1
|
Zhao X, Cui H, Zhou M, Ren X, Li Z, Liu P, Zhao D, Lin S, Kang H. A novel glycogene-related signature for prognostic prediction and immune microenvironment assessment in kidney renal clear cell carcinoma. Ann Med 2025; 57:2495762. [PMID: 40329678 PMCID: PMC12064129 DOI: 10.1080/07853890.2025.2495762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Kidney Renal Clear Cell Carcinoma (KIRC) is a prevalent urinary malignancies worldwide. Glycosylation is a key post-translational modification that is essential in cancer progression. However, its relationship with prognosis, tumour microenvironment (TME), and treatment response in KIRC remains unclear. METHOD Expression profiles and clinical data were retrieved from The Cancer Genome Atlas and Gene Expression Omnibus databases. Consensus clustering, Cox regression, and LASSO regression analyses were conducted to develop an optimal glycogene-related signature. The prognostic relevance of this molecular signature was rigorously analyzed, along with its connections to tumour microenvironment (TME), tumour mutation burden, immune checkpoint activity, cancer-immunity cycle regulation, immunomodulatory gene expression patterns, and therapeutic response profiles. Validation was performed using real-world clinical specimens, quantitative PCR (qPCR), and immunohistochemistry (IHC), supported by cohort analyses from the Human Protein Atlas (HPA) database. RESULTS A glycogene-associated prognostic scoring system was established to categorize patients into risk-stratified subgroups. Patients in the high-risk cohort exhibited significantly poorer survival outcomes (p < 0.001). By incorporating clinicopathological variables into this framework, we established a predictive nomogram demonstrating strong calibration and a concordance index (C-index) of 0.78. The high-risk subgroup displayed elevated immune infiltration scores (p < 0.001), upregulated expression of immune checkpoint-related genes (p < 0.05), and an increased frequency of somatic mutations (p = 0.043). The risk score positively correlated with cancer-immunity cycle activation and immunotherapy-related signals. The high-risk groups also showed associations with T cell exhaustion, immune-activating genes, chemokines, and receptors. Drug sensitivity analysis revealed that low-risk patients were more sensitive to sorafenib, pazopanib, and erlotinib, whereas high-risk individuals responded better to temsirolimus (p < 0.01). qPCR and IHC analyses consistently revealed distinct expression patterns of MX2 and other key genes across the risk groups, further corroborated by the HPA findings. CONCLUSION This glycogene-based signature provides a robust tool for predicting prognosis, TME characteristics, and therapeutic responses in KIRC, offering potential clinical utility in patient management.
Collapse
Affiliation(s)
- Xuyan Zhao
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Hanxiao Cui
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Mingjing Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xueting Ren
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Zihao Li
- Department of Urology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Peinan Liu
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Danni Zhao
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Shuai Lin
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Huafeng Kang
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
2
|
Jia Z, Zhang Y, Cao L, Wang J, Liang H. Research hotspots and trends of immunotherapy and melanoma: A bibliometric analysis during 2014-2024. Hum Vaccin Immunother 2025; 21:2464379. [PMID: 40012099 PMCID: PMC11869780 DOI: 10.1080/21645515.2025.2464379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/22/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
Over the last decade, the increasing global prevalence of melanoma has sparked growing interest in immunotherapies, which show significant potential against this form of skin cancer. This research aims to offer a framework to guide future studies and inspire new research directions. In this study, we used the Web of Science Core Collection to collect papers on immunotherapy and melanoma published between 2014 and 2024. With Excel and visualization tools like VOSviewer, COOC 13.2, Citespace, and Bibliometrix (R-Tool of R-Studio), we analyzed the data to spot trends and new focuses in the research. Our findings indicate a substantial surge in research activity concerning immunotherapy and melanoma between 2014 and 2024. The USA and China emerged as leading contributors, engaging in extensive and close collaborative efforts with European counterparts. Furthermore, seven of the top 10 research institutions are located in the USA, with the MD Anderson Cancer Center in Texas being the most productive. In addition, the Journal of Cancer Immunotherapy is the journal with the most articles published in the field. Professor Georgina V. Long from the Melanoma Institute at the University of Sydney was one of the most productive scholars. Keyword analysis shows that immune checkpoint inhibitors, tumor microenvironment and targeted therapies are key areas of interest for the research community. This paper uses bibliometric analysis to outline research trends and key points in immunotherapy and melanoma from 2014 to 2024, which helps understand the current research and guides future research directions.
Collapse
Affiliation(s)
- Zixuan Jia
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Youao Zhang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Luyan Cao
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, Scotland
| | - Jieyan Wang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
| | - Hui Liang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Wiecken M, Machiraju D, Chakraborty S, Mayr EM, Lenoir B, Eurich R, Richter J, Pfarr N, Halama N, Hassel JC. The immune checkpoint LAG-3 is expressed by melanoma cells and correlates with clinical progression of the melanoma. Oncoimmunology 2025; 14:2430066. [PMID: 39716918 DOI: 10.1080/2162402x.2024.2430066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 12/25/2024] Open
Abstract
Immune checkpoint blockers have substantially improved prognosis of melanoma patients, nevertheless, resistance remains a significant problem. Here, intrinsic and extrinsic factors in the tumor microenvironment are discussed, including the expression of alternative immune checkpoints such as lymphocyte activation gene 3 (LAG-3) and T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3). While most studies focus on immune cell expression of these proteins, we investigated their melanoma cell intrinsic expression by immunohistochemistry in melanoma metastases of 60 patients treated with anti-programmed cell death protein 1 (PD-1) and/or anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) therapy, and correlated it with the expression of potential ligands, RNA sequencing data and clinical outcome. LAG-3 and TIM-3 were commonly expressed in melanoma cells. In the stage IV cohort, expression of LAG-3 was associated with M1 stage (p < 0.001) and previous exposure to immune checkpoint inhibitors (p = 0.029). Moreover, in the anti-PD-1 monotherapy treatment group patients with high LAG-3 expression by tumor cells tended to have a shorter progression-free survival (p = 0.088), whereas high expression of TIM-3 was associated with a significantly longer overall survival (p = 0.007). In conclusion, we provide a systematic analysis of melanoma cell intrinsic LAG-3 and TIM-3 expression, highlighting potential implications of their expression on patient survival.
Collapse
Affiliation(s)
- Melanie Wiecken
- Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Devayani Machiraju
- Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Shounak Chakraborty
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Eva-Maria Mayr
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Bénédicte Lenoir
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit "Applied Tumor Immunity"(TME unit), Heidelberg, Germany
| | - Rosa Eurich
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit "Applied Tumor Immunity"(TME unit), Heidelberg, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Division of Translational Immunotherapy, Heidelberg, Germany
| | - Jasmin Richter
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Nicole Pfarr
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Niels Halama
- German Cancer Research Center (DKFZ) Heidelberg, Division of Translational Immunotherapy, Heidelberg, Germany
- Department of Medical Oncology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica C Hassel
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
4
|
O'Connor MH, Rhodin KE, Tyler DS, Beasley GM. Management of In-transit Disease: Regional Therapies, Intralesional Therapies, and Systemic Therapy. Surg Oncol Clin N Am 2025; 34:393-410. [PMID: 40413006 PMCID: PMC12104569 DOI: 10.1016/j.soc.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
In-transit (IT) melanoma represents a distinct, heterogeneous pattern of disease that arises as superficial tumors along the track between the primary site and the draining regional lymph node basin. Many therapies have been explored for treatment of this disease with the goal of maximizing delivery of the therapeutic agent to the tumor while minimizing systemic toxicities. These include regional chemotherapies, intralesional injections, checkpoint inhibitors, immunomodulators, and vaccines in various combinations or as monotherapy. Here, we review the general managemnt of patients with ITmelanoma, the range of currently available treatment options, and recommendations for specific therapies for individual patients.
Collapse
Affiliation(s)
- Margaret H O'Connor
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Kristen E Rhodin
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Douglas S Tyler
- Department of Surgery, Texas Medical Branch, Galveston, TX, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
5
|
Reader CS, Liao W, Potter-Landua BJ, Veyssier CS, Seal CJ, Brewis N, Morrow M. The tetravalent, bispecific properties of FS118, an anti-LAG-3/PD-L1 antibody, mediate LAG-3 shedding from CD4 + and CD8 + tumor-infiltrating lymphocytes. Anticancer Drugs 2025; 36:447-458. [PMID: 39960386 PMCID: PMC12061380 DOI: 10.1097/cad.0000000000001705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/06/2024] [Indexed: 03/01/2025]
Abstract
Tumor-infiltrating lymphocytes (TILs) often have upregulated expression of immune checkpoint receptors, such as programmed cell death 1 (PD-1) and lymphocyte-activation gene 3 (LAG-3). Patients treated with antibodies targeting PD-1 or its ligand (PD-L1) can develop resistance or relapse, with LAG-3 upregulation on T cells being one possible mechanism. FS118 is a tetravalent, bispecific antibody comprising a full-length IgG 1 anti-PD-L1 antibody with bivalent LAG-3-binding capability in the fragment crystallizable region. Here we demonstrate how the structure of FS118 is important for its function. We generated variants of FS118 and tested their ability to mediate LAG-3 shedding using staphylococcal enterotoxin B assays, antigen recall assays, and soluble LAG-3 ELISAs. Mediated by metalloproteases ADAM10 and ADAM17, FS118 induced shedding of LAG-3 from the surface of both CD4 + and CD8 + T cells. We also determined the effect of surrogate antibodies on immune cell LAG-3 expression and proliferation in syngeneic mouse models. In vivo , the bivalent LAG-3 binding sites of a mouse surrogate of FS118 and their location in the fragment crystallizable region were important for eliciting maximal reduction in LAG-3 levels on the surface of TILs, as variants with a single LAG-3 binding site in the fragment crystallizable region, or with reversed orientation of the LAG-3 and PD-L1 binding sites, were less efficient at inducing shedding. We also show that PD-L1, not PD-1, binding drives the LAG-3 reduction on TILs. We hypothesize that the LAG-3 bivalency in the fragment crystallizable region of FS118 allows LAG-3 clustering, which optimizes cleavage by ADAM10/ADAM17 and thus shedding.
Collapse
|
6
|
Seldomridge AN, Weiser R, Holder AM. Systemic Therapy for Melanoma: What Surgeons Need to Know. Surg Oncol Clin N Am 2025; 34:359-374. [PMID: 40413004 DOI: 10.1016/j.soc.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Immune checkpoint inhibitors and targeted therapies (BRAF/MEK inhibitors) have transformed the care of patients with stage IV melanoma, now with 5-year overall survival rates around 50%. Surgeons should be acquainted with these drugs, the multidisciplinary considerations of their use, and the unique immune-related adverse events (irAEs) they can cause. In this review, we discuss systemic therapies for cutaneous melanoma, including the biology of immune checkpoint inhibition, treatment indications, and toxicities. We also explain how these irAEs and other toxicities can impact surgical planning and perioperative management.
