J, Yu X, Zhang T, Zhang B, Huang J, Ni C. Neoadjuvant chemotherapy-induced remodeling of human hormonal receptor-positive breast cancer revealed by single-cell RNA sequencing. Cancer Lett 2024; 585:216656. [PMID: 38266804 DOI: 10.1016/j.canlet.2024.216656] [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: 09/19/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 01/26/2024]
Abstract
Hormone receptor-positive breast cancer (HR+ BC) is known to be relatively insensitive to chemotherapy, and since chemotherapy has remained the major neoadjuvant therapy for HR+ BC, the undetermined mechanism of chemoresistance and how chemotherapy reshapes the immune microenvironment need to be explored by high-throughput technology. By using single-cell RNA sequencing and multiplexed immunofluorescence staining analysis of HR+ BC samples (paired pre- and post-neoadjuvant chemotherapy (NAC)), the levels of previously unrecognized immune cell subsets, including CD8+ T cells with pronounced expression of T-cell development (LMNA) and cytotoxicity (FGFBP2) markers, CD4+ T cells characterized by proliferation marker (ATP1B3) expression and macrophages characterized by CD52 expression, were found to be increased post-NAC, which were predictive of chemosensitivity and their antitumor function was also validated with in vitro experiments. In terms of immune checkpoint expression of CD8+ T cells, we found their changes were inconsistent post-NAC, that LAG3, VSIR were decreased, and PDCD1, HAVCR2, CTLA4, KLRC1 and BTLA were increased. In addition, we have identified novel genomic and transcriptional patterns of chemoresistant cancer cells, both innate and acquired, and have confirmed their prognostic value with TCGA cohorts. By shedding light on the ecosystem of HR+ BC reshaped by chemotherapy, our results uncover valuable candidates for predicting chemosensitivity and overcoming chemoresistance in HR+ BC.
Collapse
Affiliation(s)
- Fang Jia
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Shanshan Sun
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China
| | - Jiaxin Li
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Wenwen Wang
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huanhuan Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Xiaoxiao Hu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng Pan
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wuzhen Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lesang Shen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Yao
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siwei Zheng
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hailong Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjie Xia
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Hongjun Yuan
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jun Zhou
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuyan Yu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Zhang
- Department of Radiotherapy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing Zhang
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Jian Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| | - Chao Ni
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China.
| |
Collapse
|
35
|
Lu C, Tan Y. Promising immunotherapy targets: TIM3, LAG3, and TIGIT joined the party. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200773. [PMID: 38596295 PMCID: PMC10905042 DOI: 10.1016/j.omton.2024.200773] [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] [Indexed: 04/11/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have shown great promise as immunotherapy for restoring T cell function and reactivating anti-tumor immunity. The US Food and Drug Administration (FDA) approved the first immune checkpoint inhibitor, ipilimumab, in 2011 for advanced melanoma patients, leading to significant improvements in survival rates. Subsequently, other immune checkpoint-targeting antibodies were tested. Currently, seven ICIs, namely ipilimumab (anti-cytotoxic T lymphocyte-associated protein 4 [CTLA4]), pembrolizumab, nivolumab (anti-programmed cell death protein 1 [PD-1]), atezolizumab, avelumab, durvalumab, and cemiplimab (anti-PD-L1), have been approved for various cancer types. However, the efficacy of antibodies targeting CTLA4 or PD-1/programmed death-ligand 1 (PD-L1) remains suboptimal. Consequently, ongoing studies are evaluating the next generation of ICIs, such as lymphocyte activation gene-3 (LAG3), T cell immunoglobulin and mucin-domain containing 3 (TIM3), and T cell immunoglobulin and ITIM domain (TIGIT). Our review provides a summary of clinical trials evaluating these novel immune checkpoints in cancer treatment.
