1
|
De Castro V, Abdellaoui O, Dehecq B, Ndao B, Mercier-Letondal P, Dauvé A, Garnache-Ottou F, Adotévi O, Loyon R, Godet Y. Characterization of the aryl hydrocarbon receptor as a potential candidate to improve cancer T cell therapies. Cancer Immunol Immunother 2025; 74:200. [PMID: 40358739 PMCID: PMC12075070 DOI: 10.1007/s00262-025-04065-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
The efficacy of T-cell-based cancer therapies can be limited by the tumor microenvironment which can lead to T cell dysfunction. Multiple studies, particularly in murine models, have demonstrated the capacity of the aryl hydrocarbon receptor (AHR) to negatively regulate antitumor T cell functions. AHR is a cytoplasmic receptor and transcription factor that was originally identified as a xenobiotic sensor, but has since been shown to play a significant role in the gene regulation of various immune cells, including T cells. Given the insights from murine studies, AHR emerges as a promising candidate to invalidate for optimizing T cell-based cancer therapies. However, the controversial role of AHR in human T cells underscores the need for a more comprehensive characterization of AHR expressing T cells. This study aims to investigate the regulatory mechanisms of AHR in human T cell biology to better understand its impact on reducing antitumor immune responses. Here, we knocked-out AHR in human T cells using CRISPR-Cas9 technology to characterize AHR's function in an in vitro chronic stimulation model. Engineered T cells exhibited enhanced effector- and memory-like profiles and expressed reduced amount of CD39 and TIGIT. AHR knockout enhanced human CAR-T cells' functionality and persistence upon tumor chronic stimulation. Collectively, these results highlight the role of AHR in human CAR-T cells efficiency.
Collapse
Affiliation(s)
- Valentine De Castro
- Université Marie et Louis Pasteur, EFS, INSERM UMR1098 RIGHT, 25000, Besançon, France
| | - Oumaïma Abdellaoui
- Université Marie et Louis Pasteur, EFS, INSERM UMR1098 RIGHT, 25000, Besançon, France
| | - Barbara Dehecq
- Université Marie et Louis Pasteur, EFS, INSERM UMR1098 RIGHT, 25000, Besançon, France
| | - Babacar Ndao
- Université Marie et Louis Pasteur, EFS, INSERM UMR1098 RIGHT, 25000, Besançon, France
| | | | - Alexandra Dauvé
- MGX-Montpellier GenomiX, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Francine Garnache-Ottou
- Université Marie et Louis Pasteur, EFS, INSERM UMR1098 RIGHT, 25000, Besançon, France
- Service d'hématologie et d'immunologie cellulaire, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Olivier Adotévi
- Université Marie et Louis Pasteur, EFS, INSERM UMR1098 RIGHT, 25000, Besançon, France
- Service d'oncologie médicale, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Romain Loyon
- Université Marie et Louis Pasteur, EFS, INSERM UMR1098 RIGHT, 25000, Besançon, France
| | - Yann Godet
- Université Marie et Louis Pasteur, EFS, INSERM UMR1098 RIGHT, 25000, Besançon, France.
| |
Collapse
|
2
|
Konstantaraki M, Berdiaki A, Neagu M, Zurac S, Krasagakis K, Nikitovic D. Understanding Merkel Cell Carcinoma: Pathogenic Signaling, Extracellular Matrix Dynamics, and Novel Treatment Approaches. Cancers (Basel) 2025; 17:1212. [PMID: 40227764 PMCID: PMC11987840 DOI: 10.3390/cancers17071212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Merkel cell carcinoma (MCC) is a rare but aggressive neuroendocrine skin cancer, driven by either Merkel cell polyomavirus (MCPyV) integration or ultraviolet (UV)-induced mutations. In MCPyV-positive tumors, viral T antigens inactivate tumor suppressors pRb and p53, while virus-negative MCCs harbor UV-induced mutations that activate similar oncogenic pathways. Key signaling cascades, including PI3K/AKT/mTOR and MAPK, support tumor proliferation, survival, and resistance to apoptosis. Histologically, MCC consists of small round blue cells with neuroendocrine features, high mitotic rate, and necrosis. The tumor microenvironment (TME) plays a central role in disease progression and immune escape. It comprises a mix of tumor-associated macrophages, regulatory and cytotoxic T cells, and elevated expression of immune checkpoint molecules such as PD-L1, contributing to an immunosuppressive niche. The extracellular matrix (ECM) within the TME is rich in proteoglycans, collagens, and matrix metalloproteinases (MMPs), facilitating tumor cell adhesion, invasion, and interaction with stromal and immune cells. ECM remodeling and integrin-mediated signaling further promote immune evasion and therapy resistance. Although immune checkpoint inhibitors targeting PD-1/PD-L1 have shown promise in treating MCC, resistance remains a major hurdle. Therapeutic strategies that concurrently target the TME-through inhibition of ECM components, MMPs, or integrin signaling-may enhance immune responses and improve clinical outcomes.
Collapse
Affiliation(s)
- Maria Konstantaraki
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
- Dermatology Department, University Hospital of Heraklion, 71110 Heraklion, Greece;
| | - Aikaterini Berdiaki
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
| | - Monica Neagu
- Immunology Laboratory, “Victor Babes” National Institute of Pathology, 99-101 Splaiul Independenței, 050096 Bucharest, Romania;
- Pathology Department, Colentina Clinical Hospital, 19-21 Sos Stefan Cel Mare, 020125 Bucharest, Romania;
| | - Sabina Zurac
- Pathology Department, Colentina Clinical Hospital, 19-21 Sos Stefan Cel Mare, 020125 Bucharest, Romania;
- Faculty of Dentistry, University of Medicine and Pharmacy, 8 Eroilor Sanitari Boulevard, 050474 Bucharest, Romania
| | | | - Dragana Nikitovic
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
| |
Collapse
|
3
|
Han JW, Kang MW, Lee SK, Yang H, Kim JH, Yoo JS, Cho HS, Jang EJ, Seo DH, Kwon JH, Nam SW, Bae SH, Jang JW, Choi JY, Yoon SK, Sung PS. Dynamic Peripheral T-Cell Analysis Identifies On-Treatment Prognostic Biomarkers of Atezolizumab plus Bevacizumab in Hepatocellular Carcinoma. Liver Cancer 2025; 14:104-116. [PMID: 40144473 PMCID: PMC11936438 DOI: 10.1159/000541181] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 08/21/2024] [Indexed: 03/28/2025] Open
Abstract
Introduction Variability in response to atezolizumab plus bevacizumab (AB) treatment of hepatocellular carcinoma (HCC) underscores the critical need for the development of effective biomarkers. We sought to identify peripheral blood biomarkers reflecting response to AB treatment. Methods We analyzed dynamic changes in peripheral blood mononuclear cells from a prospective, multicenter cohort of 65 patients with HCC, using flow cytometry to evaluate the T-cell population before and 3 weeks after the first AB treatment. Results We found a unique response of the CD8+ T cells in terms of both frequency and phenotype, in contrast to CD4+ T cells and regulatory T cells. Notably, CD8+ T cells showed significant changes in expression of Ki-67 and T-cell immunoreceptors with Ig and ITIM domains (TIGIT). These distinct responses were observed particularly in the programmed cell death receptor-1 (PD-1)+ subpopulation of CD8+ T cells. Interestingly, the baseline differentiation status of PD-1+CD8+ T cells, particularly the central memory T-cell subset, correlated positively with greater proliferation (higher Ki-67 expression) of PD-1+CD8+ T cells after treatment. Moreover, effector memory cells expressing CD45RA correlated negatively with the increase in TIGIT+/PD-1+CD8+ T cells. The increase in TIGIT+/CD8+ T cells was associated with the development of immune-related adverse events, whereas increase in Ki-67+/PD-1+CD8+ T cells was associated with the better objective response rate. Importantly, dynamic shifts of Ki-67+/PD-1+CD8+ T cells and TIGIT+/CD8+ T cells significantly predicted progression-free survival and overall survival, as confirmed by multivariate analysis. Conclusion These findings highlight the potential of dynamic changes in CD8+ T cells as an on-treatment prognostic biomarker. Our study underscores the value of peripheral blood profiling as a noninvasive and practical method for predicting the clinical outcomes of AB treatment in patients with HCC.
Collapse
Affiliation(s)
- Ji Won Han
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Sciences, Catholic University of Korea College of Medicine, POSTECH-Catholic Biomedical Engineering Institute, Seoul, Republic of Korea
| | - Min Woo Kang
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Sciences, Catholic University of Korea College of Medicine, POSTECH-Catholic Biomedical Engineering Institute, Seoul, Republic of Korea
| | - Soon Kyu Lee
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, Incheon, Republic of Korea
| | - Hyun Yang
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Hoon Kim
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Uijeongbu St. Mary’s Hospital, The Catholic University of Korea, Uijeongbu, Republic of Korea
| | - Jae-Sung Yoo
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hee Sun Cho
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Ji Jang
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Sciences, Catholic University of Korea College of Medicine, POSTECH-Catholic Biomedical Engineering Institute, Seoul, Republic of Korea
| | - Deok Hwa Seo
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Sciences, Catholic University of Korea College of Medicine, POSTECH-Catholic Biomedical Engineering Institute, Seoul, Republic of Korea
| | - Jung Hyun Kwon
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, Incheon, Republic of Korea
| | - Soon Woo Nam
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, Incheon, Republic of Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong Won Jang
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong Young Choi
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Pil Soo Sung
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Sciences, Catholic University of Korea College of Medicine, POSTECH-Catholic Biomedical Engineering Institute, Seoul, Republic of Korea
| |
Collapse
|
4
|
Naing A, McKean M, Tolcher A, Victor A, Hu P, Gao W, Nogueira Filho MAF, Kitzing T, Gleicher S, Holland D, Richter E, Tadjalli-Mehr K, Siu LL. TIGIT inhibitor M6223 as monotherapy or in combination with bintrafusp alfa in patients with advanced solid tumors: a first-in-human, phase 1, dose-escalation trial. J Immunother Cancer 2025; 13:e010584. [PMID: 39929671 PMCID: PMC11815413 DOI: 10.1136/jitc-2024-010584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/20/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND M6223 is an intravenous (IV), Fc-competent, fully human, antagonistic, anti-T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) antibody. Bintrafusp alfa (BA) is a bifunctional fusion protein that simultaneously blocks nonredundant immunosuppressive TGF-β and PD-(L)1 pathways. METHODS This first-in-human, dose-escalation study in patients with advanced solid tumors (N=58; aged ≥18 years, ECOG PS≤1) evaluated M6223 alone (Part 1A, n=40; M6223 10-2400 mg every 2 weeks, n=32; M6223 2400 mg every 3 weeks, n=8) or with BA (Part 1B, n=18; M6223 300-1600 mg with BA 1200 mg; both every 2 weeks, intravenous). Primary objectives were safety, tolerability, maximum tolerated dose (MTD) and recommended dose for expansion (RDE). Additional objectives included pharmacokinetics, pharmacodynamics and clinical activity (NCT04457778). RESULTS Two dose-limiting toxicities were observed: grade 3 adrenal insufficiency (Part 1A: M6223 900 mg every 2 weeks) and grade 3 anemia (Part 1B: M6223 300 mg, only BA related). MTD was not reached. Overall, median overall survival and progression-free survival were 7.6 (95% CI 4.9, 12.0) and 1.4 (95% CI 1.3, 1.8) months, respectively. Stable disease as best response was observed in 13 (32.5%) and 5 (27.8%) patients in parts 1A and 1B, respectively. M6223±BA displayed a linear pharmacokinetic profile. Anti-TIGIT mode-of-action-related pharmacodynamic effects were observed in peripheral blood and in tumor tissue. RDEs were 1600 mg every 2 weeks or 2400 mg every 3 weeks for M6223 monotherapy and 1600+1200 mg every 2 weeks for M6223+BA. CONCLUSIONS M6223±BA had a manageable safety profile, with RDEs defined for both monotherapy and combination therapy. Further evaluation of M6223 is ongoing in combination with the PD-L1 inhibitor avelumab in patients with advanced urothelial carcinoma (JAVELIN Bladder Medley; NCT05327530). TRIAL REGISTRATION NUMBER NCT04457778.
Collapse
Affiliation(s)
- Aung Naing
- The University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| | | | | | | | - Ping Hu
- EMD Serono, Billerica, Massachusetts, USA
| | - Wei Gao
- EMD Serono, Billerica, Massachusetts, USA
| | | | | | | | | | | | | | - Lillian L Siu
- Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Piao M, Zhang N, Li J, Li C, Xun Z, Zhang L, Wang S, Sun B, Li S, Yang X, Yang X, Wang H, Zhao H. Peripheral blood PD-1 + T lymphocytes as biomarkers in liquid biopsies for solid tumors: Clinical significance and prognostic applications. Int Immunopharmacol 2025; 147:114052. [PMID: 39799737 DOI: 10.1016/j.intimp.2025.114052] [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: 11/25/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
A shift toward a T cell exhaustion phenotype is associated with the upregulation of expression of programmed cell death protein 1 (PD-1) on T lymphocytes in patients with malignant solid tumors. The interaction between PD-1 and programmed death-ligand 1 (PD-L1) inhibits PD-1+ T lymphocyte function, impacting their anti-tumor immune activity. Immune checkpoint inhibitors targeting PD-1/PD-L1 have revolutionized the treatment of various solid malignancies, improving therapeutic efficacy and survival outcomes. Peripheral blood analysis of liquid biopsies is being increasingly used to identify populations most likely to benefit from various treatment modalities. PD-1+ T lymphocytes represent the primary cell population responsive to immunotherapeutic interventions for patients with solid malignancies, as evidenced by the altered PD-1 expression levels and proportion of cells comprising the overall population of immunocytes. PD-1+ T cells in peripheral blood exert an associative and reciprocal predictive effect on homologous intratumoral cells. Distinct subpopulations of PD-1+ T cells exhibit differential ability to proliferate in the periphery and can be characterized by tumor antigen-specific and exhaustion phenotypes. These characteristics have prognostic implications, aiding in the prediction of the efficacy of antitumor therapy and predicting survival outcomes. We highlight distinct subpopulations of PD-1+ T cells, their exhaustion and antigen-specific phenotypes, and their dynamic changes over treatment, providing insights into their utility for tailoring personalized therapies. For the first time, this review discusses the role of peripheral PD-1+ T lymphocytes as prognostic biomarkers in liquid biopsies, focusing on their clinical significance, predictive value during therapy, and future research directions.
Collapse
Affiliation(s)
- Mingjian Piao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Nan Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Jiongyuan Li
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Chengjie Li
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Ziyu Xun
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Longhao Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Shanshan Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Boyu Sun
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Shuofeng Li
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Xu Yang
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Xiaobo Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China.
| | - Hanping Wang
- Division of Pulmonary and Critical Care Medicine, State Key Laboratory of Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China.