Collapse
Affiliation(s)
- Ashlee N Seldomridge
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Roi Weiser
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Ashley M Holder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
7
|
Huang Q, Li Y, Huang Y, Wu J, Bao W, Xue C, Li X, Dong S, Dong Z, Hu S. Advances in molecular pathology and therapy of non-small cell lung cancer. Signal Transduct Target Ther 2025; 10:186. [PMID: 40517166 DOI: 10.1038/s41392-025-02243-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/02/2025] [Accepted: 03/31/2025] [Indexed: 06/16/2025] Open
Abstract
Over the past two decades, non-small cell lung cancer (NSCLC) has witnessed encouraging advancements in basic and clinical research. However, substantial unmet needs remain for patients worldwide, as drug resistance persists as an inevitable reality. Meanwhile, the journey towards amplifying the breadth and depth of the therapeutic effect requires comprehending and integrating diverse and profound progress. In this review, therefore, we aim to comprehensively present such progress that spans the various aspects of molecular pathology, encompassing elucidations of metastatic mechanisms, identification of therapeutic targets, and dissection of spatial omics. Additionally, we also highlight the numerous small molecule and antibody drugs, encompassing their application alone or in combination, across later-line, frontline, neoadjuvant or adjuvant settings. Then, we elaborate on drug resistance mechanisms, mainly involving targeted therapies and immunotherapies, revealed by our proposed theoretical models to clarify interactions between cancer cells and a variety of non-malignant cells, as well as almost all the biological regulatory pathways. Finally, we outline mechanistic perspectives to pursue innovative treatments of NSCLC, through leveraging artificial intelligence to incorporate the latest insights into the design of finely-tuned, biomarker-driven combination strategies. This review not only provides an overview of the various strategies of how to reshape available armamentarium, but also illustrates an example of clinical translation of how to develop novel targeted drugs, to revolutionize therapeutic landscape for NSCLC.
Collapse
Affiliation(s)
- Qing Huang
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Yuanxiang Li
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Yingdan Huang
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Jingyi Wu
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Wendai Bao
- Center for Neurological Disease Research, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Chang Xue
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Xiaoyu Li
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Shuang Dong
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Zhiqiang Dong
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China.
- Center for Neurological Disease Research, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| | - Sheng Hu
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China.
| |
Collapse
|
8
|
Wang J, Klein C, Cochran JR, Sockolosky J, Lippow SM. Exploring new frontiers in LAG-3 biology and therapeutics. Trends Pharmacol Sci 2025:S0165-6147(25)00098-7. [PMID: 40514283 DOI: 10.1016/j.tips.2025.05.008] [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: 03/09/2025] [Revised: 05/18/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025]
Abstract
Lymphocyte activation gene-3 (LAG-3) has emerged as a critical immune checkpoint receptor primarily modulating T-cell responses through distinct immune regulatory mechanisms. Recent advances have elucidated LAG-3's complex receptor-ligand interactions, structure-function relationships, and unique signaling pathways. LAG-3 antagonistic antibodies, such as relatlimab approved for melanoma, have shown promising efficacy with favorable toxicity profiles, though only in combinational therapies. While LAG-3's role in oncology continues to expand, it is also gaining recognition as a potential therapeutic target for other disorders. This review highlights recent progress in understanding LAG-3's molecular features, ligand regulation, signaling, and immune modulation mechanisms. Additionally, it explores emerging questions in oncology and the exciting potential of therapies targeting the LAG-3 pathway in autoimmune disease. A deeper understanding of LAG-3's confounding biology and disease relevance would drive the development of novel immunotherapies across broader clinical indications.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA.
| | | | - Jennifer R Cochran
- Department of Bioengineering, Stanford Cancer Institute, Stanford University, Stanford, CA
| | | | | |
Collapse
|
9
|
Kimura A, Mitsuda J, Yoshimura K, Saburi S, Murakami N, Sakurai N, Yoshizawa K, Morimoto H, Mukudai S, Nagao H, Ogi H, Shibata S, Miyagawa-Hayashino A, Konishi E, Itoh K, Hirano S, Tsujikawa T. Neoadjuvant therapy-induced immune dynamics and myeloid-associated resistance in advanced head and neck cancer. NPJ Precis Oncol 2025; 9:167. [PMID: 40483270 PMCID: PMC12145449 DOI: 10.1038/s41698-025-00954-1] [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: 02/13/2025] [Accepted: 05/20/2025] [Indexed: 06/11/2025] Open
Abstract
Understanding the dynamics of the tumor-immune ecosystem is crucial for advancing neoadjuvant strategies in cancer treatment. This study investigated alterations in the tumor-immune microenvironment related to the response to preoperative combination therapy with paclitaxel, carboplatin, and cetuximab in patients with advanced head and neck squamous cell carcinoma. Thirty patients underwent combination therapy. Biopsy or surgical specimens were obtained before and after treatment. Single-cell-based, 14-marker multiplex immunohistochemistry and image cytometry were employed to assess changes in immune cell densities and profiles. Three distinct immune profiles were identified: hypo-, lymphoid-, and myeloid-inflamed. Significant decreases in tumor volume and increases in CD45+ cells and programmed cell death ligand 1 (PD-L1) scores were observed in the hypo- and lymphoid-inflamed groups, whereas the myeloid-inflamed group showed minimal changes. After treatment, increased calreticulin expression in tumor cells, together with increased lymphoid cell lineages, was observed in non-recurrent cases. The myeloid-inflamed group exhibited higher expression of hypoxia inducible factor 1α and zinc finger E-box-binding homeobox 2, suggesting the presence of a hypoxic and metastasis-promoting environment. Spatial analysis revealed that responders had a high infiltration of T cells within tumor cell nests, whereas non-responders had fewer T cells, with β-catenin expression in cancer cells. Upregulated lymphocyte activation gene 3 in the myeloid-inflammation group, and PD-L1 dynamics suggest potential targets for further therapy. These findings highlight the need for targeted neoadjuvant strategies based on immune profiling.
Collapse
Affiliation(s)
- Alisa Kimura
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junichi Mitsuda
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kanako Yoshimura
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Sumiyo Saburi
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nanako Murakami
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nana Sakurai
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Yoshizawa
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroki Morimoto
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeyuki Mukudai
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hikaru Nagao
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Ogi
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- SCREEN Holdings Co. Ltd., Kyoto, Japan
| | | | - Aya Miyagawa-Hayashino
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiichi Konishi
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeru Hirano
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takahiro Tsujikawa
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan.
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
10
|
Morchón-Araujo D, Catani G, Mirallas O, Pretelli G, Sánchez-Pérez V, Vieito M, Braña I, Pujol-Borrell R, Garralda E, Hernando-Calvo A. Emerging Immunotherapy Targets in Early Drug Development. Int J Mol Sci 2025; 26:5394. [PMID: 40508202 PMCID: PMC12155519 DOI: 10.3390/ijms26115394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/24/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025] Open
Abstract
Immunotherapy has significantly changed the treatment paradigm for solid tumors, with immune checkpoint inhibitors now established in the management of many malignancies. Despite initial success, durable responses remain limited to a subset of patients, often less than 30%, due to both intrinsic and acquired resistance mechanisms. These challenges have prompted the development of next-generation immunotherapies. Recent efforts have expanded the scope of immunotherapy beyond PD-1/PD-L1 and CTLA-4 inhibition, focusing on new immune targets currently under investigation in early phase clinical trials. These include novel immune checkpoint inhibitors, immunomodulators targeting the tumor microenvironment, and bispecific antibodies. This review provides a comprehensive overview of emerging immune targets currently being investigated in early drug development, discussing their mechanisms of action, preliminary clinical outcomes, and potential future directions.
Collapse
Affiliation(s)
- Daniel Morchón-Araujo
- Department of Medical Oncology, University Hospital of Salamanca, IBSAL, 37007 Salamanca, Spain;
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
| | - Greta Catani
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Alexander Fleming Institute, Buenos Aires 1426, Argentina
| | - Oriol Mirallas
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Giulia Pretelli
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Vicky Sánchez-Pérez
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - María Vieito
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Irene Braña
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Ricardo Pujol-Borrell
- Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Tumor Immunology and Immunotherapy Group, Vall d’Hebron Institute of Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Elena Garralda
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Alberto Hernando-Calvo
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (G.C.); (O.M.); (G.P.); (V.S.-P.); (M.V.); (I.B.); (E.G.)
- Department of Medical Oncology, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| |
Collapse
|
11
|
Kado S, Komine M. Recent Advances in Molecular Research and Treatment for Melanoma in Asian Populations. Int J Mol Sci 2025; 26:5370. [PMID: 40508177 PMCID: PMC12154924 DOI: 10.3390/ijms26115370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 05/29/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025] Open
Abstract
Melanoma treatment comprised a few treatment choices with insufficient efficacy before the emergence of molecularly targeted medication and immune checkpoint inhibitors, which dramatically improved patient outcomes. B-Rapidly Accelerated Fibrosarcoma (BRAF) and Mitogen-Activated Protein Kinase (MAPK) Kinase (MEK) inhibitors significantly improved survival in BRAF-mutant melanoma and immune checkpoint inhibitors, such as anti-programmed cell death 1 (PD-1) and Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) agents, established new standards of care. Challenges remain, however, including the existence of resistance mechanisms and the reduced efficacy of immune-based therapies in Asian populations, particularly for acral and mucosal subtypes. This review highlights historical and current therapeutic advancements, discusses regional considerations, and explores emerging strategies aiming at globally optimizing melanoma management.
Collapse
Affiliation(s)
- Soichiro Kado
- Department of Dermatology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke 329-0498, Tochigi, Japan;
| | - Mayumi Komine
- Department of Dermatology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke 329-0498, Tochigi, Japan;
- Department of Biochemistry, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke 329-0498, Tochigi, Japan
| |
Collapse
|
12
|
Lipson EJ, Stephen Hodi F, Tawbi H, Schadendorf D, Ascierto PA, Matamala L, Gutierrez EC, Rutkowski P, Gogas HJ, Lao CD, Menezes JJD, Dalle S, Arance A, Gaudy-Marqueste C, Chen B, Jackson W, Mukherjee S, Dolfi S, Long GV. Nivolumab plus relatlimab in advanced melanoma: RELATIVITY-047 4-year update. Eur J Cancer 2025; 225:115547. [PMID: 40513285 DOI: 10.1016/j.ejca.2025.115547] [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: 03/31/2025] [Revised: 05/23/2025] [Accepted: 06/02/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND In phase 2/3 randomized RELATIVITY-047, nivolumab plus relatlimab demonstrated a statistically significant improvement in progression-free survival (PFS), a clinically meaningful but not statistically significant improvement in overall survival (OS), and a numerically higher objective response rate (ORR) versus nivolumab alone in patients with previously untreated advanced melanoma. METHODS Descriptive 4-year updated analyses in patients treated with nivolumab 480 mg plus relatlimab 160 mg fixed-dose combination versus nivolumab 480 mg intravenously every 4 weeks are presented. Primary endpoint was PFS by blinded independent central review (BICR). Other endpoints included melanoma-specific survival (MSS). RESULTS At 45.3 months' minimum follow-up, nivolumab plus relatlimab versus nivolumab PFS improvement was maintained: 4-year PFS rates were 30.6 % (95 % CI, 25.4-35.9) versus 23.6 % (95 % CI, 18.9-28.5); OS was numerically better with 4-year OS rates of 52.0 % (95 % CI, 46.6-57.1) versus 42.8 % (95 % CI, 37.5-47.9); and ORR difference was maintained at 43.9 % (95 % CI, 38.7-49.3) versus 33.4 % (95 % CI, 28.6-38.6), respectively. 4-year MSS rates were 59.7 % (95 % CI, 54.1-64.8) for nivolumab plus relatlimab and 49.6 % (95 % CI, 44.0-54.9) for nivolumab. Efficacy across the majority of prespecified subgroups favored the combination. No new or unexpected safety signals were identified. CONCLUSIONS With 4 years of follow-up, nivolumab plus relatlimab demonstrated durable improvement in outcomes versus nivolumab monotherapy for patients with previously untreated advanced melanoma. The durable benefit observed comes at a lower toxicity cost compared with other immuno-oncology combinations. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03470922.
