1
|
Jain SS, Burton Sojo G, Sun H, Friedland BN, McNamara ME, Schmidt MO, Wellstein A. The Role of Aging and Senescence in Immune Checkpoint Inhibitor Response and Toxicity. Int J Mol Sci 2024; 25:7013. [PMID: 39000121 PMCID: PMC11241020 DOI: 10.3390/ijms25137013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024] Open
Abstract
Cellular senescence accumulates with age and has been shown to impact numerous physiological and pathological processes, including immune function. The role of cellular senescence in cancer is multifaceted, but the impact on immune checkpoint inhibitor response and toxicity has not been fully evaluated. In this review, we evaluate the impact of cellular senescence in various biological compartments, including the tumor, the tumor microenvironment, and the immune system, on immune checkpoint inhibitor efficacy and toxicity. We provide an overview of the impact of cellular senescence in normal and pathological contexts and examine recent studies that have connected aging and cellular senescence to immune checkpoint inhibitor treatment in both the pre-clinical and clinical contexts. Overall, senescence plays a multi-faceted, context-specific role and has been shown to modulate immune-related adverse event incidence as well as immune checkpoint inhibitor response.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anton Wellstein
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA; (S.S.J.)
| |
Collapse
|
2
|
Ganesh S, Kim MJ, Lee J, Feng X, Ule K, Mahan A, Krishnan HS, Wang Z, Anzahaee MY, Singhal G, Korboukh I, Lockridge JA, Sanftner L, Rijnbrand R, Abrams M, Brown BD. RNAi mediated silencing of STAT3/PD-L1 in tumor-associated immune cells induces robust anti-tumor effects in immunotherapy resistant tumors. Mol Ther 2024; 32:1895-1916. [PMID: 38549376 PMCID: PMC11184339 DOI: 10.1016/j.ymthe.2024.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/29/2024] [Accepted: 03/26/2024] [Indexed: 04/20/2024] Open
Abstract
Malignant tumors are often associated with an immunosuppressive tumor microenvironment (TME), rendering most of them resistant to standard-of-care immune checkpoint inhibitors (CPIs). Signal transducer and activator of transcription 3 (STAT3), a ubiquitously expressed transcription factor, has well-defined immunosuppressive functions in several leukocyte populations within the TME. Since the STAT3 protein has been challenging to target using conventional pharmaceutical modalities, we investigated the feasibility of applying systemically delivered RNA interference (RNAi) agents to silence its mRNA directly in tumor-associated immune cells. In preclinical rodent tumor models, chemically stabilized acylated small interfering RNAs (siRNAs) selectively silenced Stat3 mRNA in multiple relevant cell types, reduced STAT3 protein levels, and increased cytotoxic T cell infiltration. In a murine model of CPI-resistant pancreatic cancer, RNAi-mediated Stat3 silencing resulted in tumor growth inhibition, which was further enhanced in combination with CPIs. To further exemplify the utility of RNAi for cancer immunotherapy, this technology was used to silence Cd274, the gene encoding the immune checkpoint protein programmed death-ligand 1 (PD-L1). Interestingly, silencing of Cd274 was effective in tumor models that are resistant to PD-L1 antibody therapy. These data represent the first demonstration of systemic delivery of RNAi agents to the TME and suggest applying this technology for immuno-oncology applications.
Collapse
Affiliation(s)
- Shanthi Ganesh
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA.
| | - Min Ju Kim
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| | - Jenny Lee
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| | - Xudong Feng
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| | - Krisjanis Ule
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| | - Amy Mahan
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| | | | - Zhe Wang
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| | | | - Garima Singhal
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| | - Ilia Korboukh
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| | | | - Laura Sanftner
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| | - Rene Rijnbrand
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| | - Marc Abrams
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| | - Bob D Brown
- Dicerna Pharmaceuticals, Inc, a Novo Nordisk Company, Lexington, MA 02421, USA
| |
Collapse
|
3
|
Khan B, Qahwaji RM, Alfaifi MS, Mobashir M. Nivolumab and Ipilimumab Acting as Tormentors of Advanced Tumors by Unleashing Immune Cells and Associated Collateral Damage. Pharmaceutics 2024; 16:732. [PMID: 38931856 PMCID: PMC11207028 DOI: 10.3390/pharmaceutics16060732] [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: 03/18/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 06/28/2024] Open
Abstract
Combining immune checkpoint inhibitors, specifically nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4), holds substantial promise in revolutionizing cancer treatment. This review explores the transformative impact of these combinations, emphasizing their potential for enhancing therapeutic outcomes across various cancers. Immune checkpoint proteins, such as PD1 and CTLA4, play a pivotal role in modulating immune responses. Blocking these checkpoints unleashes anticancer activity, and the synergy observed when combining multiple checkpoint inhibitors underscores their potential for enhanced efficacy. Nivolumab and ipilimumab harness the host's immune system to target cancer cells, presenting a powerful approach to prevent tumor development. Despite their efficacy, immune checkpoint inhibitors are accompanied by a distinct set of adverse effects, particularly immune-related adverse effects affecting various organs. Understanding these challenges is crucial for optimizing treatment strategies and ensuring patient well-being. Ongoing clinical trials are actively exploring the combination of checkpoint inhibitory therapies, aiming to decipher their synergistic effects and efficacy against diverse cancer types. This review discusses the mechanisms, adverse effects, and various clinical trials involving nivolumab and ipilimumab across different cancers, emphasizing their transformative impact on cancer treatment.
Collapse
Affiliation(s)
- Bushra Khan
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Rowaid M. Qahwaji
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22233, Saudi Arabia;
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mashael S. Alfaifi
- Department of Epidemiology, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Mohammad Mobashir
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Solnavägen 9, 171 65 Solna, Sweden
| |
Collapse
|
4
|
Möller M, Orth V, Umansky V, Hetjens S, Braun V, Reißfelder C, Hardt J, Seyfried S. Myeloid-derived suppressor cells in peripheral blood as predictive biomarkers in patients with solid tumors undergoing immune checkpoint therapy: systematic review and meta-analysis. Front Immunol 2024; 15:1403771. [PMID: 38855104 PMCID: PMC11157008 DOI: 10.3389/fimmu.2024.1403771] [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: 03/19/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024] Open
Abstract
Background Immunotherapeutic approaches, including immune checkpoint inhibitor (ICI) therapy, are increasingly recognized for their potential. Despite notable successes, patient responses to these treatments vary significantly. The absence of reliable predictive and prognostic biomarkers hampers the ability to foresee outcomes. This meta-analysis aims to evaluate the predictive significance of circulating myeloid-derived suppressor cells (MDSC) in patients with solid tumors undergoing ICI therapy, focusing on progression-free survival (PFS) and overall survival (OS). Methods A comprehensive literature search was performed across PubMed and EMBASE from January 2007 to November 2023, utilizing keywords related to MDSC and ICI. We extracted hazard ratios (HRs) and 95% confidence intervals (CIs) directly from the publications or calculated them based on the reported data. A hazard ratio greater than 1 indicated a beneficial effect of low MDSC levels. We assessed heterogeneity and effect size through subgroup analyses. Results Our search yielded 4,023 articles, of which 17 studies involving 1,035 patients were included. The analysis revealed that patients with lower levels of circulating MDSC experienced significantly improved OS (HR=2.13 [95% CI 1.51-2.99]) and PFS (HR=1.87 [95% CI 1.29-2.72]) in response to ICI therapy. Notably, heterogeneity across these outcomes was primarily attributed to differences in polymorphonuclear MDSC (PMN-MDSC) subpopulations and varying cutoff methodologies used in the studies. The monocytic MDSC (M-MDSC) subpopulation emerged as a consistent and significant prognostic marker across various subgroup analyses, including ethnicity, tumor type, ICI target, sample size, and cutoff methodology. Conclusions Our findings suggest that standardized assessment of MDSC, particularly M-MDSC, should be integral to ICI therapy strategies. These cells hold the promise of identifying patients at risk of poor response to ICI therapy, enabling tailored treatment approaches. Further research focusing on the standardization of markers and validation of cutoff methods is crucial for integrating MDSC into clinical practice. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023420095, identifier CRD42023420095.
Collapse
Affiliation(s)
- Maximilian Möller
- Department of Surgery, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Vanessa Orth
- Department of Surgery, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Viktor Umansky
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ)-Hector Cancer Institute, University Medical Centre Mannheim, Mannheim, Germany
| | - Svetlana Hetjens
- Department of Biometry and Statistics, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Volker Braun
- Department of Library and Information Sciences, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Reißfelder
- Department of Surgery, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- German Cancer Research Center (DKFZ)-Hector Cancer Institute, University Medical Centre Mannheim, Mannheim, Germany
| | - Julia Hardt
- Department of Surgery, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Steffen Seyfried
- Department of Surgery, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
5
|
Wu LY, Park SH, Jakobsson H, Shackleton M, Möller A. Immune Regulation and Immune Therapy in Melanoma: Review with Emphasis on CD155 Signalling. Cancers (Basel) 2024; 16:1950. [PMID: 38893071 PMCID: PMC11171058 DOI: 10.3390/cancers16111950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Melanoma is commonly diagnosed in a younger population than most other solid malignancies and, in Australia and most of the world, is the leading cause of skin-cancer-related death. Melanoma is a cancer type with high immunogenicity; thus, immunotherapies are used as first-line treatment for advanced melanoma patients. Although immunotherapies are working well, not all the patients are benefitting from them. A lack of a comprehensive understanding of immune regulation in the melanoma tumour microenvironment is a major challenge of patient stratification. Overexpression of CD155 has been reported as a key factor in melanoma immune regulation for the development of therapy resistance. A more thorough understanding of the actions of current immunotherapy strategies, their effects on immune cell subsets, and the roles that CD155 plays are essential for a rational design of novel targets of anti-cancer immunotherapies. In this review, we comprehensively discuss current anti-melanoma immunotherapy strategies and the immune response contribution of different cell lineages, including tumour endothelial cells, myeloid-derived suppressor cells, cytotoxic T cells, cancer-associated fibroblast, and nature killer cells. Finally, we explore the impact of CD155 and its receptors DNAM-1, TIGIT, and CD96 on immune cells, especially in the context of the melanoma tumour microenvironment and anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Li-Ying Wu
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Su-Ho Park
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haakan Jakobsson
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
| | - Mark Shackleton
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
- School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Andreas Möller
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
6
|
Dyikanov D, Zaitsev A, Vasileva T, Wang I, Sokolov AA, Bolshakov ES, Frank A, Turova P, Golubeva O, Gantseva A, Kamysheva A, Shpudeiko P, Krauz I, Abdou M, Chasse M, Conroy T, Merriam NR, Alesse JE, English N, Shpak B, Shchetsova A, Tikhonov E, Filatov I, Radko A, Bolshakova A, Kachalova A, Lugovykh N, Bulahov A, Kilina A, Asanbekov S, Zheleznyak I, Skoptsov P, Alekseeva E, Johnson JM, Curry JM, Linnenbach AJ, South AP, Yang E, Morozov K, Terenteva A, Nigmatullina L, Fastovetz D, Bobe A, Balabanian L, Nomie K, Yong ST, Davitt CJH, Ryabykh A, Kudryashova O, Tazearslan C, Bagaev A, Fowler N, Luginbuhl AJ, Ataullakhanov RI, Goldberg MF. Comprehensive peripheral blood immunoprofiling reveals five immunotypes with immunotherapy response characteristics in patients with cancer. Cancer Cell 2024; 42:759-779.e12. [PMID: 38744245 DOI: 10.1016/j.ccell.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/20/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024]
Abstract
The lack of comprehensive diagnostics and consensus analytical models for evaluating the status of a patient's immune system has hindered a wider adoption of immunoprofiling for treatment monitoring and response prediction in cancer patients. To address this unmet need, we developed an immunoprofiling platform that uses multiparameter flow cytometry to characterize immune cell heterogeneity in the peripheral blood of healthy donors and patients with advanced cancers. Using unsupervised clustering, we identified five immunotypes with unique distributions of different cell types and gene expression profiles. An independent analysis of 17,800 open-source transcriptomes with the same approach corroborated these findings. Continuous immunotype-based signature scores were developed to correlate systemic immunity with patient responses to different cancer treatments, including immunotherapy, prognostically and predictively. Our approach and findings illustrate the potential utility of a simple blood test as a flexible tool for stratifying cancer patients into therapy response groups based on systemic immunoprofiling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jennifer M Johnson
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Joseph M Curry
- Department of Otolaryngology Head and Neck Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Alban J Linnenbach
- Department of Otolaryngology Head and Neck Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrew P South
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - EnJun Yang
- The Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Adam J Luginbuhl
- Department of Otolaryngology Head and Neck Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | |
Collapse
|
7
|
Li Y, Liu W, Xu H, Zhou Y, Xie W, Guo Y, Liao Z, Jiang X, Liu J, Ren C. Aptamers combined with immune checkpoints for cancer detection and targeted therapy: A review. Int J Biol Macromol 2024; 262:130032. [PMID: 38342267 DOI: 10.1016/j.ijbiomac.2024.130032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
In recent years, remarkable strides have been made in the field of immunotherapy, which has emerged as a standard treatment for many cancers. As a kind of immunotherapy drug, monoclonal antibodies employed in immune checkpoint therapy have proven beneficial for patients with diverse cancer types. However, owing to the extensive heterogeneity of clinical responses and the complexity and variability of the immune system and tumor microenvironment (TME), accurately predicting its efficacy remains a challenge. Recent advances in aptamers provide a promising approach for monitoring alterations within the immune system and TME, thereby facilitating targeted immunotherapy, particularly focused on immune checkpoint blockade, with enhanced antitumor efficiency. Aptamers have been widely used in tumor cell detection, biosensors, drug discovery, and biomarker screening due to their high specificity and high affinity with their targets. This review aims to comprehensively examine the research status and progress of aptamers in cancer diagnosis and immunotherapy, with a specific emphasis on those related to immune checkpoints. Additionally, we will discuss the future research directions and potential therapeutic targets for aptamer-based immune checkpoint therapy, aiming to provide a theoretical basis for targeting immunotherapy molecules and blocking tumor immune escape.