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Gu X, Li D, Wu P, Zhang C, Cui X, Shang D, Ma R, Liu J, Sun N, He J. Revisiting the CXCL13/CXCR5 axis in the tumor microenvironment in the era of single-cell omics: Implications for immunotherapy. Cancer Lett 2024; 605:217278. [PMID: 39332588 DOI: 10.1016/j.canlet.2024.217278] [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: 07/15/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
As one of the important members of the family of chemokines and their receptors, the CXCL13/CXCR5 axis is involved in follicle formation in normal lymphoid tissues and the establishment of somatic cavity immunity under physiological conditions, as well as being associated with a wide range of infectious, autoimmune, and tumoral diseases. Here in this review, we focus on its role in tumors. Traditional studies have found the axis to be both pro- and anti-tumorigenic, involving a variety of immune cells, including the tumor cells themselves and those in the tumor microenvironment (TME), and the prognostic significance of this axis is clinical context-dependent. With the development of techniques at the single-cell level, we were able to explain in detail the status of the CXCL13/CXCR5 axis in the TME based on real clinical samples and found that it involves a range of crucial intrinsic anti-tumor immune processes in the TME and is therefore important in tumor immunotherapy. We summarize the cellular subsets, physiological functions, and prognostic significance associated with this axis in the most promising immune checkpoint inhibitor (ICI) therapies of the day and summarize possible therapeutic ideas based on this axis. As with any TME study, the most important takeaway is that the complexity of the CXCL13/CXCR5 axis in TME suggests the importance of personalized therapy in tumor therapy.
Collapse
Affiliation(s)
- Xuanyu Gu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dongyu Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Wu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinyu Cui
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dexin Shang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ruijie Ma
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jingjing Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
7
|
Guo H, Guo L, Wang B, Jiang X, Wu Z, Mo X, Sun Y, Zhang Y, Wang Z, Kong J, Yan C, Huang X. Distinct Immune Homeostasis Remodeling Patterns after HLA-Matched and Haploidentical Transplantation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400544. [PMID: 39225336 PMCID: PMC11497014 DOI: 10.1002/advs.202400544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/21/2024] [Indexed: 09/04/2024]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a widely used treatment for a variety of hematopoietic disorders, and also provides a valuable platform for investigating the development of donor-derived immune cells in recipients post-HSCT. The immune system remodels from the donor to the recipient during allo-HSCT. However, little is known about the cell profile alterations as donor homeostasis rebalances to recipient homeostasis following HSCT. Here, multi-omics technology is applied at both the single cell and bulk sample levels, as well as spectrum flow cytometry and fluorescent transgenic mouse models, to dissect the dynamics of the rebalanced homeostatic immune system in recipients after allo-HSCT. The data reveal that all immune subpopulations observed in donors are successfully restored in recipients, though with varying levels of abundance. The remodeling of immune homeostasis exhibits different patterns in HLA-matched and haploidentical HSCT, highlighting distinct biases in T cell reconstitution from the central and peripheral pathways. Furthermore, ZNF683 is critical for maintaining the persistence and quiescence of CD8 T-cell in haploidentical HSCT. The research can serve as a foundation for developing novel strategies to induce immune tolerance.
Collapse
Affiliation(s)
- Huidong Guo
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
| | - Liping Guo
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
| | - Bixia Wang
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
| | - Xinya Jiang
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic MalignanciesChinese Academy of Medical SciencesBeijing2019RU029China
| | - Zhigui Wu
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
- Peking‐Tsinghua Center for Life SciencesAcademy for Advanced Interdisciplinary StudiesPeking UniversityBeijing100871China
| | - Xiao‐Dong Mo
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
| | - Yu‐Qian Sun
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
| | - Yuan‐Yuan Zhang
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
| | - Zhi‐Dong Wang
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
| | - Jun Kong
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
| | - Chen‐Hua Yan
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
| | - Xiao‐Jun Huang
- National Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalPeking University Institute of HematologyPeking UniversityBeijing100044China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic MalignanciesChinese Academy of Medical SciencesBeijing2019RU029China
- Peking‐Tsinghua Center for Life SciencesAcademy for Advanced Interdisciplinary StudiesPeking UniversityBeijing100871China
| |
Collapse
|
8
|
Goswami M, Toney NJ, Pitts SC, Celades C, Schlom J, Donahue RN. Peripheral immune biomarkers for immune checkpoint inhibition of solid tumours. Clin Transl Med 2024; 14:e1814. [PMID: 39162097 PMCID: PMC11333946 DOI: 10.1002/ctm2.1814] [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: 05/30/2024] [Revised: 07/29/2024] [Accepted: 08/04/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND With the rapid adoption of immunotherapy for the treatment of cancer comes the pressing need for readily accessible biomarkers to guide immunotherapeutic strategies and offer insights into outcomes with specific treatments. Regular sampling of solid tumour tissues outside of melanoma for immune monitoring is not often feasible; conversely, routine, frequent interrogation of circulating immune biomarkers is entirely possible. As immunotherapies and immune checkpoint inhibitors, in particular, are more widely used in first-line, neoadjuvant, and metastatic settings, the discovery and validation of peripheral immune biomarkers are urgently needed across solid tumour types for improved prediction and prognostication of clinical outcomes in response to immunotherapy, as well as elucidation of mechanistic underpinnings of the intervention. Careful experimental design, encompassing both retrospective and prospective studies, is required in such biomarker identification studies, and concerted efforts are essential for their advancement into clinical settings. CONCLUSION In this review, we summarize shared immune features between the tumour microenvironment and systemic circulation, evaluate exploratory peripheral immune biomarker studies, and discuss associations between candidate biomarkers with clinical outcomes. We also consider integration of multiple peripheral immune parameters for better prediction and prognostication and discuss considerations in study design to further evaluate the clinical utility of candidate peripheral immune biomarkers for immunotherapy of solid tumours. HIGHLIGHTS Peripheral immune biomarkers are critical for improved prediction and prognostication of clinical outcomes for patients with solid tumours treated with immune checkpoint inhibition. Candidate peripheral biomarkers, such as cytokines, soluble factors, and immune cells, have potential as biomarkers to guide immunotherapy of solid tumours. Multiple peripheral immune parameters may be integrated to improve prediction and prognostication. The potential of peripheral immune biomarkers to guide immunotherapy of solid tumours requires critical work in biomarker discovery, validation, and standardization.
Collapse
Affiliation(s)
- Meghali Goswami
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Nicole J. Toney
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Stephanie C. Pitts
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Carolina Celades
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Jeffrey Schlom
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Renee N. Donahue
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
9
|
Wondergem NE, Miedema IHC, van de Ven R, Zwezerijnen GJC, de Graaf P, Karagozoglu KH, Hendrickx JJ, Eerenstein SEJ, Bun RJ, Mulder DC, Voortman J, Boellaard R, Windhorst AD, Hagers JP, Peferoen LAN, de Gruijl TD, Bloemena E, Brakenhoff RH, Leemans CR, Menke-van der Houven van Oordt CW. Circulating T cell status and molecular imaging may predict clinical benefit of neoadjuvant PD-1 blockade in oral cancer. J Immunother Cancer 2024; 12:e009278. [PMID: 39038919 PMCID: PMC11268040 DOI: 10.1136/jitc-2024-009278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Addition of neoadjuvant immune checkpoint inhibition to standard-of-care interventions for locally advanced oral cancer could improve clinical outcome. METHODS In this study, 16 evaluable patients with stage III/IV oral cancer were treated with one dose of 480 mg nivolumab 3 weeks prior to surgery. Primary objectives were safety, feasibility, and suitability of programmed death receptor ligand-1 positron emission tomography (PD-L1 PET) as a biomarker for response. Imaging included 18F-BMS-986192 (PD-L1) PET and 18F-fluorodeoxyglucose (FDG) PET before and after nivolumab treatment. Secondary objectives included clinical and pathological response, and immune profiling of peripheral blood mononuclear cells (PBMCs) for response prediction. Baseline tumor biopsies and postnivolumab resection specimens were evaluated by histopathology. RESULTS Grade III or higher adverse events were not observed and treatment was not delayed in relation to nivolumab administration and other study procedures. Six patients (38%) had a pathological response, of whom three (19%) had a major (≥90%) pathological response (MPR). Tumor PD-L1 PET uptake (quantified using standard uptake value) was not statistically different in patients with or without MPR (median 5.3 vs 3.4). All major responders showed a significantly postnivolumab decreased signal on FDG PET. PBMC immune phenotyping showed higher levels of CD8+ T cell activation in MPR patients, evidenced by higher baseline expression levels of PD-1, TIGIT, IFNγ and lower levels of PD-L1. CONCLUSION Together these data support that neoadjuvant treatment of advanced-stage oral cancers with nivolumab was safe and induced an MPR in a promising 19% of patients. Response was associated with decreased FDG PET uptake as well as activation status of peripheral T cell populations.
Collapse
Affiliation(s)
- Niels E Wondergem
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Iris H C Miedema
- Amsterdam UMC location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Rieneke van de Ven
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, Netherlands
| | - Gerben J C Zwezerijnen
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Pim de Graaf
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - K Hakki Karagozoglu
- Amsterdam UMC and Academic Centre for Dentistry Amsterdam (ACTA), Oral and Maxillofacial Surgery/Oral Pathology, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Jan-Jaap Hendrickx
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Simone E J Eerenstein
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Rolf J Bun
- Oral and Maxillofacial Surgery, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | - Dorien C Mulder
- Oral and Maxillofacial Surgery, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | - Jens Voortman
- Amsterdam UMC location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - J Pascal Hagers
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Laura A N Peferoen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Pathology, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Amsterdam UMC location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Elisabeth Bloemena
- Amsterdam UMC location Vrije Universiteit Amsterdam, Pathology, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Ruud H Brakenhoff
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - C René Leemans
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - C Willemien Menke-van der Houven van Oordt
- Amsterdam UMC location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Cadiou G, Beauvais T, Marotte L, Lambot S, Deleine C, Vignes C, Gantier M, Hussong M, Rulli S, Jarry A, Simon S, Malissen B, Labarriere N. Differential impact of genetic deletion of TIGIT or PD-1 on melanoma-specific T-lymphocytes. Oncoimmunology 2024; 13:2376782. [PMID: 38983599 PMCID: PMC11232637 DOI: 10.1080/2162402x.2024.2376782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024] Open
Abstract
Immune checkpoint (IC) blockade and adoptive transfer of tumor-specific T-cells (ACT) are two major strategies to treat metastatic melanoma. Their combination can potentiate T-cell activation in the suppressive tumor microenvironment, but the autoimmune adverse effects associated with systemic injection of IC blockers persist with this strategy. ACT of tumor-reactive T-cells defective for IC expression would overcome this issue. For this purpose, PD-1 and TIGIT appear to be relevant candidates, because their co-expression on highly tumor-reactive lymphocytes limits their therapeutic efficacy within the tumor microenvironme,nt. Our study compares the consequences of PDCD1 or TIGIT genetic deletion on anti-tumor properties and T-cell fitness of melanoma-specific T lymphocytes. Transcriptomic analyses revealed down-regulation of cell cycle-related genes in PD-1KO T-cells, consistent with biological observations, whereas proliferative pathways were preserved in TIGITKO T-cells. Functional analyses showed that PD-1KO and TIGITKO T-cells displayed superior antitumor reactivity than their wild-type counterpart in vitro and in a preclinical melanoma model using immunodeficient mice. Interestingly, it appears that TIGITKO T-cells were more effective at inhibiting tumor cell proliferation in vivo, and persist longer within tumors than PD-1KO T-cells, consistent with the absence of impact of TIGIT deletion on T-cell fitness. Taken together, these results suggest that TIGIT deletion, over PD-1 deletion, in melanoma-specific T-cells is a compelling option for future immunotherapeutic strategies.
Collapse
Affiliation(s)
- Gwenann Cadiou
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Tiffany Beauvais
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Lucine Marotte
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
| | - Sylvia Lambot
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Cécile Deleine
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Caroline Vignes
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Malika Gantier
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, Nantes, France
| | - Melanie Hussong
- QIAGEN Sciences, Frederick, MD, USA
- NeoGenomics, Research Triangle Park, Durham, NC, USA
| | | | - Anne Jarry
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Sylvain Simon
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bernard Malissen
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
| | - Nathalie Labarriere
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
| |
Collapse
|
11
|
Monteran L, Ershaid N, Scharff Y, Zoabi Y, Sanalla T, Ding Y, Pavlovsky A, Zait Y, Langer M, Caller T, Eldar-Boock A, Avivi C, Sonnenblick A, Satchi-Fainaro R, Barshack I, Shomron N, Zhang XHF, Erez N. Combining TIGIT Blockade with MDSC Inhibition Hinders Breast Cancer Bone Metastasis by Activating Antitumor Immunity. Cancer Discov 2024; 14:1252-1275. [PMID: 38427556 DOI: 10.1158/2159-8290.cd-23-0762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 01/17/2024] [Accepted: 02/28/2024] [Indexed: 03/03/2024]
Abstract
Bone is the most common site of breast cancer metastasis. Bone metastasis is incurable and is associated with severe morbidity. Utilizing an immunocompetent mouse model of spontaneous breast cancer bone metastasis, we profiled the immune transcriptome of bone metastatic lesions and peripheral bone marrow at distinct metastatic stages, revealing dynamic changes during the metastatic process. We show that cross-talk between granulocytes and T cells is central to shaping an immunosuppressive microenvironment. Specifically, we identified the PD-1 and TIGIT signaling axes and the proinflammatory cytokine IL1β as central players in the interactions between granulocytes and T cells. Targeting these pathways in vivo resulted in attenuated bone metastasis and improved survival, by reactivating antitumor immunity. Analysis of patient samples revealed that TIGIT and IL1β are prominent in human bone metastasis. Our findings suggest that cotargeting immunosuppressive granulocytes and dysfunctional T cells may be a promising novel therapeutic strategy to inhibit bone metastasis. Significance: Temporal transcriptome profiling of the immune microenvironment in breast cancer bone metastasis revealed key communication pathways between dysfunctional T cells and myeloid derived suppressor cells. Cotargeting of TIGIT and IL1β inhibited bone metastasis and improved survival. Validation in patient data implicated these targets as a novel promising approach to treat human bone metastasis.