Collapse
Affiliation(s)
- Evan J Lipson
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | | | - Hussein Tawbi
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dirk Schadendorf
- University of Essen and the German Cancer Consortium, Essen, Germany
| | - Paolo A Ascierto
- Istituto Nazionale dei Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Luis Matamala
- Instituto Oncológico Fundación Arturo López Pérez and Department of Oncology, Instituto Nacional del Cáncer, Santiago, Chile
| | | | - Piotr Rutkowski
- Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Helen J Gogas
- National and Kapodistrian University of Athens, Athens, Greece
| | - Christopher D Lao
- Michigan Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | | | - Stephane Dalle
- Hospices Civils de Lyon, Cancer Research Center of Lyon, Pierre-Bénite, France
| | - Ana Arance
- Hospital Clinic Barcelona and IDIBAPS, Barcelona, Spain
| | | | | | | | | | | | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| |
Collapse
|
13
|
Joshi UM, Hundal J, Mata JR, Schollenberger MD, Warrier G, Luke JJ, Lipson EJ, Funchain P. Beyond Checkpoint Inhibition: Keeping Therapeutic Options Open. Am Soc Clin Oncol Educ Book 2025; 45:e473856. [PMID: 40233298 DOI: 10.1200/edbk-25-473856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Combination immune checkpoint inhibitor therapy (ICI) with ipilimumab (anti-cytotoxic T-lymphocyte-associated protein 4) + nivolumab (anti-PD-1) in untreated, metastatic melanoma has achieved a ten-year melanoma-specific survival of 52%. However, approximately 40%-55% of patients with metastatic melanoma have primary resistance and do not initially respond to anti-PD-1, and an additional 25% of patients develop secondary resistance, exhibiting an initial response followed by disease progression. In PD-1-refractory melanoma, treatment options are limited. Addition of ipilimumab, relatlimab (anti-LAG3), or lenvatinib (VEGFR TKI) has minimal to modest efficacy. Switching to targeted BRAF/MEK inhibition improves survival for BRAF-mutant disease. MEK and KIT inhibitors have limited activity in NRAS- and KIT-mutant metastatic melanoma, respectively. Recently, personalized, autologous tumor-infiltrating lymphocyte therapy has become a US Food and Drug Administration-approved second-line option; lifileucel demonstrates durable response (approximately 30%) in heavily pretreated, metastatic melanoma. Emerging therapeutics that show promising clinical benefit in ongoing clinical trials include novel engineered oncolytic viral and human leukocyte antigen (HLA)-restricted immune-mediated T-cell therapies. As a therapy which is limited to patients who are HLA-A*02:01, T-cell receptor (TCR) engineered T cells (TCR-T) iterates on personalized adoptive cell transfer, and immune mobilizing monoclonal TCRs against cancer are CD3 bispecifics that bind glycoprotein 100 (tebentafusp, approved for metastatic uveal melanoma) or PRAME to activate T cells. Finally, in patients at high risk for immune-related adverse events (irAEs), ICI should still be considered. ICI may be given with modified immunosuppression in patients with autoimmune disease or previous organ transplantation. Cumulative data support safe administration in older patients and in ICI rechallenge for patients with previous irAE.
Collapse
Affiliation(s)
- Urvashi Mitbander Joshi
- Division of Malignant Hematology and Medical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Jasmin Hundal
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | | | - Megan D Schollenberger
- Department of Oncology, Johns Hopkins University, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Govind Warrier
- Department of Oncology, Johns Hopkins University, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jason J Luke
- Division of Malignant Hematology and Medical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Evan J Lipson
- Department of Oncology, Johns Hopkins University, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Pauline Funchain
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
14
|
Robert C, Kicinski M, Dutriaux C, Routier É, Govaerts AS, Bührer E, Neidhardt EM, Durando X, Baroudjian B, Saiag P, Gaudy-Marqueste C, Ascierto PA, Arance A, Russillo M, Perrot JL, Mortier L, Aubin F, Dalle S, Grange F, Muñoz-Couselo E, Mary-Prey S, Amini-Adle M, Mansard S, Lebbe C, Funck-Brentano E, Monestier S, Eggermont AMM, Oppong F, Wijnen L, Schilling B, MandalÁ M, Lorigan P, van Akkooi ACJ. Combination of encorafenib and binimetinib followed by ipilimumab and nivolumab versus ipilimumab and nivolumab in patients with advanced melanoma with BRAF V600E or BRAF V600K mutations (EBIN): an international, open-label, randomised, controlled, phase 2 study. Lancet Oncol 2025; 26:781-794. [PMID: 40449497 DOI: 10.1016/s1470-2045(25)00133-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND Current first-line treatment for patients with metastatic melanoma with BRAFV600E or BRAFV600K mutations includes immunotherapy with immune checkpoint inhibitors and targeted therapy; however, the optimal sequencing of these treatments is unclear. We aimed to investigate the use of a targeted-therapy induction regimen before treatment with immune checkpoint inhibitors. METHODS This open-label, randomised, controlled, phase 2 trial (EBIN) was conducted at 37 centres in eight European countries. Eligible patients were 18 years or older and had previously untreated, unresectable, stage III or IV melanoma with BRAFV600E or BRAFV600K mutations and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned (1:1) to one of two groups. Those in the induction group received targeted therapy (oral encorafenib 450 mg once a day plus oral binimetinib 45 mg twice a day for 12 weeks) followed by immune checkpoint inhibitors (intravenous nivolumab 3 mg/kg plus intravenous ipilimumab 1 mg/kg once every 3 weeks for four doses, followed by intravenous nivolumab 480 mg once every 4 weeks until unacceptable toxicity, disease progression, or 2 years of treatment). Patients in the control group received immune checkpoint inhibitors as above without any induction targeted therapy. Randomisation was conducted using a minimisation technique and was stratified by centre and a variable defined using stage and lactate dehydrogenase activity. The primary outcome was progression-free survival in the intention-to-treat population. Safety was assessed in all patients who initiated the protocol treatment. In this Article we report the primary analysis. The study is registered with ClinicalTrials.gov, NCT03235245, and is ongoing. FINDINGS Between Nov 12, 2018, and July 11, 2022, 271 patients were randomly assigned: 136 to the induction group and 135 to the control group. 103 (38%) patients were female, 168 (62%) were male, and the median age was 55 years (IQR 43-66). The median follow-up time was 21 months (IQR 13-33). There was no evidence of a longer progression-free survival in the induction group than in the control group (hazard ratio 0·87, 90% CI 0·67-1·12; p=0·36). The median progression-free survival was 9 months (95% CI 7-13) in the induction group and 9 months (5-14) in the control group. Grade 3-5 treatment-related adverse events occurred in 57 (42%) of 136 patients who started treatment in the induction group and in 42 (32%) of 131 patients who started treatment in the control group. The most common grade 3-4 treatment-related adverse event was hepatitis (17 [13%] of 136 patients in the induction group and nine [7%] of 131 patients in the control group). Serious treatment-related adverse events occurred in 45 (33%) of 136 patients in the induction group and 33 (25%) of 131 patients in the control group. There were three treatment-related deaths: two from cardiac events (heart failure and arrhythmia) in the induction group and one from meningitis in the control group. INTERPRETATION The targeted-therapy induction regimen did not improve progression-free survival compared with first-line treatment with immune checkpoint inhibitors in unselected patients with advanced melanoma with BRAFV600E or BRAFV600K mutations. FUNDING Bristol Myers Squibb and Pierre Fabre.
Collapse
Affiliation(s)
- Caroline Robert
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, University of Paris-Saclay, Villejuif, France.
| | | | - Caroline Dutriaux
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France
| | - Émilie Routier
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, University of Paris-Saclay, Villejuif, France
| | | | | | | | - Xavier Durando
- INSERM U1240 IMoST, Université Clermont Auvergne, Clermont-Ferrand, France; Département de Recherche Clinique, Délégation Recherche Clinique et Innovation, Centre Jean Perrin, Clermont-Ferrand, France; Département d'Oncologie Médicale, Centre Jean Perrin, Clermont-Ferrand, France; Centre d'Investigation Clinique UMR501, Clermont-Ferrand, France
| | - Barouyr Baroudjian
- Université Paris Cité, AP-HP Dermato-oncology, Cancer Institute AP-HP, Nord Paris Cité, INSERM U976, Saint Louis Hospital, Paris, France
| | - Philippe Saiag
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, APHP & EA 4340 "Biomarkers in cancerology and hemato-oncology", UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Caroline Gaudy-Marqueste
- Dermatology and Skin Cancer Department, Aix Marseille Univ, APHM, La Timone Hospital, Marseille, France
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS 'Fondazione G Pascale', Naples, Italy
| | - Ana Arance
- Department of Medical Oncology and IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Michelangelo Russillo
- Sarcoma and Rare Tumours Departmental Unit, IRCCS Regina Elena National Cancer Institute Rome, Rome, Italy
| | - Jean-Luc Perrot
- Groupe d'Imagerie Cutanée Non Invasive (GICNI), Société Française de Dermatologie (SFD), Paris, France; Department of Dermatology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Laurent Mortier
- Department of Dermatology, INSERM U1189, CHU Lille, CARADERM, Lille University, Lille, France
| | - Francois Aubin
- Department of Dermatology, UHC and INSERM 1098, Besançon, France
| | - Stéphane Dalle
- Dermatology Department, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Florent Grange
- Dermatology/Oncology, CHU Reims-Hôpital Robert Debre, Reims, France; Dermatology Department, Valence Hospital, Valence, France
| | - Eva Muñoz-Couselo
- Department of Oncology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Sorilla Mary-Prey
- Service de Dermatologie, CHU de Bordeaux, Bordeaux, France; BRIC (Bordeaux Institute of Oncology), INSERM UMR1312, Université de Bordeaux, Bordeaux, France
| | | | - Sandrine Mansard
- Service de Dermatologie, Centre Hospitalo-Universitaire de Clermont Auvergne, Clermont-Ferrand, France
| | - Céleste Lebbe
- Université Paris Cité, AP-HP Dermato-oncology, Cancer Institute AP-HP, Nord Paris Cité, INSERM U976, Saint Louis Hospital, Paris, France
| | - Elisa Funck-Brentano
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, APHP & EA 4340 "Biomarkers in cancerology and hemato-oncology", UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Sandrine Monestier
- Dermatology and Skin Cancer Department Aix Marseille Univ, APHM, La Timone Hospital, Marseille, France
| | - Alexander M M Eggermont
- Board of Comprehensive Cancer Center Munich of the Technical University Munich and the Ludwig Maximilians University, Munich, Germany; Princess Máxima Center, Utrecht, Netherlands
| | | | | | - Bastian Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany; Department of Dermatology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Mario MandalÁ
- Unit of Oncology, Santa Maria Misericordia Hospital, University of Perugia, Perugia, Italy
| | - Paul Lorigan
- Division of Cancer Sciences, University of Manchester and Christie NHS Foundation Trust, Manchester, UK
| | - Alexander C J van Akkooi
- Melanoma Institute Australia, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
15
|
Day D, Ganju V, Chung K, Si L, Mao L, Aghmesheh M, Hoyer R, Brewin K, Zeng S, Zhang M, Lu Q, Jiang C, Ren F, Zhu Y, Guo J. First-in-human phase I study of EMB-02, a bispecific antibody targeting PD-1 and LAG-3 in patients with advanced solid tumors. Br J Cancer 2025; 132:905-912. [PMID: 40234667 DOI: 10.1038/s41416-025-02990-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/08/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND EMB-02 is a symmetric bispecific antibody targeting programmed cell death protein-1 and lymphocyte-activation gene 3 simultaneously. Here, we present the first-in-human study results of EMB-02 in patients with advanced solid tumors. METHODS Patients were treated with intravenous infusions of EMB-02 at doses of 6-900 mg. The primary objective was to evaluate the safety and tolerability and to determine the maximum tolerated dose and/or recommended phase II dose(s). Secondary objectives included characterizing the pharmacokinetic (PK) profile, assessing preliminary antitumor activity and the immunogenicity. RESULTS A total of 47 patients were enrolled. All grade and grade 3/4 treatment-emergent and treatment related adverse events occurred in 97.9%, 48.9%, 68.1% and 12.8% patients, respectively. The objective response rate (ORR) was 6.4% and clinical benefit rate at 24 weeks (CBR-24) was 25.5% in overall population. The CBR-24 was 33.3% in checkpoint inhibitor (CPI)-naïve patients, and 15% in CPI-treated. No clear relationship was observed between the efficacy and PD-L1, LAG-3, or MHC II expression level. Doses 360 mg or higher resulted in sustained saturation of PD-1 receptors on circulating CD3 + T cells. CONCLUSIONS EMB-02 demonstrated a favorable safety profile and early efficacy signals in multiple solid tumors, warranting further development. (NCT04618393).