Collapse
Affiliation(s)
- Yihan Li
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Weidong Liu
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Hongjuan Xu
- NHC Key Laboratory of Biological Nanotechnology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yao Zhou
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Wen Xie
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Youwei Guo
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Ziling Liao
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Xingjun Jiang
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Liu
- Department of Critical care medicine, Hainan Hospital of Chinese PLA General Hosptial; project supported by Hainan Province Clinical Medical Center, China.
| | - Caiping Ren
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
8
|
Jin Y, Huang Y, Ren H, Huang H, Lai C, Wang W, Tong Z, Zhang H, Wu W, Liu C, Bao X, Fang W, Li H, Zhao P, Dai X. Nano-enhanced immunotherapy: Targeting the immunosuppressive tumor microenvironment. Biomaterials 2024; 305:122463. [PMID: 38232643 DOI: 10.1016/j.biomaterials.2023.122463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/27/2023] [Accepted: 12/31/2023] [Indexed: 01/19/2024]
Abstract
The tumor microenvironment (TME), which is mostly composed of tumor cells, immune cells, signaling molecules, stromal tissue, and the vascular system, is an integrated system that is conducive to the formation of tumors. TME heterogeneity makes the response to immunotherapy different in different tumors, such as "immune-cold" and "immune-hot" tumors. Tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells are the major suppressive immune cells and their different phenotypes interact and influence cancer cells by secreting different signaling factors, thus playing a key role in the formation of the TME as well as in the initiation, growth, and metastasis of cancer cells. Nanotechnology development has facilitated overcoming the obstacles that limit the further development of conventional immunotherapy, such as toxic side effects and lack of targeting. In this review, we focus on the role of three major suppressive immune cells in the TME as well as in tumor development, clinical trials of different drugs targeting immune cells, and different attempts to combine drugs with nanomaterials. The aim is to reveal the relationship between immunotherapy, immunosuppressive TME and nanomedicine, thus laying the foundation for further development of immunotherapy.
Collapse
Affiliation(s)
- Yuzhi Jin
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Yangyue Huang
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China; Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Hui Ren
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Huanhuan Huang
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China; Postgraduate Training Base Alliance of Wenzhou Medical University, Hangzhou, 310022, China
| | - Chunyu Lai
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Wenjun Wang
- Department of Plastic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Zhou Tong
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Hangyu Zhang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Wei Wu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Chuan Liu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xuanwen Bao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Hongjun Li
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China; Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China; Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
| | - Xiaomeng Dai
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
9
|
Di Giacomo AM, Schenker M, Medioni J, Mandziuk S, Majem M, Gravis G, Cornfeld M, Ranganathan S, Lou S, Csoszi T. A phase II study of retifanlimab, a humanized anti-PD-1 monoclonal antibody, in patients with solid tumors (POD1UM-203). ESMO Open 2024; 9:102387. [PMID: 38401247 PMCID: PMC10982862 DOI: 10.1016/j.esmoop.2024.102387] [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: 06/09/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND POD1UM-203, an open-label, multicenter, phase II study, evaluated retifanlimab, a humanized monoclonal antibody targeting programmed cell death protein-1 (PD-1) in patients with selected solid tumors where immune checkpoint inhibitor therapies have previously shown efficacy. PATIENTS AND METHODS Eligible patients (≥18 years) had measurable disease and included unresectable or metastatic melanoma, treatment-naive metastatic non-small-cell lung cancer (NSCLC) with high programmed death-ligand 1 (PD-L1) expression (tumor proportion score ≥50%), cisplatin-ineligible locally advanced/metastatic urothelial carcinoma (UC) with PD-L1 expression (combined positive score ≥10%), or treatment-naive locally advanced/metastatic clear-cell renal cell carcinoma (RCC). Retifanlimab 500 mg was administered intravenously every 4 weeks as a 30-min infusion. The primary endpoint was investigator-assessed overall response rate. RESULTS Overall, 121 patients (35 melanoma, 23 NSCLC, 29 UC, 34 RCC) were enrolled and treated. The overall response rate [95% confidence interval (CI)] was 40.0% (23.9-57.9) in the melanoma cohort, 34.8% (16.4-57.3) in the NSCLC cohort, 37.9% (20.7-57.7) in the UC cohort, and 23.5% (10.7-41.2) in the RCC cohort. Median duration of response was 11.5 months (95% CI 2.2-not reached) in the UC cohort, and was not reached in the other cohorts. Retifanlimab safety was consistent with previous experience for PD-(L)1 inhibitors. CONCLUSIONS Retifanlimab demonstrated durable antitumor activity in patients with melanoma, NSCLC, UC, or RCC. The efficacy and safety of retifanlimab were as expected for a PD-(L)1 inhibitor. These data support further study of retifanlimab in solid tumors.
Collapse
Affiliation(s)
- A M Di Giacomo
- University of Siena, Siena, Italy; Center for Immuno-Oncology, University Hospital of Siena, Siena, Italy
| | - M Schenker
- Centrul de Oncologie Sf. Nectarie, Oncologie Medicala, Craiova, Romania
| | - J Medioni
- Centre of Early Clinical Trials in Cancer, Hôpital Européen Georges-Pompidou, Université Paris Cité, Paris, France
| | - S Mandziuk
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - M Majem
- Medical Oncology Department, Hospital de Sant Pau, Barcelona, Spain
| | - G Gravis
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, CRCM, Marseille, France
| | | | | | - S Lou
- Incyte Corporation, Wilmington, USA
| | - T Csoszi
- Hetényi Géza Kórház Onkológiai Központ, Szolnok, Hungary.
| |
Collapse
|
10
|
Feng J, Chen X, Wei J, Weng Y, Wang J, Wang T, Song Q, Min P. Safety and efficacy of immune checkpoint inhibitor rechallenge in advanced non-small cell lung cancer: a retrospective study. Sci Rep 2024; 14:2315. [PMID: 38281979 PMCID: PMC10822846 DOI: 10.1038/s41598-024-52034-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 01/12/2024] [Indexed: 01/30/2024] Open
Abstract
We conducted a retrospective study to evaluate the efficacy of immune checkpoint inhibitor (ICI) rechallenge in patients with advanced non-small cell lung cancer (NSCLC). The study included 111 patients who had previously received ICI therapy and experienced disease progression. The primary endpoints assessed were overall survival (OS), progression-free survival (PFS), and objective response rate (ORR). Our findings revealed that the ICI rechallenge showed promising results in improving patient outcomes. OS (r) is the time from rechallenging with immune checkpoint inhibitors to the last follow-up or death from any cause. The median OS (r) was 14.3 months (95% CI 11.3-17.3 months), with a median PFS (r) of 5.9 months (95% CI 4.1-7.7 months). The ORR was 17.1%; the DCR was 82.3%. Subgroup analysis demonstrated that patients without brain or liver metastases had a longer OS (r) compared to those with metastases (21.6 vs. 13.8 months, χ2 = 3.873, P = 0.046; 20.8 vs. 9.1 months, χ2 = 10.733, P = 0.001, respectively). Moreover, patients without driver gene mutations exhibited significantly longer OS than those with mutations or wild-type patients (22.9 vs. 16.1 vs. 7.5 months, χ2 = 10.710, P = 0.005). Notably, patients who switched to a different ICI during the rechallenge had shorter OS than those who did not change medications (10.4 vs. 21.1 months, χ2 = 9.014, P = 0.003). The incidence of immune-related adverse events did not significantly differ between the two treatment phases. These findings suggest that ICI rechallenge may be a viable therapeutic strategy for select NSCLC patients. Further prospective studies are needed to validate these results and guide treatment decisions for advanced NSCLC.
Collapse
Affiliation(s)
- Jia Feng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xinyi Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiayan Wei
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yiming Weng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jingsong Wang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Tong Wang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Peng Min
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
11
|
Vitiello L, Lixi F, Coco G, Giannaccare G. Ocular Surface Side Effects of Novel Anticancer Drugs. Cancers (Basel) 2024; 16:344. [PMID: 38254833 PMCID: PMC10814578 DOI: 10.3390/cancers16020344] [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: 12/24/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Surgery, anticancer drugs (chemotherapy, hormonal medicines, and targeted treatments), and/or radiation are common treatment strategies for neoplastic diseases. Anticancer drugs eliminate malignant cells through the inhibition of specific pathways that contribute to the formation and development of cancer. Given the ability of such pharmacological medications to combat cancerous cells, their role in the management of neoplastic diseases has become essential. However, these drugs may also lead to undesirable systemic and ocular adverse effects due to cyto/neuro-toxicity and inflammatory reactions. Ocular surface side effects are recognized to significantly impact patient's quality of life and quality of vision. Blepharoconjunctivitis is known to be a common side effect caused by oxaliplatin, cyclophosphamide, cytarabine, and docetaxel, while anastrozole, methotrexate, and 5-fluorouracil can all determine dry eye disease. However, the potential processes involved in the development of these alterations are yet not fully understood, especially for novel drugs currently available for cancer treatment. This review aims at analyzing the potential ocular surface and adnexal side effects of novel anticancer medications, trying to provide a better understanding of the underlying pharmacological processes and useful insights on the choice of proper management.
Collapse
Affiliation(s)
- Livio Vitiello
- Eye Unit, “Luigi Curto” Hospital, Azienda Sanitaria Locale Salerno, 84035 Polla, SA, Italy;
| | - Filippo Lixi
- Eye Clinic, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, CA, Italy;
| | - Giulia Coco
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, RM, Italy;
| | - Giuseppe Giannaccare
- Eye Clinic, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, CA, Italy;
| |
Collapse
|
12
|
Curkovic NB, Bai K, Ye F, Johnson DB. Incidence of Cutaneous Immune-Related Adverse Events and Outcomes in Immune Checkpoint Inhibitor-Containing Regimens: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024; 16:340. [PMID: 38254829 PMCID: PMC10814132 DOI: 10.3390/cancers16020340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/19/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are used to treat many cancers, and cutaneous immune-related adverse events (cirAEs) are among the most frequently encountered toxic effects. Understanding the incidence and prognostic associations of cirAEs is of importance as their uses in different settings, combinations, and tumor types expand. To evaluate the incidence of cirAEs and their association with outcome measures across a variety of ICI regimens and cancers, we performed a systematic review and meta-analysis of published trials of anti-programmed death-1/ligand-1 (PD-1/PD-L1) and anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) ICIs, both alone and in combination with chemotherapy, antiangiogenic agents, or other ICIs in patients with melanoma, renal cell carcinoma, non-small cell lung cancer, and urothelial carcinoma. Key findings of our study include variable cirAE incidence among tumors and ICI regimens, positive association with increased cirAE incidence and response rate, as well as significant association between increased vitiligo incidence and overall survival. Across 174 studies, rash, pruritis, and vitiligo were the most reported cirAEs, with incidences of 16.7%, 18.0%, and 6.6%, respectively. Higher incidence of cirAEs was associated with ICI combination regimens and with CTLA-4-containing regimens, particularly with higher doses of ipilimumab, as compared to PD-1/L1 monotherapies. Outcome measures including response rate and progression-free survival were positively correlated with incidence of cirAEs. The response rate and incidence of pruritis, vitiligo, and rash were associated with expected rises in incidence of 0.17% (p = 0.0238), 0.40% (p = 0.0010), and 0.18% (p = 0.0413), respectively. Overall survival was positively correlated with the incidence of pruritis, vitiligo, and rash; this association was significant for vitiligo (p = 0.0483). Our analysis provides benchmark incidence rates for cirAEs and links cirAEs with favorable treatment outcomes at a study level across diverse solid tumors and multiple ICI regimens.
Collapse
Affiliation(s)
- Nina B. Curkovic
- School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Kun Bai
- Vanderbilt Ingram Cancer Center, Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Fei Ye
- Vanderbilt Ingram Cancer Center, Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Douglas B. Johnson
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| |
Collapse
|
13
|
Wiencke JK, Nissen E, Koestler DC, Tamaki SJ, Tamaki CM, Hansen HM, Warrier G, Hadad S, McCoy L, Rice T, Clarke J, Taylor JW, Salas LA, Christensen BC, Kelsey KT, Butler R, Molinaro AM. Enrichment of a neutrophil-like monocyte transcriptional state in glioblastoma myeloid suppressor cells. RESEARCH SQUARE 2023:rs.3.rs-3793353. [PMID: 38234734 PMCID: PMC10793488 DOI: 10.21203/rs.3.rs-3793353/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Glioblastomas (GBM) are lethal central nervous system cancers associated with tumor and systemic immunosuppression. Heterogeneous monocyte myeloid-derived suppressor cells (M-MDSC) are implicated in the altered immune response in GBM, but M-MDSC ontogeny and definitive phenotypic markers are unknown. Using single-cell transcriptomics, we revealed heterogeneity in blood M-MDSC from GBM subjects and an enrichment in a transcriptional state reminiscent of neutrophil-like monocytes (NeuMo), a newly described pathway of monopoiesis in mice. Human NeuMo gene expression and Neu-like deconvolution fraction algorithms were created to quantitate the enrichment of this transcriptional state in GBM subjects. NeuMo populations were also observed in M-MDSCs from lung and head and neck cancer subjects. Dexamethasone (DEX) and prednisone exposures increased the usage of Neu-like states, which were inversely associated with tumor purity and survival in isocitrate dehydrogenase wildtype (IDH WT) gliomas. Anti-inflammatory ZC3HA12/Regnase-1 transcripts were highly correlated with NeuMo expression in tumors and in blood M-MDSC from GBM, lung, and head and neck cancer subjects. Additional novel transcripts of immune-modulating proteins were identified. Collectively, these findings provide a framework for understanding the heterogeneity of M-MDSCs in GBM as cells with different clonal histories and may reshape approaches to study and therapeutically target these cells.