Collapse
Affiliation(s)
- Lea Monteran
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nour Ershaid
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ye'ela Scharff
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yazeed Zoabi
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamer Sanalla
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yunfeng Ding
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, Texas
| | - Anna Pavlovsky
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Zait
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Marva Langer
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tal Caller
- Tamman Cardiovascular Research Institute, Sheba Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anat Eldar-Boock
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Camila Avivi
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amir Sonnenblick
- Oncology Division, Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noam Shomron
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, Texas
| | - Neta Erez
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
12
|
Becker JC, Stang A, Schrama D, Ugurel S. Merkel Cell Carcinoma: Integrating Epidemiology, Immunology, and Therapeutic Updates. Am J Clin Dermatol 2024; 25:541-557. [PMID: 38649621 PMCID: PMC11193695 DOI: 10.1007/s40257-024-00858-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2024] [Indexed: 04/25/2024]
Abstract
Merkel cell carcinoma (MCC) is a rare skin cancer characterized by neuroendocrine differentiation. Its carcinogenesis is based either on the integration of the Merkel cell polyomavirus or on ultraviolet (UV) mutagenesis, both of which lead to high immunogenicity either through the expression of viral proteins or neoantigens. Despite this immunogenicity resulting from viral or UV-associated carcinogenesis, it exhibits highly aggressive behavior. However, owing to the rarity of MCC and the lack of epidemiologic registries with detailed clinical data, there is some uncertainty regarding the spontaneous course of the disease. Historically, advanced MCC patients were treated with conventional cytotoxic chemotherapy yielding a median response duration of only 3 months. Starting in 2017, four programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) immune checkpoint inhibitors-avelumab, pembrolizumab, nivolumab (utilized in both neoadjuvant and adjuvant settings), and retifanlimab-have demonstrated efficacy in treating patients with disseminated MCC on the basis of prospective clinical trials. However, generating clinical evidence for rare cancers, such as MCC, is challenging owing to difficulties in conducting large-scale trials, resulting in small sample sizes and therefore lacking statistical power. Thus, to comprehensively understand the available clinical evidence on various immunotherapy approaches for MCC, we also delve into the epidemiology and immune biology of this cancer. Nevertheless, while randomized studies directly comparing immune checkpoint inhibitors and chemotherapy in MCC are lacking, immunotherapy shows response rates comparable to those previously reported with chemotherapy but with more enduring responses. Notably, adjuvant nivolumab has proven superiority to the standard-of-care therapy (observation) in the adjuvant setting.
Collapse
Affiliation(s)
- Jürgen C Becker
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), partner site Essen, University Duisburg-Essen, Universitätsstrasse 1, 45141, Essen, Germany.
- Department of Dermatology, University Medicine Essen, Essen, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Andreas Stang
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany
- Cancer Registry of North Rhine-Westphalia, Bochum, Germany
| | - David Schrama
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Selma Ugurel
- Department of Dermatology, University Medicine Essen, Essen, Germany
| |
Collapse
|
13
|
Liu X, Zhao A, Xiao S, Li H, Li M, Guo W, Han Q. PD-1: A critical player and target for immune normalization. Immunology 2024; 172:181-197. [PMID: 38269617 DOI: 10.1111/imm.13755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/05/2024] [Indexed: 01/26/2024] Open
Abstract
Immune system imbalances contribute to the pathogenesis of several different diseases, and immunotherapy shows great therapeutic efficacy against tumours and infectious diseases with immune-mediated derivations. In recent years, molecules targeting the programmed cell death protein 1 (PD-1) immune checkpoint have attracted much attention, and related signalling pathways have been studied clearly. At present, several inhibitors and antibodies targeting PD-1 have been utilized as anti-tumour therapies. However, increasing evidence indicates that PD-1 blockade also has different degrees of adverse side effects, and these new explorations into the therapeutic safety of PD-1 inhibitors contribute to the emerging concept that immune normalization, rather than immune enhancement, is the ultimate goal of disease treatment. In this review, we summarize recent advancements in PD-1 research with regard to immune normalization and targeted therapy.
Collapse
Affiliation(s)
- Xuening Liu
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Alison Zhao
- Cleveland Clinic Lerner College of Medicine at Case Western Reserve School of Medicine, Cleveland, Ohio, USA
| | - Su Xiao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
- People's Hospital of Zhoucun, Zibo, Shandong, China
| | - Haohao Li
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Menghua Li
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Wei Guo
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| |
Collapse
|
14
|
Dong Y, Chen Z, Yang F, Wei J, Huang J, Long X. Prediction of immunotherapy responsiveness in melanoma through single-cell sequencing-based characterization of the tumor immune microenvironment. Transl Oncol 2024; 43:101910. [PMID: 38417293 PMCID: PMC10907870 DOI: 10.1016/j.tranon.2024.101910] [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: 11/27/2023] [Revised: 01/13/2024] [Accepted: 02/08/2024] [Indexed: 03/01/2024] Open
Abstract
Immune checkpoint inhibitors (ICB) therapy have emerged as effective treatments for melanomas. However, the response of melanoma patients to ICB has been highly heterogenous. Here, by analyzing integrated scRNA-seq datasets from melanoma patients, we revealed significant differences in the TiME composition between ICB-resistant and responsive tissues, with resistant or responsive tissues characterized by an abundance of myeloid cells and CD8+ T cells or CD4+ T cell predominance, respectively. Among CD4+ T cells, CD4+ CXCL13+ Tfh-like cells were associated with an immunosuppressive phenotype linked to immune escape-related genes and negative regulation of T cell activation. We also develop an immunotherapy response prediction model based on the composition of the immune compartment. Our predictive model was validated using CIBERSORTx on bulk RNA-seq datasets from melanoma patients pre- and post-ICB treatment and showed a better performance than other existing models. Our study presents an effective immunotherapy response prediction model with potential for further translation, as well as underscores the critical role of the TiME in influencing the response of melanomas to immunotherapy.
Collapse
Affiliation(s)
- Yucheng Dong
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Zhizhuo Chen
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Fan Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaxin Wei
- Department of Emergency Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiuzuo Huang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| | - Xiao Long
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
15
|
Hou J, Yang X, Xie S, Zhu B, Zha H. Circulating T cells: a promising biomarker of anti-PD-(L)1 therapy. Front Immunol 2024; 15:1371559. [PMID: 38576625 PMCID: PMC10991692 DOI: 10.3389/fimmu.2024.1371559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
Anti-PD-(L)1 therapy has shown great efficacy in some patients with cancer. However, a significant proportion of patients with cancer do not respond to it. Another unmet clinical need for anti-PD-(L)1 therapy is the dynamic monitoring of treatment effects. Therefore, identifying biomarkers that can stratify potential responders before PD-(L)1 treatment and timely monitoring of the efficacy of PD-(L)1 treatment are crucial in the clinical setting. The identification of biomarkers by liquid biopsy has attracted considerable attention. Among the identified biomarkers, circulating T cells are one of the most promising because of their indispensable contribution to anti-PD-(L)1 therapy. The present review aimed to thoroughly explore the potential of circulating T cells as biomarkers of anti-PD-(L)1 therapy and its advantages and limitations.
Collapse
Affiliation(s)
- Junlei Hou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xuezhi Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shuanglong Xie
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Haoran Zha
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| |
Collapse
|
16
|
Davoudi F, Moradi A, Sadeghirad H, Kulasinghe A. Tissue biomarkers of immune checkpoint inhibitor therapy. Immunol Cell Biol 2024; 102:179-193. [PMID: 38228572 DOI: 10.1111/imcb.12723] [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: 09/21/2023] [Revised: 12/19/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
Cancer immunotherapy has been rejuvenated by the growing understanding of the immune system's role in tumor activity over the past two decades. During cancer initiation and progression, tumor cells employ various mechanisms that resemble peripheral immune tolerance to evade the antitumor responses of the immune system. Immune checkpoint molecules are the major mechanism of immune resistance that are exploited by tumor cells to inhibit T-cell activation and suppress immune responses. The targeting of immune checkpoint pathways has led to substantial improvements in survival rates in a number of solid cancers. However, a lack of understanding of the heterogeneity of the tumor microenvironment (TME) has resulted in inefficient therapy responses. A greater understanding of the TME is needed to identify patients likely to respond, and those that will have resistance to immune checkpoint inhibitors (ICIs). Advancement in spatial single-cell technologies has allowed deeper insight into the phenotypic and functional diversities of cells in the TME. In this review, we provide an overview of ICI biomarkers and highlight how high-dimensional spatially resolved, single-cell approaches provide deep molecular insights into the TME and allow for the discovery of biomarkers of clinical benefit.
Collapse
Affiliation(s)
- Fatemeh Davoudi
- Department of Medical Genetics, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Afshin Moradi
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Habib Sadeghirad
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
17
|
Kiem D, Ocker M, Greil R, Neureiter D, Melchardt T. Enhancing anti-CD274 (PD-L1) targeting through combinatorial immunotherapy with bispecific antibodies and fusion proteins: from preclinical to phase II clinical trials. Expert Opin Investig Drugs 2024; 33:229-242. [PMID: 38354028 DOI: 10.1080/13543784.2024.2319317] [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: 12/03/2023] [Accepted: 02/12/2024] [Indexed: 02/24/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitors have achieved great success in the treatment of many different types of cancer. Programmed cell death protein ligand 1 (PD-L1, CD274) is a major immunosuppressive immune checkpoint and a target for several already approved monoclonal antibodies. Despite this, novel strategies are under development, as the overall response remains low. AREAS COVERED In this review, an overview of the current biomarkers for response to PD-L1 inhibitor treatment is given, followed by a discussion of potential novel biomarkers, including tumor mutational burden and circulating tumor DNA. Combinatorial immunotherapy is a potential novel strategy to increase the response to PD-L1 inhibitor treatment and currently, several interesting bispecific antibodies as well as bispecific fusion proteins are undergoing early clinical investigation. We focus on substances targeting PD-L1 and a secondary target, and a secondary immunomodulatory target like CTLA-4, TIGIT, or CD47. EXPERT OPINION Overall, the presented studies show anti-tumor activity of these combinatorial immunotherapeutic approaches. However, still relatively low response rates suggest a need for better biomarkers.
Collapse
Affiliation(s)
- Dominik Kiem
- III Medical Department, Paracelsus Medical University, Salzburg, Austria
| | - Matthias Ocker
- Medical Department, Division of Hematology, Oncology, and Cancer Immunology, Campus, Charité Mitte, Charité University Medicine Berlin, Berlin, Germany
- EO Translational Insights Consulting GmbH, Berlin, Germany
- Tacalyx GmbH, Berlin, Germany
| | - Richard Greil
- III Medical Department, Paracelsus Medical University, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Daniel Neureiter
- Cancer Cluster Salzburg, Salzburg, Austria
- Institute of Pathology, Paracelsus Medical University, University Hospital Salzburg (SALK), Salzburg, Austria
| | - Thomas Melchardt
- III Medical Department, Paracelsus Medical University, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
18
|
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
|
19
|
Spehner L, Orillard E, Falcoz A, Lepiller Q, Bouard A, Almotlak H, Kim S, Curtit E, Meynard G, Jary M, Nardin C, Asgarov K, Abdeljaoued S, Chartral U, Mougey V, Ben Khelil M, Lopez M, Loyon R, Vernerey D, Adotevi O, Borg C, Mansi L, Kroemer M. Predictive biomarkers and specific immune responses of COVID-19 mRNA vaccine in patients with cancer: prospective results from the CACOV-VAC trial. BMJ ONCOLOGY 2023; 2:e000054. [PMID: 39886486 PMCID: PMC11235023 DOI: 10.1136/bmjonc-2023-000054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 10/17/2023] [Indexed: 02/01/2025]
Abstract
Objective Vaccinated patients with cancer in follow-up studies showed a high seropositivity rate but impaired antibody titres and T cell responses following mRNA vaccine against COVID-19. Besides clinical characteristics and the type of anticancer treatment before vaccination, the identification of patients susceptible to non-response following vaccination using immunological markers is worth to be investigated. Methods and analysis All patients (n=138, solid cancers) were included in the CACOV-VAC Study comprising three cohorts ((neo)-adjuvant, metastatic and surveillance). Immune responses were assessed using, respectively, anti-receptor-binding domain (RBD) SARS-CoV-S-IgG assay and interferon-γ ELISpot assay 3 months following the prime vaccination dose. Immunophenotyping of T cells and immunosuppressive cells from peripheral blood was performed before the prime dose. The serological threshold 3563 AU/mL was used to discriminate non-responders or suboptimal responders versus responders. Results Most patients achieved seroconversion after receiving the two doses of vaccine (97.6%). The median serological level of anti-RBD SARS-CoV-S-IgG was equal to 3029 for patients at the metastatic stage. The patient's age was the main demographic characteristic that influenced vaccine efficacy. Among the immunological parameters measured at baseline, lower TIGIT (T cell immunoreceptor with Ig and ITIM domains) expression on CD8 T cells was associated with a better vaccine immunogenicity both in terms of humoral and cellular immune responses. Conclusion Despite a high seroconversion rate, median serological levels of patients with cancer, particularly elderly patients, were below the threshold equal to 3563 AU/mL considered as a humoral correlate of protection against SARS-CoV-2. Our findings suggest that the inhibitory receptor TIGIT might be an interesting predictive biomarker of COVID-19 vaccine immunogenicity and beyond in an anticancer vaccine context. Trial registration number ClinicalTrials.gov Registry (NCT04836793).