Collapse
Affiliation(s)
- Daphne Day
- Medical Oncology Department, Monash Health-Monash MedicalCentre, Clayton, VIC, Australia
| | - Vinod Ganju
- Oncology Department, Peninsula And Southeast Oncology, Frankston, VIC, Australia
| | - Ki Chung
- Department of Medicine, Prisma Health Cancer Institute, Greenville, SC, USA
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Collaborative Innovation Center for Cancer Medicine, Peking UniversityCancer Hospital and Institute, Beijing, China
| | - Lili Mao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Collaborative Innovation Center for Cancer Medicine, Peking UniversityCancer Hospital and Institute, Beijing, China
| | - Morteza Aghmesheh
- Medical Oncology Department, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Robert Hoyer
- Medical Oncology Department, UCHealth Memorial Hospital Central, Colorado Springs, CO, USA
| | - Kim Brewin
- Medical Oncology Department, Monash Health-Monash MedicalCentre, Clayton, VIC, Australia
| | - Shuqi Zeng
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Mingfei Zhang
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Qiaoyang Lu
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Chengjun Jiang
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Fang Ren
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Yonghong Zhu
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Collaborative Innovation Center for Cancer Medicine, Peking UniversityCancer Hospital and Institute, Beijing, China.
| |
Collapse
|
16
|
Elkrief A, Routy B, Derosa L, Bolte L, Wargo JA, McQuade JL, Zitvogel L. Gut Microbiota in Immuno-Oncology: A Practical Guide for Medical Oncologists With a Focus on Antibiotics Stewardship. Am Soc Clin Oncol Educ Book 2025; 45:e472902. [PMID: 40262063 DOI: 10.1200/edbk-25-472902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The gut microbiota has emerged as a critical determinant of immune checkpoint inhibitor (ICI) efficacy, resistance, and toxicity. Retrospective and prospective studies profiling the taxonomic composition of intestinal microbes of patients treated with ICI have revealed specific gut microbial signatures associated with response. By contrast, dysbiosis, which can be caused by chronic inflammatory processes (such as cancer) or comedications, is a risk factor of resistance to ICI. Recent large-scale meta-analyses have confirmed that antibiotic (ATB) use before or during ICI therapy alters the microbiota repertoire and significantly shortens overall survival, even after adjusting for prognostic factors. These results underscore the importance of implementing ATB stewardship recommendations in routine oncology practice. Microbiota-centered interventions are now being explored to treat gut dysbiosis and optimize ICI responses. Early-phase clinical trials evaluating fecal microbiota transplantation (FMT) from ICI responders or healthy donors have shown that this approach is safe and provided preliminary data on potential efficacy to overcome both primary and secondary resistance to ICI in melanoma, non-small cell lung cancer, and renal cell carcinoma. More targeted interventions including live bacterial products including Clostridium butyricum and Akkermansia massiliensis represent novel microbiome-based adjunct therapies. Likewise, dietary interventions, such as high-fiber diets, have shown promise in enhancing ICI activity. In this ASCO Educational Book, we summarize the current state-of-the-evidence of the clinical relevance of the intestinal microbiota in cancer immunotherapy and provide a practical guide for ATB stewardship.
Collapse
Affiliation(s)
- Arielle Elkrief
- University of Montreal Hospital Research Centre, Cancer Axis, Montreal, Canada
- University of Montreal Hospital Centre, Department of Hematology-Oncology, Montreal, Canada
| | - Bertrand Routy
- University of Montreal Hospital Research Centre, Cancer Axis, Montreal, Canada
- University of Montreal Hospital Centre, Department of Hematology-Oncology, Montreal, Canada
| | - Lisa Derosa
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Gustave Roussy, ClinicObiome, Villejuif, France
- Université Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France
| | - Laura Bolte
- Department of Medical Oncology, University Groningen and University Medical Center, Groningen, the Netherlands
- Department of Gastroenterology and Hepatology, University Groningen and University Medical Center, Groningen, the Netherlands
| | | | | | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Gustave Roussy, ClinicObiome, Villejuif, France
- Université Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| |
Collapse
|
17
|
Shao YF, Baca Y, Hinton A, Xiu J, VanderWalde A, Hadfield M, Park SJ, Darabi S, Sato T, Moser JC. Immune Profiling of Uveal Melanoma Liver Metastases and Response to Checkpoint Inhibitors. J Immunother 2025; 48:189-195. [PMID: 40231356 PMCID: PMC12052074 DOI: 10.1097/cji.0000000000000558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/13/2025] [Indexed: 04/16/2025]
Abstract
Responses to immune checkpoint inhibitors (ICIs) differ significantly between uveal melanoma (UM) and cutaneous melanoma (CM) patients. We investigated the immune profile of metastatic UM(mUM) patients and identified markers that are predictive of improved survival. Metastatic liver samples from 189 UM patients and 48 CM patients were analyzed at genomic and transcriptional levels, and survival analysis was performed on a subgroup of 76 ICI-treated mUM patients. UM liver metastases seem to preserve the genomic and immune characteristics of primary UM (pUM), with a low prevalence of ICI markers and high mutation rates of GNA11, GNAQ, BAP1, and SF3B. Compared with mCM, UM liver metastasis showed lower infiltration of several immune cells, but a greater proportion of M2 macrophages. Compared with UM liver metastases, CM liver metastases showed higher expression of G2M checkpoint and EF2 target genes. Among the mUM and mCM samples, expression of G2M and E2F pathway genes was highest in tumors with high TMB values and T-cell inflamed scores. A longer median overall survival (OS) was observed in mUM patients with higher expression of LAG3, HLA class I, or HLA class II; which may represent a small proportion of immune hot tumors. Expression patterns of G2M checkpoint and E2F targets suggest a possible contribution to immunotherapy response.
Collapse
Affiliation(s)
- Yusra F. Shao
- Karmanos Cancer Institute and Wayne State University, Detroit, MI
| | - Yasmine Baca
- Medical Affairs, Caris Life Sciences, Phoenix, AZ
| | | | - Joanne Xiu
- Medical Affairs, Caris Life Sciences, Phoenix, AZ
| | | | | | - Soo J. Park
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | | | - Takami Sato
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | |
Collapse
|
18
|
Amrane K, Le Noac'h P, Hemon P, Abgral R, Le Meur C, Pradier O, Misery L, Legoupil D, Berthou C, Uguen A. MHC class II: a predictor of outcome in melanoma treated with immune checkpoint inhibitors. Melanoma Res 2025; 35:176-186. [PMID: 39945603 DOI: 10.1097/cmr.0000000000001022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
This study aimed to evaluate the predictive value of MHC class II (MHC-II) expression by melanoma cells in a large cohort of metastatic cutaneous melanoma patients treated with immune checkpoint inhibitors (ICIs). We conducted a single-center, retrospective study involving stage IV cutaneous melanoma patients who received ICI as first-line therapy. MHC-II expression in melanoma cells was quantified using dual-color anti-SOX10 and anti-MHC-II immunohistochemistry on tumor samples from 95 patients. The primary endpoint was event-free survival (EFS), with secondary endpoints including 1-year EFS, 1-year overall survival (OS), disease control rate (DCR), and the correlation between MHC-II expression and clinico-biological characteristics. The cohort had a median age of 67 years (range, 33-90), with a male-to-female ratio of 50 : 45. Thirty-three percent of patients received the ipilimumab-nivolumab combination. The median follow-up was 16.8 months. Disease progression occurred in 58 patients (61%), with a median time to progression of 4.8 months. Forty-six patients (48.4%) experienced an event within the first year, and 52 patients (54.7%) died during follow-up. MHC-II positivity was observed in ≥10% of melanoma cells in 6.3% of patients. MHC-II expression was significantly associated with 1-year EFS ( P = 0.037) and DCR ( P = 0.032), but not with EFS or 1-year OS. Age, phototype, and brain metastases were correlated with MHC-II expression status. Our findings suggest that MHC-II expression by melanoma cells may serve as a favorable predictive biomarker for survival in metastatic cutaneous melanoma patients treated with ICIs.
Collapse
Affiliation(s)
- Karim Amrane
- Department of Oncology, Regional Hospital of Morlaix, Morlaix
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
| | - Pierre Le Noac'h
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- Department of Pathology, University Hospital of Brest
| | - Patrice Hemon
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
| | - Ronan Abgral
- Department of Nuclear Medicine, University Hospital of Brest
- UMR Inserm 1304 GETBO, IFR 148, University of Western Brittany
| | - Coline Le Meur
- Department of Radiotherapy, University Hospital of Brest
| | - Olivier Pradier
- Department of Radiotherapy, University Hospital of Brest
- Inserm, UMR1101, LaTIM, University of Western Brittany
| | - Laurent Misery
- Department of Dermatology, University Hospital of Brest
- Laboratoire sur les Interactions Épithéliums-Neurones (LIEN-EA4685), Université de Bretagne Occidentale
| | - Delphine Legoupil
- Department of Dermatology, University Hospital of Brest
- Laboratoire sur les Interactions Épithéliums-Neurones (LIEN-EA4685), Université de Bretagne Occidentale
| | - Christian Berthou
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- Department of Hematology, University Hospital of Brest, Brest, France
| | - Arnaud Uguen
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- UMR Inserm 1304 GETBO, IFR 148, University of Western Brittany
| |
Collapse
|
19
|
Zenderowski V, Schreieder L, Drexler K, Haferkamp S. [New therapeutic approaches in the neoadjuvant/adjuvant treatment of melanoma]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2025; 76:354-360. [PMID: 40407846 DOI: 10.1007/s00105-025-05508-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/07/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND Although effective treatment options for malignant melanoma already exist, currently available adjuvant and neoadjuvant therapies are not always sufficient to prevent relapse or ensure a long-term treatment response. Many patients develop resistance or show inadequate responses to existing therapies. Therefore, there is an urgent need for new and more effective therapeutic approaches in the adjuvant and neoadjuvant settings to sustainably improve patient prognosis. OBJECTIVE This study provides an overview of current developments in melanoma treatment, with a particular focus on the adjuvant and neoadjuvant application of novel immunotherapies. MATERIAL AND METHODS A literature search and discussion of relevant recent studies was carried out. RESULTS Adjuvant mRNA-based adjuvant treatment combined with pembrolizumab demonstrated a significantly improved relapse-free survival compared to pembrolizumab monotherapy (79% vs. 62%). The value of adjuvant LAG‑3 antibodies in combination with PD‑1 blockers remains inconclusive. A neoadjuvant intralesional treatment with daromun after complete removal of tumor tissue reduced the risk of recurrence by 41% and led to a significant extension in relapse-free survival (16.7 months vs. 6.8 months). Tebentafusp improved overall survival in metastatic uveal melanoma and is currently being investigated in the adjuvant and neoadjuvant setting. DISCUSSION The mRNA-based treatment combined with checkpoint inhibitors have the potential to induce long-term immune responses. The efficacy of LAG-3 inhibitors in the adjuvant setting is currently under evaluation in clinical studies. The immunocytokine treatment with daromun shows promising results in the neoadjuvant setting by stimulating both local and systemic immune responses. Future studies should focus on identifying optimal combinations of treatment to improve the long-term prognosis of patients.