Collapse
Affiliation(s)
- Chenyu Lu
- Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Yuanyan Tan
- Institute of Advanced Biotechnology and School of Medicine, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, Shenzhen 518061, Guangdong, China
| |
Collapse
|
36
|
Yang Y, Xin D, Guan L, Luo X, Wu H, Chu J, Xing J, Liu C, Wang F. Dual immunotherapy in advanced or metastatic non-small cell lung cancer: A network meta-analysis. Heliyon 2024; 10:e27576. [PMID: 38463838 PMCID: PMC10923855 DOI: 10.1016/j.heliyon.2024.e27576] [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: 11/24/2023] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 03/12/2024] Open
Abstract
Objectives Recently, there has been extensive research on dual immunotherapy for advanced or metastatic non-small cell lung cancer (NSCLC), yet a comprehensive evaluation is lacking. This study aimed to rank the available treatment options and assess the efficacy and safety of dual immunotherapy regimens through the implementation of a Bayesian network meta-analysis (NMA). Materials and methods A thorough search was conducted to recognize eligible randomized controlled trials (RCTs) on March 20, 2023. Overall survival (OS), progression-free survival (PFS), treatment-related adverse events (TRAEs) and grade ≥3 TRAEs were evaluated to identify the efficacy and safety of dual immunotherapy regimens. The surface under the cumulative ranking curve (SUCRA) and P score were employed to rank the treatments. Results Eleven clinical trials involving six different regimens were included in this study. The combination of anti-programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) antibodies with anti-T-cell immunoglobulin and ITIM domain (TIGIT) antibodies emerged as the most promising regimen for improving OS and PFS, followed by anti-PD-1/PD-L1 + anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) + chemotherapy treatment and anti-PD-1/PD-L1 + anti-CTLA-4 treatment. The forest plots demonstrated that these three regimens were all superior to chemotherapy. The above results were observed in both unselected treatment line and first-line settings. The least likely to be associated with TRAEs and grade ≥3 TRAEs were respectively anti-CTLA-4 treatment and anti-PD-1/PD-L1 + anti-TIGIT treatment, with anti-PD-1/PD-L1 + anti-CTLA-4 + chemotherapy treatment to be the worst. Conclusions This NMA validated the promising efficacy and safety of dual immunotherapy in advanced or metastatic NSCLC. Among them, anti-PD-1/PD-L1 + anti-TIGIT regimen emerges as a highly potential therapeutic approach. Ongoing research efforts should focus on improving treatment regimens, identifying biomarkers, and managing TRAEs to optimize the patient benefits of dual immunotherapy.
Collapse
Affiliation(s)
- Yuanyuan Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Dao Xin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Lulu Guan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xi Luo
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Han Wu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jingwen Chu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jianxiang Xing
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Chengjiang Liu
- Department of General Medicine, Affiliated Anqing First People's Hospital of Anhui Medical University, Anqing 246000, China
| | - Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
37
|
La'ah AS, Chiou SH. Cutting-Edge Therapies for Lung Cancer. Cells 2024; 13:436. [PMID: 38474400 DOI: 10.3390/cells13050436] [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: 01/22/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Lung cancer remains a formidable global health challenge that necessitates inventive strategies to improve its therapeutic outcomes. The conventional treatments, including surgery, chemotherapy, and radiation, have demonstrated limitations in achieving sustained responses. Therefore, exploring novel approaches encompasses a range of interventions that show promise in enhancing the outcomes for patients with advanced or refractory cases of lung cancer. These groundbreaking interventions can potentially overcome cancer resistance and offer personalized solutions. Despite the rapid evolution of emerging lung cancer therapies, persistent challenges such as resistance, toxicity, and patient selection underscore the need for continued development. Consequently, the landscape of lung cancer therapy is transforming with the introduction of precision medicine, immunotherapy, and innovative therapeutic modalities. Additionally, a multifaceted approach involving combination therapies integrating targeted agents, immunotherapies, or traditional cytotoxic treatments addresses the heterogeneity of lung cancer while minimizing its adverse effects. This review provides a brief overview of the latest emerging therapies that are reshaping the landscape of lung cancer treatment. As these novel treatments progress through clinical trials are integrated into standard care, the potential for more effective, targeted, and personalized lung cancer therapies comes into focus, instilling renewed hope for patients facing challenging diagnoses.
Collapse
Affiliation(s)
- Anita Silas La'ah
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
38
|
Gomez-Randulfe I, Leporati R, Gupta B, Liu S, Califano R. Recent advances and future strategies in first-line treatment of ES-SCLC. Eur J Cancer 2024; 200:113581. [PMID: 38301317 DOI: 10.1016/j.ejca.2024.113581] [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: 12/05/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
Recent advancements in treating extensive-stage small-cell lung cancer (ES-SCLC) have been significantly marked by incorporating immune checkpoint inhibitors (ICIs) into platinum-based chemotherapy, leading to modest yet notable improvements in patient outcomes, which become more evident with longer follow-up. However, critical challenges persist, such as identifying effective biomarkers for accurate patient selection or finding more effective drugs. This review delves into the current and evolving treatment landscape for ES-SCLC, focusing on the most promising therapeutic strategies under investigation. We discuss the latest developments in the use of newer ICIs, antiangiogenic agents, PARP inhibitors (PARPi), lurbinectedin, and anti-DLL3 agents, offering insights into potential future directions in the management of this aggressive cancer.
Collapse
Affiliation(s)
- Igor Gomez-Randulfe
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Rita Leporati
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Brinda Gupta
- Division of Hematology and Oncology, Department of Medicine, Georgetown University, Washington, DC, USA
| | - Stephen Liu
- Division of Hematology and Oncology, Department of Medicine, Georgetown University, Washington, DC, USA
| | - Raffaele Califano
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, The University of Manchester, Manchester, UK.