Collapse
Affiliation(s)
- Laurie Spehner
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Emeline Orillard
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Antoine Falcoz
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Methodology and Quality of Life Unit in Oncology, CHU Besançon, Besançon, France
| | | | - Adeline Bouard
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- ITAC Platform, University of Franche-Comté, Besançon, France
| | - Hamadi Almotlak
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Stefano Kim
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Elsa Curtit
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | | | - Marine Jary
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Medical Oncology, Hôpital Jean Minjoz, Besançon, France
| | - Charlee Nardin
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Dermatology, CHU Besançon, Besançon, France
| | - Kamal Asgarov
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- ITAC Platform, University of Franche-Comté, Besançon, France
| | - Syrine Abdeljaoued
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Ugo Chartral
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Virginie Mougey
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Myriam Ben Khelil
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Morgane Lopez
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Romain Loyon
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Dewi Vernerey
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Methodology and Quality of Life Unit in Oncology, CHU Besançon, Besançon, France
| | - Olivier Adotevi
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Christophe Borg
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
- ITAC Platform, University of Franche-Comté, Besançon, France
| | - Laura Mansi
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Marie Kroemer
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- ITAC Platform, University of Franche-Comté, Besançon, France
- Department of Pharmacy, University Hospital Centre Besançon, Besançon, France
| |
Collapse
|
20
|
van Eijs MJM, Verheijden RJ, van der Wees SA, Nierkens S, van Lindert ASR, Suijkerbuijk KPM, van Wijk F. Toxicity-specific peripheral blood T and B cell dynamics in anti-PD-1 and combined immune checkpoint inhibition. Cancer Immunol Immunother 2023; 72:4049-4064. [PMID: 37794264 PMCID: PMC10700442 DOI: 10.1007/s00262-023-03541-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 09/03/2023] [Indexed: 10/06/2023]
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized the treatment landscape of advanced malignancies, but come with a diverse spectrum of immune-related adverse events (irAEs). Mechanistic studies can aid the transition from expert-opinion to evidence-based irAE treatment strategies. We aimed to longitudinally characterize peripheral blood T and B cell dynamics in ICI-treated patients by multicolor flow cytometry and serum multiplex immunoassay at baseline, ± 3 weeks and ± 6 weeks or upon clinically relevant irAEs. We analyzed samples from 44 ICI-treated patients (24 anti-PD-1 monotherapy, 20 combined anti-PD-1/anti-CTLA-4; cICI), of whom 21 developed irAEs, and 10 healthy donors. IrAEs after cICI were characterized by significantly enhanced proliferation of Th1-associated, mainly (CD4+) CD27- effector memory T cells, as well as Th17-associated immune responses and germinal center activation (reflected by CXCL13 and IL-21 increases). We observed no changes in CD21lo, memory, class-switched or newly activated B cell subsets. Particularly double-positive PD-1+LAG-3+ CD8+ T cells showed enhanced cytotoxic capacity in patients with irAEs after cICI. Within anti-PD-1 monotherapy, irAEs were associated with modestly enhanced Th1-associated responses reflected by increased serum CXCL9 and CXCL10. In conclusion, ICI-induced toxicity is dominated by enhanced Th1-associated responses, but in cICI we also found evidence for Th17-associated responses and germinal center activation. Together, our data add to the growing body of evidence that irAEs may be driven by newly activated CD4+ helper T cells, specifically after cICI. This study also supports tailored irAE treatment, based on ICI regimen, and to deploy specific strategies such as Th17 inhibition especially in cICI-associated irAEs.
Collapse
Affiliation(s)
- Mick J M van Eijs
- Department of Medical Oncology, University Medical Center Utrecht, KC.02.085.2, P.O. Box 85090, 3508 AB, Utrecht, the Netherlands.
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Rik J Verheijden
- Department of Medical Oncology, University Medical Center Utrecht, KC.02.085.2, P.O. Box 85090, 3508 AB, Utrecht, the Netherlands
| | - Stefanie A van der Wees
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Stefan Nierkens
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Anne S R van Lindert
- Department of Pulmonology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, KC.02.085.2, P.O. Box 85090, 3508 AB, Utrecht, the Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
21
|
Luo H, Wang W, Mai J, Yin R, Cai X, Li Q. The nexus of dynamic T cell states and immune checkpoint blockade therapy in the periphery and tumor microenvironment. Front Immunol 2023; 14:1267918. [PMID: 37881432 PMCID: PMC10597640 DOI: 10.3389/fimmu.2023.1267918] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/18/2023] [Indexed: 10/27/2023] Open
Abstract
Immune checkpoint blockade (ICB) therapies, that is, using monoclonal antibodies to reinvigorate tumor-reactive, antigen-specific T cells from the inhibitory effects of CTLA-4, PD-1 and PD-L1 immune checkpoints, have revolutionized the therapeutic landscape of modern oncology. However, only a subset of patients can benefit from the ICB therapy. Biomarkers associated with ICB response, resistance and prognosis have been subjected to intensive research in the past decade. Early studies focused on the analysis of tumor specimens and their residing microenvironment. However, biopsies can be challenging to obtain in clinical practice, and do not reflect the dynamic changes of immunological parameters during the ICB therapy. Recent studies have investigated profiles of antigen-specific T cells derived from the peripheral compartment using multi-omics approaches. By tracking the clonotype and diversity of tumor-reactive T cell receptor repertoire, these studies collectively establish that de novo priming of antigen-specific T cells in peripheral blood occurs throughout the course of ICB, whereas preexisting T cells prior to ICB are exhausted to various degrees. Here, we review what is known about ICB-induced T cell phenotypic and functional changes in cancer patients both within the tumor microenvironment and in the peripheral compartment. A better understanding of parameters influencing the response to ICBs will provide rationales for developing novel diagnostics and combinatorial therapeutic strategies to maximize the clinical efficacies of ICB therapies.
Collapse
Affiliation(s)
- Hong Luo
- Department of Obstetrics & Gynecology, Laboratory Medicine and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenxiang Wang
- Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, China
| | - Jia Mai
- Department of Obstetrics & Gynecology, Laboratory Medicine and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rutie Yin
- Department of Obstetrics & Gynecology, Laboratory Medicine and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuyu Cai
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qintong Li
- Department of Obstetrics & Gynecology, Laboratory Medicine and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
22
|
Fitzgerald KJ, Schoenfeld JD. Radiotherapy Dose in Patients Receiving Immunotherapy. Semin Radiat Oncol 2023; 33:327-335. [PMID: 37331787 DOI: 10.1016/j.semradonc.2023.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
There is significant rationale for combining radiation therapy (RT) and immuno-oncology (IO) agents, but the optimal radiation parameters are unknown. This review summarizes key trials in the RT and IO space with a focus on RT dose. Very low RT doses solely modulate the tumor immune microenvironment, intermediate doses both modulate the tumor immune microenvironment and kill some fraction of tumor cells, and ablative doses eliminate the majority of target tumor cells and also possess immunomodulatory effects. Ablative RT doses may have high toxicity if targets are adjacent to radiosensitive normal organs. The majority of completed trials have been conducted in the setting of metastatic disease and direct RT to a single lesion with the goal of generating systemic antitumor immunity termed the abscopal effect. Unfortunately, reliable generation of an abscopal effect has proved elusive over a range of radiation doses. Newer trials are exploring the effects of delivering RT to all or most sites of metastatic disease, with dose personalization based on the number and location of lesions. Additional directions include testing RT and IO in earlier stages of disease, sometimes in further combination with chemotherapy and surgery, where lower doses of RT may still contribute substantially to pathologic responses.
Collapse
|
23
|
Chu X, Tian W, Wang Z, Zhang J, Zhou R. Co-inhibition of TIGIT and PD-1/PD-L1 in Cancer Immunotherapy: Mechanisms and Clinical Trials. Mol Cancer 2023; 22:93. [PMID: 37291608 DOI: 10.1186/s12943-023-01800-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/02/2023] [Indexed: 06/10/2023] Open
Abstract
Over the past decade, immune checkpoint inhibitors (ICIs) have emerged as a revolutionary cancer treatment modality, offering long-lasting responses and survival benefits for a substantial number of cancer patients. However, the response rates to ICIs vary significantly among individuals and cancer types, with a notable proportion of patients exhibiting resistance or showing no response. Therefore, dual ICI combination therapy has been proposed as a potential strategy to address these challenges. One of the targets is TIGIT, an inhibitory receptor associated with T-cell exhaustion. TIGIT has diverse immunosuppressive effects on the cancer immunity cycle, including the inhibition of natural killer cell effector function, suppression of dendritic cell maturation, promotion of macrophage polarization to the M2 phenotype, and differentiation of T cells to regulatory T cells. Furthermore, TIGIT is linked with PD-1 expression, and it can synergize with PD-1/PD-L1 blockade to enhance tumor rejection. Preclinical studies have demonstrated the potential benefits of co-inhibition of TIGIT and PD-1/PD-L1 in enhancing anti-tumor immunity and improving treatment outcomes in several cancer types. Several clinical trials are underway to evaluate the safety and efficacy of TIGIT and PD-1/PD-L1 co-inhibition in various cancer types, and the results are awaited. This review provides an overview of the mechanisms of TIGIT and PD-1/PD-L1 co-inhibition in anti-tumor treatment, summarizes the latest clinical trials investigating this combination therapy, and discusses its prospects. Overall, co-inhibition of TIGIT and PD-1/PD-L1 represents a promising therapeutic approach for cancer treatment that has the potential to improve the outcomes of cancer patients treated with ICIs.
Collapse
Affiliation(s)
- Xianjing Chu
- Department of Oncology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, China
| | - Wentao Tian
- Department of Oncology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, China
| | - Ziqi Wang
- Department of Oncology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, China
| | - Jing Zhang
- Department of Oncology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, China
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P.R. China.
| |
Collapse
|
24
|
Chen YL, Ng JSW, Ottakandathil Babu R, Woo J, Nahler J, Hardman CS, Kurupati P, Nussbaum L, Gao F, Dong T, Ladell K, Price DA, Duncan DA, Johnson D, Gileadi U, Koohy H, Ogg GS. Group A Streptococcus induces CD1a-autoreactive T cells and promotes psoriatic inflammation. Sci Immunol 2023; 8:eadd9232. [PMID: 37267382 PMCID: PMC7615662 DOI: 10.1126/sciimmunol.add9232] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 04/26/2023] [Indexed: 06/04/2023]
Abstract
Group A Streptococcus (GAS) infection is associated with multiple clinical sequelae, including different subtypes of psoriasis. Such post-streptococcal disorders have been long known but are largely unexplained. CD1a is expressed at constitutively high levels by Langerhans cells and presents lipid antigens to T cells, but the potential relevance to GAS infection has not been studied. Here, we investigated whether GAS-responsive CD1a-restricted T cells contribute to the pathogenesis of psoriasis. Healthy individuals had high frequencies of circulating and cutaneous GAS-responsive CD4+ and CD8+ T cells with rapid effector functions, including the production of interleukin-22 (IL-22). Human skin and blood single-cell CITE-seq analyses of IL-22-producing T cells showed a type 17 signature with proliferative potential, whereas IFN-γ-producing T cells displayed cytotoxic T lymphocyte characteristics. Furthermore, individuals with psoriasis had significantly higher frequencies of circulating GAS-reactive T cells, enriched for markers of activation, cytolytic potential, and tissue association. In addition to responding to GAS, subsets of expanded GAS-reactive T cell clones/lines were found to be autoreactive, which included the recognition of the self-lipid antigen lysophosphatidylcholine. CD8+ T cell clones/lines produced cytolytic mediators and lysed infected CD1a-expressing cells. Furthermore, we established cutaneous models of GAS infection in a humanized CD1a transgenic mouse model and identified enhanced and prolonged local and systemic inflammation, with resolution through a psoriasis-like phenotype. Together, these findings link GAS infection to the CD1a pathway and show that GAS infection promotes the proliferation and activation of CD1a-autoreactive T cells, with relevance to post-streptococcal disease, including the pathogenesis and treatment of psoriasis.
Collapse
Affiliation(s)
- Yi-Ling Chen
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Jessica Soo Weei Ng
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Rosana Ottakandathil Babu
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Jeongmin Woo
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Janina Nahler
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Clare S Hardman
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Prathiba Kurupati
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Lea Nussbaum
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Fei Gao
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- CAMS-Oxford International Centre for Translational Immunology, University of Oxford, Oxford, UK
| | - Tao Dong
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- CAMS-Oxford International Centre for Translational Immunology, University of Oxford, Oxford, UK
| | - Kristin Ladell
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - David A Price
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - David A Duncan
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK
| | - David Johnson
- Department of Plastic and Reconstructive Surgery, John Radcliffe Hospital, Oxford University Hospitals National Health Services Foundation Trust, Oxford, UK
| | - Uzi Gileadi
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Hashem Koohy
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Alan Turing Fellow in Health and Medicine, Oxford, UK
| | - Graham S Ogg
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- CAMS-Oxford International Centre for Translational Immunology, University of Oxford, Oxford, UK
| |
Collapse
|
25
|
Patel AJ, Middleton GW. TIGIT-based immunotherapeutics in lung cancer. IMMUNOTHERAPY ADVANCES 2023; 3:ltad009. [PMID: 37325585 PMCID: PMC10266577 DOI: 10.1093/immadv/ltad009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
In this review, we explore the biology of the TIGIT checkpoint and its potential as a therapeutic target in lung cancer. We briefly review a highly selected set of clinical trials that have reported or are currently recruiting in non-small cell and small cell lung cancer, a disease transformed by the advent of PD-1/PD-L1 checkpoint blockade immunotherapy. We explore the murine data underlying TIGIT blockade and further explore the reliance of effective anti-TIGIT therapy on DNAM-1(CD226)-positive activated effector CD8+ T cells. The synergism with anti-PD-1 therapy is also explored. Future directions in the realm of overcoming resistance to checkpoint blockade and extending the repertoire of other checkpoints are also briefly explored.
Collapse
Affiliation(s)
- Akshay J Patel
- Correspondence: Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, B15 2TT, Birmingham, UK.
| | - Gary W Middleton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Department of Medical Oncology, University Hospitals Birmingham, Birmingham, UK
| |
Collapse
|
26
|
Ziogas DC, Theocharopoulos C, Lialios PP, Foteinou D, Koumprentziotis IA, Xynos G, Gogas H. Beyond CTLA-4 and PD-1 Inhibition: Novel Immune Checkpoint Molecules for Melanoma Treatment. Cancers (Basel) 2023; 15:2718. [PMID: 37345056 PMCID: PMC10216291 DOI: 10.3390/cancers15102718] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
More than ten years after the approval of ipilimumab, immune checkpoint inhibitors (ICIs) against PD-1 and CTLA-4 have been established as the most effective treatment for locally advanced or metastatic melanoma, achieving durable responses either as monotherapies or in combinatorial regimens. However, a considerable proportion of patients do not respond or experience early relapse, due to multiple parameters that contribute to melanoma resistance. The expression of other immune checkpoints beyond the PD-1 and CTLA-4 molecules remains a major mechanism of immune evasion. The recent approval of anti-LAG-3 ICI, relatlimab, in combination with nivolumab for metastatic disease, has capitalized on the extensive research in the field and has highlighted the potential for further improvement of melanoma prognosis by synergistically blocking additional immune targets with new ICI-doublets, antibody-drug conjugates, or other novel modalities. Herein, we provide a comprehensive overview of presently published immune checkpoint molecules, including LAG-3, TIGIT, TIM-3, VISTA, IDO1/IDO2/TDO, CD27/CD70, CD39/73, HVEM/BTLA/CD160 and B7-H3. Beginning from their immunomodulatory properties as co-inhibitory or co-stimulatory receptors, we present all therapeutic modalities targeting these molecules that have been tested in melanoma treatment either in preclinical or clinical settings. Better understanding of the checkpoint-mediated crosstalk between melanoma and immune effector cells is essential for generating more effective strategies with augmented immune response.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Helen Gogas
- First Department of Medicine, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.T.); (P.-P.L.); (D.F.); (I.-A.K.); (G.X.)