Collapse
Affiliation(s)
- J K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA
| | - Emily Nissen
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS
| | - Devin C Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS
| | - Stan J Tamaki
- Parnassus Flow Cytometry CoLab, University of California San Francisco, San Francisco, CA 94143-0511, USA
| | - Courtney M Tamaki
- Parnassus Flow Cytometry CoLab, University of California San Francisco, San Francisco, CA 94143-0511, USA
| | - Helen M Hansen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA
| | - Gayathri Warrier
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA
| | - Sara Hadad
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA
| | - Lucie McCoy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA
| | - Terri Rice
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA
| | - Jennifer Clarke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA
- Department of Neurology, University of California San Francisco, San Francisco, CA
| | - Jennie W Taylor
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA
- Department of Neurology, University of California San Francisco, San Francisco, CA
| | - Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH
- Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH
| | - Karl T Kelsey
- Department of Epidemiology, Brown University, Providence, RI
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI
| | - Rondi Butler
- Department of Epidemiology, Brown University, Providence, RI
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA
| |
Collapse
|
14
|
Boutros A, Carosio R, Campanella D, Spagnolo F, Banelli B, Morabito A, Pistillo M, Croce E, Cecchi F, Pronzato P, Queirolo P, Raposio E, Fontana V, Tanda E. The predictive and prognostic role of single nucleotide gene variants of PD-1 and PD-L1 in patients with advanced melanoma treated with PD-1 inhibitors. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 20:100408. [PMID: 38192613 PMCID: PMC10772261 DOI: 10.1016/j.iotech.2023.100408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Background Despite having revolutionized the treatment paradigm for advanced melanoma, not all patients benefit from immune checkpoint inhibitor therapy. To date, there are no predictive biomarkers for response or the occurrence of immune-related adverse events (irAEs) to programmed cell death protein 1 (PD-1) inhibitors. Our aim was to investigate the predictive and prognostic role of single nucleotide variants (SNVs) of genes involved in the PD-1 axis. Methods We analysed, in metastatic melanoma patients treated with nivolumab or pembrolizumab, five PD-1 SNVs, namely PD1.3 G>A (rs11568821), PD1.5 C>T (rs2227981), PD1.6 G>A (rs10204525), PD1.7 T>C(rs7421861), PD1.10 C>G (rs5582977) and three programmed death-ligand 1 (PD-L1) SNVs: +8293 C>A (rs2890658), PD-L1 C>T (rs2297136) and PD-L1 G>C (rs4143815). Association of SNV genotypic frequencies with best overall response to PD-1 inhibitors and development of irAEs were estimated through a modified Poisson regression. A Cox regression modelling approach was applied to evaluate the SNV association with OS. Results A total of 125 patients with advanced melanoma were included in the analysis. A reduction in irAEs risk was observed in patients carrying the PD-L1 +8293 C/A genotype compared with those carrying the C/C genotype (risk ratio = 0.45; 95% CL 0.22-0.93; P = 0.031). A trend for a reduction in irAEs was also observed with the PD1.5 T allele (risk ratio = 0.70, 95% confidence limits 0.48-1.01 versus C allele). None of the SNVs was associated with response to therapy. Finally, a survival benefit was observed in patients harbouring the PD1.7 C/C genotype (hazard ratio = 0.37; 95% confidence limits 0.14-0.96; P = 0.028) in the homozygous model. Conclusions Our study showed that PD-1.5 and PD-L1 +8293 SNVs may play a role as a predictive biomarker of development of irAEs to PD-1 inhibitors. PD1.7 SNV may also be associated with a reduction of the risk of death, although further translational research is needed to confirm these results.
Collapse
Affiliation(s)
- A. Boutros
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa
| | - R. Carosio
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - D. Campanella
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - F. Spagnolo
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Plastic Surgery Division, University of Genova, Genoa
| | - B. Banelli
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - A. Morabito
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - M.P. Pistillo
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - E. Croce
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa
| | - F. Cecchi
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
| | - P. Pronzato
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
| | - P. Queirolo
- Division of Melanoma Sarcoma and Rare Tumors, IRCCS European Institute of Oncology, Milan, Italy
| | - E. Raposio
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Plastic Surgery Division, University of Genova, Genoa
| | - V. Fontana
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - E.T. Tanda
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
| |
Collapse
|
15
|
Singh S, Singh N, Baranwal M, Sharma S, Devi SSK, Kumar S. Understanding immune checkpoints and PD-1/PD-L1-mediated immune resistance towards tumour immunotherapy. 3 Biotech 2023; 13:411. [PMID: 37997595 PMCID: PMC10663421 DOI: 10.1007/s13205-023-03826-2] [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: 12/28/2022] [Accepted: 10/18/2023] [Indexed: 11/25/2023] Open
Abstract
Immunotherapy has emerged as a transformative approach in the treatment of various cancers, offering new hope for patients previously faced with limited treatment options. A cornerstone of cancer immunotherapy lies in targeting immune checkpoints, particularly the programmed cell death protein-1 (PD-1) and programmed death-ligand 1 (PD-L1) pathway. Immune checkpoints serve as crucial regulators of the immune response, preventing excessive immune activity and maintaining self-tolerance. PD-1, expressed on the surface of T cells, and its ligand PD-L1, expressed on various cell types, including cancer cells and immune cells, play a central role in this regulatory process. Although the success rate associated with these immunotherapies is very promising, most patients still show intrinsic or acquired resistance. Since the mechanisms related to PD-1/PD-L1 resistance are not well understood, an in-depth analysis is necessary to improve the success rate of anti-PD-1/PD-L1 therapy. Hence, here we provide an overview of PD-1, its ligand PD-L1, and the resistance mechanism towards PD-1/PD-L1. Furthermore, we have discussed the plausible solution to increase efficacy and clinical response. For the following research, joint endeavours of clinicians and basic scientists are essential to address the limitation of resistance towards immunotherapy.
Collapse
Affiliation(s)
- Sidhartha Singh
- School of Bioscience and Bioengineering, D Y Patil International University, Pune, Maharastra 411051 India
| | - Navneet Singh
- Department of Pulmonary Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012 India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, 147004 India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, 147004 India
| | - S. S. Kirthiga Devi
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037 India
| | - Sandeep Kumar
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037 India
| |
Collapse
|
16
|
Zhang Q, Chen X, Palen K, Johnson B, Bui D, Xiong D, Pan J, Hu M, Wang Y, You M. Cancer chemoprevention with PV-1, a novel Prunella vulgaris-containing herbal mixture that remodels the tumor immune microenvironment in mice. Front Immunol 2023; 14:1196434. [PMID: 38077406 PMCID: PMC10704350 DOI: 10.3389/fimmu.2023.1196434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/20/2023] [Indexed: 12/18/2023] Open
Abstract
The herb Prunella vulgaris has shown significant immune-stimulatory and anti-inflammatory effects in mouse models. Here, the effects of a novel Prunella vulgaris-containing herbal mixture, PV-1, were examined in several mouse models for cancer, including chemically induced models of lung and oral cancers as well as syngraft models for lung cancer and melanoma. PV-1, consisting of extracts from Prunella vulgaris, Polygonum bistorta, Sonchus brachyotus and Dictamnus dasycarpus, exhibited no toxicity in a dose escalation study in A/J mice. PV-1 significantly inhibited mouse lung tumor development induced by the lung carcinogens vinyl carbamate and benzo[a]pyrene. PV-1 also hindered the induction of oral squamous cell carcinomas in C57BL/6 mice caused by 4-nitroquinoline-1-oxide. Flow cytometry analysis showed that PV-1 increased the numbers of CD8+ tumor-infiltrating lymphocytes (TILs) and increased the production of granzyme B, TNF-α, and IFN-γ by CD8+ TILs. PV-1 also suppressed granulocytic myeloid-derived suppressor cell numbers (g-MDSCs) and improved the anti-cancer activity of anti-PD-1 immunotherapy. These results indicate that PV-1 remodels the tumor immune microenvironment by selectively inhibiting g-MDSCs and increasing CD8+ TILs within tumors, resulting in decreased immune suppression and enhanced cancer chemopreventive efficacy.
Collapse
Affiliation(s)
- Qi Zhang
- Center for Cancer Prevention, Dr. Mary and Ron Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, United States
| | - Xu Chen
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Katie Palen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bryon Johnson
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Dinh Bui
- College of Pharmacy, University of Houston, Houston, TX, United States
| | - Donghai Xiong
- Center for Cancer Prevention, Dr. Mary and Ron Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, United States
| | - Jing Pan
- Center for Cancer Prevention, Dr. Mary and Ron Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, United States
| | - Ming Hu
- College of Pharmacy, University of Houston, Houston, TX, United States
| | - Yian Wang
- Center for Cancer Prevention, Dr. Mary and Ron Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, United States
| | - Ming You
- Center for Cancer Prevention, Dr. Mary and Ron Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
17
|
Chen N, Xu X, Fan Y. Immune checkpoint inhibitors in the treatment of oesophageal squamous cell carcinoma: where are we and where are we going? Ther Adv Med Oncol 2023; 15:17588359231189420. [PMID: 37547447 PMCID: PMC10399266 DOI: 10.1177/17588359231189420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/05/2023] [Indexed: 08/08/2023] Open
Abstract
Oesophageal squamous cell carcinoma (ESCC) is a kind of malignant tumour with high invasiveness and a poor prognosis. Immunotherapy, especially immune checkpoint inhibitors (ICIs), is a rapidly growing therapeutic method that activates and enhances anti-tumour immunity to treat patients with malignancy. Several clinical trials have confirmed the efficacy of ICIs in the treatment of ESCC. ICIs have been approved for the treatment of patients with ESCC. However, only a subset of patients can obtain excellent benefits from ICI therapy. In recent years, there has been a growing interest in exploring predictive biomarkers of immunotherapy response. In this review, we highlighted the predictive biomarkers for the prognosis of ESCC patients treated with ICIs and pointed out the existing problems and the direction of future research in this field.
Collapse
Affiliation(s)
- Ning Chen
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiaoling Xu
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| | - Yun Fan
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
18
|
Ma KSK, Tsai PF, Hsieh TYJ, Chodosh J. Ocular surface complications following biological therapy for cancer. FRONTIERS IN TOXICOLOGY 2023; 5:1137637. [PMID: 37424746 PMCID: PMC10324604 DOI: 10.3389/ftox.2023.1137637] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Novel and highly effective biological agents developed to treat cancer over the past two decades have also been linked to multiple adverse outcomes, including unanticipated consequences for the cornea. This review provides an overview of adverse corneal complications of biological agents currently in use for the treatment of cancer. Epidermal growth factor receptor inhibitors and immune checkpoint inhibitors are the two classes of biological agents most frequently associated with corneal adverse events. Dry eye, Stevens-Johnson syndrome, and corneal transplant rejection have all been reported following the use of immune checkpoint inhibitors. The management of these adverse events requires close collaboration between ophthalmologists, dermatologists, and oncologists. This review focuses in depth on the epidemiology, pathophysiology, and management of ocular surface complications of biological therapies against cancer.
Collapse
Affiliation(s)
- Kevin Sheng-Kai Ma
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Ping-Feng Tsai
- Department of Ophthalmology, Tri-Service General Hospital, Taipei, Taiwan
| | - Tina Yi-Jin Hsieh
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - James Chodosh
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| |
Collapse
|
19
|
Peng Z, Li M, Li H, Gao Q. PD-1/PD-L1 immune checkpoint blockade in ovarian cancer: dilemmas and opportunities. Drug Discov Today 2023:103666. [PMID: 37302543 DOI: 10.1016/j.drudis.2023.103666] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized treatment in oncology. Antibodies against PD-1/PD-L1 and ICI-based combinations are under clinical investigations in multiple cancers, including ovarian cancer. However, the success of ICIs has not materialized in ovarian cancer, which remains one of the few malignancies where ICIs exhibit modest efficacy as either monotherapy or combination therapy. In this review, we summarize completed and ongoing clinical trials of PD-1/PD-L1 blockade in ovarian cancer, categorize the underlying mechanisms of resistance emergence, and introduce candidate approaches to rewire the tumor microenvironment (TME) to potentiate anti-PD-1/PD-L1 antibodies. Teaser: The intrinsic resistance of ovarian cancer to PD-1/PD-L1 blockade could be overcome by advanced understanding of underlying mechanisms and discoveries of new actionable targets for combinatory treatment.