| |
Collapse
|
39
|
Libert D, Zhao S, Younes S, Mosquera AP, Bharadwaj S, Ferreira C, Natkunam Y. TIGIT is Frequently Expressed in the Tumor Microenvironment of Select Lymphomas: Implications for Targeted Therapy. Am J Surg Pathol 2024; 48:337-352. [PMID: 38148663 PMCID: PMC10876169 DOI: 10.1097/pas.0000000000002168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Immune checkpoint inhibitors against Programmed Cell Death Protein 1/Programmed Cell (PD-1/PD-L1) and CTLA-4/B7 axes have had limited success in hematologic malignancies, requiring the need to explore alternative targets such as T-cell immunoreceptor with Ig and ITIM domains (TIGIT)/CD155 to improve durable clinical responses. We undertook this study to investigate the expression profile of TIGIT such that the potential efficacy of TIGIT blockade could be mapped among lymphoma subtypes. We validated an immunohistochemical assay for TIGIT and evaluated its expression in lymphoma and tumor microenvironment (TME) cells in 661 lymphoma/leukemia biopsies. Multiplex immunofluorescence was used for correlation with normal TME cell subsets. Tumor or TME TIGIT-positivity was defined as moderate to strong membrane staining in at least 10% of tumor or TME cells, respectively. TME TIGIT expression was correlated with overall survival and progression-free survival and comparison with PD-L1 expression. In most cases, lymphoma cells were TIGIT-negative except for angioimmunoblastic and peripheral T-cell lymphomas, which showed 91% and 47% positivity, respectively. A high proportion of small B-cell lymphoma and anaplastic large cell lymphoma cases had TIGIT-positive TME cells. Chronic lymphocytic leukemia/small lymphocytic lymphoma patients with TIGIT-negative TME cells showed significantly shorter overall survival ( P =0.04). No other statistically significant differences were found. When TIGIT was expressed in TME cells, there were a comparable number of TIGIT-positive only and dual TIGIT/PD-L1 positive cases except for more TIGIT-positive only cases in CLL/SLL. TIGIT expression shows distinctive profiles among lymphoma subtypes. Chronic lymphocytic leukemia/small lymphocytic lymphoma and anaplastic large cell lymphoma demonstrated high TME TIGIT expression compared with PD-L1, with a high proportion of dual TIGIT and PD-L1-positivity. Our results are likely to contribute to the design and correlative study of therapeutic response in clinical trials targeting TIGIT alone or in combination with PD1/PDL1.
Collapse
|
40
|
Lin X, Xie M, Yao J, Ma X, Qin L, Zhang X, Song J, Bao X, Zhang X, Zhang Y, Liu Y, Han W, Liang Y, Jing Y, Xue X. Immune-related adverse events in non-small cell lung cancer: Occurrence, mechanisms and therapeutic strategies. Clin Transl Med 2024; 14:e1613. [PMID: 38451000 PMCID: PMC10918746 DOI: 10.1002/ctm2.1613] [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: 10/17/2023] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 03/08/2024] Open
Abstract
The emergence of immune checkpoint inhibitors (ICIs) has heralded a transformative era in the therapeutic landscape of non-small cell lung cancer (NSCLC). While ICIs have demonstrated clinical efficacy in a portion of patients with NSCLC, these treatments concurrently precipitate a spectrum of immune-related adverse events (irAEs), encompassing mild to severe manifestations, collectively posing a risk of significant organ damage. Consequently, there exists an imperative to augment our comprehension of the pathophysiological underpinnings of irAEs and to formulate more efficacious preventive and ameliorative strategies. In this comprehensive review, we delineate the clinical presentation of organ-specific irAEs in patients with NSCLC and provide an in-depth analysis of recent advancements in understanding the mechanisms driving ICI-induced toxicity. Furthermore, we discuss potential strategies and targets for ameliorating these irAEs. Ultimately, this review aims to furnish valuable insights to guide further research endeavours in the context of irAEs in NSCLC patients.