| |
Collapse
|
27
|
Noel S, Lee K, Gharaie S, Kurzhagen JT, Pierorazio PM, Arend LJ, Kuchroo VK, Cahan P, Rabb H. Immune Checkpoint Molecule TIGIT Regulates Kidney T Cell Functions and Contributes to AKI. J Am Soc Nephrol 2023; 34:755-771. [PMID: 36747315 PMCID: PMC10125646 DOI: 10.1681/asn.0000000000000063] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 12/02/2022] [Indexed: 01/22/2023] Open
Abstract
SIGNIFICANCE STATEMENT T cells mediate pathogenic and reparative processes during AKI, but the exact mechanisms regulating kidney T cell functions are unclear. This study identified upregulation of the novel immune checkpoint molecule, TIGIT, on mouse and human kidney T cells after AKI. TIGIT-expressing kidney T cells produced proinflammatory cytokines and had effector (EM) and central memory (CM) phenotypes. TIGIT-deficient mice had protection from both ischemic and nephrotoxic AKI. Single-cell RNA sequencing led to the discovery of possible downstream targets of TIGIT. TIGIT mediates AKI pathophysiology, is a promising novel target for AKI therapy, and is being increasingly studied in human cancer therapy trials. BACKGROUND T cells play pathogenic and reparative roles during AKI. However, mechanisms regulating T cell responses are relatively unknown. We investigated the roles of the novel immune checkpoint molecule T cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) in kidney T cells and AKI outcomes. METHODS TIGIT expression and functional effects were evaluated in mouse kidney T cells using RNA sequencing (RNA-Seq) and flow cytometry. TIGIT effect on AKI outcomes was studied with TIGIT knockout (TIGIT-KO) mice in ischemia reperfusion (IR) and cisplatin AKI models. Human kidney T cells from nephrectomy samples and single cell RNA sequencing (scRNA-Seq) data from the Kidney Precision Medicine Project were used to assess TIGIT's role in humans. RESULTS RNA-Seq and flow cytometry analysis of mouse kidney CD4+ T cells revealed increased expression of TIGIT after IR injury. Ischemic injury also increased TIGIT expression in human kidney T cells, and TIGIT expression was restricted to T/natural killer cell subsets in patients with AKI. TIGIT-expressing kidney T cells in wild type (WT) mice had an effector/central memory phenotype and proinflammatory profile at baseline and post-IR. Kidney regulatory T cells were predominantly TIGIT+ and significantly reduced post-IR. TIGIT-KO mice had significantly reduced kidney injury after IR and nephrotoxic injury compared with WT mice. scRNA-Seq analysis showed enrichment of genes related to oxidative phosphorylation and mTORC1 signaling in Th17 cells from TIGIT-KO mice. CONCLUSIONS TIGIT expression increases in mouse and human kidney T cells during AKI, worsens AKI outcomes, and is a novel therapeutic target for AKI.
Collapse
Affiliation(s)
- Sanjeev Noel
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Kyungho Lee
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Sepideh Gharaie
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | | | - Philip M. Pierorazio
- Department of Surgery, Division of Urology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lois J. Arend
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Vijay K. Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Patrick Cahan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Department of Molecular Biology & Genetics, Johns Hopkins University, Baltimore, Maryland
| | - Hamid Rabb
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
28
|
Geng R, Tang H, You T, Xu X, Li S, Li Z, Liu Y, Qiu W, Zhou N, Li N, Ge Y, Guo F, Sun Y, Wang Y, Li T, Bai C. Peripheral CD8+CD28+ T lymphocytes predict the efficacy and safety of PD-1/PD-L1 inhibitors in cancer patients. Front Immunol 2023; 14:1125876. [PMID: 36969245 PMCID: PMC10038730 DOI: 10.3389/fimmu.2023.1125876] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
BackgroundProgrammed cell death protein-1/programmed cell death ligand-1 (PD-1/PD-L1) inhibitors works by reactivating immune cells. Considering the accessibility of noninvasive liquid biopsies, it is advisable to employ peripheral blood lymphocyte subsets to predict immunotherapy outcomes.MethodsWe retrospectively enrolled 87 patients with available baseline circulating lymphocyte subset data who received first-line PD-1/PD-L1 inhibitors at Peking Union Medical College Hospital between May 2018 and April 2022. Immune cell counts were determined by flow cytometry.ResultsPatients who responded to PD-1/PD-L1 inhibitors had significantly higher circulating CD8+CD28+ T-cell counts (median [range] count: 236 [30-536] versus 138 [36-460]/μL, p < 0.001). Using 190/μL as the cutoff value, the sensitivity and specificity of CD8+CD28+ T cells for predicting immunotherapy response were 0.689 and 0.714, respectively. Furthermore, the median progression-free survival (PFS, not reached versus 8.7 months, p < 0.001) and overall survival (OS, not reached versus 16.2 months, p < 0.001) were significantly longer in the patients with higher CD8+CD28+ T-cell counts. However, the CD8+CD28+ T-cell level was also associated with the incidence of grade 3-4 immune-related adverse events (irAEs). The sensitivity and specificity of CD8+CD28+ T cells for predicting irAEs of grade 3-4 were 0.846 and 0.667, respectively, at the threshold of CD8+CD28+ T cells ≥ 309/μL.ConclusionsHigh circulating CD8+CD28+ T-cell levels is a potential biomarker for immunotherapy response and better prognosis, while excessive CD8+CD28+ T cells (≥ 309/μL) may also indicate the emergence of severe irAEs.
Collapse
Affiliation(s)
- Ruixuan Geng
- Department of International Medical Services, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Tang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tingting You
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiuxiu Xu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sijian Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Obstetric and Gynecologic Diseases, Beijing, China
| | - Zepeng Li
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Liu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Qiu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Zhou
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ningning Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuping Ge
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fuping Guo
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuhong Sun
- Department of Radiation Oncology, Dandong First Hospital, Dandong, Liaoning, China
| | - Yingyi Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yingyi Wang, ; Taisheng Li,
| | - Taisheng Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yingyi Wang, ; Taisheng Li,
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
29
|
Guo Z, Zhang K, Wei X, Li Y, Ma X, Li Y, Han D, Du Q, Zhang T, Chen X, Wei H, Yan C, Zhang W, Pang Q, Wang P. Radiotherapy plus camrelizumab affects peripheral CD8 T-cell differentiation subsets expressing PD-1, TIGIT, and CTLA-4 in esophageal squamous cell carcinoma. J Leukoc Biol 2023; 113:11-17. [PMID: 36822161 DOI: 10.1093/jleuko/qiac018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Indexed: 01/11/2023] Open
Abstract
Our previous phase Ib trial (NCT03222440) showed that radiotherapy plus the anti-PD-1 antibody camrelizumab is a safe and feasible first-line therapy for locally advanced esophageal squamous cell carcinoma. In this study, we divided peripheral CD8 T-cell differentiation subsets into 4 subpopulations (naive T cells, central memory T cells, effector memory T cells, and CD45RA+ effector memory T cells). We then investigated the influence of radiotherapy plus camrelizumab therapy on the proportions of the 4 subsets and their PD-1, TIGIT, and CTLA-4 expression as well as their proliferative activity and compared the effects with those of concurrent chemoradiotherapy. Nineteen and 15 patients with esophageal squamous cell carcinoma who received radiotherapy plus camrelizumab therapy and concurrent chemoradiotherapy, respectively, were enrolled in this study. We isolated peripheral blood mononuclear cells from these patients before treatment and longitudinally after the delivery of 40 Gy radiotherapy. Flow cytometry was conducted to detect peripheral CD8 T-cell subsets and PD-1, TIGIT, CTLA-4, and Ki67 expression levels in patients with esophageal squamous cell carcinoma. We found that radiotherapy plus camrelizumab therapy did not change the proportions of the 4 subsets or the expression of CTLA-4, but this therapy decreased PD-1 expression by the 4 subsets and TIGIT expression by effector memory T cells, as well as significantly enhanced the proliferative activity of CD8 T cells, whereas concurrent chemoradiotherapy produced different effects. In addition, we further identified peripheral biomarkers that potentially predict the outcome of radiotherapy plus camrelizumab therapy.
Collapse
Affiliation(s)
- Zhoubo Guo
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Kunning Zhang
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Xiaoying Wei
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Yanqi Li
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Xiaoxue Ma
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Yang Li
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Dong Han
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Qingwu Du
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Tian Zhang
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Xi Chen
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Hui Wei
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Wencheng Zhang
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Qingsong Pang
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Ping Wang
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| |
Collapse
|
30
|
Colligan SH, Amitrano AM, Zollo RA, Peresie J, Kramer ED, Morreale B, Barbi J, Singh PK, Yu H, Wang J, Opyrchal M, Sykes DB, Nemeth MJ, Abrams SI. Inhibiting the biogenesis of myeloid-derived suppressor cells enhances immunotherapy efficacy against mammary tumor progression. J Clin Invest 2022; 132:e158661. [PMID: 36453551 PMCID: PMC9711879 DOI: 10.1172/jci158661] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 10/05/2022] [Indexed: 12/05/2022] Open
Abstract
While immune checkpoint inhibitors (ICIs) have transformed the therapeutic landscape in oncology, they are effective in select subsets of patients. Efficacy may be limited by tumor-driven immune suppression, of which 1 key mechanism is the development of myeloid-derived suppressor cells (MDSCs). A fundamental gap in MDSC therapeutics is the lack of approaches that target MDSC biogenesis. We hypothesized that targeting MDSC biogenesis would mitigate MDSC burden and bolster tumor responses to ICIs. We tested a class of agents, dihydroorotate dehydrogenase (DHODH) inhibitors, that have been previously shown to restore the terminal differentiation of leukemic myeloid progenitors. DHODH inhibitors have demonstrated preclinical safety and are under clinical study for hematologic malignancies. Using mouse models of mammary cancer that elicit robust MDSC responses, we demonstrated that the DHODH inhibitor brequinar (a) suppressed MDSC production from early-stage myeloid progenitors, which was accompanied by enhanced myeloid maturation; (b) augmented the antitumor and antimetastatic activities of programmed cell death 1-based (PD-1-based) ICI therapy in ICI-resistant mammary cancer models; and (c) acted in concert with PD-1 blockade through modulation of MDSC and CD8+ T cell responses. Moreover, brequinar facilitated myeloid maturation and inhibited immune-suppressive features in human bone marrow culture systems. These findings advance the concept of MDSC differentiation therapy in immuno-oncology.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Joseph Barbi
- Department of Immunology
- Department of Thoracic Surgery
| | | | - Han Yu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Mateusz Opyrchal
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - David B. Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | | | | |
Collapse
|
31
|
Pawłowska A, Skiba W, Suszczyk D, Kuryło W, Jakubowicz-Gil J, Paduch R, Wertel I. The Dual Blockade of the TIGIT and PD-1/PD-L1 Pathway as a New Hope for Ovarian Cancer Patients. Cancers (Basel) 2022; 14:5757. [PMID: 36497240 PMCID: PMC9740841 DOI: 10.3390/cancers14235757] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
The prognosis for ovarian cancer (OC) patients is poor and the five-year survival rate is only 47%. Immune checkpoints (ICPs) appear to be the potential targets in up-and-coming OC treatment. However, the response of OC patients to immunotherapy based on programmed cell death pathway (PD-1/PD-L1) inhibitors totals only 6-15%. The promising approach is a combined therapy, including other ICPs such as the T-cell immunoglobulin and ITIM domain/CD155/DNAX accessory molecule-1 (TIGIT/CD155/DNAM-1) axis. Preclinical studies in a murine model of colorectal cancer showed that the dual blockade of PD-1/PD-L1 and TIGIT led to remission in the whole studied group vs. the regression of the tumors with the blockade of a single pathway. The approach stimulates the effector activity of T cells and NK cells, and redirects the immune system activity against the tumor. The understanding of the synergistic action of the TIGIT and PD-1/PD-L1 blockade is, however, poor. Thus, the aim of this review is to summarize the current knowledge about the mode of action of the dual TIGIT and PD-1/PD-L1 blockade and its potential benefits for OC patients. Considering the positive impact of this combined therapy in malignancies, including lung and colorectal cancer, it appears to be a promising approach in OC treatment.
Collapse
Affiliation(s)
- Anna Pawłowska
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Wiktoria Skiba
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Dorota Suszczyk
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Weronika Kuryło
- Students’ Scientific Association, Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Joanna Jakubowicz-Gil
- Department of Functional Anatomy and Cytobiology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Roman Paduch
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| |
Collapse
|
32
|
Wei H, Li Y, Guo Z, Ma X, Li Y, Wei X, Han D, Zhang T, Chen X, Yan C, Zhou J, Pang Q, Wang P, Zhang W. Comparison of dynamic changes in the peripheral CD8 + T cells function and differentiation in ESCC patients treated with radiotherapy combined with anti-PD-1 antibody or concurrent chemoradiotherapy. Front Immunol 2022; 13:1060695. [PMID: 36479110 PMCID: PMC9720318 DOI: 10.3389/fimmu.2022.1060695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
Objective The systematic immune status of cancer patients undergoing immunotherapy is little known. We prospectively identified the function and differentiation traits of peripheral CD8+ T cells based on our phase 1b clinical trial (NCT03222440) of radiotherapy combined with camrelizumab in patients with locally advanced esophageal squamous cell carcinoma (ESCC) and compared it with concurrent chemoradiotherapy (CCRT). Methods 19 and 18 patients were included in the cohort of radiotherapy plus camrelizumab and cohort of CCRT treatment. By using flow cytometry, we evaluated the expression levels of PD-1, Eomes, T-bet and IFN-γ (function), CD38 and HLA-DR (activation), and differentiation subsets classified according to the expression levels of CD45RA and CD62L in peripheral CD8+ T cells before and during treatment. Results Effective binding of anti-PD-1 antibody camrelizumab with PD-1 on CD8+ T cells was detected during treatment. Both two treatments elevated the expression levels of activation molecules CD38 and HLA-DR on CD8+ T cells. PD-1+CD8+ T cells had more activation features than PD-1-CD8+ T cells in two groups and the treatments did not alter these differences. The two treatments activated both PD-1+ and PD-1- CD8+ T cells. PD-1+CD8+ T cells had less Naïve and TEMRA but more Tcm and Tem than PD-1-CD8+ T cells in two groups and both two treatments changed the ratio of memory T cells in PD-1+ and PD-1- cells. RT plus camrelizumab treatment reduced Naïve T cells and TEMRA subsets both in PD-1+ and PD-1- CD8+ T cells while elevated Tcm subset in PD-1+CD8+ T cells and Tem subset in PD-1-CD8+ T cells. CCRT elevated Tcm subset and reduced TEMRA subset in PD-1-CD8+ T cells while did not change any subset in PD-1+CD8+ T cells. Furthermore, patients undergoing radiotherapy plus immunotherapy were found to obtain better prognosis than those receiving CCRT. Conclusions This study identified the dynamic changes of systematic immune status of patients undergoing treatment. The two treatments had similar activation effects on peripheral CD8+ T cells with different PD-1 properties but had different effects on their differentiation status. These results provided potential clues to the reasons underlying the difference in prognosis of the two treatments.