Collapse
Affiliation(s)
- Zikun Peng
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Li
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huayi Li
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinglei Gao
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
20
|
Gaißler A, Bochem J, Spreuer J, Ottmann S, Martens A, Amaral T, Wagner NB, Claassen M, Meier F, Terheyden P, Garbe C, Eigentler T, Weide B, Pawelec G, Wistuba-Hamprecht K. Early decrease of blood myeloid-derived suppressor cells during checkpoint inhibition is a favorable biomarker in metastatic melanoma. J Immunother Cancer 2023; 11:e006802. [PMID: 37286306 PMCID: PMC10254874 DOI: 10.1136/jitc-2023-006802] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND The need for reliable clinical biomarkers to predict which patients with melanoma will benefit from immune checkpoint blockade (ICB) remains unmet. Several different parameters have been considered in the past, including routine differential blood counts, T cell subset distribution patterns and quantification of peripheral myeloid-derived suppressor cells (MDSC), but none has yet achieved sufficient accuracy for clinical utility. METHODS Here, we investigated potential cellular biomarkers from clinical routine blood counts as well as several myeloid and T cell subsets, using flow cytometry, in two independent cohorts of a total of 141 patients with stage IV M1c melanoma before and during ICB. RESULTS Elevated baseline frequencies of monocytic MDSCs (M-MDSC) in the blood were confirmed to predict shorter overall survival (OS) (HR 2.086, p=0.030) and progression-free survival (HR 2.425, p=0.001) in the whole patient cohort. However, we identified a subgroup of patients with highly elevated baseline M-MDSC frequencies that fell below a defined cut-off during therapy and found that these patients had a longer OS that was similar to that of patients with low baseline M-MDSC frequencies. Importantly, patients with high M-MDSC frequencies exhibited a skewed baseline distribution of certain other immune cells but these did not influence patient survival, illustrating the paramount utility of MDSC assessment. CONCLUSION We confirmed that in general, highly elevated frequencies of peripheral M-MDSC are associated with poorer outcomes of ICB in metastatic melanoma. However, one reason for an imperfect correlation between high baseline MDSCs and outcome for individual patients may be the subgroup of patients identified here, with rapidly decreasing M-MDSCs on therapy, in whom the negative effect of high M-MDSC frequencies was lost. These findings might contribute to developing more reliable predictors of late-stage melanoma response to ICB at the individual patient level. A multifactorial model seeking such markers yielded only MDSC behavior and serum lactate dehydrogenase as predictors of treatment outcome.
Collapse
Affiliation(s)
- Andrea Gaißler
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Jonas Bochem
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Janine Spreuer
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Shannon Ottmann
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Alexander Martens
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Teresa Amaral
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Tübingen, Germany
| | - Nikolaus Benjamin Wagner
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Department of Dermatology, Venereology and Allergology, Kantonsspital St Gallen, Sankt Gallen, Switzerland
| | - Manfred Claassen
- Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Department of Computer Science, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Friedegund Meier
- Skin Cancer Center at the University Cancer Centre and National Center for Tumor Diseases Dresden; Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Claus Garbe
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Thomas Eigentler
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Dermatology, Venereology and Allergology, Berlin, Germany
| | - Benjamin Weide
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Graham Pawelec
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany
- Health Sciences North Research Institute, Sudbury, Ontario, Canada
| | - Kilian Wistuba-Hamprecht
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
21
|
Magagnoli J, Narendran S, Pereira F, Cummings TH, Hardin JW, Sutton SS, Ambati J. Association between Fluoxetine Use and Overall Survival among Patients with Cancer Treated with PD-1/L1 Immunotherapy. Pharmaceuticals (Basel) 2023; 16:ph16050640. [PMID: 37242422 DOI: 10.3390/ph16050640] [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: 03/14/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/28/2023] Open
Abstract
Checkpoint inhibitors can be a highly effective antitumor therapy but only to a subset of patients, presumably due to immunotherapy resistance. Fluoxetine was recently revealed to inhibit the NLRP3 inflammasome, and NLRP3 inhibition could serve as a target for immunotherapy resistance. Therefore, we evaluated the overall survival (OS) in patients with cancer receiving checkpoint inhibitors combined with fluoxetine. A cohort study was conducted among patients diagnosed with lung, throat (pharynx or larynx), skin, or kidney/urinary cancer treated with checkpoint inhibitor therapy. Utilizing the Veterans Affairs Informatics and Computing Infrastructure, patients were retrospectively evaluated during the period from October 2015 to June 2021. The primary outcome was overall survival (OS). Patients were followed until death or the end of the study period. There were 2316 patients evaluated, including 34 patients who were exposed to checkpoint inhibitors and fluoxetine. Propensity score weighted Cox proportional hazards demonstrated a better OS in fluoxetine-exposed patients than unexposed (HR: 0.59, 95% CI 0.371-0.936). This cohort study among cancer patients treated with checkpoint inhibitor therapy showed a significant improvement in the OS when fluoxetine was used. Because of this study's potential for selection bias, randomized trials are needed to assess the efficacy of the association of fluoxetine or another anti-NLRP3 drug to checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Joseph Magagnoli
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC 29209, USA
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Siddharth Narendran
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Felipe Pereira
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Tammy H Cummings
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC 29209, USA
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - James W Hardin
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC 29209, USA
- Department of Epidemiology & Biostatistics, University of South Carolina, Columbia, SC 29208, USA
| | - S Scott Sutton
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC 29209, USA
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Jayakrishna Ambati
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| |
Collapse
|
22
|
Tobin RP, Cogswell DT, Cates VM, Davis DM, Borgers JS, Van Gulick RJ, Katsnelson E, Couts KL, Jordan KR, Gao D, Davila E, Medina TM, Lewis KD, Gonzalez R, McFarland RW, Robinson WA, McCarter MD. Targeting MDSC Differentiation Using ATRA: A Phase I/II Clinical Trial Combining Pembrolizumab and All-Trans Retinoic Acid for Metastatic Melanoma. Clin Cancer Res 2023; 29:1209-1219. [PMID: 36378549 PMCID: PMC10073240 DOI: 10.1158/1078-0432.ccr-22-2495] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/03/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE A phase Ib/II clinical trial was conducted to evaluate the safety and efficacy of the combination of all-trans retinoic acid (ATRA) with pembrolizumab in patients with stage IV melanoma. PATIENTS AND METHODS Anti-PD-1 naïve patients with stage IV melanoma were treated with pembrolizumab plus supplemental ATRA for three days surrounding each of the first four pembrolizumab infusions. The primary objective was to establish the MTD and recommended phase II dose (RP2D) of the combination. The secondary objectives were to describe the safety and toxicity of the combined treatment and to assess antitumor activity in terms of (i) the reduction in circulating myeloid-derived suppressor cell (MDSC) frequency and (ii) progression-free survival (PFS). RESULTS Twenty-four patients were enrolled, 46% diagnosed with M1a and 29% with M1c stage disease at enrollment. All patients had an ECOG status ≤1, and 75% had received no prior therapies. The combination was well tolerated, with the most common ATRA-related adverse events being headache, fatigue, and nausea. The RP2D was established at 150 mg/m2 ATRA + 200 mg Q3W pembrolizumab. Median PFS was 20.3 months, and the overall response rate was 71%, with 50% of patients experiencing a complete response, and the 1-year overall survival was 80%. The combination effectively lowered the frequency of circulating MDSCs. CONCLUSIONS With a favorable tolerability and high response rate, this combination is a promising frontline treatment strategy for advanced melanoma. Targeting MDSCs remains an attractive mechanism to enhance the efficacy of immunotherapies, and this combination merits further investigation. See related commentary by Olson and Luke, p. 1167.
Collapse
Affiliation(s)
- Richard P. Tobin
- University of Colorado Anschutz Medical Campus, Department of Surgery, Division of Surgical Oncology, Aurora, Colorado, USA
| | - Dasha T. Cogswell
- University of Colorado Anschutz Medical Campus, Department of Surgery, Division of Surgical Oncology, Aurora, Colorado, USA
| | - Victoria M. Cates
- University of Colorado Anschutz Medical Campus, Department of Surgery, Division of Surgical Oncology, Aurora, Colorado, USA
| | - Dana M. Davis
- University of Colorado Anschutz Medical Campus, Department of Surgery, Division of Surgical Oncology, Aurora, Colorado, USA
| | - Jessica S.W. Borgers
- University of Colorado Anschutz Medical Campus, Department of Surgery, Division of Surgical Oncology, Aurora, Colorado, USA
- Netherlands Cancer Institute, Department of Medical Oncology, Amsterdam, The Netherlands
| | - Robert J. Van Gulick
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Medical Oncology, Aurora, Colorado, USA
| | - Elizabeth Katsnelson
- University of Colorado Anschutz Medical Campus, Department of Surgery, Division of Surgical Oncology, Aurora, Colorado, USA
| | - Kasey L. Couts
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Medical Oncology, Aurora, Colorado, USA
| | - Kimberly R. Jordan
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, Colorado, USA
| | - Dexiang Gao
- University of Colorado Anschutz Medical Campus, Pediatrics, Biostatistics and Informatics, Cancer Center Biostatistics Core, Aurora, Colorado, USA
| | - Eduardo Davila
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Medical Oncology, Aurora, Colorado, USA
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, Colorado, USA
| | - Theresa M. Medina
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Medical Oncology, Aurora, Colorado, USA
| | - Karl D. Lewis
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Medical Oncology, Aurora, Colorado, USA
| | - Rene Gonzalez
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Medical Oncology, Aurora, Colorado, USA
| | - Ross W. McFarland
- UCHealth Cancer Care and Hematology Clinic - Harmony Campus, Fort Collins, Colorado, USA
| | - William A. Robinson
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Medical Oncology, Aurora, Colorado, USA
| | - Martin D. McCarter
- University of Colorado Anschutz Medical Campus, Department of Surgery, Division of Surgical Oncology, Aurora, Colorado, USA
| |
Collapse
|
23
|
Zhao Y, Du J, Shen X. Targeting myeloid-derived suppressor cells in tumor immunotherapy: Current, future and beyond. Front Immunol 2023; 14:1157537. [PMID: 37006306 PMCID: PMC10063857 DOI: 10.3389/fimmu.2023.1157537] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are one of the major negative regulators in tumor microenvironment (TME) due to their potent immunosuppressive capacity. MDSCs are the products of myeloid progenitor abnormal differentiation in bone marrow, which inhibits the immune response mediated by T cells, natural killer cells and dendritic cells; promotes the generation of regulatory T cells and tumor-associated macrophages; drives the immune escape; and finally leads to tumor progression and metastasis. In this review, we highlight key features of MDSCs biology in TME that are being explored as potential targets for tumor immunotherapy. We discuss the therapies and approaches that aim to reprogram TME from immunosuppressive to immunostimulatory circumstance, which prevents MDSC immunosuppression activity; promotes MDSC differentiation; and impacts MDSC recruitment and abundance in tumor site. We also summarize current advances in the identification of rational combinatorial strategies to improve clinical efficacy and outcomes of cancer patients, via deeply understanding and pursuing the mechanisms and characterization of MDSCs generation and suppression in TME.
Collapse
Affiliation(s)
- Yang Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Junfeng Du
- Department of General Surgery, The 7th Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: Junfeng Du, ; Xiaofei Shen,
| | - Xiaofei Shen
- Department of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Junfeng Du, ; Xiaofei Shen,
| |
Collapse
|
24
|
Tomela K, Pietrzak B, Galus Ł, Mackiewicz J, Schmidt M, Mackiewicz AA, Kaczmarek M. Myeloid-Derived Suppressor Cells (MDSC) in Melanoma Patients Treated with Anti-PD-1 Immunotherapy. Cells 2023; 12:cells12050789. [PMID: 36899926 PMCID: PMC10000540 DOI: 10.3390/cells12050789] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/18/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are a subset of immature myeloid cells with suppressive activity well described in the context of cancer. They inhibit anti-tumour immunity, promote metastasis formation and can lead to immune therapy resistance. In a retrospective study, blood probes of 46 advanced melanoma patients were analysed before the first administration of anti-PD-1 immunotherapy and in the third month of treatment for MDSC, immature monocytic (ImMC), monocytic MDSC (MoMDSC) and granulocytic MDSC (GrMDSC) by multi-channel flow cytometry. Cell frequencies were correlated with response to immunotherapy, progression-free survival (PFS) and lactate dehydrogenase (LDH) serum level. Responders to anti-PD-1 therapy had higher MoMDSC levels (4.1 ± 1.2%) compared to non-responders (3.0 ± 1.2%) (p = 0.0333) before the first administration of anti-PD-1. No significant changes in MDSCs frequencies were observed in the groups of patients before and in the third month of therapy. The cut-off values of MDSCs, MoMDSCs, GrMDSCs and ImMCs for favourable 2- and 3-year PFS were established. Elevated LDH level is a negative prognostic factor of response to the treatment and is related to an elevated ratio of GrMDSCs and ImMCs level compared to patients' LDH level below the cut-off. Our data may provide a new perspective for more careful consideration of MDSCs, and specially MoMDSCs, as a tool for monitoring the immune status of melanoma patients. Changes in MDSC levels may have a potential prognostic value, however a correlation with other parameters must be established.