Collapse
Affiliation(s)
- Veronika Zenderowski
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Deutschland.
| | - Laura Schreieder
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Deutschland
| | - Konstantin Drexler
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Deutschland
| | - Sebastian Haferkamp
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Deutschland
| |
Collapse
|
20
|
Zaemes J, Gibney GT. Cellular Therapy for Advanced Melanoma. Surg Clin North Am 2025; 105:681-690. [PMID: 40412894 DOI: 10.1016/j.suc.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Cellular therapy has been an evolving therapeutic approach in advanced melanoma over the past 40 y. The first tumor infiltrating lymphocyte (TIL) therapy, lifileucil, was Food and Drug Administration-approved in 2024 for patients with metastatic melanoma who have previously been treated with an anti-PD-1 therapy and BRAF inhibitor (BRAF V600 mutant disease). Further clinical development of TIL therapy will hopefully lead to safer and more effective strategies. Cellular therapy in melanoma has also expanded beyond TIL therapy with anti-tumor activity demonstrated for TCR-transduced T-cell products and T-cell engager bi-specific agents that target melanoma antigens.
Collapse
Affiliation(s)
- Jacob Zaemes
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, USA
| | - Geoffrey T Gibney
- Lombardi Comprehensive Cancer Center, Medstar Georgetown University Hospital, Washington, DC, USA.
| |
Collapse
|
21
|
Yao Y, Liu Y, Lu B, Ji G, Wang L, Dong K, Zhao Z, Lyu D, Wei M, Tu S, Lyu X, Li Y, Huang R, Zhou W, Xu G, Pan X, Cui X. Construction and validation of a regulatory T cells-based classification of renal cell carcinoma: an integrated bioinformatic analysis and clinical cohort study. Cell Oncol (Dordr) 2025; 48:591-615. [PMID: 39714755 PMCID: PMC12119669 DOI: 10.1007/s13402-024-01030-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 12/24/2024] Open
Abstract
PURPOSE Renal cell carcinoma (RCC), exhibiting remarkable heterogeneity, can be highly infiltrated by regulatory T cells (Tregs). However, the relationship between Treg and the heterogeneity of RCC remains to be explored. METHODS We acquired single-cell RNA-seq profiles and 537 bulk RNA-seq profiles of TCGA-KIRC cohort. Through clustering, monocle2 pseudotime and prognostic analyses, we identified Treg states-related prognostic genes (TSRPGs), then constructing the RCC Treg states-related prognostic classification (RCC-TSC). We also explored its prognostic significance and multi-omics landmarks. Additionally, we utilized correlation analysis to establish regulatory networks, and predicted candidate inhibitors. More importantly, in Xinhua cohort of 370 patients with kidney neoplasm, we used immunohistochemical (IHC) staining for classification, then employing statistical analyses including Chi-square tests and multivariate Cox proportional hazards regression analysis to explore its clinical relevance. RESULTS We defined 44 TSRPGs in four different monocle states, and identified high immune infiltration RCC (HIRC, LAG3+, Mki67+) as the highly exhausted subtype with the worst prognosis in RCC-TSC (p < 0.001). BATF-LAG3-immune cells axis might be its underlying metastasis-related mechanism. Immunotherapy and inhibitors including sunitinib potentially conferred best therapeutic effects for HIRC. Furthermore, we successfully validated HIRC subtype as an independent prognostic factor within the Xinhua cohort (OS, HR = 16.68, 95% CI = 1.88-148.1, p = 0.011; PFS, HR = 4.43, 95% CI = 1.55-12.6, p = 0.005). CONCLUSION Through integrated bioinformatics analysis and a large-sample retrospective clinical study, we successfully established RCC-TSC and a diagnostic kit, which could stratify RCC patients with different prognosis and to guide personalized treatment.
Collapse
Affiliation(s)
- Yuntao Yao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yifan Liu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Bingnan Lu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guo Ji
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lei Wang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Keqin Dong
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zihui Zhao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Donghao Lyu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Maodong Wei
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Siqi Tu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xukun Lyu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuanan Li
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| | - Wang Zhou
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Guofeng Xu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Xingang Cui
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
22
|
Sperduto PW, Marqueen KE, Chang E, Li J, Davies MA, Ebner DK, Breen WG, Lamba N, Shih HA, Edwards D, Kim MM, Mahal A, Rahman R, Ankrah N, Boggs DH, Lewis C, Hyer D, Buatti JM, Johri F, Soliman H, Masucci L, Roberge D, Aneja S, Chiang V, Phuong C, Braunstein S, Dajani S, Sachdev S, Wan Z, Niedzwiecki D, Vaios E, Kirkpatrick JP, Pasetsky J, Wang TJ, Kutuk T, Kotecha R, Ross RB, Rusthoven CG, Nakano T, Tawbi HA, Mehta MP. Improved Survival and Prognostication in Melanoma Patients With Brain Metastases: An Update of the Melanoma Graded Prognostic Assessment. J Clin Oncol 2025; 43:1910-1919. [PMID: 40245362 PMCID: PMC12119226 DOI: 10.1200/jco-24-01351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/24/2025] [Accepted: 02/21/2025] [Indexed: 04/19/2025] Open
Abstract
PURPOSE Survival for patients with melanoma has recently improved. The propensity of melanoma to metastasize to the brain remains a common and serious feature of this disease. The purposes of this study were to evaluate prognostic factors for patients with newly diagnosed melanoma brain metastases (MBMs) in a large cohort treated with modern multimodal therapies, compare those results with those in prior eras, and update the Melanoma Graded Prognostic Assessment (GPA). METHODS Univariable and multivariable (MVA) analyses of prognostic factors and treatments associated with survival were performed on 1,796 patients with newly diagnosed MBM treated between January 01, 2015, and December 31, 2021, using a multi-institutional retrospective database. Multiple imputation was used to address missingness of potential predictors. Significant variables in combined MVA were used to update the Melanoma GPA. Comparisons were made with legacy cohorts. RESULTS Median survivals for cohorts A (1985-2007, n = 481), B (2006-2015, n = 823), and C (2015-2021, n = 1,796) were 6.7, 9.8, and 16.6 months and median follow-up times were 40.1, 43.6, and 48.8 months, respectively. In combined MVA, significant prognostic factors for survival were higher Karnofsky Performance Status, fewer MBMs, absence of extracranial metastases, lower serum lactate dehydrogenase, and no immunotherapy before MBM. These factors were incorporated into the updated Melanoma GPA. The combined median and 3-year survivals for patients with GPA 0-1, 1.5-2, and 2.5-4.0 were 5.4, 13.2, and 43.2 months and 12.4%, 28.8%, and 51.6%, respectively. CONCLUSION Prognostic factors have changed and survival has improved for patients with MBM but varies widely by GPA. The updated Melanoma GPA calculator (BrainMetGPA), available free online, can be used to estimate survival, individualize treatment, stratify clinical trials, guide surveillance, and augment clinical trial eligibility. Multidisciplinary treatment is essential. Trials are needed to elucidate the optimal sequencing of various therapeutic modalities.
Collapse
Affiliation(s)
| | | | - Enoch Chang
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jing Li
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | | | | | | | | - Nii Ankrah
- University of Alabama, Birmingham, Birmingham, AL
| | | | | | | | | | - Fasila Johri
- Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Canada
| | - Hany Soliman
- Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Canada
| | - Laura Masucci
- Centre Hospitalier de l’Universite de Montreal, Montreal, Canada
| | - David Roberge
- Centre Hospitalier de l’Universite de Montreal, Montreal, Canada
| | | | | | | | | | | | | | - Zihan Wan
- Duke Cancer Institute-Biostatistics Shared Resource, Durham, NC
| | | | | | | | | | | | - Tugce Kutuk
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | - Rupesh Kotecha
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | | | | | | | | | - Minesh P. Mehta
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| |
Collapse
|
23
|
Williams TP, Vaghjiani RG, Tyler DS. Melanoma and Merkel Cell Carcinoma Clinical Trial Status. Surg Clin North Am 2025; 105:691-699. [PMID: 40412895 DOI: 10.1016/j.suc.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
In the last decade, the management of melanoma and Merkel cell carcinoma has changed dramatically following the results of landmark clinical trials in surgical and medical management. This review will provide a brief background for the clinical surgical oncologist as to how certain trials are conducted and designated by regulatory bodies. Additionally, we will herein review the landscape of ongoing multicenter interventional trials for cutaneous melanoma and Merkel cell carcinoma.
Collapse
Affiliation(s)
- Taylor P Williams
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Raj G Vaghjiani
- Division of Surgical Oncology, Department of Surgery, The University of Texas Medical Branch, Galveston, TX 77555-0527, USA
| | - Douglas S Tyler
- Division of Surgical Oncology, Department of Surgery, The University of Texas Medical Branch, Galveston, TX 77555-0527, USA.
| |
Collapse
|
24
|
Farooq MS, Tidwell JC, Karakousis GC. Neoadjuvant Therapy in Melanoma: Current Evidence and Future Directions of Investigation. Surg Clin North Am 2025; 105:569-589. [PMID: 40412887 DOI: 10.1016/j.suc.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Neoadjuvant therapy for advance-stage melanoma has had increasing momentum over the past decade owing to several landmark clinical trials. Neoadjuvant therapy has now been shown to confer multiple advantages over adjuvant therapy, including more robust antitumor immunity, improved prognostication, and ability to personalize surgical and medical therapy based on therapeutic response. Neoadjuvant therapy has led to a major shift in clinical and surgical practice for melanoma, and future trials will give further insight into improving patient outcomes.
Collapse
Affiliation(s)
- Mohammad S Farooq
- Division of Endocrine and Oncologic Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| | - Jerica C Tidwell
- Department of Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, 4 Maloney, Philadelphia, PA 19104, USA
| | - Giorgos C Karakousis
- Division of Endocrine and Oncologic Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
25
|
Braverman EL, Mognol GP, Minn AJ, Vignali DAA, Varner JA. One Step Ahead: Preventing Tumor Adaptation to Immune Therapy. Am Soc Clin Oncol Educ Book 2025; 45:e481556. [PMID: 40334183 DOI: 10.1200/edbk-25-481556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Immune checkpoint inhibitors are cancer therapeutics that have shown remarkable success in extending lives in many cancers, including melanoma, MSI-high cancers, and other cancers. However, these therapeutics have not shown benefit for many patients with cancer, especially those with advanced cancer diagnoses. In addition, many patients develop resistance to these therapeutics and/or life-altering adverse events that can include cardiotoxicity, pneumonitis, thyroiditis, pancreatitis, and hepatitis. Extensive efforts to improve cancer care by uncovering mechanisms of resistance to immune therapy in solid tumors have led to identification of new sources of resistance and to the development of new approaches to activate or sustain antitumor immunity. Chronic stimulation of T cells by tumors and by checkpoint inhibitors can lead to a progressive state of T-cell exhaustion. Chronic T-cell activation by the tumor microenvironment (TME) or immune therapeutics can upregulate the expression and function of alternate checkpoints, including the T-cell protein LAG-3. Persistent interferon signaling in the TME can drive epigenetic changes in cancer cells that enable tumors to counter immune activation and disrupt tumor cell elimination. In addition, immune-suppressive macrophages can flood tumors in response to signals from dying tumor cells, further preventing effective immune responses. New clinical developments and/or approvals for therapies that target alternate immune checkpoints, such as the T-cell checkpoint LAG-3; myeloid cell proteins, such as the kinase phosphoinositide 3-kinase gamma isoform; and chronic interferon signaling, such as Jak 1 inhibitors, have been approved for cancer care or shown promise in recent clinical trials.