Collapse
Affiliation(s)
- Hui Wei
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yanqi Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Zhoubo Guo
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xiaoxue Ma
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yang Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xiaoying Wei
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Dong Han
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Tian Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xi Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jiahuan Zhou
- Department of Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Qingsong Pang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,*Correspondence: Qingsong Pang, ; Ping Wang, ; Wencheng Zhang,
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,*Correspondence: Qingsong Pang, ; Ping Wang, ; Wencheng Zhang,
| | - Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,*Correspondence: Qingsong Pang, ; Ping Wang, ; Wencheng Zhang,
| |
Collapse
|
33
|
Turner CN, Mullins GN, Hoyer KK. CXCR5 +CD8 T cells: Potential immunotherapy targets or drivers of immune-mediated adverse events? Front Med (Lausanne) 2022; 9:1034764. [PMID: 36314014 PMCID: PMC9606409 DOI: 10.3389/fmed.2022.1034764] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/23/2022] [Indexed: 11/15/2022] Open
Abstract
CXCR5+CD8 T cells have attracted significant interest within multiple areas of immunology, cancer, and infection. This is in part due to their apparent dual functionality. These cells perform as cytotoxic cells in a variety of infection states including LCMV, HBV, HIV and SIV. However, CXCR5+CD8 T cells also associate with B cells in peripheral organs and function to stimulate B cell proliferation, antibody/B cell receptor class-switch, and antibody production. CXCR5+CD8 T cells are similar to CXCR5+CD4 T follicular helpers in their genetic make-up, B cell interactions, and functionality despite possessing elevated programmed cell death 1 and cytotoxic proteins. Within cancer CXCR5+CD8 T cells have risen as potential prognostic markers for overall survival and are functionally cytotoxic within tumor microenvironments. In inflammatory disease and autoimmunity, CXCR5+CD8 T cells are implicated in disease progression. During viral infection and cancer, CXCR5 expression on CD8 T cells generally is indicative of progenitor memory stem-like exhausted cells, which are more responsive to immune checkpoint blockade therapy. The use of immune checkpoint inhibitors to overcome immune exhaustion in cancer, and subsequent consequence of immune adverse events, highlights the dual nature of the cellular immune response. This review will detail the functionality of CXCR5+CD8 T cells in cancer and autoimmunity with potential repercussions during immune checkpoint blockade therapy discussed.
Collapse
Affiliation(s)
- Christi N. Turner
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Genevieve N. Mullins
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Katrina K. Hoyer
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States,Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States,Health Sciences Research Institute, University of California, Merced, Merced, CA, United States,*Correspondence: Katrina K. Hoyer
| |
Collapse
|
34
|
Luo Q, Fu P, Guo Y, Fu B, Guo Y, Huang Q, Huang Z, Li J. Increased TIGIT +PD‑1 +CXCR5 ‑CD4 +T cells are associated with disease activity in rheumatoid arthritis. Exp Ther Med 2022; 24:642. [PMID: 36160887 PMCID: PMC9468811 DOI: 10.3892/etm.2022.11579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/29/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Qing Luo
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Peng Fu
- School of Public Health, Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yongqin Guo
- School of Public Health, Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Biqi Fu
- Department of Rheumatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yang Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qingshui Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zikun Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Junming Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
35
|
Annese T, Tamma R, Ribatti D. Update in TIGIT Immune-Checkpoint Role in Cancer. Front Oncol 2022; 12:871085. [PMID: 35656508 PMCID: PMC9152184 DOI: 10.3389/fonc.2022.871085] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
The in-depth characterization of cross-talk between tumor cells and T cells in solid and hematological malignancies will have to be considered to develop new therapeutical strategies concerning the reactivation and maintenance of patient-specific antitumor responses within the patient tumor microenvironment. Activation of immune cells depends on a delicate balance between activating and inhibitory signals mediated by different receptors. T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) is an inhibitory receptor expressed by regulatory T cells (Tregs), activated T cells, and natural killer (NK) cells. TIGIT pathway regulates T cell-mediated tumor recognition in vivo and in vitro and represents an exciting target for checkpoint blockade immunotherapy. TIGIT blockade as monotherapy or in combination with other inhibitor receptors or drugs is emerging in clinical trials in patients with cancer. The purpose of this review is to update the role of TIGIT in cancer progression, looking at TIGIT pathways that are often upregulated in immune cells and at possible therapeutic strategies to avoid tumor aggressiveness, drug resistance, and treatment side effects. However, in the first part, we overviewed the role of immune checkpoints in immunoediting, the TIGIT structure and ligands, and summarized the key immune cells that express TIGIT.
Collapse
Affiliation(s)
- Tiziana Annese
- Department of Medicine and Surgery, Libera Università del Mediterraneo (LUM) Giuseppe Degennaro University, Bari, Italy.,Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| |
Collapse
|
36
|
Zhang J, Huang D, Saw PE, Song E. Turning cold tumors hot: from molecular mechanisms to clinical applications. Trends Immunol 2022; 43:523-545. [PMID: 35624021 DOI: 10.1016/j.it.2022.04.010] [Citation(s) in RCA: 272] [Impact Index Per Article: 90.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022]
Abstract
Immune checkpoint blockade (ICB) therapies have achieved clinical benefit, but most 'immune-cold' solid tumors are not responsive. The diversity of immune evasion mechanisms remains a key obstacle in turning nonresponsive 'cold' tumors into responsive 'hot' ones. Therefore, exploring the mechanisms of such transitions and tumor immunotyping can provide significant insights into designing effective therapeutic strategies against cancer. Here, we focus on the latest advances regarding local and systemic regulatory mechanisms of immune responses in cold and hot tumors. We also highlight the necessity for tumor immunotyping through the assessment of multiple immunological variables using various diagnostic techniques and biomarkers. Finally, we discuss the challenges and potential clinical applications of immunophenotyping to turn cold tumors hot, which may further guide combined immunotherapies.
Collapse
Affiliation(s)
- Jiahui Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Di Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
37
|
Huyghe N, Benidovskaya E, Stevens P, Van den Eynde M. Biomarkers of Response and Resistance to Immunotherapy in Microsatellite Stable Colorectal Cancer: Toward a New Personalized Medicine. Cancers (Basel) 2022; 14:2241. [PMID: 35565369 PMCID: PMC9105843 DOI: 10.3390/cancers14092241] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 11/28/2022] Open
Abstract
Immune Checkpoint Inhibitors (ICIs) are well recognized as a major immune treatment modality for multiple types of solid cancers. However, for colorectal cancer (CRC), ICIs are only approved for the treatment of Mismatch-Repair-Deficient and Microsatellite Instability-High (dMMR/MSI-H) tumors. For the vast majority of CRC, that are not dMMR/MSI-H, ICIs alone provide limited to no clinical benefit. This discrepancy of response between CRC and other solid cancers suggests that CRC may be inherently resistant to ICIs alone. In translational research, efforts are underway to thoroughly characterize the immune microenvironment of CRC to better understand the mechanisms behind this resistance and to find new biomarkers of response. In the clinic, trials are being set up to study biomarkers along with treatments targeting newly discovered immune checkpoint molecules or treatments combining ICIs with other existing therapies to improve response in MSS CRC. In this review, we will focus on the characteristics of response and resistance to ICIs in CRC, and discuss promising biomarkers studied in recent clinical trials combining ICIs with other therapies.
Collapse
Affiliation(s)
- Nicolas Huyghe
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
| | - Elena Benidovskaya
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
| | - Philippe Stevens
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
| | - Marc Van den Eynde
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
- Institut Roi Albert II, Department of Medical Oncology and Gastroenterology, Cliniques Universitaires St-Luc, 1200 Brussels, Belgium
| |
Collapse
|
38
|
Niebel D, Fröhlich A, Zarbl R, Fietz S, de Vos L, Vogt TJ, Dietrich J, Sirokay J, Kuster P, Saavedra G, Ramírez Valladolid S, Hoffmann F, Strieth S, Landsberg J, Dietrich D. DNA methylation regulates TIGIT expression within the melanoma microenvironment, is prognostic for overall survival, and predicts progression-free survival in patients treated with anti-PD-1 immunotherapy. Clin Epigenetics 2022; 14:50. [PMID: 35410311 PMCID: PMC9004005 DOI: 10.1186/s13148-022-01270-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/02/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND TIGIT is an immune checkpoint under investigation as therapeutic target. Understanding the regulation of TIGIT on an epigenetic level might support the development of companion biomarkers. METHODS We correlated TIGIT DNA methylation of single CpG sites with gene expression, signatures of immune infiltrates and interferon-γ, and survival in melanoma. We further analyzed methylation levels in immune cell subsets, melanocyte and melanoma cell lines. TIGIT expression patterns within components of the melanoma microenvironment were analyzed by single cell sequencing. We used quantitative methylation-specific PCR, flow cytometry, and immunohistochemistry for correlations between expression and methylation and to assess the effect of pharmacological demethylation of melanoma cells treated with 5-aza-2-deoxycytidine (decitabine). Finally, we investigated the association of patients' survival with TIGIT mRNA and methylation. RESULTS Depending on the sequence context of the analyzed CpG site, we found a cell type-specific TIGIT gene locus methylation pattern and significant correlations of TIGIT methylation with mRNA expression, an interferon γ signature, and distinct immune cell infiltrates, including TIGIT+ lymphocytes. We detected a melanoma cell-intrinsic TIGIT protein expression. Pharmacological demethylation of the A375 melanoma cell line led to a constitutive TIGIT expression. Low promoter flank methylation and high mRNA expression was associated with patients' prognosis and predicted progression-free survival in patients treated with anti-PD-1 immunotherapy. A high TIGIT+ lymphocyte score was associated with better progression-free survival under anti-PD-1 immunotherapy. CONCLUSIONS Our data demonstrate an epigenetic regulation of TIGIT expression via DNA methylation within the melanoma microenvironment. TIGIT DNA methylation and expression may serve as predictive biomarkers in the context of immunotherapies in melanoma.
Collapse
Affiliation(s)
- Dennis Niebel
- Department of Dermatology and Allergy, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Anne Fröhlich
- Department of Dermatology and Allergy, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Romina Zarbl
- Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Simon Fietz
- Department of Dermatology and Allergy, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany.,Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Luka de Vos
- Department of Dermatology and Allergy, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Timo J Vogt
- Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Jörn Dietrich
- Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Judith Sirokay
- Department of Dermatology and Allergy, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Pia Kuster
- Department of Dermatology and Allergy, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Gonzalo Saavedra
- Department of Dermatology and Allergy, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany.,Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Susana Ramírez Valladolid
- Department of Dermatology and Allergy, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Friederike Hoffmann
- Department of Dermatology and Allergy, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Sebastian Strieth
- Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Jennifer Landsberg
- Department of Dermatology and Allergy, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Dimo Dietrich
- Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
39
|
Wen T, Barham W, Li Y, Zhang H, Gicobi JK, Hirdler JB, Liu X, Ham H, Peterson Martinez KE, Lucien F, Lavoie RR, Li H, Correia C, Monie DD, An Z, Harrington SM, Wu X, Guo R, Dronca RS, Mansfield AS, Yan Y, Markovic SN, Park SS, Sun J, Qin H, Liu MC, Vasmatzis G, Billadeau DD, Dong H. NKG7 Is a T-cell-Intrinsic Therapeutic Target for Improving Antitumor Cytotoxicity and Cancer Immunotherapy. Cancer Immunol Res 2022; 10:162-181. [PMID: 34911739 PMCID: PMC8816890 DOI: 10.1158/2326-6066.cir-21-0539] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/19/2021] [Accepted: 12/09/2021] [Indexed: 01/22/2023]
Abstract
Cytotoxic CD8+ T cells (CTL) are a crucial component of the immune system notable for their ability to eliminate rapidly proliferating malignant cells. However, the T-cell intrinsic factors required for human CTLs to accomplish highly efficient antitumor cytotoxicity are not well defined. By evaluating human CD8+ T cells from responders versus nonresponders to treatment with immune checkpoint inhibitors, we sought to identify key factors associated with effective CTL function. Single-cell RNA-sequencing analysis of peripheral CD8+ T cells from patients treated with anti-PD-1 therapy showed that cells from nonresponders exhibited decreased expression of the cytolytic granule-associated molecule natural killer cell granule protein-7 (NKG7). Functional assays revealed that reduced NKG7 expression altered cytolytic granule number, trafficking, and calcium release, resulting in decreased CD8+ T-cell-mediated killing of tumor cells. Transfection of T cells with NKG7 mRNA was sufficient to improve the tumor-cell killing ability of human T cells isolated from nonresponders and increase their response to anti-PD-1 or anti-PD-L1 therapy in vitro. NKG7 mRNA therapy also improved the antitumor activity of murine tumor antigen-specific CD8+ T cells in an in vivo model of adoptive cell therapy. Finally, we showed that the transcription factor ETS1 played a role in regulating NKG7 expression. Together, our results identify NKG7 as a necessary component for the cytotoxic function of CD8+ T cells and establish NKG7 as a T-cell-intrinsic therapeutic target for enhancing cancer immunotherapy.See related article by Li et al., p. 154.