Collapse
Affiliation(s)
- Xuwen Lin
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Mei Xie
- Department of Respiratory and Critical CareChinese PLA General HospitalBeijingChina
| | - Jie Yao
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Xidong Ma
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Lin Qin
- Department of Endoscopic Diagnosis and TreatmentTuberculosis and Thoracic Tumor InstituteBeijing Chest HospitalCapital Medical UniversityBeijingChina
| | - Xu‐Mei Zhang
- Department of PathologyAffiliated Hospital of Weifang Medical UniversityWeifangShandongChina
| | - Jialin Song
- Department of Respiratory and Critical CareShandong Second Medical UniversityShandongChina
| | - Xinyu Bao
- Department of Respiratory and Critical CareShandong Second Medical UniversityShandongChina
| | - Xin Zhang
- Department of Respiratory and Critical CareShandong Second Medical UniversityShandongChina
| | - Yinguang Zhang
- Department of Thoracic SurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yiming Liu
- Department of Thoracic SurgeryChinese PLA General HospitalBeijingChina
| | - Wenya Han
- Department of Respiratory and Critical CareTaihe HospitalHubei University of MedicineShiyanChina
| | - Yiran Liang
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Ying Jing
- Center for Intelligent MedicineGreater Bay Area Institute of Precision Medicine (Guangzhou)School of Life SciencesFudan UniversityGuangzhouGuangdongChina
| | - Xinying Xue
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
- Department of Respiratory and Critical CareShandong Second Medical UniversityShandongChina
| |
Collapse
|
41
|
Sahu P, Mitra A, Ganguly A. Targeting KRAS and SHP2 signaling pathways for immunomodulation and improving treatment outcomes in solid tumors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 386:167-222. [PMID: 38782499 DOI: 10.1016/bs.ircmb.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Historically, KRAS has been considered 'undruggable' inspite of being one of the most frequently altered oncogenic proteins in solid tumors, primarily due to the paucity of pharmacologically 'druggable' pockets within the mutant isoforms. However, pioneering developments in drug design capable of targeting the mutant KRAS isoforms especially KRASG12C-mutant cancers, have opened the doors for emergence of combination therapies comprising of a plethora of inhibitors targeting different signaling pathways. SHP2 signaling pathway, primarily known for activation of intracellular signaling pathways such as KRAS has come up as a potential target for such combination therapies as it emerged to be the signaling protein connecting KRAS and the immune signaling pathways and providing the link for understanding the overlapping regions of RAS/ERK/MAPK signaling cascade. Thus, SHP2 inhibitors having potent tumoricidal activity as well as role in immunomodulation have generated keen interest in researchers to explore its potential as combination therapy in KRAS mutant solid tumors. However, the excitement with these combination therapies need to overcome challenges thrown up by drug resistance and enhanced toxicity. In this review, we will discuss KRAS and SHP2 signaling pathways and their roles in immunomodulation and regulation of tumor microenvironment and also analyze the positive effects and drawbacks of the different combination therapies targeted at these signaling pathways along with their present and future potential to treat solid tumors.
Collapse
Affiliation(s)
- Priyanka Sahu
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, United States
| | - Ankita Mitra
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, United States
| | - Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar, Jharkhand, India.
| |
Collapse
|
42
|
Wienke J, Visser LL, Kholosy WM, Keller KM, Barisa M, Poon E, Munnings-Tomes S, Himsworth C, Calton E, Rodriguez A, Bernardi R, van den Ham F, van Hooff SR, Matser YAH, Tas ML, Langenberg KPS, Lijnzaad P, Borst AL, Zappa E, Bergsma FJ, Strijker JGM, Verhoeven BM, Mei S, Kramdi A, Restuadi R, Sanchez-Bernabeu A, Cornel AM, Holstege FCP, Gray JC, Tytgat GAM, Scheijde-Vermeulen MA, Wijnen MHWA, Dierselhuis MP, Straathof K, Behjati S, Wu W, Heck AJR, Koster J, Nierkens S, Janoueix-Lerosey I, de Krijger RR, Baryawno N, Chesler L, Anderson J, Caron HN, Margaritis T, van Noesel MM, Molenaar JJ. Integrative analysis of neuroblastoma by single-cell RNA sequencing identifies the NECTIN2-TIGIT axis as a target for immunotherapy. Cancer Cell 2024; 42:283-300.e8. [PMID: 38181797 PMCID: PMC10864003 DOI: 10.1016/j.ccell.2023.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 11/10/2023] [Accepted: 12/11/2023] [Indexed: 01/07/2024]
Abstract
Pediatric patients with high-risk neuroblastoma have poor survival rates and urgently need more effective treatment options with less side effects. Since novel and improved immunotherapies may fill this need, we dissect the immunoregulatory interactions in neuroblastoma by single-cell RNA-sequencing of 24 tumors (10 pre- and 14 post-chemotherapy, including 5 pairs) to identify strategies for optimizing immunotherapy efficacy. Neuroblastomas are infiltrated by natural killer (NK), T and B cells, and immunosuppressive myeloid populations. NK cells show reduced cytotoxicity and T cells have a dysfunctional profile. Interaction analysis reveals a vast immunoregulatory network and identifies NECTIN2-TIGIT as a crucial immune checkpoint. Combined blockade of TIGIT and PD-L1 significantly reduces neuroblastoma growth, with complete responses (CR) in vivo. Moreover, addition of TIGIT+PD-L1 blockade to standard relapse treatment in a chemotherapy-resistant Th-ALKF1174L/MYCN 129/SvJ syngeneic model induces CR. In conclusion, our integrative analysis provides promising targets and a rationale for immunotherapeutic combination strategies.