Collapse
Affiliation(s)
- Katarzyna Tomela
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
- Correspondence:
| | - Bernadeta Pietrzak
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland
| | - Łukasz Galus
- Department of Medical and Experimental Oncology, Institute of Oncology, University of Medical Sciences, 60-355 Poznan, Poland
| | - Jacek Mackiewicz
- Department of Medical and Experimental Oncology, Institute of Oncology, University of Medical Sciences, 60-355 Poznan, Poland
| | - Marcin Schmidt
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland
| | - Andrzej Adam Mackiewicz
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| |
Collapse
|
25
|
Matos I, Barvalia M, Chehal MK, Robertson AG, Kulic I, Silva JAFD, Ranganathan A, Short A, Huang YH, Long E, Priatel JJ, Dhanji S, Nelson BH, Krebs DL, Harder KW. Tumor-derived GCSF Alters Tumor and Systemic Immune System Cell Subset Composition and Signaling. CANCER RESEARCH COMMUNICATIONS 2023; 3:404-419. [PMID: 36911097 PMCID: PMC9997410 DOI: 10.1158/2767-9764.crc-22-0278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/01/2022] [Accepted: 02/09/2023] [Indexed: 02/17/2023]
Abstract
While immunotherapies such as immune checkpoint blockade and adoptive T-cell therapy improve survival for a subset of human malignancies, many patients fail to respond. Phagocytes including dendritic cells (DC), monocytes, and macrophages (MF) orchestrate innate and adaptive immune responses against tumors. However, tumor-derived factors may limit immunotherapy effectiveness by altering phagocyte signal transduction, development, and activity. Using Cytometry by Time-of-Flight, we found that tumor-derived GCSF altered myeloid cell distribution both locally and systemically. We distinguished a large number of GCSF-induced immune cell subset and signal transduction pathway perturbations in tumor-bearing mice, including a prominent increase in immature neutrophil/myeloid-derived suppressor cell (Neut/MDSC) subsets and tumor-resident PD-L1+ Neut/MDSCs. GCSF expression was also linked to distinct tumor-associated MF populations, decreased conventional DCs, and splenomegaly characterized by increased splenic progenitors with diminished DC differentiation potential. GCSF-dependent dysregulation of DC development was recapitulated in bone marrow cultures in vitro, using medium derived from GCSF-expressing tumor cell cultures. Importantly, tumor-derived GCSF impaired T-cell adoptive cell therapy effectiveness and was associated with increased tumor volume and diminished survival of mice with mammary cancer. Treatment with neutralizing anti-GCSF antibodies reduced colonic and circulatory Neut/MDSCs, normalized colonic immune cell composition and diminished tumor burden in a spontaneous model of mouse colon cancer. Analysis of human colorectal cancer patient gene expression data revealed a significant correlation between survival and low GCSF and Neut/MDSC gene expression. Our data suggest that normalizing GCSF bioactivity may improve immunotherapy in cancers associated with GCSF overexpression. Significance Tumor-derived GCSF leads to systemic immune population changes. GCSF blockade restores immune populations, improves immunotherapy, and reduces tumor size, paralleling human colorectal cancer data. GCSF inhibition may synergize with current immunotherapies to treat GCSF-secreting tumors.
Collapse
Affiliation(s)
- Israel Matos
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Maunish Barvalia
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Manreet K Chehal
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - A Gordon Robertson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency. Vancouver, British Columbia, Canada
| | - Iva Kulic
- ME Therapeutics Inc. Vancouver, British Columbia, Canada
| | - Jessica A F D Silva
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Abhinandan Ranganathan
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Amy Short
- ME Therapeutics Inc. Vancouver, British Columbia, Canada
| | - Yu-Hsuan Huang
- ME Therapeutics Inc. Vancouver, British Columbia, Canada
| | - Erin Long
- ME Therapeutics Inc. Vancouver, British Columbia, Canada
| | - John J Priatel
- ME Therapeutics Inc. Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Salim Dhanji
- ME Therapeutics Inc. Vancouver, British Columbia, Canada
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - Danielle L Krebs
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Kenneth W Harder
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada.,ME Therapeutics Inc. Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
The Receptor for Advanced Glycation Endproducts (RAGE) and Its Ligands S100A8/A9 and High Mobility Group Box Protein 1 (HMGB1) Are Key Regulators of Myeloid-Derived Suppressor Cells. Cancers (Basel) 2023; 15:cancers15041026. [PMID: 36831371 PMCID: PMC9954573 DOI: 10.3390/cancers15041026] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/27/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Immunotherapies including checkpoint blockade immunotherapy (CBI) and chimeric antigen receptor T cells (CAR-T) have revolutionized cancer treatment for patients with certain cancers. However, these treatments are not effective for all cancers, and even for those cancers that do respond, not all patients benefit. Most cancer patients have elevated levels of myeloid-derived suppressor cells (MDSCs) that are potent inhibitors of antitumor immunity, and clinical and animal studies have demonstrated that neutralization of MDSCs may restore immune reactivity and enhance CBI and CAR-T immunotherapies. MDSCs are homeostatically regulated in that elimination of mature circulating and intratumoral MDSCs results in increased production of MDSCs from bone marrow progenitor cells. Therefore, targeting MDSC development may provide therapeutic benefit. The pro-inflammatory molecules S100A8/A9 and high mobility group box protein 1 (HMGB1) and their receptor RAGE are strongly associated with the initiation and progression of most cancers. This article summarizes the literature demonstrating that these molecules are integrally involved in the early development, accumulation, and suppressive activity of MDSCs, and postulates that S100A8/A9 and HMGB1 serve as early biomarkers of disease and in conjunction with RAGE are potential targets for reducing MDSC levels and enhancing CBI and CAR-T immunotherapies.
Collapse
|
27
|
Petrova V, Groth C, Bitsch R, Arkhypov I, Simon SCS, Hetjens S, Müller V, Utikal J, Umansky V. Immunosuppressive capacity of circulating MDSC predicts response to immune checkpoint inhibitors in melanoma patients. Front Immunol 2023; 14:1065767. [PMID: 36860876 PMCID: PMC9968744 DOI: 10.3389/fimmu.2023.1065767] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/27/2023] [Indexed: 02/15/2023] Open
Abstract
Purpose Although the treatment of advanced melanoma patients with immune checkpoint inhibitors (ICI) significantly increased the therapeutic efficiency, many patients remain resistant to ICI that could be due to immunosuppression mediated by myeloid-derived suppressor cells (MDSC). These cells are enriched and activated in melanoma patients and could be considered as therapeutic targets. Here we studied dynamic changes in immunosuppressive pattern and activity of circulating MDSC from melanoma patients treated with ICI. Experimental design MDSC frequency, immunosuppressive markers and function were evaluated in freshly isolated peripheral blood mononuclear cells (PBMC) from 29 melanoma patients receiving ICI. Blood samples were taken prior and during the treatment and analyzed by flow cytometry and bio-plex assay. Results MDSC frequency was significantly increased before the therapy and through three months of treatment in non-responders as compared to responders. Prior to the ICI therapy, MDSC from non-responders displayed high levels of immunosuppression measured by the inhibition of T cell proliferation assay, whereas MDSC from responding patients failed to inhibit T cells. Patients without visible metastasis were characterized by the absence of MDSC immunosuppressive activity during the ICI treatment. Moreover, non-responders showed significantly higher IL-6 and IL-8 concentrations before therapy and after the first ICI application as compared to responders. Conclusions Our findings highlight the role of MDSC during melanoma progression and suggest that frequency and immunosuppressive activity of circulating MDSC before and during the ICI treatment of melanoma patients could be used as biomarkers of response to ICI therapy.
Collapse
Affiliation(s)
- Vera Petrova
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,DKFZ-Hector Cancer Institute at the University Medical Centre Mannheim, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christopher Groth
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,DKFZ-Hector Cancer Institute at the University Medical Centre Mannheim, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rebekka Bitsch
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,DKFZ-Hector Cancer Institute at the University Medical Centre Mannheim, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ihor Arkhypov
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,DKFZ-Hector Cancer Institute at the University Medical Centre Mannheim, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Sonja C S Simon
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Svetlana Hetjens
- Department of Medical Statistics and Biomathematics, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Verena Müller
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,DKFZ-Hector Cancer Institute at the University Medical Centre Mannheim, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,DKFZ-Hector Cancer Institute at the University Medical Centre Mannheim, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
28
|
Zhang F, Hu K, Liu W, Quan B, Li M, Lu S, Chen R, Ren Z, Yin X. Oxaliplatin-Resistant Hepatocellular Carcinoma Drives Immune Evasion Through PD-L1 Up-Regulation and PMN-Singular Recruitment. Cell Mol Gastroenterol Hepatol 2023; 15:573-591. [PMID: 36513250 PMCID: PMC9868681 DOI: 10.1016/j.jcmgh.2022.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Previously, we showed the inhibitor of differentiation or DNA binding 1 (ID1)/Myc signaling is highly expressed in oxaliplatin-resistant hepatocellular carcinoma (HCC). This study sought to investigate the role of ID1/Myc signaling on immune evasion in oxaliplatin-resistant HCC. METHODS The oxaliplatin (OXA)-resistant HCC cell lines (Hepa 1-6-OXA, 97H-OXA, and 3B-OXA) were established and their oxaliplatin tolerance was confirmed in vitro and in vivo. The relationship between ID1/Myc and programmed death-ligand 1 (PD-L1) up-regulation and polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC) accumulation was explored. The underlying mechanism in which ID1/Myc signaling regulated PD-L1 expression and PMN-MDSC accumulation was investigated in vitro and vivo. RESULTS Increased ID1/Myc expression was identified in oxaliplatin-resistant HCC and correlated with PD-L1 up-regulation and PMN-MDSC accumulation. The knockdown of Myc sensitized oxaliplatin-resistant HCC cells to oxaliplatin and resulted in a decrease of PMN-MDSCs and an increase of interferon-γ+ CD8+ T cells in a tumor microenvironment. Polymerase chain reaction array, enzyme-linked immunosorbent assay, and MDSC Transwell migration assay indicated that oxaliplatin-resistant HCC cells recruited PMN-MDSCs through chemokine (C-C motif) ligand 5 (CCL5). The dual luciferase reporter assay and chromatin immunoprecipitation assay indicated that Myc could directly increase the transcriptions of PD-L1 and CCL5. Furthermore, anti-PD-L1 antibody combined with CCL5 blockade showed significant antitumor effects in oxaliplatin-resistant HCC. CONCLUSIONS ID1/Myc signaling drives immune evasion in oxaliplatin-resistant HCC via PD-L1 up-regulation and PMN-MDSC recruitment. Blocking the ID1/Myc-induced immune tolerance represents a promising treatment target to conquer chemoresistance in HCC.
Collapse
Affiliation(s)
- Feng Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Keshu Hu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenfeng Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bing Quan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Miao Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shenxin Lu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongxin Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenggang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Yin
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
29
|
Ding Z, Zhang Y. Differentiation and Immunological Function of MDSC-Derived Dendritic Cells. Glob Med Genet 2022; 9:290-299. [PMID: 36567953 PMCID: PMC9771685 DOI: 10.1055/s-0042-1756659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/06/2022] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells (DCs) play a key role in initiating and regulating immune responses, and in addition to their roles in vivo, DCs are used as natural adjuvants for various tumor vaccines. In vitro, monocytes can be used to induce DCs, but in tumor patients, due to insufficient bone marrow hematopoiesis, extramedullary hematopoiesis and tumor-associated myeloid cells expand, and monocytes mainly exist in the form of myeloid-derived suppressor cells (MDSCs). The purpose of this experiment was to explore the differences in the differentiation and immune function of DCs induced by MDSCs in tumor patients. In a mouse model, we used normal mouse bone marrow cell-derived DCs as control cells, and in a tumor-bearing model, we induced MDSCs in the spleen to generate DCs (MDSC-DCs). Through flow cytometry, we found that the production of MDSC-DCs was significantly higher than that of control mice, and the secretion of interferon-γ of MDSC-DCs was significantly reduced. Through OVA antigen presentation experiments, we found that the antigen presentation ability of MDSC-DCs was significantly decreased. Through adoptive treatment of tumor-bearing mice cells, we found that the antitumor immune function of MDSC-DCs was significantly reduced. After that, we explored the mechanism of the decrease of immune function activity of MDSC-DCs. We determined that the surface markers of MDSC-DCs were changed by flow cytometry. Through flow sorting and RNA sequencing, we found that some pathways and key gene expression in MDSC-DCs were changed. In conclusion, this study found that the immune function of MDSC-DCs decreased and explored the mechanism of the decreased immune function activity.
Collapse
Affiliation(s)
- Zequn Ding
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China,Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China,Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China,Address for correspondence Yan Zhang, PhD Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong UniversityNo.1954 Huashan Road, ShanghaiChina, 200127
| |
Collapse
|
30
|
Burkert SC, He X, Shurin GV, Nefedova Y, Kagan VE, Shurin MR, Star A. Nitrogen-Doped Carbon Nanotube Cups for Cancer Therapy. ACS APPLIED NANO MATERIALS 2022; 5:13685-13696. [PMID: 36711215 PMCID: PMC9879341 DOI: 10.1021/acsanm.1c03245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Carbon nanomaterials have attracted significant attention for a variety of biomedical applications including sensing and detection, photothermal therapy, and delivery of therapeutic cargo. The ease of chemical functionalization, tunable length scales and morphologies, and ability to undergo complete enzymatic degradation make carbon nanomaterials an ideal drug delivery system. Much work has been done to synthesize carbon nanomaterials ranging from carbon dots, graphene, and carbon nanotubes to carbon nanocapsules, specifically carbon nanohorns or nitrogen-doped carbon nanocups. Here, we analyze specific properties of nitrogen-doped carbon nanotube cups which have been designed and utilized as drug delivery systems with the focus on the loading of these nanocapsules with specific therapeutic cargo and the targeted delivery for cancer therapy. We also summarize our targeted synthesis of gold nanoparticles on the open edge of nitrogen-doped carbon nanotube cups to create loaded and sealed nanocarriers for the delivery of chemotherapeutic agents to myeloid regulatory cells responsible for the immunosuppressive properties of the tumor microenvironment and thus tumor immune escape.