Collapse
Affiliation(s)
- Ti Wen
- Department of Urology, Mayo Clinic, Rochester, MN
| | - Whitney Barham
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Ying Li
- Division of Computational Biology, Mayo Clinic, Rochester, MN
| | - Henan Zhang
- Department of Urology, Mayo Clinic, Rochester, MN
| | - Joanina K. Gicobi
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | | | - Xin Liu
- Department of Urology, Mayo Clinic, Rochester, MN
| | - Hyoungjun Ham
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | | | | | | | - Hu Li
- Center for Individualized Medicine, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Cristina Correia
- Center for Individualized Medicine, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Dileep D. Monie
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Zesheng An
- Department of Urology, Mayo Clinic, Rochester, MN
| | | | - Xiaosheng Wu
- Division of Hematology, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Ruifeng Guo
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology, Mayo Clinic, Rochester, MN
| | | | | | - Yiyi Yan
- Division of Medical Oncology, Mayo Clinic, Rochester, MN
| | | | - Sean S. Park
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| | - Jie Sun
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN.,Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Hong Qin
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL
| | - Minetta C. Liu
- Division of Medical Oncology, Mayo Clinic, Rochester, MN
| | | | - Daniel D. Billadeau
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Haidong Dong
- Department of Urology, Mayo Clinic, Rochester, MN.,Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN.,Corresponding Author: Haidong Dong, M.D., Ph.D., 200 First Street SW, Rochester, MN 55905; Phone: 507-284-5482;
| |
Collapse
|
40
|
Semmrich M, Marchand JB, Fend L, Rehn M, Remy C, Holmkvist P, Silvestre N, Svensson C, Kleinpeter P, Deforges J, Junghus F, Cleary KL, Bodén M, Mårtensson L, Foloppe J, Teige I, Quéméneur E, Frendéus B. Vectorized Treg-depleting αCTLA-4 elicits antigen cross-presentation and CD8+ T cell immunity to reject ‘cold’ tumors. J Immunother Cancer 2022; 10:jitc-2021-003488. [PMID: 35058324 PMCID: PMC8783833 DOI: 10.1136/jitc-2021-003488] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2021] [Indexed: 12/12/2022] Open
Abstract
BackgroundImmune checkpoint blockade (ICB) is a clinically proven concept to treat cancer. Still, a majority of patients with cancer including those with poorly immune infiltrated ‘cold’ tumors are resistant to currently available ICB therapies. Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) is one of few clinically validated targets for ICB, but toxicities linked to efficacy in approved αCTLA-4 regimens have restricted their use and precluded full therapeutic dosing. At a mechanistic level, accumulating preclinical and clinical data indicate dual mechanisms for αCTLA-4; ICB and regulatory T cell (Treg) depletion are both thought to contribute efficacy and toxicity in available, systemic, αCTLA-4 regimens. Accordingly, strategies to deliver highly effective, yet safe αCTLA-4 therapies have been lacking. Here we assess and identify spatially restricted exposure to a novel strongly Treg-depleting, checkpoint-blocking, vectorized αCTLA-4, as a highly efficacious and potentially safe strategy to target CTLA-4.MethodsA novel human IgG1 CTLA-4 antibody (4-E03) was identified using function-first screening for monoclonal antibodies (mAbs) and targets associated with superior Treg-depleting activity. A tumor-selective oncolytic vaccinia vector was then engineered to encode this novel, strongly Treg-depleting, checkpoint-blocking, αCTLA-4 antibody or a matching surrogate antibody, and Granulocyte-macrophage colony-stimulating factor (GM-CSF) (VVGM-αCTLA-4).ResultsThe identified 4-E03 antibody showed significantly stronger Treg depletion, but equipotent checkpoint blockade, compared with clinically validated αCTLA-4 ipilimumab against CTLA-4-expressing Treg cells in a humanized mouse model in vivo. Intratumoral administration of VVGM-αCTLA-4 achieved tumor-restricted CTLA-4 receptor saturation and Treg depletion, which elicited antigen cross-presentation and stronger systemic expansion of tumor-specific CD8+ T cells and antitumor immunity compared with systemic αCTLA-4 antibody therapy. Efficacy correlated with FcγR-mediated intratumoral Treg depletion. Remarkably, in a clinically relevant mouse model resistant to systemic ICB, intratumoral VVGM-αCTLA-4 synergized with αPD-1 to reject cold tumors.ConclusionOur findings demonstrate in vivo proof of concept for spatial restriction of Treg depletion-optimized immune checkpoint blocking, vectorized αCTLA-4 as a highly effective and safe strategy to target CTLA-4. A clinical trial evaluating intratumoral VVGM-αhCTLA-4 (BT-001) alone and in combination with αPD-1 in metastatic or advanced solid tumors has commenced.
Collapse
Affiliation(s)
- Monika Semmrich
- Department of Research, BioInvent International AB, Lund, Sweden
| | | | - Laetitia Fend
- Department of Research, Transgene SA, Illkirch-Graffenstaden, France
| | - Matilda Rehn
- Department of Research, BioInvent International AB, Lund, Sweden
| | - Christelle Remy
- Department of Research, Transgene SA, Illkirch-Graffenstaden, France
| | - Petra Holmkvist
- Department of Research, BioInvent International AB, Lund, Sweden
| | | | - Carolin Svensson
- Department of Research, BioInvent International AB, Lund, Sweden
| | | | - Jules Deforges
- Department of Research, Transgene SA, Illkirch-Graffenstaden, France
| | - Fred Junghus
- Department of Research, BioInvent International AB, Lund, Sweden
| | - Kirstie L Cleary
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Mimoza Bodén
- Department of Research, BioInvent International AB, Lund, Sweden
| | - Linda Mårtensson
- Department of Research, BioInvent International AB, Lund, Sweden
| | - Johann Foloppe
- Department of Research, Transgene SA, Illkirch-Graffenstaden, France
| | - Ingrid Teige
- Department of Research, BioInvent International AB, Lund, Sweden
| | - Eric Quéméneur
- Department of Research, Transgene SA, Illkirch-Graffenstaden, France
| | - Björn Frendéus
- Department of Research, BioInvent International AB, Lund, Sweden
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
41
|
Zhou C, Liu Q, Xiang Y, Gou X, Li W. Role of the tumor immune microenvironment in tumor immunotherapy. Oncol Lett 2022; 23:53. [PMID: 34992685 PMCID: PMC8721848 DOI: 10.3892/ol.2021.13171] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/02/2021] [Indexed: 12/18/2022] Open
Abstract
Tumor immunotherapy is considered to be a novel and promising therapy for tumors and it has recently become a hot research topic. The clinical success of tumor immunotherapy has been notable, but it has been less than totally satisfactory because tumor immunotherapy has performed poorly in numerous patients although it has shown appreciable efficacy in some patients. A minority of patients demonstrate durable responses but the majority of patients do not respond to tumor immunotherapy as the tumor immune microenvironment is different in different patients for different tumor types. The success of tumor immunotherapy may be affected by the heterogeneity of the tumor immune microenvironment and its components, as these vary widely during neoplastic progression. The deepening of research and the development of technology have improved our understanding of the complexity and heterogeneity of the tumor immune microenvironment and its components, and their effects on response to tumor immunotherapy. Therefore, investigating the tumor immune microenvironment and its components and elucidating their association with tumor immunotherapy should improve the ability to study, predict and guide immunotherapeutic responsiveness, and uncover new therapeutic targets.
Collapse
Affiliation(s)
- Changsheng Zhou
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China.,Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China.,School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China.,Cancer Research Center of Xiamen University, Xiamen University, Xiamen, Fujian 361102, P.R. China.,Retroperitoneal Tumor Research Center of Oncology Chapter of Chinese Medical Association, Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Qianqian Liu
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China.,School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Yi Xiang
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Xin Gou
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Wengang Li
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China.,School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China.,Cancer Research Center of Xiamen University, Xiamen University, Xiamen, Fujian 361102, P.R. China.,Retroperitoneal Tumor Research Center of Oncology Chapter of Chinese Medical Association, Xiamen University, Xiamen, Fujian 361102, P.R. China
| |
Collapse
|
42
|
Mettu NB, Ulahannan SV, Bendell JC, Garrido-Laguna I, Strickler JH, Moore KN, Stagg R, Kapoun AM, Faoro L, Sharma S. A Phase 1a/b Open‑Label, Dose‑Escalation Study of Etigilimab Alone or in Combination with Nivolumab in Patients with Locally Advanced or Metastatic Solid Tumors. Clin Cancer Res 2021; 28:882-892. [PMID: 34844977 DOI: 10.1158/1078-0432.ccr-21-2780] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/13/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE TIGIT is a co-inhibitory receptor of T cell and natural killer cell activity. Targeting TIGIT with or without PD-1/PD-L1 checkpoint inhibition may enhance anti-tumor immunity. PATIENTS AND METHODS This Phase 1a/b trial was a first-in-human, open label, multicenter, dose escalation and expansion study in patients with locally advanced or metastatic solid tumors. Using 3+3 design, patients underwent 14-day treatment cycles with anti-TIGIT antibody etigilimab alone (phase 1a; 0.3, 1.0, 3.0, 10.0, 20.0 mg/kg intravenously) or in combination with anti-PD-1 antibody nivolumab (phase 1b; 3.0, 10.0, 20.0 mg/kg etigilimab and 240 mg nivolumab). Primary objective was safety and tolerability. RESULTS Thirty-three patients were enrolled (Phase 1a, n=23; Phase 1b, n=10). There were no DLTs. MTD for single and combination therapy was not determined; maximum administered dose was 20 mg/kg. The most commonly reported adverse events (AEs) were rash (43.5%), nausea (34.8%) and fatigue (30.4%) in Phase 1a and decreased appetite (50.0%), nausea (50.0%) and rash (40%) in Phase 1b. Six patients experienced Grade {greater than or equal to}3 treatment-related AEs. In phase 1a, 7 patients (30.0%) had stable disease. In Phase 1b, 1 patient had a partial response; 1 patient had prolonged stable disease of nearly 8 months. Median progression-free survival was 56.0 days (Phase 1a) and 57.5 days (Phase 1b). Biomarker correlative analyses demonstrated evidence of clear dose-dependent target engagement by etigilimab. CONCLUSION Etigilimab had an acceptable safety profile with preliminary evidence of clinical benefit alone and in combination with nivolumab and warrants further investigation in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Kathleen N Moore
- Obstetrics and Gynecology, Stephenson Cancer Center, Stephenson Cancer Center at the University of Oklahoma Health Sciences Center/Sarah Cannon Research Institute
| | | | | | | | | |
Collapse
|
43
|
Dong C, Dang D, Zhao X, Wang Y, Wang Z, Zhang C. Integrative Characterization of the Role of IL27 In Melanoma Using Bioinformatics Analysis. Front Immunol 2021; 12:713001. [PMID: 34733272 PMCID: PMC8558420 DOI: 10.3389/fimmu.2021.713001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/24/2021] [Indexed: 12/11/2022] Open
Abstract
Background IL27 has been reported to play dual roles in cancer; however, its effects on the tumor microenvironment (TME), immunotherapy, and prognosis in melanoma remain largely unclear. This study was aimed to uncover the effects of IL27 on TME, immunotherapy and prognosis in patients with melanoma. Methods RNA-seq data, drug sensitivity data, and clinical data were obtained from TCGA, GEO, CCLE, and CTRP. Log-rank test was used to determine the survival value of IL27. Univariate and multivariate Cox regression analyses were employed to determine the independent predictors of survival outcomes. DAVID and GSEA were used to perform gene set functional annotations. ssGSEA was used to explore the association between IL27 and immune infiltrates. ConsensusClusterPlus was used to classify melanoma tissues into hot tumors or cold tumors. Results Clinically, IL27 was negatively correlated with Breslow depth (P = 0.00042) and positively associated with response to radiotherapy (P = 0.038). High IL27 expression showed an improved survival outcome (P = 0.00016), and could serve as an independent predictor of survival outcomes (hazard ratio: 0.32 - 0.88, P = 0.015). Functionally, elevated IL27 expression could induce an enhanced immune response and pyroptosis (R = 0.64, P = 1.2e-55), autophagy (R = 0.37, P = 7.1e-17) and apoptosis (R = 0.47, P = 1.1e-27) in patients with melanoma. Mechanistically, elevated IL27 expression was positively correlated with cytotoxic cytokines (including INFG and GZMB), enhanced immune infiltrates, and elevated CD8/Treg ratio (R = 0.14, P = 0.02), possibly driving CD8+ T cell infiltration by suppressing β-catenin signaling in the TME. Furthermore, IL27 was significantly associated with hot tumor state, multiple predictors of response to immunotherapy, and improved drug response in patients with melanoma. Conclusions IL27 was correlated with enriched CD8+ T cells, desirable therapeutic response and improved prognosis. It thus can be utilized as a promising modulator in the development of cytokine-based immunotherapy for melanoma.
Collapse
Affiliation(s)
- Chunyu Dong
- Department of Pediatric Surgery, The First Hospital of Jilin University, Changchun, China
| | - Dan Dang
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China
| | - Xuesong Zhao
- Department of Pediatric Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yuanyuan Wang
- Department of Pediatric Ultrasound, The First Hospital of Jilin University, Changchun, China
| | - Zhijun Wang
- Department of Pediatric Surgery, The First Hospital of Jilin University, Changchun, China
| | - Chuan Zhang
- Department of Pediatric Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
44
|
Kalinin RS, Ukrainskaya VM, Chumakov SP, Moysenovich AM, Tereshchuk VM, Volkov DV, Pershin DS, Maksimov EG, Zhang H, Maschan MA, Rubtsov YP, Stepanov AV. Engineered Removal of PD-1 From the Surface of CD19 CAR-T Cells Results in Increased Activation and Diminished Survival. Front Mol Biosci 2021; 8:745286. [PMID: 34722633 PMCID: PMC8548718 DOI: 10.3389/fmolb.2021.745286] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
CAR-T cell therapy is the most advanced way to treat therapy resistant hematologic cancers, in particular B cell lymphomas and leukemias, with high efficiency. Donor T cells equipped ex vivo with chimeric receptor recognize target tumor cells and kill them using lytic granules. CAR-T cells that recognize CD19 marker of B cells (CD19 CAR-T) are considered the gold standard of CAR-T therapy and are approved by FDA. But in some cases, CD19 CAR-T cell therapy fails due to immune suppressive microenvironment. It is shown that tumor cells upregulate expression of PD-L1 surface molecule that binds and increases level and signal provided by PD-1 receptor on the surface of therapeutic CAR-T cells. Induction of this negative signaling results in functional impairment of cytotoxic program in CAR-T cells. Multiple attempts were made to block PD-1 signaling by reducing binding or surface level of PD-1 in CAR-T cells by various means. In this study we co-expressed CD19-CAR with PD-1-specific VHH domain of anti-PD-1 nanobody to block PD-1/PD-L1 signaling in CD19 CAR-T cells. Unexpectedly, despite increased activation of CAR-T cells with low level of PD-1, these T cells had reduced survival and diminished cytotoxicity. Functional impairment caused by disrupted PD-1 signaling was accompanied by faster maturation and upregulation of exhaustion marker TIGIT in CAR-T cells. We conclude that PD-1 in addition to its direct negative effect on CAR-induced signaling is required for attenuation of strong stimulation leading to cell death and functional exhaustion. These observations suggest that PD-1 downregulation should not be considered as the way to improve the quality of therapeutic CAR-T cells.