Collapse
Affiliation(s)
- Judith Wienke
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| | - Lindy L Visser
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Waleed M Kholosy
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Kaylee M Keller
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Marta Barisa
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Evon Poon
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Sophie Munnings-Tomes
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Courtney Himsworth
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Elizabeth Calton
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | | | - Ronald Bernardi
- Genentech, A Member of the Roche Group, South San Francisco, CA, USA
| | - Femke van den Ham
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Yvette A H Matser
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Michelle L Tas
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Philip Lijnzaad
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Anne L Borst
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Elisa Zappa
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | | | - Bronte M Verhoeven
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Shenglin Mei
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Amira Kramdi
- Institut Curie, Inserm U830, PSL Research University, Diversity and Plasticity of Childhood Tumors Lab, Paris, France; SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Restuadi Restuadi
- Infection, Immunity and Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK; NIHR Biomedical Research Centre, Great Ormond Street Hospital, London, UK
| | - Alvaro Sanchez-Bernabeu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Netherlands Proteomics Centre, Utrecht University, Utrecht, the Netherlands
| | - Annelisa M Cornel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Juliet C Gray
- Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | | | | | - Marc H W A Wijnen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Karin Straathof
- University College London (UCL) Great Ormond Street Institute of Child Health, London, UK; UCL Cancer Institute, London, UK
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Netherlands Proteomics Centre, Utrecht University, Utrecht, the Netherlands; Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Netherlands Proteomics Centre, Utrecht University, Utrecht, the Netherlands
| | - Jan Koster
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, the Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Isabelle Janoueix-Lerosey
- Institut Curie, Inserm U830, PSL Research University, Diversity and Plasticity of Childhood Tumors Lab, Paris, France; SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Louis Chesler
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, England, UK
| | | | | | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Division Imaging & Cancer, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
43
|
Hoffman-Censits J, Grivas P, Powles T, Hawley J, Tyroller K, Seeberger S, Guenther S, Jacob N, Mehr KT, Hahn NM. The JAVELIN Bladder Medley trial: avelumab-based combinations as first-line maintenance in advanced urothelial carcinoma. Future Oncol 2024; 20:179-190. [PMID: 37671748 DOI: 10.2217/fon-2023-0492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023] Open
Abstract
Results from JAVELIN Bladder 100 established avelumab (anti-PD-L1) first-line maintenance as the standard-of-care treatment for patients with advanced urothelial carcinoma (UC) that has not progressed with first-line platinum-based chemotherapy. We describe the design of JAVELIN Bladder Medley (NCT05327530), an ongoing phase II, multicenter, randomized, open-label, parallel-arm, umbrella trial. Overall, 252 patients with advanced UC who are progression-free following first-line platinum-based chemotherapy will be randomized 1:2:2:2 to receive maintenance therapy with avelumab alone (control group) or combined with sacituzumab govitecan (anti-Trop-2/topoisomerase inhibitor conjugate), M6223 (anti-TIGIT) or NKTR-255 (recombinant human IL-15). Primary end points are progression-free survival per investigator and safety/tolerability of the combination regimens. Secondary end points include overall survival, objective response and duration of response per investigator, and pharmacokinetics.
Collapse
Affiliation(s)
- Jean Hoffman-Censits
- Departments of Medical Oncology & Urology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Petros Grivas
- University of Washington, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Thomas Powles
- Department of Genitourinary Oncology, Barts Cancer Institute, Experimental Cancer Medicine Centre, Queen Mary University of London, St Bartholomew's Hospital, London, UK
| | - Jessica Hawley
- University of Washington, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Karin Tyroller
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA, an affiliate of Merck KGaA
| | | | | | | | | | - Noah M Hahn
- Departments of Medical Oncology & Urology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|
44
|
Nguyen CB, Oh E, Bahar P, Vaishampayan UN, Else T, Alva AS. Novel Approaches with HIF-2α Targeted Therapies in Metastatic Renal Cell Carcinoma. Cancers (Basel) 2024; 16:601. [PMID: 38339352 PMCID: PMC10854987 DOI: 10.3390/cancers16030601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Germline inactivation of the Von Hippel-Lindau (VHL) tumor suppressor is the defining hallmark in hereditary VHL disease and VHL-associated renal cell carcinoma (RCC). However, somatic VHL mutations are also observed in patients with sporadic RCC. Loss of function VHL mutations result in constitutive activation of hypoxia-inducible factor-2 alpha (HIF-2α), which leads to increased expression of HIF target genes that promote angiogenesis and tumor growth. As of 2023, belzutifan is currently the only approved HIF-2α inhibitor for both VHL-associated and sporadic metastatic RCC (mRCC). However, there is potential for resistance with HIF-2α inhibitors which warrants novel HIF-2α-targeting strategies. In this review, we discuss the potential resistance mechanisms with belzutifan and current clinical trials evaluating novel combinations of belzutifan with other targeted therapies and immune checkpoint inhibitors which may enhance the efficacy of HIF-2α targeting. Lastly, we also discuss newer generation HIF-2α inhibitors that are currently under early investigation and outline future directions and challenges with HIF-2α inhibitors for mRCC.
Collapse
Affiliation(s)
- Charles B. Nguyen
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (U.N.V.); (T.E.); (A.S.A.)
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eugene Oh
- University of Michigan Medical School, Ann Arbor, MI 48109, USA; (E.O.); (P.B.)
| | - Piroz Bahar
- University of Michigan Medical School, Ann Arbor, MI 48109, USA; (E.O.); (P.B.)
| | - Ulka N. Vaishampayan
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (U.N.V.); (T.E.); (A.S.A.)
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tobias Else
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (U.N.V.); (T.E.); (A.S.A.)
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ajjai S. Alva
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (U.N.V.); (T.E.); (A.S.A.)