Collapse
Affiliation(s)
- Seth C. Burkert
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department of Chemistry, Franklin & Marshall College, Lancaster, Pennsylvania 17604, United States
| | - Xiaoyun He
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Galina V. Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15261, United States
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15261, United States
| | - Yulia Nefedova
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Valerian E. Kagan
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Michael R. Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15261, United States
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15261, United States
| | - Alexander Star
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department of Bioengineering, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15261, United States
- Corresponding author: Alexander Star —Department of Chemistry and Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States;
| |
Collapse
|
31
|
Li J, Li X, Guo Q. Drug Resistance in Cancers: A Free Pass for Bullying. Cells 2022; 11:3383. [PMID: 36359776 PMCID: PMC9654341 DOI: 10.3390/cells11213383] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 08/13/2023] Open
Abstract
The cancer burden continues to grow globally, and drug resistance remains a substantial challenge in cancer therapy. It is well established that cancerous cells with clonal dysplasia generate the same carcinogenic lesions. Tumor cells pass on genetic templates to subsequent generations in evolutionary terms and exhibit drug resistance simply by accumulating genetic alterations. However, recent evidence has implied that tumor cells accumulate genetic alterations by progressively adapting. As a result, intratumor heterogeneity (ITH) is generated due to genetically distinct subclonal populations of cells coexisting. The genetic adaptive mechanisms of action of ITH include activating "cellular plasticity", through which tumor cells create a tumor-supportive microenvironment in which they can proliferate and cause increased damage. These highly plastic cells are located in the tumor microenvironment (TME) and undergo extreme changes to resist therapeutic drugs. Accordingly, the underlying mechanisms involved in drug resistance have been re-evaluated. Herein, we will reveal new themes emerging from initial studies of drug resistance and outline the findings regarding drug resistance from the perspective of the TME; the themes include exosomes, metabolic reprogramming, protein glycosylation and autophagy, and the relates studies aim to provide new targets and strategies for reversing drug resistance in cancers.
Collapse
Affiliation(s)
| | | | - Qie Guo
- The Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
32
|
Arkhypov I, Özbay Kurt FG, Bitsch R, Novak D, Petrova V, Lasser S, Hielscher T, Groth C, Lepper A, Hu X, Li W, Utikal J, Altevogt P, Umansky V. HSP90α induces immunosuppressive myeloid cells in melanoma via TLR4 signaling. J Immunother Cancer 2022; 10:e005551. [PMID: 36113897 PMCID: PMC9486388 DOI: 10.1136/jitc-2022-005551] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Tumor cells modulate host immunity by secreting extracellular vesicles (EV) and soluble factors. Their interactions with myeloid cells lead to the generation of myeloid-derived suppressor cells (MDSC), which inhibit the antitumor function of T and NK cells. We demonstrated previously that EV derived from mouse and human melanoma cells induced immunosuppressive activity via increased expression of programmed cell death ligand 1 (PD-L1) on myeloid cells that was dependent on the heat-shock protein 90α (HSP90α) in EV. Here, we investigated whether soluble HSP90α could convert monocytes into MDSC. METHODS CD14 monocytes were isolated from the peripheral blood of healthy donors, incubated with human recombinant HSP90α (rHSP90α) alone or in the presence of inhibitors of TLR4 signaling and analyzed by flow cytometry. Inhibition of T cell proliferation assay was applied to assess the immunosuppressive function of rHSP90α-treated monocytes. HSP90α levels were measured by ELISA in plasma of patients with advanced melanoma and correlated with clinical outcome. RESULTS We found that the incubation of monocytes with rHSP90α resulted in a strong upregulation of PD-L1 expression, whereas reactive oxygen species (ROS) and nitric oxide (NO) production as well as the expression of arginase-1, ectoenzymes CD39 and CD73 remained unchanged. The PD-L1 upregulation was blocked by anti-TLR4 antibodies and a nuclear factor-κB inhibitor. rHSP90α-treated monocytes displayed the downregulation of HLA-DR expression and acquired the resistance to apoptosis. Moreover, these monocytes were converted into MDSC as indicated by their capacity to inhibit T cell proliferation, which was mediated by TLR4 signaling as well as PD-L1 and indoleamine 2,3-dioxygenase (IDO) 1 expression. Higher levels of HSP90α in plasma of patients with melanoma correlated with augmented PD-L1 expression on circulating monocytic (M)-MDSC. Patients with melanoma with high levels of HSP90α displayed shorter progression-free survival (PFS) on the treatment with immune checkpoint inhibitors (ICIs). CONCLUSION Our findings demonstrated that soluble rHSP90α increased the resistance of normal human monocytes to apoptosis and converted them into immunosuppressive MDSC via TLR4 signaling that stimulated PD-L1 and IDO-1 expression. Furthermore, patients with melanoma with high concentrations of HSP90α displayed increased PD-L1 expression on M-MDSC and reduced PFS after ICI therapy, suggesting HSP90α as a promising therapeutic target for overcoming immunosuppression in melanoma.
Collapse
Affiliation(s)
- Ihor Arkhypov
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Feyza Gül Özbay Kurt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rebekka Bitsch
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Daniel Novak
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Vera Petrova
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Samantha Lasser
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christopher Groth
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Alisa Lepper
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Xiaoying Hu
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Wei Li
- Department of Dermatology and the USC-Norris Comprehensive Cancer Centre, University of Southern California Keck Medical Center, Los Angeles, California, USA
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
33
|
Wu Y, Yu S, Qiao H. Understanding the functional inflammatory factors involved in therapeutic response to immune checkpoint inhibitors for pan-cancer. Front Pharmacol 2022; 13:990445. [PMID: 36120342 PMCID: PMC9474995 DOI: 10.3389/fphar.2022.990445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) fight tumor progression by activating immune conditions. The inflammatory factors are playing a functional role in programmed death-1 (PD-1) or other immune checkpoints. They are involved in regulating the expression of programmed death ligand-1 (PD-L1), the only predictor recognized by the guidelines in response to ICIs. In addition, abundant components of the tumor microenvironment (TME) all interact with various immune factors contributing to the response to ICIs, including infiltration of various immune cells, extracellular matrix, and fibroblasts. Notably, the occurrence of immune-related adverse events (irAEs) in patients receiving ICIs is increasingly observed in sundry organs. IrAEs are often regarded as an inflammatory factor-mediated positive feedback loop associated with better response to ICIs. It deserves attention because inflammatory factors were observed to be different when targeting different immune checkpoints or in the presence of different irAEs. In the present review, we address the research progresses on regulating inflammatory factors for an intentional controlling anti-cancer response with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Yanmeizhi Wu
- Department of Endocrinology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shan Yu
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Shan Yu, ; Hong Qiao,
| | - Hong Qiao
- Department of Endocrinology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Shan Yu, ; Hong Qiao,
| |
Collapse
|
34
|
Jakub JW, Weaver AL, Meves A. Association of tumor molecular factors with in-transit metastasis in primary cutaneous melanoma. Int J Dermatol 2022; 61:1117-1123. [PMID: 35246838 PMCID: PMC9391269 DOI: 10.1111/ijd.16141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/19/2021] [Accepted: 02/06/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND In-transit metastases (ITM) are a form of locoregional relapse representing intralymphatic metastatic spread and occur in approximately 4-9% of patients with melanoma >1 mm Breslow thickness. Our objective was to evaluate a combination of clinicopathologic risk factors and gene expression biomarkers predictive of ITM risk. METHODS We used PCR to quantify gene expression in diagnostic biopsy tissue across a prospectively designed archival cohort of 854 consecutive thin and intermediate thickness primary cutaneous melanomas. The outcome of interest was ITM >90 days after a melanoma diagnosis. Cox proportional hazard models were fit to estimate each clinicopathologic and molecular characteristic's association with the risk of ITM. RESULTS The 5-year cumulative incidence of ITM was 3.2%. Clinical factors univariately associated with an increased risk of ITM were older age, greater Breslow thickness, greater mitotic rate, lower extremity location, ulceration, and a positive SLN biopsy. Of 108 genes tested, five were significantly upregulated and five significantly downregulated when evaluated in Cox models adjusted for age, Breslow thickness, mitotic rate, and lower extremity location. Among the upregulated genes, the strongest association was observed for interleukin-8 (IL8). CONCLUSION A subset of gene expression biomarkers was identified as independently associated with the risk of ITM after adjusting for key covariates. Once sufficiently validated, our results may lead the way to regional therapy trials for a small, selected group of high-risk patients.
Collapse
Affiliation(s)
- James W. Jakub
- Department of Surgery, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Amy L. Weaver
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Alexander Meves
- Department of Dermatology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
35
|
Mandula JK, Rodriguez PC. Tumor-directed dysregulation of erythroid progenitors drives immunosuppressive myeloid cells. Cancer Cell 2022; 40:597-599. [PMID: 35594864 DOI: 10.1016/j.ccell.2022.04.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
In this issue of Cancer Cell, Long et al. demonstrate that tumors can reprogram erythroid progenitors into myeloid-erythroid cells that promote immunosuppression. Erythroid-differentiated myeloid cells (EDMCs) expand in cancer-bearing individuals, resemble the functionality of myeloid-derived suppressor cells (MDSCs), and correlate with poor response to immune-checkpoint inhibitors (ICIs) and tumor-related anemia.
Collapse
Affiliation(s)
- Jessica K Mandula
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
36
|
Wong SW, McCarroll J, Hsu K, Geczy CL, Tedla N. Intranasal Delivery of Recombinant S100A8 Protein Delays Lung Cancer Growth by Remodeling the Lung Immune Microenvironment. Front Immunol 2022; 13:826391. [PMID: 35655772 PMCID: PMC9152328 DOI: 10.3389/fimmu.2022.826391] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/30/2022] [Indexed: 12/03/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Increasing evidence indicates a critical role for chronic inflammation in lung carcinogenesis. S100A8 is a protein with reported pro- and anti-inflammatory functions. It is highly expressed in myeloid-derived suppressor cells (MDSC) that accumulate in the tumor microenvironment and abrogate effective anti-cancer immune responses. Mechanisms of MDSC-mediated immunosuppression include production of reactive oxygen species and nitric oxide, and depletion of L-arginine required for T cell function. Although S100A8 is expressed in MDSC, its role in the lung tumor microenvironment is largely unknown. To address this, mouse recombinant S100A8 was repeatedly administered intranasally to mice bearing orthotopic lung cancers. S100A8 treatment prolonged survival from 19 days to 28 days (p < 0.001). At midpoint of survival, whole lungs and bronchoalveolar lavage fluid (BALF) were collected and relevant genes/proteins measured. We found that S100A8 significantly lowered expression of cytokine genes and proteins that promote expansion and activation of MDSC in lungs and BALF from cancer-bearing mice. Moreover, S100A8 enhanced activities of antioxidant enzymes and suppressed production of nitrite to create a lung microenvironment conducive to cytotoxic lymphocyte expansion and function. In support of this, we found decreased MDSC numbers, and increased numbers of CD4+ T cells and natural killer T (NK-T) cells in lungs from cancer-bearing mice treated with S100A8. Ex-vivo treatment of splenocytes with S100A8 protein activated NK cells. Our results indicate that treatment with S100A8 may favourably modify the lung microenvironment to promote an effective immune response in lungs, thereby representing a new strategy that could complement current immunotherapies in lung cancer.
Collapse
Affiliation(s)
- Sze Wing Wong
- School of Medical Sciences and the Kirby Institute, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia.,Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Joshua McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,Australian Centre for Nanomedicine, UNSW Sydney, Sydney, NSW, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Kenneth Hsu
- School of Medical Sciences and the Kirby Institute, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Carolyn L Geczy
- School of Medical Sciences and the Kirby Institute, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Nicodemus Tedla
- School of Medical Sciences and the Kirby Institute, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
37
|
Immunometabolic Markers in a Small Patient Cohort Undergoing Immunotherapy. Biomolecules 2022; 12:biom12050716. [PMID: 35625643 PMCID: PMC9139165 DOI: 10.3390/biom12050716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 11/16/2022] Open
Abstract
Although the discovery of immune checkpoints was hailed as a major breakthrough in cancer therapy, generating a sufficient response to immunotherapy is still limited. Thus, the objective of this exploratory, hypothesis-generating study was to identify potentially novel peripheral biomarkers and discuss the possible predictive relevance of combining scarcely investigated metabolic and hormonal markers with immune subsets. Sixteen markers that differed significantly between responders and non-responders were identified. In a further step, the correlation with progression-free survival (PFS) and false discovery correction (Benjamini and Hochberg) revealed potential predictive roles for the immune subset absolute lymphocyte count (rs = 0.51; p = 0.0224 *), absolute basophil count (rs = 0.43; p = 0.04 *), PD-1+ monocytes (rs = −0.49; p = 0.04 *), hemoglobin (rs = 0.44; p = 0.04 *), metabolic markers LDL (rs = 0.53; p = 0.0224 *), free androgen index (rs = 0.57; p = 0.0224 *) and CRP (rs = −0.46; p = 0.0352 *). The absolute lymphocyte count, LDL and free androgen index were the most significant individual markers, and combining the immune subsets with the metabolic markers into a biomarker ratio enhanced correlation with PFS (rs = −0.74; p ≤ 0.0001 ****). In summary, in addition to well-established markers, we identified PD-1+ monocytes and the free androgen index as potentially novel peripheral markers in the context of immunotherapy. Furthermore, the combination of immune subsets with metabolic and hormonal markers may have the potential to enhance the power of future predictive scores and should, therefore, be investigated further in larger trials.