Collapse
Affiliation(s)
- R S Kalinin
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - V M Ukrainskaya
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - S P Chumakov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - A M Moysenovich
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - V M Tereshchuk
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - D V Volkov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - D S Pershin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - E G Maksimov
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - H Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, China
| | - M A Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Y P Rubtsov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - A V Stepanov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia.,Department of Chemistry, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
45
|
Flatmark K, Torgunrud A, Fleten KG, Davidson B, Juul HV, Mensali N, Lund-Andersen C, Inderberg EM. Peptide vaccine targeting mutated GNAS: a potential novel treatment for pseudomyxoma peritonei. J Immunother Cancer 2021; 9:jitc-2021-003109. [PMID: 34711663 PMCID: PMC8557294 DOI: 10.1136/jitc-2021-003109] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
Background Pseudomyxoma peritonei (PMP) is a rare, slow-growing abdominal cancer with no efficacious treatment options in non-resectable and recurrent cases. Otherwise, rare activating mutations in the GNAS oncogene are remarkably frequent in PMP and the mutated gene product, guanine nucleotide-binding protein α subunit (Gsα), is a potential tumor neoantigen, presenting an opportunity for targeting by a therapeutic cancer vaccine. Methods Tumor and blood samples were collected from 25 patients undergoing surgery for PMP (NCT02073500). GNAS mutation analysis was performed by next-generation targeted sequencing or digital droplet PCR. Responses to stimulation with Gsα mutated (point mutations R201H and R201C) 30 mer peptides were analyzed in peripheral blood T cells derived from patients with PMP and healthy donors. Fresh PMP tumor samples were analyzed by mass cytometry using a panel of 35 extracellular markers, and cellular subpopulations were clustered and visualized using the visual stochastic network embedding analysis tool. Results GNAS mutations were detected in 22/25 tumor samples (88%; R201H and R201C mutations detected in 16 and 6 cases, respectively). Strong T cell proliferation against Gsα mutated peptides was observed in 18/24 patients with PMP. Mass cytometry analysis of tumor revealed infiltration of CD3 +T cells in most samples, with variable CD4+:CD8 + ratios. A large proportion of T cells expressed immune checkpoint molecules, in particular programmed death receptor-1 and T cell immunoreceptor with Ig and ITIM, indicating that these T cells were antigen experienced. Conclusion These findings point to the existence of a pre-existing immunity in patients with PMP towards mutated Gsα, which has been insufficient to control tumor growth, possibly because of inhibition of antitumor T cells by upregulation of immune checkpoint molecules. The results form a rationale for exploring peptide vaccination with Gsα peptides in combination with immune checkpoint inhibiton as a possible curative treatment for PMP and other GNAS mutated cancers.
Collapse
Affiliation(s)
- Kjersti Flatmark
- Department of Gastroenterological Surgery, Oslo University Hospital, Oslo, Norway .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Annette Torgunrud
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Karianne G Fleten
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Ben Davidson
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Hedvig V Juul
- Department of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Nadia Mensali
- Department of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Christin Lund-Andersen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Else Marit Inderberg
- Department of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
46
|
Kondou R, Akiyama Y, Iizuka A, Miyata H, Maeda C, Kanematsu A, Watanabe K, Ashizawa T, Nagashima T, Urakami K, Shimoda Y, Ohshima K, Shiomi A, Ohde Y, Terashima M, Uesaka K, Onitsuka T, Nishimura S, Hirashima Y, Hayashi N, Kiyohara Y, Tsubosa Y, Katagiri H, Niwakawa M, Takahashi K, Kashiwagi H, Nakagawa M, Ishida Y, Sugino T, Notsu A, Mori K, Takahashi M, Kenmotsu H, Yamaguchi K. Identification of tumor microenvironment-associated immunological genes as potent prognostic markers in the cancer genome analysis project HOPE. Mol Clin Oncol 2021; 15:232. [PMID: 34631056 PMCID: PMC8461598 DOI: 10.3892/mco.2021.2395] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 08/05/2021] [Indexed: 01/02/2023] Open
Abstract
Project High-tech Omics-based Patient Evaluation (HOPE), which used whole-exome sequencing and gene expression profiling, was launched in 2014. A total of ~2,000 patients were enrolled until March 2016, and the survival time was observed up to July 2019. In our previous study, a tumor microenvironment immune type classification based on the expression levels of the programmed death-ligand 1 (PD-L1) and CD8B genes was performed based on four types: A, adaptive immune resistance; B, intrinsic induction; C, immunological ignorance; and D, tolerance. Type A (PD-L1+ and CD8B+) exhibited upregulated features of T helper 1 antitumor responses. In the present study, survival time analysis at 5 years revealed that patients in type A had a better prognosis than those in other categories [5 year survival rate (%); A (80.5) vs. B (73.9), C (73.4) and D (72.6), P=0.0005]. Based on the expression data of 293 immune response-associated genes, 62 specific genes were upregulated in the type A group. Among these genes, 18 specific genes, such as activated effector T-cell markers (CD8/CD40LG/GZMB), effector memory T-cell markers (PD-1/CD27/ICOS), chemokine markers (CXCL9/CXCL10) and activated dendritic cell markers (CD80/CD274/SLAMF1), were significantly associated with a good prognosis using overall survival time analysis. Finally, multivariate Cox proportional hazard regression analyses of overall survival demonstrated that four genes (GZMB, HAVCR2, CXCL9 and CD40LG) were independent prognostic markers, and GZMB, CXCL9 and CD40LG may contribute to the survival benefit of patients in the immune type A group.
Collapse
Affiliation(s)
- Ryota Kondou
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Yasuto Akiyama
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Akira Iizuka
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Haruo Miyata
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Chie Maeda
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Akari Kanematsu
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Kyoko Watanabe
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Tadashi Ashizawa
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Takeshi Nagashima
- Division of Cancer Diagnostics Research, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan.,Special Reference Laboratory, Tokyo 191-0002, Japan
| | - Kenichi Urakami
- Division of Cancer Diagnostics Research, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Yuji Shimoda
- Division of Cancer Diagnostics Research, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan.,Special Reference Laboratory, Tokyo 191-0002, Japan
| | - Keiichi Ohshima
- Medical Genetics Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Akio Shiomi
- Division of Colon and Rectal Surgery, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Yasuhisa Ohde
- Division of Thoracic Surgery, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Masanori Terashima
- Division of Gastric Surgery, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Katsuhiko Uesaka
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Tetsuro Onitsuka
- Division of Head and Neck Surgery, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Seiichiro Nishimura
- Division of Breast Surgery, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Yasuyuki Hirashima
- Division of Gynecology, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Nakamasa Hayashi
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Yoshio Kiyohara
- Division of Dermatology, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Yasuhiro Tsubosa
- Division of Esophageal Surgery, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Hirohisa Katagiri
- Division of Orthopedic Oncology, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Masashi Niwakawa
- Division of Urology, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Kaoru Takahashi
- Division of Breast Oncology Center, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Hiroya Kashiwagi
- Division of Ophthalmology, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Masahiro Nakagawa
- Division of Plastic and Reconstructive Surgery, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Yuji Ishida
- Division of Pediatrics, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Takashi Sugino
- Division of Pathology, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Akifumi Notsu
- Clinical Trial Coordination Office, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Keita Mori
- Clinical Trial Coordination Office, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Mitsuru Takahashi
- Division of Orthopedic Oncology, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Hirotsugu Kenmotsu
- Division of Thoracic Oncology, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | | |
Collapse
|
47
|
van Wilpe S, Wosika V, Ciarloni L, Hosseinian Ehrensberger S, Jeitziner R, Angelino P, Duiveman-de Boer T, Koornstra RHT, de Vries IJM, Gerritsen WR, Schalken J, Mehra N. Whole Blood Transcriptome Profiling Identifies DNA Replication and Cell Cycle Regulation as Early Marker of Response to Anti-PD-1 in Patients with Urothelial Cancer. Cancers (Basel) 2021; 13:cancers13184660. [PMID: 34572887 PMCID: PMC8465885 DOI: 10.3390/cancers13184660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Unfortunately, not all patients with urothelial cancer benefit from checkpoint inhibitors (ICIs). Currently, the first radiological response evaluation is not performed until after 9 to 12 weeks of ICI therapy. Early response biomarkers might enable an early switch to more effective therapies in patients that do not respond. In this study, we aimed to identify early response biomarkers in the blood of patients treated with ICIs. In whole blood of patients with clinical benefit, genes involved in DNA replication and cell cycle regulation were upregulated after 2 to 6 weeks of treatment. This appeared to be a result of T cell proliferation and was not observed in patients without clinical benefit. Our results suggest that whole blood RNA sequencing can contribute to early response prediction in patients treated with ICIs and warrants further research. Abstract Although immune checkpoint inhibitors improve median overall survival in patients with metastatic urothelial cancer (mUC), only a minority of patients benefit from it. Early blood-based response biomarkers may provide a reliable way to assess response weeks before imaging is available, enabling an early switch to other therapies. We conducted an exploratory study aimed at the identification of early markers of response to anti-PD-1 in patients with mUC. Whole blood RNA sequencing and phenotyping of peripheral blood mononuclear cells were performed on samples of 26 patients obtained before and after 2 to 6 weeks of anti-PD-1. Between baseline and on-treatment samples of patients with clinical benefit, 51 differentially expressed genes (DEGs) were identified, of which 37 were upregulated during treatment. Among the upregulated genes was PDCD1, the gene encoding PD-1. STRING network analysis revealed a cluster of five interconnected DEGs which were all involved in DNA replication or cell cycle regulation. We hypothesized that the upregulation of DNA replication/cell cycle genes is a result of T cell proliferation and we were able to detect an increase in Ki-67+ CD8+ T cells in patients with clinical benefit (median increase: 1.65%, range −0.63 to 7.06%, p = 0.012). In patients without clinical benefit, no DEGs were identified and no increase in Ki-67+ CD8+ T cells was observed. In conclusion, whole blood transcriptome profiling identified early changes in DNA replication and cell cycle regulation genes as markers of clinical benefit to anti-PD-1 in patients with urothelial cancer. Although promising, our findings require further validation before implementation in the clinic.
Collapse
Affiliation(s)
- Sandra van Wilpe
- Department of Medical Oncology, The Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; (S.v.W.); (W.R.G.)
| | - Victoria Wosika
- Novigenix SA, 1066 Epalinges, Switzerland; (V.W.); (L.C.); (S.H.E.)
| | - Laura Ciarloni
- Novigenix SA, 1066 Epalinges, Switzerland; (V.W.); (L.C.); (S.H.E.)
| | | | - Rachel Jeitziner
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland; (R.J.); (P.A.)
| | - Paolo Angelino
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland; (R.J.); (P.A.)
| | - Tjitske Duiveman-de Boer
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (T.D.-d.B.); (I.J.M.d.V.)
| | | | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (T.D.-d.B.); (I.J.M.d.V.)
| | - Winald R. Gerritsen
- Department of Medical Oncology, The Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; (S.v.W.); (W.R.G.)
| | - Jack Schalken
- Department of Urology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands;
| | - Niven Mehra
- Department of Medical Oncology, The Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; (S.v.W.); (W.R.G.)
- Correspondence:
| |
Collapse
|
48
|
T-Cell Responses in Merkel Cell Carcinoma: Implications for Improved Immune Checkpoint Blockade and Other Therapeutic Options. Int J Mol Sci 2021; 22:ijms22168679. [PMID: 34445385 PMCID: PMC8395396 DOI: 10.3390/ijms22168679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023] Open
Abstract
Merkel cell carcinoma (MCC) is a rare and aggressive skin cancer with rising incidence and high mortality. Approximately 80% of the cases are caused by the human Merkel cell polyomavirus, while the remaining 20% are induced by UV light leading to mutations. The standard treatment of metastatic MCC is the use of anti-PD-1/-PD-L1-immune checkpoint inhibitors (ICI) such as Pembrolizumab or Avelumab, which in comparison with conventional chemotherapy show better overall response rates and longer duration of responses in patients. Nevertheless, 50% of the patients do not respond or develop ICI-induced, immune-related adverse events (irAEs), due to diverse mechanisms, such as down-regulation of MHC complexes or the induction of anti-inflammatory cytokines. Other immunotherapeutic options such as cytokines and pro-inflammatory agents or the use of therapeutic vaccination offer great ameliorations to ICI. Cytotoxic T-cells play a major role in the effectiveness of ICI, and tumour-infiltrating CD8+ T-cells and their phenotype contribute to the clinical outcome. This literature review presents a summary of current and future checkpoint inhibitor therapies in MCC and demonstrates alternative therapeutic options. Moreover, the importance of T-cell responses and their beneficial role in MCC treatment is discussed.
Collapse
|
49
|
Dréno B, Khammari A, Fortun A, Vignard V, Saiagh S, Beauvais T, Jouand N, Bercegay S, Simon S, Lang F, Labarrière N. Phase I/II clinical trial of adoptive cell transfer of sorted specific T cells for metastatic melanoma patients. Cancer Immunol Immunother 2021; 70:3015-3030. [PMID: 34120214 PMCID: PMC8423703 DOI: 10.1007/s00262-021-02961-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
Adoptive cell transfer (ACT) of tumor-specific T lymphocytes represents a relevant therapeutic strategy to treat metastatic melanoma patients. Ideal T-cells should combine tumor specificity and reactivity with survival in vivo, while avoiding autoimmune side effects. Here we report results from a Phase I/II clinical trial (NCT02424916, performed between 2015 and 2018) in which 6 metastatic HLA-A2 melanoma patients received autologous antigen-specific T-cells produced from PBMC, after peptide stimulation in vitro, followed by sorting with HLA-peptide multimers and amplification. Each patient received a combination of Melan-A and MELOE-1 polyclonal specific T-cells, whose specificity and anti-tumor reactivity were checked prior to injection, with subcutaneous IL-2. Transferred T-cells were also characterized in terms of functional avidity, diversity and phenotype and their blood persistence was evaluated. An increase in specific T-cells was detected in the blood of all patients at day 1 and progressively disappeared from day 7 onwards. No serious adverse events occurred after this ACT. Clinically, five patients progressed and one patient experienced a partial response following therapy. Melan-A and MELOE-1 specific T-cells infused to this patient were diverse, of high avidity, with a high proportion of T lymphocytes co-expressing PD-1 and TIGIT but few other exhaustion markers. In conclusion, we demonstrated the feasibility and safety of ACT with multimer-sorted Melan-A and MELOE-1 specific T cells to metastatic melanoma patients. The clinical efficacy of such therapeutic strategy could be further enhanced by the selection of highly reactive T-cells, based on PD-1 and TIGIT co-expression, and a combination with ICI, such as anti-PD-1.
Collapse
Affiliation(s)
- Brigitte Dréno
- Dermato-Cancerology Department, CIC 1413, CHU Nantes, Nantes, France.,UTCG, CHU Nantes, Nantes, France.,CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes, France
| | - Amir Khammari
- Dermato-Cancerology Department, CIC 1413, CHU Nantes, Nantes, France.,CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes, France
| | - Agnès Fortun
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Virginie Vignard
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes, France
| | | | - Tiffany Beauvais
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,CHU Nantes, Nantes, France
| | - Nicolas Jouand
- LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.,SFR Santé, CNRS, Inserm, Inserm UMS 016, CNRS UMS 3556, Université de Nantes, CHU Nantes, 44000, Nantes, France
| | | | - Sylvain Simon
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - François Lang
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France. .,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.
| | - Nathalie Labarrière
- CRCINA, Inserm, Université de Nantes, 44000, Nantes, France. .,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France. .,SFR Santé, CNRS, Inserm, Inserm UMS 016, CNRS UMS 3556, Université de Nantes, CHU Nantes, 44000, Nantes, France.
| |
Collapse
|