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
45
|
Jiang F, Mao M, Jiang S, Jiao Y, Cao D, Xiang Y. PD-1 and TIGIT coexpressing CD8 + CD103 + tissue-resident memory cells in endometrial cancer as potential targets for immunotherapy. Int Immunopharmacol 2024; 127:111381. [PMID: 38150880 DOI: 10.1016/j.intimp.2023.111381] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Immunotherapy has shown promise in treating various cancers; however, its efficacy in endometrial cancer (EC) remains suboptimal owing to the complex dynamics of the tumour immune microenvironment. This study focuses on exploring the potential of targeting the programmed cell death protein 1 gene (PD-1) and the T cell Immunoreceptor with Ig and ITIM domains gene (TIGIT) coexpressing tissue-resident memory cells in EC. METHODS A comprehensive approach, utilizing RNA sequencing, single-cell RNA sequencing, mass cytometry, and flow cytometry, was employed to analyse the expression patterns of PD-1 and TIGIT in the EC tumor environment and to characterize the phenotypic properties of tumor-infiltrating lymphocytes (TILs), particularly tissue-resident memory (TRM) cells. Additionally, in vitro cell experiments were conducted to assess the functional impact of PD-1 and TIGIT blockade on T-cell activity. RESULTS Our analysis identified a significant co-expression of PD-1 and TIGIT in TRM cells within the EC tumor microenvironment. These TRM cells displayed an exhausted phenotype with impaired cytotoxicity, enhanced proliferative capacity, and diminished cytotoxic activity. In vitro T-cell assays showed that a dual blockade of PD-1 and TIGIT more effectively restored T-cell functionality compared to single blockade, suggesting enhanced therapeutic potential. CONCLUSIONS TRM cells co-expressing PD-1 and TIGIT represent potential targets for EC immunotherapy. Dual immune checkpoint blockade targeting PD-1 and TIGIT may offer an effective therapeutic strategy for EC, providing valuable insights for the development of immunotherapeutic approaches.
Collapse
Affiliation(s)
- Fang Jiang
- Department of Obstetrics and Gynaecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynaecologic Diseases, Beijing, China
| | - Mingyi Mao
- Department of Obstetrics and Gynaecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynaecologic Diseases, Beijing, China
| | - Shiyang Jiang
- Ovarian Cancer Program, Department of Gynaecologic Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuhao Jiao
- Department of Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Dongyan Cao
- Department of Obstetrics and Gynaecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynaecologic Diseases, Beijing, China
| | - Yang Xiang
- Department of Obstetrics and Gynaecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynaecologic Diseases, Beijing, China.
| |
Collapse
|
46
|
Zhang P, Liu X, Gu Z, Jiang Z, Zhao S, Song Y, Yu J. Targeting TIGIT for cancer immunotherapy: recent advances and future directions. Biomark Res 2024; 12:7. [PMID: 38229100 PMCID: PMC10790541 DOI: 10.1186/s40364-023-00543-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/08/2023] [Indexed: 01/18/2024] Open
Abstract
As a newly identified checkpoint, T cell immunoreceptor with immunoglobulin and tyrosine-based inhibitory motif (ITIM) domain (TIGIT) is highly expressed on CD4+ T cells, CD8+ T cells, natural killer (NK) cells, regulatory T cells (Tregs), and tumor-infiltrating lymphocytes (TILs). TIGIT has been associated with NK cell exhaustion in vivo and in individuals with various cancers. It not only modulates NK cell survival but also mediates T cell exhaustion. As the primary ligand of TIGIT in humans, CD155 may be the main target for immunotherapy due to its interaction with TIGIT. It has been found that the anti-programmed cell death protein 1 (PD-1) treatment response in cancer immunotherapy is correlated with CD155 but not TIGIT. Anti-TIGIT alone and in combination with anti-PD-1 agents have been tested for cancer immunotherapy. Although two clinical studies on advanced lung cancer had positive results, the TIGIT-targeted antibody, tiragolumab, recently failed in two new trials. In this review, we highlight the current developments on TIGIT for cancer immunotherapy and discuss the characteristics and functions of TIGIT.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Medical Key Laboratory of Thoracic Oncology, Zhengzhou, 450052, Henan, China
| | - Xinyuan Liu
- Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Zhuoyu Gu
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Medical Key Laboratory of Thoracic Oncology, Zhengzhou, 450052, Henan, China
| | - Zhongxing Jiang
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Song Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Yongping Song
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Jifeng Yu
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Henan International Joint Laboratory of Nuclear Protein Gene Regulation, Henan University College of Medicine, Kaifeng, 475004, Henan, China.