Collapse
Affiliation(s)
- Erica L Braverman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Giuliana P Mognol
- Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Andy J Minn
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Judith A Varner
- Moores Cancer Center, University of California, San Diego, La Jolla, CA
- Department of Pathology, University of California, San Diego, La Jolla, CA
| |
Collapse
|
26
|
Lens M, Schachter J. Immune Checkpoint Inhibitors in the Treatment of Advanced Melanoma in Older Patients: An Overview of Published Data. Cancers (Basel) 2025; 17:1835. [PMID: 40507314 PMCID: PMC12153560 DOI: 10.3390/cancers17111835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2025] [Revised: 05/26/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
Melanoma has important burden in older populations due to high incidence and aggressive biology. The emergence of immunotherapy with immune checkpoint inhibitors and targeted therapy (BRAF/MEK inhibitors) significantly improved melanoma prognosis. Currently, the body of knowledge on the efficacy and tolerability of these treatments in geriatric patients is primarily based on the results outside of clinical trials since the majority of clinical studies do not include older patients. We present a comprehensive narrative review of published data regarding efficacy and safety of therapeutic modalities using immune checkpoint inhibitors in patients age 65-75 years and >75 years: the anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4) inhibitor (ipilimumab), the anti-programmed death-ligand 1 (PD-1) inhibitors (nivolumab and pembrolizumab), and the lymphocyte activation gene-3 (LAG-3) inhibitor (relatlimab). We carefully address difficulties in multi-disciplinary clinical decision-making in care of older melanoma patients. Although many older patients may not be offered immunotherapy, the available evidence indicates that immunotherapy is equally beneficial in the older patients and does not have higher incidence of adverse events in this group of patients compared to younger population.
Collapse
Affiliation(s)
- Marko Lens
- Ella Lemelbaum Institute for Immuno Oncology, Chaim Sheba Medical Center, Tel Aviv 6997801, Israel
| | | |
Collapse
|
27
|
Kim YS. Gastric Carcinoma. Curr Top Microbiol Immunol 2025. [PMID: 40423781 DOI: 10.1007/82_2025_303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Epstein-Barr virus (EBV)-associated gastric cancers (EBVaGCs) account for about 10% of gastric cancers globally, with higher prevalence in East Asia and Latin America. These cancers develop through a "gastritis-infection-cancer sequence" and are characterized by unique molecular signatures, including CpG island methylator phenotype and mutations in ARID1A and PIK3CA genes. EBVaGCs typically present in the proximal stomach with diffuse-type histology and dense lymphocytic infiltration. Key viral proteins EBNA1 and LMP2A drive oncogenesis by altering cellular processes and immune responses. The IFN-γ signature and extensive epigenetic modifications contribute to their distinct profile. Despite often presenting at advanced stages, EBVaGCs generally have a more favorable prognosis. EBV employs sophisticated strategies to evade immune detection, utilizing latent proteins and noncoding RNAs. Paradoxically, despite an immune-hot environment, EBVaGCs demonstrate effective immune evasion, partly due to the expression of immune checkpoint molecules like PD-L1 and LAG3. Treatment approaches vary based on disease stage, from endoscopic resection for early-stage cancers to systemic therapies for advanced cases. Immunotherapy, particularly PD-1/PD-L1 inhibitors, shows promising results. Emerging research suggests combining these with LAG3 inhibitors may enhance efficacy. Ongoing research and advanced genomic techniques continue to reveal new insights, paving the way for personalized therapies and novel diagnostic approaches.
Collapse
Affiliation(s)
- Young-Sik Kim
- Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
28
|
Zhang Y, Zhou Z, Rui Y, Kong F, Guo Z, Zhao G, Wang J, Li J, Zhao F, Huang H, Fang F, Zhang J, Zhang T, Zhang W, Wang P, Chen X, Zhen P, Pang Q. LAG3 as a marker of immune activation in esophageal squamous carcinoma treated with concurrent chemoradiotherapy. Cancer Immunol Immunother 2025; 74:215. [PMID: 40411616 PMCID: PMC12103390 DOI: 10.1007/s00262-025-04076-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/30/2025] [Indexed: 05/26/2025]
Abstract
INTRODUCTION Esophageal squamous carcinoma (ESCC) is a common malignant tumor of the gastrointestinal tract with high morbidity and mortality rates. Lymphocyte activation gene-3 (LAG3), an important suppressive immune checkpoint in tumor immunity, exhibits a wobbling effect in the prediction of ESCC efficacy. METHODS Tumor bite paraffin-embedded specimens from 84 patients diagnosed with ESCC, all of whom received radical concurrent chemoradiotherapy (CCRT) at our institution, were screened. For each tissue, we delineated the partitions and analyzed the spatial distribution of the tumor in an in situ immune microenvironment. The density and regional characteristics of immune factor-positive cells, together with the dynamics of various cells based on treatment regimens, were considered important factors influencing the prognostic significance of cancer. RESULTS Compared with baseline tissues, the density of CD4 + and CD8 + T cells in the tumor microenvironment of the on-treatment tissues decreased, but the expression of IFN-γ in CD4 + and CD8 + T cells increased. The density of LAG3 positive cells was correlated significantly with the density of CD4 + and CD8 + T cells in both baseline and on-treatment tissues. The density of LAG3 + T cells and the rate of LAG3 positivity in activated CD4 + and CD8 + T cells were associated with elevated Ki67 expression. There was a significant correlation between high LAG3 expression and active CD4 + and CD8 + T cells in tumor cells. Elevated densities and tighter spatial relationships of both CD4 + and CD8 + T cells were associated with longer overall survival with ESCC. CONCLUSION Concurrent chemoradiotherapy without combined immunotherapy inhibited tumor-infiltrating T cells to a certain extent, and elevated immune checkpoint LAG3 was closely associated with immune activation in the ESCC tumor microenvironment.
Collapse
Affiliation(s)
- Yuxuan Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Zijing Zhou
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Yuanyuan Rui
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Fanhao Kong
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Zhoubo Guo
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Gang Zhao
- Department of Pathology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Jun Wang
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Hebei Clinical Research Center for Radiation Oncology, No. 12 Jian Kang Road, Shijiazhuang, 050010, Hebei, China
| | - Jiacheng Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Fangdong Zhao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Hui Huang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Fang Fang
- Department of Radiation Oncology, Chifeng Tumor Hospital, Second Affiliated Hospital of Chifeng University, 45 Jiefang Street, Hongshan District, Chifeng, 024000, Inner Mongolia, China
| | - Jiarui Zhang
- Department of Radiation Oncology, Chifeng Tumor Hospital, Second Affiliated Hospital of Chifeng University, 45 Jiefang Street, Hongshan District, Chifeng, 024000, Inner Mongolia, China
| | - Tian Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Xi Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Peng Zhen
- Department of Radiation Oncology, Chifeng Tumor Hospital, Second Affiliated Hospital of Chifeng University, 45 Jiefang Street, Hongshan District, Chifeng, 024000, Inner Mongolia, China.
| | - Qingsong Pang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, Tianjin, 300060, China.
| |
Collapse
|
29
|
Chehade L, Abbas N, Dagher K, Mourad M, Amhez G, Moumneh MB, Kreidieh L, Kreidieh F, Pereira MM, Shamseddine A. Unmasking the Rare but Lethal Cardiac Complications of Immune Checkpoint Inhibitor Therapy: A Review of Mechanisms, Risk Factors, and Management Strategies. Curr Treat Options Oncol 2025:10.1007/s11864-025-01329-1. [PMID: 40411721 DOI: 10.1007/s11864-025-01329-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2025] [Indexed: 05/26/2025]
Abstract
OPINION STATEMENT Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by enabling the immune system to effectively target and destroy cancer cells. While ICIs offer significant survival benefits across various malignancies, their use is associated with a unique profile of immune-related adverse events, including potentially fatal cardiovascular toxicities. Recent studies have highlighted various cardiac complications associated with ICIs, such as myocarditis, arrhythmias, heart failure, pericarditis, atherosclerosis, and hypertension. These complications arise from mechanisms involving T-cell activation and cytokine release. Patient-related factors such as pre-existing cardiovascular disease, diabetes mellitus, age, gender, and genetic predisposition, along with treatment-related factors like specific ICI regimens, contribute to these toxicities. To manage these complications effectively, comprehensive cardiovascular risk assessment and monitoring before, during, and after ICI therapy are crucial. Adhering to guidelines from the European Society of Cardiology (ESC) and other international organizations allows for early recognition of cardiovascular toxicities and tailored interventions. This review emphasizes the importance of cardioprotective measures, regular monitoring, and multidisciplinary collaboration between oncologists and cardiologists to mitigate cardiovascular risk and optimize patient outcomes. Ongoing research is essential to better understand the mechanisms of ICI-induced cardiovascular toxicities and to develop effective management strategies for affected patients. As we continue to expand the use of ICIs in oncology, balancing oncologic efficacy with cardiovascular safety remains critical.
Collapse
Affiliation(s)
- Laudy Chehade
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Riad El Solh, Beirut, Lebanon
| | - Noura Abbas
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Riad El Solh, Beirut, Lebanon
| | - Kristel Dagher
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Riad El Solh, Beirut, Lebanon
| | - Mohamad Mourad
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Riad El Solh, Beirut, Lebanon
| | - Ghid Amhez
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Riad El Solh, Beirut, Lebanon
| | - Mohamad B Moumneh
- Inova Center of Outcomes Research, Inova Heart and Vascular, Fairfax, VA, USA
| | - Lara Kreidieh
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Riad El Solh, Beirut, Lebanon
| | - Firas Kreidieh
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Riad El Solh, Beirut, Lebanon
| | - Maria Manuel Pereira
- Department of Hematology-Oncology, Unidade Local de Saúde (ULS) de Braga, Braga, Portugal
- School of Medicine, University of Minho, Braga, Portugal
| | - Ali Shamseddine
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Riad El Solh, Beirut, Lebanon.