Collapse
|
38
|
Qin Z, Cao H, Ni C, Han L, Wang R, Blasig R, Haseloff R, Qin Y, Lan J, Lou X, Ma P, Yao X, Wang L, Wang F, Zhu L, Lei N, Blasig IE. Claudin-12 deficiency inhibits tumor growth by impairing transendothelial migration of myeloid-derived suppressor cells. Cancer Res 2022; 82:2472-2484. [PMID: 35580275 DOI: 10.1158/0008-5472.can-21-3896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/23/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022]
Abstract
Migration of myeloid-derived suppressor cells (MDSCs) out of the circulation, across vascular walls, and into tumor is crucial for their immunosuppressive activity. A deeper understanding of critical junctional molecules and the regulatory mechanisms that mediate the extravasation of MDSCs could identify approaches to overcome cancer immunosuppression. In this study we used mice deficient in tight-junction protein Claudin-12 (Cldn12) compared to wild-type mice and found that loss of host Cldn12 inhibited the growth of transplanted tumors, reduced intratumoral accumulation of MDSCs, increased anti-tumor immune responses, and decreased tumor vescular density. Further studies revealed that Cldn12 expression on the cell surface of both MDSCs and endothelial cells is required for MDSCs transit across tumor vascular endothelial cells (ECs). Importantly, expression of Cldn12 in MDSCs was modulated by granulocyte-macrophage colony-stimulating factor (GM-CSF) in an AKT-dependent manner. Therefore, our results indicate that Cldn12 could serve as a promising target for restoring the anti-tumor response by interfering with MDSCs transendothelial migration.
Collapse
Affiliation(s)
- Zhihai Qin
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hong Cao
- First Affiliated Hospital of Zhengzhou University, Zhenzhou, China
| | - Chen Ni
- First Affiliated Hospital of Zhengzhou University, ZhengZhou, Henan, China
| | - Le Han
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Ruoqi Wang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Rosel Blasig
- Leibniz Institute of Molecular Pharmacology, Berlin, Germany
| | - Reiner Haseloff
- Leibniz Institute of Molecular Pharmacology, Berlin, Germany
| | - Yue Qin
- First Affiliated Hospital of Zhengzhou University, Zhenzhou, China
| | - Jie Lan
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiaohan Lou
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pan Ma
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaohan Yao
- First Affiliated Hospital of Zhengzhou University, China
| | - Linlin Wang
- First Affiliated Hospital of Zhengzhou University, Zhenzhou, Henan, China
| | - Fei Wang
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Linyu Zhu
- Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Ingolf E Blasig
- Leibniz Institute of Molecular Pharmacology, Berlin, Germany
| |
Collapse
|
39
|
Gorris MAJ, van der Woude LL, Kroeze LI, Bol K, Verrijp K, Amir AL, Meek J, Textor J, Figdor CG, de Vries IJM. Paired primary and metastatic lesions of patients with ipilimumab-treated melanoma: high variation in lymphocyte infiltration and HLA-ABC expression whereas tumor mutational load is similar and correlates with clinical outcome. J Immunother Cancer 2022; 10:e004329. [PMID: 35550553 PMCID: PMC9109111 DOI: 10.1136/jitc-2021-004329] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) can lead to long-term responses in patients with metastatic melanoma. Still many patients with melanoma are intrinsically resistant or acquire secondary resistance. Previous studies have used primary or metastatic tumor tissue for biomarker assessment. Especially in melanoma, metastatic lesions are often present at different anatomical sites such as skin, lymph nodes, and visceral organs. The anatomical site may directly affect the tumor microenvironment (TME). To evaluate the impact of tumor evolution on the TME and on ICI treatment outcome, we directly compared paired primary and metastatic melanoma lesions for tumor mutational burden (TMB), HLA-ABC status, and tumor infiltrating lymphocytes (TILs) of patients that received ipilimumab. METHODS TMB was analyzed by sequencing primary and metastatic melanoma lesions using the TruSight Oncology 500 assay. Tumor tissues were subjected to multiplex immunohistochemistry to assess HLA-ABC status and for the detection of TIL subsets (B cells, cytotoxic T cells, helper T cells, and regulatory T cells), by using a machine-learning algorithm. RESULTS While we observed a very good agreement between TMB of matched primary and metastatic melanoma lesions (intraclass coefficient=0.921), such association was absent for HLA-ABC status, TIL density, and subsets thereof. Interestingly, analyses of different metastatic melanoma lesions within a single patient revealed that TIL density and composition agreed remarkably well, rejecting the hypothesis that the TME of different anatomical sites affects TIL infiltration. Similarly, the HLA-ABC status between different metastatic lesions within patients was also comparable. Furthermore, high TMB, of either primary or metastatic melanoma tissue, directly correlated with response to ipilimumab, whereas lymphocyte density or composition did not. Loss of HLA-ABC in the metastatic lesion correlated to a shorter progression-free survival on ipilimumab. CONCLUSIONS We confirm the link between TMB and HLA-ABC status and the response to ipilimumab-based immunotherapy in melanoma, but no correlation was found for TIL density, neither in primary nor metastatic lesions. Our finding that TMB between paired primary and metastatic melanoma lesions is highly stable, demonstrates its independency of the time point and location of acquisition. TIL and HLA-ABC status in metastatic lesions of different anatomical sites are highly similar within an individual patient.
Collapse
Affiliation(s)
- Mark A J Gorris
- Tumor Immunology, Radboudumc, Nijmegen, The Netherlands
- Oncode Institute, Nijmegen, The Netherlands
| | - Lieke L van der Woude
- Tumor Immunology, Radboudumc, Nijmegen, The Netherlands
- Oncode Institute, Nijmegen, The Netherlands
- Pathology, Radboudumc, Nijmegen, The Netherlands
| | | | - Kalijn Bol
- Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | - Kiek Verrijp
- Oncode Institute, Nijmegen, The Netherlands
- Pathology, Radboudumc, Nijmegen, The Netherlands
| | | | - Jelena Meek
- Tumor Immunology, Radboudumc, Nijmegen, The Netherlands
| | - Johannes Textor
- Department of Tumor Immunology, Radboudumc, Nijmegen, The Netherlands
- Data Science Group, Institute for Computing and Information Sciences, Radboud Universiteit, Nijmegen, The Netherlands
| | - Carl G Figdor
- Tumor Immunology, Radboudumc, Nijmegen, The Netherlands
- Oncode Institute, Nijmegen, The Netherlands
| | | |
Collapse
|
40
|
Plazy C, Hannani D, Gobbini E. Immune Checkpoint Inhibitor Rechallenge and Resumption: a Systematic Review. Curr Oncol Rep 2022; 24:1095-1106. [PMID: 35389138 DOI: 10.1007/s11912-022-01241-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE OF THE REVIEW The reintroduction of immune checkpoint inhibitors (ICIs) after disease progression (rechallenge) or immune-related adverse events (irAEs) recovering (resumption) raises questions in terms of efficacy and safety. RECENT FINDINGS Here, we reviewed literature data about ICIs rechallenge/resumption in cancer patients along with their clinical characteristics to explore those factors associated with better outcomes. Heterogenous results were pointed out across rechallenge studies with an overall response rate between 0 and 54%, and a progression free survival ranged from 1.5 to 12.9 months and an overall survival between 6.5 and 23.8 months. Better outcomes have been recorded in patients with good ECOG PS, longer duration of initial ICI, discontinuation reason of initial ICI other than progression, and those who received ICI sequence other than the switch between anti-PD1 and anti-PDL1. Studies about ICI resumption highlighted that certain types of irAEs were more likely to relapse at retreatment. These results suggest that ICI rechallenge/resumption can be an interesting strategy for selected patients.
Collapse
Affiliation(s)
- Caroline Plazy
- CHU Grenoble-Alpes, Biological and Pathological Institute, Avenue Maquis du Gresivaudan, 38700, La Tronche, France
- Univ. Grenoble Alpes, CNRS, Grenoble INP, CHU Grenoble Alpes, TIMC, UMR 5525, VetAgro Sup, 38000, Grenoble, France
| | - Dalil Hannani
- Univ. Grenoble Alpes, CNRS, Grenoble INP, CHU Grenoble Alpes, TIMC, UMR 5525, VetAgro Sup, 38000, Grenoble, France
| | - Elisa Gobbini
- Thoracic Oncology Unit, CHU Grenoble-Alpes, Avenue Maquis du Gresivaudan, 38700, La Tronche, France.
- Cancer Research Center of Lyon, CISTAR Research Team, 28 rue Laennec, 69008, Lyon, France.
| |
Collapse
|
41
|
宋 攀, 颜 晓, 姜 燕, 叶 煜, 王 静, 韩 萍. [The research progress of PD-1/L1 inhibitors application in the treatment of head and neck squamous cell carcinoma]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2022; 36:315-320. [PMID: 35511629 PMCID: PMC10128179 DOI: 10.13201/j.issn.2096-7993.2022.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Indexed: 04/30/2023]
Abstract
Over the past few years, the FDA has approved PD-1/L1 inhibitor for the treatment of advanced head and neck squamous cell carcinoma, involving PD-1/L1 inhibitor monotherapy, PD-1/L1 inhibitor combined with chemoradiotherapy, combined with targeted therapy, combined with neoadjuvant immunotherapy and duplex-block of immune checkpoints and so on. Herein, we briefly review the latest research results in this field, and summarize the application and efficacy of immunotherapy in the treatment of head and neck squamous cell carcinoma, which will benefits such patients to develop more precise and individualized treatment plans.
Collapse
Affiliation(s)
- 攀 宋
- 中山大学孙逸仙纪念医院耳鼻咽喉头颈外科(广州, 510220)
| | - 晓晴 颜
- 中山大学孙逸仙纪念医院耳鼻咽喉头颈外科(广州, 510220)
| | - 燕慧 姜
- 中山大学孙逸仙纪念医院耳鼻咽喉头颈外科(广州, 510220)
| | - 煜初 叶
- 中山大学孙逸仙纪念医院耳鼻咽喉头颈外科(广州, 510220)
| | - 静怡 王
- 中山大学孙逸仙纪念医院耳鼻咽喉头颈外科(广州, 510220)
| | - 萍 韩
- 中山大学孙逸仙纪念医院耳鼻咽喉头颈外科(广州, 510220)
- 韩萍,
| |
Collapse
|
42
|
Girardi DM, Niglio SA, Mortazavi A, Nadal R, Lara P, Pal SK, Saraiya B, Cordes L, Ley L, Ortiz OS, Cadena J, Diaz C, Bagheri H, Redd B, Steinberg SM, Costello R, Chan KS, Lee MJ, Lee S, Yu Y, Gurram S, Chalfin HJ, Valera V, Figg WD, Merino M, Toubaji A, Streicher H, Wright JJ, Sharon E, Parnes HL, Ning YM, Bottaro DP, Cao L, Trepel JB, Apolo AB. Cabozantinib plus Nivolumab Phase I Expansion Study in Patients with Metastatic Urothelial Carcinoma Refractory to Immune Checkpoint Inhibitor Therapy. Clin Cancer Res 2022; 28:1353-1362. [PMID: 35031545 PMCID: PMC9365339 DOI: 10.1158/1078-0432.ccr-21-3726] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/17/2021] [Accepted: 01/12/2022] [Indexed: 01/26/2023]
Abstract
PURPOSE This study investigated the efficacy and tolerability of cabozantinib plus nivolumab (CaboNivo) in patients with metastatic urothelial carcinoma (mUC) that progressed on checkpoint inhibition (CPI). PATIENTS AND METHODS A phase I expansion cohort of patients with mUC who received prior CPI was treated with cabozantinib 40 mg/day and nivolumab 3 mg/kg every 2 weeks until disease progression/unacceptable toxicity. The primary goal was objective response rate (ORR) per RECIST v.1.1. Secondary objectives included progression-free survival (PFS), duration of response (DoR), overall survival (OS), safety, and tolerability. RESULTS Twenty-nine out of 30 patients enrolled were evaluable for efficacy. Median follow-up was 22.2 months. Most patients (86.7%) received prior chemotherapy and all patients received prior CPI (median seven cycles). ORR was 16.0%, with one complete response and three partial responses (PR). Among 4 responders, 2 were primary refractory, 1 had a PR, and 1 had stable disease on prior CPI. Median DoR was 33.5 months [95% confidence interval (CI), 3.7-33.5], median PFS was 3.6 months (95% CI, 2.1-5.5), and median OS was 10.4 months (95% CI, 5.8-19.5). CaboNivo decreased immunosuppressive subsets such as regulatory T cells (Tregs) and increased potential antitumor immune subsets such as nonclassical monocytes and effector T cells. A lower percentage of monocytic myeloid-derived suppressor cells (M-MDSC) and polymorphonuclear MDSCs, lower CTLA-4 and TIM-3 expression on Tregs, and higher effector CD4+ T cells at baseline were associated with better PFS and/or OS. CONCLUSIONS CaboNivo was clinically active, well tolerated, and favorably modulated peripheral blood immune subsets in patients with mUC refractory to CPI.