| |
Collapse
|
47
|
Jiang YJ, Guo NT, Xia XP, Ji Y, Huo JG. Immunotherapy strategies and traditional Chinese medicine treatment for microsatellite stable metastatic colorectal cancer. Shijie Huaren Xiaohua Zazhi 2023; 31:1007-1013. [DOI: 10.11569/wcjd.v31.i24.1007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/24/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023] Open
Abstract
The incidence and mortality of colorectal cancer (CRC) have increased year by year. In addition to traditional radiotherapy, chemotherapy, and targeted therapy, immunotherapy also brings hope to more patients with metastatic colorectal cancer (mCRC). However, these treatments are limited to patients with high microsatellite instability, and about 95% of mCRC patients with microsatellite stability (MSS) can not benefit from them. How to enhance the response of MSS mCRC patients to immunotherapy is the focus of current research. In recent years, it has been found that immunotherapy strategies are expected to improve the clinical efficacy for such patients, and the research reports of TCM combined with immunotherapy are increasing day by day. Therefore, this article aims to review the immunotherapy and traditional Chinese medicine treatment for MSS colorectal cancer.
Collapse
Affiliation(s)
- Yu-Jing Jiang
- The Third Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| | - Nai-Ting Guo
- The Third Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| | - Xue-Ping Xia
- The Third Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| | - Yi Ji
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| | - Jie-Ge Huo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| |
Collapse
|
48
|
Bredel D, Tihic E, Mouraud S, Danlos FX, Susini S, Aglave M, Alfaro A, Mohamed-Djalim C, Rouanne M, Halse H, Bigorgne A, Tselikas L, Dalle S, Hartl DM, Baudin E, Guettier C, Vibert E, Rosmorduc O, Robert C, Ferlicot S, Parier B, Albiges L, de Montpreville VT, Besse B, Mercier O, Even C, Breuskin I, Classe M, Radulescu C, Lebret T, Pautier P, Gouy S, Scoazec JY, Zitvogel L, Marabelle A, Bonvalet M. Immune checkpoints are predominantly co-expressed by clonally expanded CD4 +FoxP3 + intratumoral T-cells in primary human cancers. J Exp Clin Cancer Res 2023; 42:333. [PMID: 38057799 DOI: 10.1186/s13046-023-02897-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/11/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND In addition to anti-PD(L)1, anti-CTLA-4 and anti-LAG-3, novel immune checkpoint proteins (ICP)-targeted antibodies have recently failed to demonstrate significant efficacy in clinical trials. In these trials, patients were enrolled without screening for drug target expression. Although these novel ICP-targeted antibodies were expected to stimulate anti-tumor CD8 + T-cells, the rationale for their target expression in human tumors relied on pre-clinical IHC stainings and transcriptomic data, which are poorly sensitive and specific techniques for assessing membrane protein expression on immune cell subsets. Our aim was to describe ICP expression on intratumoral T-cells from primary solid tumors to better design upcoming neoadjuvant cancer immunotherapy trials. METHODS We prospectively performed multiparameter flow cytometry and single-cell RNA sequencing (scRNA-Seq) paired with TCR sequencing on freshly resected human primary tumors of various histological types to precisely determine ICP expression levels within T-cell subsets. RESULTS Within a given tumor type, we found high inter-individual variability for tumor infiltrating CD45 + cells and for T-cells subsets. The proportions of CD8+ T-cells (~ 40%), CD4+ FoxP3- T-cells (~ 40%) and CD4+ FoxP3+ T-cells (~ 10%) were consistent across patients and indications. Intriguingly, both stimulatory (CD25, CD28, 4-1BB, ICOS, OX40) and inhibitory (PD-1, CTLA-4, PD-L1, CD39 and TIGIT) checkpoint proteins were predominantly co-expressed by intratumoral CD4+FoxP3+ T-cells. ScRNA-Seq paired with TCR sequencing revealed that T-cells with high clonality and high ICP expressions comprised over 80% of FoxP3+ cells among CD4+ T-cells. Unsupervised clustering of flow cytometry and scRNAseq data identified subsets of CD8+ T-cells and of CD4+ FoxP3- T-cells expressing certain checkpoints, though these expressions were generally lower than in CD4+ FoxP3+ T-cell subsets, both in terms of proportions among total T-cells and ICP expression levels. CONCLUSIONS Tumor histology alone does not reveal the complete picture of the tumor immune contexture. In clinical trials, assumptions regarding target expression should rely on more sensitive and specific techniques than conventional IHC or transcriptomics. Flow cytometry and scRNAseq accurately characterize ICP expression within immune cell subsets. Much like in hematology, flow cytometry can better describe the immune contexture of solid tumors, offering the opportunity to guide patient treatment according to drug target expression rather than tumor histological type.