| |
Collapse
|
30
|
Hanna KS, Palaia J, Patel D, Moshyk A, Zhou ZY, Yang F, Xin Y, Garcia-Horton V. Cost per outcome of nivolumab + relatlimab vs BRAF + MEK inhibitor combinations for first-line treatment of BRAF-mutant advanced melanoma. J Manag Care Spec Pharm 2025:1-9. [PMID: 40391872 DOI: 10.18553/jmcp.2025.25015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
BACKGROUND The National Comprehensive Cancer Network guidelines list combination immunotherapy as the preferred first-line (1L) treatment for unresectable or metastatic melanoma over BRAF and MEK inhibitor (BRAFi/MEKi) therapy, regardless of BRAF mutation status. However, the economic impact of 1L treatment with nivolumab plus relatlimab (NIVO + RELA) vs BRAFi/MEKi therapies for BRAF-mutated advanced melanoma has not been assessed. OBJECTIVE To compare the health care costs, cost per progression-free life-year (PFLY), and cost per life-year (LY) of NIVO + RELA vs dabrafenib plus trametinib (DAB + TRAM), encorafenib plus binimetinib (ENCO + BINI), and vemurafenib plus cobimetinib (VEM + COBI) as 1L treatment for BRAF-mutated, unresectable or metastatic melanoma. METHODS A cost-per-outcome model compared the economic value of NIVO + RELA vs each BRAFi/MEKi therapy. Clinical inputs were derived from previous matching-adjusted indirect comparisons using individual patient data from the BRAF-mutant subgroup of RELATIVITY-047 and published data pooled from COMBI-d, COMBI-v, COLUMBUS, and coBRIM. LYs, PFLYs per investigator, and treatment duration were estimated using the restricted mean survival time. Health care costs (2024 US dollars), including drug acquisition and administration costs, disease management costs over the preprogression and postprogression periods, and adverse event management costs, were calculated over 5 years. Several scenario analyses were performed, including adding subsequent treatment costs. RESULTS Over 5 years, NIVO + RELA was associated with improved PFLYs and LYs compared with DAB + TRAM (mean PFLY: 1.94 vs 1.82 years, mean LY: 3.41 vs 2.77 years), ENCO + BINI (1.87 vs 1.78 years and 3.40 vs 2.91 years, respectively), and VEM + COBI (2.12 vs 1.80 years and 3.39 vs 2.63 years). The estimated total costs over 5 years were lower for NIVO + RELA vs DAB + TRAM ($300,479 vs $519,770), ENCO + BINI ($343,996 vs $572,556), and VEM + COBI ($296,361 vs $317,851). Main cost drivers were drug acquisition and administration costs. NIVO + RELA had lower costs per PFLY and per LY than DAB + TRAM ($155,107 vs $285,617 and $88,203 vs $187,699, respectively); ENCO + BINI ($183,628 vs $322,113 and $101,151 vs $196,924); and VEM + COBI ($139,688 vs $176,645 and $87,315 vs $121,086). The sensitivity analyses' results supported the base-case results. CONCLUSIONS NIVO + RELA showed improved LYs and PFLYs at lower cost than all 3 BRAFi/MEKi comparators over 5 years. These results support the economic value of NIVO + RELA for patients with previously untreated, BRAF-mutated, unresectable or metastatic melanoma.
Collapse
Affiliation(s)
| | | | | | | | | | - Fan Yang
- Analysis Group, Inc., London, United Kingdom
| | - Yiqiao Xin
- Analysis Group, Inc., London, United Kingdom
| | | |
Collapse
|
31
|
Billard K, Mortier L, Dereure O, Dalac S, Montaudié H, Legoupil D, Dutriaux C, De Quatrebarbes J, Maubec E, Leccia MT, Granel-Brocard F, Brunet-Possenti F, Arnault JP, Gaudy-Marqueste C, Pages C, Saiag P, L'Orphelin JM, Zehou O, Lesimple T, Allayous C, Porcher R, Oriano B, Dalle S, Lebbé C. The efficacy and safety of first-line metastatic melanoma treatment with ipilimumab + nivolumab vs. nivolumab in a real-world setting. Br J Dermatol 2025; 192:1096-1105. [PMID: 39605282 DOI: 10.1093/bjd/ljae470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/21/2024] [Accepted: 10/20/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND The Checkmate 067 randomized controlled trial, published in 2015, demonstrated improved progression-free survival (PFS) and numerically, although not statistically, superior overall survival (OS) for ipilimumab + nivolumab (I + N). OBJECTIVES The objective of this study was to compare the efficacy and safety of N with I + N as first-line treatment for metastatic melanoma in a real-world setting. METHODS Patients were prospectively included in the French MelBase cohort from 2013 to 2022. Eligible patients were those in first-line treatment for stage IIIc or IV melanoma, undergoing immunotherapy with N or I + N. The primary endpoint was OS at 36 months. The secondary endpoints included PFS at 36 months, best radiological response, and safety analyses. We conducted a propensity score using the inverse probability of treatment weighting (IPTW) method to overcome the various confounding factors and also a subgroup analysis (brain metastasis, lactate dehydrogenase levels and BRAF mutation status). RESULTS Patients were treated with N (n = 406) or I + N (n = 416). OS at 36 months was higher in the I + N group at 57.1% [95% confidence interval (CI) 50.7-64.2] than in the N group [46.6% (95% CI 41.6-52.1)]; hazard ratio (HR) 1.4 (95% CI 1.1-1.8). PFS at 36 months was significantly improved in the I + N group (42.3%) compared with the N group (21.9%), with a HR of 1.6 (95% CI 1.4-1.9). The objective response rate (ORR) was similar for the two groups (44%). The overall incidence of side-effects was comparable (82% vs. 84%), and severe toxicity (grade ≥ 3) was more frequent, although not significantly so, in the I + N arm vs. the N arm (41% vs. 29%). CONCLUSIONS Our results are consistent with those from the Checkmate 067 study, except for the ORR and the incidence of toxicities, which proved to be lower in our analysis.
Collapse
Affiliation(s)
- Karine Billard
- Service d'onco-dermatologie, Hôpital Saint Louis APHP, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Clara Allayous
- Service d'onco-dermatologie, Hôpital Saint Louis APHP, Paris, France
| | | | - Bastien Oriano
- Service d'onco-dermatologie, Hôpital Saint Louis APHP, Paris, France
| | | | - Céleste Lebbé
- Service d'onco-dermatologie, Hôpital Saint Louis APHP, Paris, France
| |
Collapse
|
32
|
Yang A, Zhou M, Gao Y, Zhang Y. Mechanisms of CD8 + T cell exhaustion and its clinical significance in prognosis of anti-tumor therapies: A review. Int Immunopharmacol 2025; 159:114843. [PMID: 40394796 DOI: 10.1016/j.intimp.2025.114843] [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: 12/09/2024] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025]
Abstract
In recent years, immunotherapy has gradually become one of the main strategies for cancer treatment, with immune checkpoint inhibitors (ICIs) offering new possibilities for tumor therapy. However, some cancer patients exhibit low responses and resistance to ICIs treatment. T cell exhaustion, a process associated with tumor progression, refers to a subset of T cells that progressively lose effector functions and exhibit increased expression of inhibitory receptors. These exhausted T cells are considered key players in the therapeutic efficacy of immune checkpoint inhibitors. Therefore, understanding the impact of T cell exhaustion on tumor immunotherapy and the underlying mechanisms is critical for improving clinical treatment outcomes. Several elegant studies have provided insights into the prognostic value of exhausted T cells in cancers. In this review, we highlight the process of exhausted T cells and its predictive value in various cancers, as well as the relevant mechanisms behind it, providing new insights into the immunotherapy of cancer.
Collapse
Affiliation(s)
- Anrui Yang
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Meng Zhou
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yixuan Gao
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Ying Zhang
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
33
|
Giri VK, McDermott DF, Zaemes J. The emerging role of lymphocyte-activation gene 3 targeting in the treatment of solid malignancies. Cancer 2025; 131:e35892. [PMID: 40344213 DOI: 10.1002/cncr.35892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
PD-(L)1-based immune checkpoint inhibitor therapies have profoundly impacted the treatment of many solid malignancies. Although the addition of CTLA-4 checkpoint inhibitors can enhance anticancer activity, it also significantly increases the rate of immune-related adverse events. Therefore, there has been much interest in identifying additional immune checkpoints to improve the outcomes seen with PD-1-based therapy while minimizing additional side effects. One such target, lymphocyte-activation gene 3 (LAG-3), has long been recognized as an important inhibitor of T-cell function via modulation of the T-cell receptor pathway. Several drugs targeting LAG-3 have been developed, including most prominently the monoclonal antibody relatlimab. To date, the most significant demonstration of efficacy in targeting LAG-3 has been the use of relatlimab with the PD-1 inhibitor nivolumab in the treatment of advanced melanoma. The combination of nivolumab plus relatlimab is more efficacious compared to PD-1 inhibition alone, as has been previously seen with the combination of CTLA-4 inhibitor ipilimumab with nivolumab. However, nivolumab plus relatlimab offers a potentially more favorable toxicity profile. Here, the authors review the mechanism of the LAG-3 pathway and its rationale as a target for anticancer therapy as well as currently available data regarding the use of LAG-3 agents in treating melanoma and other solid tumors. Other investigational agents that target LAG-3 via novel mechanisms are also reviewed.
Collapse
Affiliation(s)
- Vinay K Giri
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - David F McDermott
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jacob Zaemes
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Hosseini MS, Jadidi-Niaragh F. An updated systematic review and meta-analysis on the efficacy and safety of nivolumab/relatlimab combination therapy in melanoma patients. Arch Dermatol Res 2025; 317:755. [PMID: 40358773 DOI: 10.1007/s00403-025-04270-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/21/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Affiliation(s)
- Mohammad-Salar Hosseini
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 51666, EA, Iran.
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Evidence-Based Medicine, Iranian EBM Center: A JBI Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran.
- Iranian Cancer Control Center (MACSA) - Tabriz Branch, Tabriz, Iran.
| | - Farhad Jadidi-Niaragh
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 51666, EA, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
35
|
Alchaikh Hassan R, Vu A, Tsai H, Dasanu CA. Complete color vision loss in a patient with metastatic melanoma of the skin treated with nivolumab-relatlimab. J Oncol Pharm Pract 2025:10781552251340014. [PMID: 40356467 DOI: 10.1177/10781552251340014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
IntroductionThe combination nivolumab-relatlimab has demonstrated therapeutic benefits for patients with metastatic melanoma. However, its adverse effects can affect various organs, including the eye.Case reportWe present the case of an 80-year-old man who developed complete color blindness after the 2nd dose of nivolumab-relatlimab for advanced cutaneous melanoma. He also had significant blurry vision. Fundoscopy identified bilateral serous macular detachment. Ocular coherence tomography confirmed serous infiltration within the macula. After a thorough investigation, causality assessment linked this retinal effect to nivolumab-relatlimab (probable relationship via Naranjo criteria).Management and outcomeThe patient was treated with corticosteroids, intravenous immunoglobulin (IVIG), rituximab, and plasmapheresis, with improvement in blurry vision. However, he continued to experience persistent absence of color perception ("black and white vision") at a follow-up visit six months later.Discussion/conclusionFurther studies are necessary to understand the exact pathophysiology of this process. We hypothesize that it involves direct toxicity to the photoreceptors or retinal ganglion cells, leading to irreversible color vision loss. Effective strategies for preventing this significant, life-changing toxic effect of nivolumab-relatlimab should be sought.