Collapse
Affiliation(s)
- Daniel M. Girardi
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Scot A. Niglio
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, and the Comprehensive Cancer Center, Columbus, Ohio
| | - Rosa Nadal
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Primo Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Sumanta K. Pal
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Biren Saraiya
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Lisa Cordes
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Lisa Ley
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Olena Sierra Ortiz
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Jacqueline Cadena
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Carlos Diaz
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Hadi Bagheri
- Clinical Image Processing Service, Department of Radiology and Imaging Sciences, Clinical Center, NIH, Bethesda, Maryland
| | - Bernadette Redd
- Clinical Image Processing Service, Department of Radiology and Imaging Sciences, Clinical Center, NIH, Bethesda, Maryland
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Office of the Clinical Director, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Rene Costello
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Keith S. Chan
- Samuel Oschin Cancer Center, Cedars Sinai Medical Center, Los Angeles, California
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Sunmin Lee
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Yunkai Yu
- Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Sandeep Gurram
- Urologic Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Heather J. Chalfin
- Urologic Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Vladimir Valera
- Urologic Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - William D. Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Maria Merino
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Antoun Toubaji
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Howard Streicher
- Investigational Drug Branch, Cancer Therapy Evaluation Program, NCI, NIH, Rockville, Maryland
| | - John J. Wright
- Investigational Drug Branch, Cancer Therapy Evaluation Program, NCI, NIH, Rockville, Maryland
| | - Elad Sharon
- Investigational Drug Branch, Cancer Therapy Evaluation Program, NCI, NIH, Rockville, Maryland
| | - Howard L. Parnes
- Division of Cancer Prevention, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Yang-Min Ning
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Donald P. Bottaro
- Urologic Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Jane B. Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Andrea B. Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland.,Corresponding Author: Andrea B. Apolo, Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, MD 20892. E-mail:
| |
Collapse
|
43
|
Li H, van der Merwe PA, Sivakumar S. Biomarkers of response to PD-1 pathway blockade. Br J Cancer 2022; 126:1663-1675. [PMID: 35228677 PMCID: PMC9174485 DOI: 10.1038/s41416-022-01743-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/17/2022] [Accepted: 02/03/2022] [Indexed: 12/15/2022] Open
Abstract
The binding of T cell immune checkpoint proteins programmed death 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) to their ligands allows immune evasion by tumours. The development of therapeutic antibodies, termed checkpoint inhibitors, that bind these molecules or their ligands, has provided a means to release this brake on the host anti-tumour immune response. However, these drugs are costly, are associated with potentially severe side effects, and only benefit a small subset of patients. It is therefore important to identify biomarkers that discriminate between responders and non-responders. This review discusses the determinants for a successful response to antibodies that bind PD-1 or its ligand PD-L1, dividing them into markers found in the tumour biopsy and those in non-tumour samples. It provides an update on the established predictive biomarkers (tumour PD-L1 expression, tumour mismatch repair deficiency and tumour mutational burden) and assesses the evidence for new potential biomarkers.
Collapse
Affiliation(s)
- Hanxiao Li
- Green Templeton College, University of Oxford, Oxford, UK.
| | | | | |
Collapse
|
44
|
Cancer cell-expressed BTNL2 facilitates tumour immune escape via engagement with IL-17A-producing γδ T cells. Nat Commun 2022; 13:231. [PMID: 35017553 PMCID: PMC8752682 DOI: 10.1038/s41467-021-27936-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 12/21/2021] [Indexed: 12/21/2022] Open
Abstract
Therapeutic blockade of the immune checkpoint proteins programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA4) has transformed cancer treatment. However, the overall response rate to these treatments is low, suggesting that immune checkpoint activation is not the only mechanism leading to dysfunctional anti-tumour immunity. Here we show that butyrophilin-like protein 2 (BTNL2) is a potent suppressor of the anti-tumour immune response. Antibody-mediated blockade of BTNL2 attenuates tumour progression in multiple in vivo murine tumour models, resulting in prolonged survival of tumour-bearing mice. Mechanistically, BTNL2 interacts with local γδ T cell populations to promote IL-17A production in the tumour microenvironment. Inhibition of BTNL2 reduces the number of tumour-infiltrating IL-17A-producing γδ T cells and myeloid-derived suppressor cells, while facilitating cytotoxic CD8+ T cell accumulation. Furthermore, we find high BTNL2 expression in several human tumour samples from highly prevalent cancer types, which negatively correlates with overall patient survival. Thus, our results suggest that BTNL2 is a negative regulator of anti-tumour immunity and a potential target for cancer immunotherapy. Cancer cells producing ligands for the immune checkpoint molecules PD-1 and CTLA-4 is an important mechanism of tumour immune resistance. Here authors show that BTNL2 expression on cancer cells generates a dysfunctional tumour immune microenvironment via promoting IL-17A-producing γδ T cells.
Collapse
|
45
|
Wang Q, Xie B, Liu S, Shi Y, Tao Y, Xiao D, Wang W. What Happens to the Immune Microenvironment After PD-1 Inhibitor Therapy? Front Immunol 2022; 12:773168. [PMID: 35003090 PMCID: PMC8733588 DOI: 10.3389/fimmu.2021.773168] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/23/2021] [Indexed: 12/11/2022] Open
Abstract
The fruitful results of tumor immunotherapy establish its indispensable status in the regulation of the tumorous immune context. It seems that the treatment of programmed cell death receptor 1 (PD-1) blockade is one of the most promising approaches for cancer control. The significant efficacy of PD-1 inhibitor therapy has been made in several cancer types, such as breast cancer, lung cancer, and multiple myeloma. Even so, the mechanisms of how anti-PD-1 therapy takes effect by impacting the immune microenvironment and how partial patients acquire the resistance to PD-1 blockade have yet to be studied. In this review, we discuss the cross talk between immune cells and how they promote PD-1 blockade efficacy. In addition, we also depict factors that may underlie tumor resistance to PD-1 blockade and feasible solutions in combination with it.
Collapse
Affiliation(s)
- Qingyi Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Department of Pathology, School of Basic Medicine, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Central South University, Changsha, China
| | - Bin Xie
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Shuang Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Department of Pathology, School of Basic Medicine, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Central South University, Changsha, China
| | - Ying Shi
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Department of Pathology, School of Basic Medicine, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Central South University, Changsha, China
| | - Yongguang Tao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Department of Pathology, School of Basic Medicine, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Central South University, Changsha, China.,National Health Commission (NHC) Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Second Xiangya Hospital, Central South University, Changsha, China
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Department of Pathology, School of Basic Medicine, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Central South University, Changsha, China
| | - Wenxiang Wang
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
46
|
Geng S, Pradhan K, Li L. Signal-Strength and History-Dependent Innate Immune Memory Dynamics in Health and Disease. Handb Exp Pharmacol 2022; 276:23-41. [PMID: 34085119 DOI: 10.1007/164_2021_485] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Innate immunity exhibits memory characteristics, reflected not only in selective recognition of external microbial or internal damage signals, but more importantly in history and signal-strength dependent reprogramming of innate leukocytes characterized by priming, tolerance, and exhaustion. Key innate immune cells such as monocytes and neutrophils can finely discern and attune to the duration and intensity of external signals through rewiring of internal signaling circuitries, giving rise to a vast array of discreet memory phenotypes critically relevant to managing tissue homeostasis as well as diverse repertoires of inflammatory conditions. This review will highlight recent advances in this rapidly expanding field of innate immune programming and memory, as well as its translational implication in the pathophysiology of selected inflammatory diseases.
Collapse
Affiliation(s)
- Shuo Geng
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Kisha Pradhan
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
47
|
Abstract
Melanoma is the most lethal skin cancer that originates from the malignant transformation of melanocytes. Although melanoma has long been regarded as a cancerous malignancy with few therapeutic options, increased biological understanding and unprecedented innovations in therapies targeting mutated driver genes and immune checkpoints have substantially improved the prognosis of patients. However, the low response rate and inevitable occurrence of resistance to currently available targeted therapies have posed the obstacle in the path of melanoma management to obtain further amelioration. Therefore, it is necessary to understand the mechanisms underlying melanoma pathogenesis more comprehensively, which might lead to more substantial progress in therapeutic approaches and expand clinical options for melanoma therapy. In this review, we firstly make a brief introduction to melanoma epidemiology, clinical subtypes, risk factors, and current therapies. Then, the signal pathways orchestrating melanoma pathogenesis, including genetic mutations, key transcriptional regulators, epigenetic dysregulations, metabolic reprogramming, crucial metastasis-related signals, tumor-promoting inflammatory pathways, and pro-angiogenic factors, have been systemically reviewed and discussed. Subsequently, we outline current progresses in therapies targeting mutated driver genes and immune checkpoints, as well as the mechanisms underlying the treatment resistance. Finally, the prospects and challenges in the development of melanoma therapy, especially immunotherapy and related ongoing clinical trials, are summarized and discussed.
Collapse
Affiliation(s)
- Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Huina Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China.
| |
Collapse
|
48
|
Predictive Biomarkers for Outcomes of Immune Checkpoint Inhibitors (ICIs) in Melanoma: A Systematic Review. Cancers (Basel) 2021; 13:cancers13246366. [PMID: 34944986 PMCID: PMC8699321 DOI: 10.3390/cancers13246366] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have strongly improved the survival of melanoma patients. However, as durable response to ICIs are only seen in a minority, there is an unmet need to identify biomarkers that predict response. Therefore, we provide a systematic review that evaluates all biomarkers studied in association with outcomes of melanoma patients receiving ICIs. We searched Pubmed, COCHRANE Library, Embase, Emcare, and Web of Science for relevant articles that were published before June 2020 and studied blood, tumor, or fecal biomarkers that predicted response or survival in melanoma patients treated with ICIs. Of the 2536 identified reports, 177 were included in our review. Risk of bias was high in 40%, moderate in 50% and low in 10% of all studies. Biomarkers that correlated with response were myeloid-derived suppressor cells (MDSCs), circulating tumor cells (CTCs), CD8+ memory T-cells, T-cell receptor (TCR) diversity, tumor-infiltrating lymphocytes (TILs), gene expression profiling (GEP), and a favorable gut microbiome. This review shows that biomarkers for ICIs in melanoma patients are widely studied, but heterogeneity between studies is high, average sample sizes are low, and validation is often lacking. Future studies are needed to further investigate the predictive utility of some promising candidate biomarkers.
Collapse
|
49
|
Suppressing MDSC Recruitment to the Tumor Microenvironment by Antagonizing CXCR2 to Enhance the Efficacy of Immunotherapy. Cancers (Basel) 2021; 13:cancers13246293. [PMID: 34944914 PMCID: PMC8699249 DOI: 10.3390/cancers13246293] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary While the development of immunotherapy has greatly advanced cancer treatment, many patients do not benefit from immunotherapy. Numerous strategies have been developed to improve response to immunotherapy across cancer types, including blocking the activity of immunosuppressive immune cells, cytokines, and signaling pathways that are linked to poor responses. Myeloid-derived suppressor cells (MDSCs) are associated with poor responses to immunotherapy, and the chemokine receptor, CXCR2, is involved in recruiting MDSCs to the tumor. In this review, we present studies that explore the potential of inhibiting MDSCs through blocking CXCR2 as a strategy to enhance response to existing and novel immunotherapies. Abstract Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of cells derived from immature myeloid cells. These cells are often associated with poor responses to cancer therapy, including immunotherapy, in a variety of tumor types. The C-X-C chemokine receptor 2 (CXCR2) signaling axis plays a key role in the migration of immunosuppressive MDSCs into the tumor microenvironment (TME) and the pre-metastatic niche. MDSCs impede the efficacy of immunotherapy through a variety of mechanisms. Efforts to target MDSCs by blocking CXCR2 is an active area of research as a method for improving existing and novel immunotherapy strategies. As immunotherapies gain approval for a wider array of clinical indications, it will become even more important to understand the efficacy of CXCR2 inhibition in combating immunotherapy resistance at different stages of tumor progression.
Collapse
|
50
|
Chen N, Higashiyama N, Hoyos V. Predictive Biomarkers of Immune Checkpoint Inhibitor Response in Breast Cancer: Looking beyond Tumoral PD-L1. Biomedicines 2021; 9:1863. [PMID: 34944679 PMCID: PMC8698415 DOI: 10.3390/biomedicines9121863] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/12/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors utilize the immune system to kill cancer cells and are now widely applied across numerous malignancies. Pembrolizumab has two breast-specific indications in triple-negative disease. Currently, programmed death ligand-1 (PD-L1) expression on tumor and surrounding immune cells is the only validated predictive biomarker for immune checkpoint inhibitors (ICIs) in breast cancer; however, it can be imprecise. Additional biomarkers are needed to identify the patient population who will derive the most benefit from these therapies. The tumor immune microenvironment contains many biomarker candidates. In tumor cells, tumor mutational burden has emerged as a robust biomarker across malignancies in general, with higher burden cancers demonstrating improved response, but will need further refinement for less mutated cancers. Preliminary studies suggest that mutations in breast cancer gene 2 (BRCA-2) are associated with increased immune infiltration and response to ICI therapy. Other genomic alterations are also being investigated as potential predictive biomarkers. In immune cells, increased quantity of tumor-infiltrating lymphocytes and CD8+ cytotoxic T cells have correlated with response to immunotherapy treatment. The role of other immune cell phenotypes is being investigated. Peripherally, many liquid-based biomarker strategies such as PD-L1 expression on circulating tumor cells and peripheral immune cell quantification are being studied; however, these strategies require further standardization and refinement prior to large-scale testing. Ultimately, multiple biomarkers utilized together may be needed to best identify the appropriate patients for these treatments.
Collapse
Affiliation(s)
- Nan Chen
- Baylor College of Medicine, Houston, TX 77030, USA; (N.H.); (V.H.)
| | | | - Valentina Hoyos
- Baylor College of Medicine, Houston, TX 77030, USA; (N.H.); (V.H.)
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|