Collapse
Affiliation(s)
- Delphine Bredel
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
| | - Edi Tihic
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - Séverine Mouraud
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - François-Xavier Danlos
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Innovation Thérapeutique Et d'Essais Précoces (DITEP), 94805, Villejuif, France
| | - Sandrine Susini
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - Marine Aglave
- Gustave Roussy, Plateforme de bioinformatique, F-94805, Villejuif, France
| | - Alexia Alfaro
- Gustave Roussy, Université Paris-Saclay, UMS 23/3655, Plateforme Imagerie Et Cytométrie, Villejuif, France
| | - Chifaou Mohamed-Djalim
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - Mathieu Rouanne
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, USA
| | - Héloise Halse
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1163, Institut Imagine, Université Paris Descartes, 75015, Paris, France
| | - Amélie Bigorgne
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1163, Institut Imagine, Université Paris Descartes, 75015, Paris, France
| | - Lambros Tselikas
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Stéphane Dalle
- Department of Dermatology, HCL Cancer Institute, Lyon Cancer Research Center, 69495, Lyon, France
| | - Dana M Hartl
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Eric Baudin
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Catherine Guettier
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Service d'Anatomie Pathologique, Hôpital Bicêtre, AP-HP, 94270, Le Kremlin-Bicêtre, France
- UMR-S 1193, Hôpital Paul Brousse Université Paris Saclay, 94800, Villejuif, France
| | - Eric Vibert
- UMR-S 1193, Hôpital Paul Brousse Université Paris Saclay, 94800, Villejuif, France
- Centre Hépato-Biliaire, Hôpital Paul Brousse, AP-HP, 94800, Villejuif, France
| | - Olivier Rosmorduc
- Centre Hépato-Biliaire, Hôpital Paul Brousse, AP-HP, 94800, Villejuif, France
| | - Caroline Robert
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U981, Gustave Roussy, 94805, Villejuif, France
| | - Sophie Ferlicot
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Service d'Anatomie Pathologique, Hôpital Bicêtre, AP-HP, 94270, Le Kremlin-Bicêtre, France
- Centre National de Recherche Scientifique (CNRS), Gustave Roussy, Université Paris-Saclay, UMR 9019, 94805, Villejuif, France
| | - Bastien Parier
- Service de Chirurgie Urologique, Hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France
| | - Laurence Albiges
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | | | - Benjamin Besse
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Olaf Mercier
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Service de Chirurgie Thoracique Et Transplantation Cardio-Pulmonaire, Hôpital Marie-Lannelongue, UMR_S 999 INSERM, Université Paris-Saclay, GHPSJ, 92350, Le Plessis-Robinson, France
| | - Caroline Even
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Ingrid Breuskin
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Marion Classe
- Gustave Roussy, Département de Biopathologie, F-94805, Villejuif, France
| | - Camélia Radulescu
- Département de Pathologie, Hôpital Foch, UVSQ, Université Paris-Saclay, 92150, Suresnes, France
| | - Thierry Lebret
- Département d'Urologie, Hôpital Foch, UVSQ-Université Paris-Saclay, 92150, Suresnes, France
| | - Patricia Pautier
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Sébastien Gouy
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Jean-Yves Scoazec
- Gustave Roussy, Département de Biopathologie, F-94805, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Aurélien Marabelle
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France.
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France.
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France.
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France.
- Gustave Roussy, Département d'Innovation Thérapeutique Et d'Essais Précoces (DITEP), 94805, Villejuif, France.
| | - Mélodie Bonvalet
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| |
Collapse
|
49
|
Nuvola G, Mollica V, Massari F, Suárez C. The future of immunotherapy in advanced renal cell carcinoma: beyond PD-1/PD-L1 inhibitors. Immunotherapy 2023; 15:1429-1433. [PMID: 37718694 DOI: 10.2217/imt-2023-0218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023] Open
Affiliation(s)
- Giacomo Nuvola
- Medical Oncology, SCIAS Hospital de Barcelona, Barcelona, 08034, Spain
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, 40138, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, 40138, Italy
| | - Cristina Suárez
- Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitario Vall d'Hebron, Barcelona, 08035, Spain
| |
Collapse
|
50
|
Kou L, Xie X, Chen X, Li B, Li J, Li Y. The progress of research on immune checkpoint inhibitor resistance and reversal strategies for hepatocellular carcinoma. Cancer Immunol Immunother 2023; 72:3953-3969. [PMID: 37917364 PMCID: PMC10992589 DOI: 10.1007/s00262-023-03568-3] [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: 07/22/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in humans, which is prone to recurrence and metastasis and has a poor prognosis. The occurrence and progression of HCC are closely related to immune elimination, immune homeostasis, and immune escape of the immune system. In recent years, immunotherapy, represented by immune checkpoint inhibitors (ICIs), has shown powerful anti-tumor capabilities in HCC patients. However, there are still some HCC patients who cannot benefit from ICIs treatment due to their innate or acquired drug resistance. Therefore, it is of great practical significance to explore the possible mechanisms of resistance to ICIs in HCC and to use them as a target to design strategies to reverse resistance, to overcome drug resistance in HCC and to improve the prognosis of patients. This article summarizes the possible primary (tumor microenvironment alteration, and signaling pathways, etc.) and acquired (immune checkpoint upregulation) resistance mechanisms in patients with HCC treated with ICIs, and based on this, discusses the status and effectiveness of combination drug strategy to reverse drug resistance, to provide a reference for subsequent related studies and decisions.
Collapse
Affiliation(s)
- Liqiu Kou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaolu Xie
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiu Chen
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Bo Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jun Li
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|