Collapse
Affiliation(s)
| | - Anderson Vu
- Department of Internal Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Henry Tsai
- Lucy Curci Cancer Center, Eisenhower Health, Rancho Mirage, CA, USA
- Department of Medical Oncology and Hematology, University of California in San Diego Health System, San Diego, CA, USA
| | - Constantin A Dasanu
- Lucy Curci Cancer Center, Eisenhower Health, Rancho Mirage, CA, USA
- Department of Medical Oncology and Hematology, University of California in San Diego Health System, San Diego, CA, USA
| |
Collapse
|
36
|
Quandt Z, Lucas A, Liang SI, Yang E, Stone S, Fadlullah MZH, Bayless NL, Marr SS, Thompson MA, Padron LJ, Bucktrout S, Butterfield LH, Tan AC, Herold KC, Bluestone JA, Anderson MS, Spencer CN, Young A, Connolly JE. Associations between immune checkpoint inhibitor response, immune-related adverse events, and steroid use in RADIOHEAD: a prospective pan-tumor cohort study. J Immunother Cancer 2025; 13:e011545. [PMID: 40355283 PMCID: PMC12083316 DOI: 10.1136/jitc-2025-011545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/17/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have led to enduring responses in subsets of patients with cancer. However, these responses carry the risk of immune-related adverse events (irAEs), which can diminish the overall benefit of ICI treatment. While associations between irAE development and overall survival have been increasingly documented, there is a need for further understanding of these connections in large prospective real-world cohorts. METHODS The Resistance Drivers for Immuno-Oncology Patients Interrogated by Harmonized Molecular Datasets (RADIOHEAD) study, a pan-tumor, prospective cohort of 1,070 individuals undergoing standard of care first-line ICI treatment, aims to identify factors driving irAEs and clinical response. Clinical data and longitudinal blood samples were collected prospectively at multiple time points from 49 community-based oncology clinics across the USA. Structured, harmonized clinical data underwent unbiased statistical analysis to uncover predictors of real-world overall survival (rwOS) and risk factors for irAEs. RESULTS Across 1,070 participants' treatment courses, RADIOHEAD accumulated over 4,500 clinical data points. Patients experiencing any irAE (25.4%, n=272) exhibited significantly improved rwOS in the pan-tumor cohort (n=1,028, HR=0.41, 95% CI=(0.31, 0.55)). This association persisted when adjusting for age and metastatic disease in multivariate time-dependent Cox proportional hazard analysis, and was consistent across major tumor subtypes, including lung cancer and melanoma. Skin and endocrine irAEs of any grade were strongly associated with improved rwOS (Cox proportional hazard analysis, skin, p=2.03e-05; endocrine, p=0.0006). In this real-world cohort, the irAE rate appeared lower than those reported in clinical trials. Patients receiving corticosteroids prior to initiation of ICI treatment had significantly worse survival outcomes than non-users (HR 1.37, p=0.0054), with a stronger association with systemic steroid use (HR 1.75, p=0.0022). The risk of irAE was increased by exposure to combination immunotherapy relative to monotherapy (OR 4.17, p=2.8e-7), zoster vaccine (OR 2.4, p=5.2e-05), and decreased by prior chemotherapy (OR 1.69, p=0.0005). CONCLUSION The RADIOHEAD cohort is a well-powered, real-world cohort that clearly demonstrates the association between irAE development with improved response and baseline steroid use with worse response to ICI treatment after adjustment for survival bias.
Collapse
Affiliation(s)
- Zoe Quandt
- Department of Medicine, Division of Endocrinology and Metabolism, UCSF, San Francisco, California, USA
- Diabetes Center, UCSF, San Francisco, California, USA
| | - Anastasia Lucas
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Samantha I Liang
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - EnJun Yang
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Samantha Stone
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Muhammad Zaki Hidayatullah Fadlullah
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Departments of Oncological Sciences and Biomedical Informatics, University of Utah, Salt Lake City, Utah, USA
| | - Nicholas L Bayless
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Sara Siebel Marr
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | | | - Lacey J Padron
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Samantha Bucktrout
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Lisa H Butterfield
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Microbiology and Immunology, University of California, San Francisco, California, USA
| | - Aik Choon Tan
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Departments of Oncological Sciences and Biomedical Informatics, University of Utah, Salt Lake City, Utah, USA
| | - Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, Connecticut, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Yale University, New Haven, Connecticut, USA
| | | | - Mark S Anderson
- Department of Medicine, Division of Endocrinology and Metabolism, UCSF, San Francisco, California, USA
- Diabetes Center, UCSF, San Francisco, California, USA
| | | | - Arabella Young
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - John E Connolly
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| |
Collapse
|
37
|
Pedroso CM, de Pauli Paglioni M, Normando AGC, Chaves ALF, Kowalski LP, de Castro Júnior G, Matos LL, Willian Junior WN, de Oliveira TB, de Marchi P, Harada G, Mak MP, Lima CSP, Viani GA, Moraes FY, Gouveia AG, Santos-Silva AR, Marta GN, Latin American Cooperative Oncology Group (LACOG) - Head and Neck and Brazilian Group of Head and Neck Cancer (GBCP). Preoperative neoadjuvant chemotherapy or immunotherapy in head and neck cancer: A systematic review and meta-analysis of surgical risk and pathologic response. Crit Rev Oncol Hematol 2025; 212:104742. [PMID: 40348212 DOI: 10.1016/j.critrevonc.2025.104742] [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: 03/14/2025] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 05/14/2025] Open
Abstract
OBJECTIVE This systematic review and meta-analysis aimed to evaluate surgical complications, pathologic responses, and disease progression in patients with head and neck squamous cell carcinoma treated with neoadjuvant chemotherapy or immunotherapy. METHODS A comprehensive literature search was conduct across four databases (PubMed, Embase, Cochrane Library, and Scopus) and grey literature sources to identify neoadjuvant therapies in head and neck cancer patients. Only prospective clinical trials were included. The certainty of evidence was appraised using GRADE tool. RESULTS A total of 12 clinical trials me the inclusion criteria, comprising, six studies on neoadjuvant chemotherapy (Cisplatin and 5-FU) and six on immunotherapy (Nivolumab, Nivolumab plus Ipilimumab, Pembrolizumab) were analyzed. The mean time from drug administration to surgery ranged from 18 to 29 days. The overall surgical complication rate was 32.8 %, with the lowest observed in the Pembrolizumab group (9 %) and the highest in the Nivolumab plus Ipilimumab group (36.7 %). However, risk ratios for surgical complications were not statistically significant for Nivolumab (RR = 1.68, p = 0.078) or chemotherapy (RR = 1.1, p = 0.70). The complete pathologic response (pCR) rate was low (4 %), highest in the Cisplatin and 5-FU group (11 %). In contrast, the partial pathologic response (pPR) rate reached 58 % with Nivolumab plus Ipilimumab. Disease progression after surgery occurred in 19.4 %, with the lowest progression rate observed in the Nivolumab plus Ipilimumab group (7.7 %). The certainty of evidence was rated as very low for chemotherapy and low for immunotherapy. CONCLUSION Combination immunotherapy, particularly Nivolumab with Ipilimumab, demonstrated favorable pPR rates and reduced disease progression but was increased surgical complications. The overall low pCR across all regimen treatments highlight the need for improved therapeutic strategies.
Collapse
Affiliation(s)
- Caique Mariano Pedroso
- Oral Diagnosis Department, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil.
| | - Mariana de Pauli Paglioni
- Oral Diagnosis Department, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Ana Gabriela Costa Normando
- Oral Diagnosis Department, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Aline Lauda Freitas Chaves
- DOM Clinica de Oncologia, Divinopolis, MG, Brazil; Grupo Oncoclínicas, São Paulo, Brazil; Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil
| | - Luiz Paulo Kowalski
- Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil; Head and Neck Surgery Department, University of São Paulo Medical School (FMUSP), São Paulo, São Paulo, Brazil; Department of Head and Neck Surgery and Otorhinolaryngology, A.C. Camargo Cancer Center, São Paulo, São Paulo, Brazil
| | - Gilberto de Castro Júnior
- Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil; Medical Oncology, Instituto do Câncer do Estado de São Paulo (ICESP) - Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Leandro Luongo Matos
- Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil; Department of Head and Neck Surgery, Instituto do Cancer do Estado de São Paulo (ICESP), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - William Nassib Willian Junior
- Grupo Oncoclínicas, São Paulo, Brazil; Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil
| | - Thiago Bueno de Oliveira
- Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil; Medical Oncology Department, AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - Pedro de Marchi
- Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil; Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil; Oncoclinicas, Rio De Janeiro, Brazil, Barretos Cancer Hospital, Barretos, Brazil
| | - Guilherme Harada
- Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil; Department of Medical Oncology; Hospital Sírio-Libanês, São Paulo, Brazil
| | - Milena Perez Mak
- Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil
| | - Carmen Silvia Passos Lima
- Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil; Department of Radiology and Oncology, School of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Gustavo Arruda Viani
- Department of Medical Imagings, Hematology and Oncology, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Fabio Ynoe Moraes
- Division of Radiation Oncology, Department of Oncology, Kingston General Hospital, Queen's University, Ontario, Kingston, Canada
| | - Andre Guimaraes Gouveia
- Department of Oncology - Division of Radiation Oncology, Juravinski Cancer Centre, McMaster University, Hamilton, ON, Canada
| | - Alan Roger Santos-Silva
- Oral Diagnosis Department, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil
| | - Gustavo Nader Marta
- Latin American Cooperative Oncology Group, Brazilian Group of Head and Neck Cancer, Brazil; Department of Radiation Oncology; Hospital Sírio-Libanês, São Paulo, Brazil; Post-Graduation Program, Radiology and Oncology Department, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | | |
Collapse
|
38
|
Sánchez-Camacho A, Torres-Zurita A, Gallego-López L, Hernández-Pacheco R, Silva-Romeiro S, Álamo de la Gala MDC, Peral-Gutiérrez de Ceballos E, de la Cruz-Merino L. Management of immune-related myocarditis, myositis and myasthenia gravis (MMM) overlap syndrome: a single institution case series and literature review. Front Immunol 2025; 16:1597259. [PMID: 40406130 PMCID: PMC12095175 DOI: 10.3389/fimmu.2025.1597259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/17/2025] [Indexed: 05/26/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of various malignancies, particularly melanoma. However, immune-related adverse events (irAEs) pose significant challenges, particularly in cases of severe toxicity syndromes. One such life-threatening irAE is the myocarditis, myositis, and myasthenia gravis (MMM) overlap syndrome, which occurs in less than 1% of patients but has in-hospital mortality rates ranging from 40 to 60%. Due to its rarity and complexity, early recognition and a multidisciplinary approach are critical to improving outcomes. Methods We present a single-institution case series of four patients diagnosed with MMM overlap syndrome following ICI therapy. Clinical presentation, laboratory findings, imaging, and electrophysiological tests were analyzed to confirm the diagnosis. Therapeutic interventions-including corticosteroids, intravenous immunoglobulins (IVIG), plasma exchange (PLEX), tocilizumab, and rituximab- were evaluated in terms of efficacy and clinical outcomes. Results The onset of MMM syndrome varied from 2 to 4 weeks after initiating ICI therapy. Patients presented with rapidly progressive symptoms, including ptosis, bulbar dysfunction, respiratory distress, myopathy, and cardiac conduction abnormalities. Immunosuppressive therapy with high-dose corticosteroids was initiated in all cases. Additional immunomodulatory treatments (IVIG, tocilizumab, PLEX, and rituximab) were administered based on clinical deterioration and autoimmune profile. Two patients achieved complete recovery, one remains on maintenance immunosuppression, and one died due to respiratory failure despite aggressive treatment. Conclusion MMM overlap syndrome is a severe and often fatal irAE associated with ICI therapy. Early identification, aggressive immunosuppressive treatment, and individualized therapeutic strategies are essential to optimize patient outcomes. Further research is needed to refine diagnostic criteria, identify predictive biomarkers, and establish standardized treatment protocols.
Collapse
Affiliation(s)
| | - Alberto Torres-Zurita
- Department of Medical Oncology, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Laura Gallego-López
- Autoimmune Disease Unit, Department of Internal Medicine, Hospital Universitario Virgen Macarena, Seville, Spain
| | | | - Silvia Silva-Romeiro
- Department of Medical Oncology, Hospital Universitario Virgen Macarena, Seville, Spain
- Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen Macarena, CSIC, University of Seville, Seville, Spain
| | | | | | - Luis de la Cruz-Merino
- Department of Medical Oncology, Hospital Universitario Virgen Macarena, Seville, Spain
- Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen Macarena, CSIC, University of Seville, Seville, Spain
- Department of Medicine, University of Seville, Seville, Spain
| |
Collapse
|
39
|
Jeon WJ, Son SY, Abodunrin F, Khanna A, Patel JD, Thawani R. Dosing immune-checkpoint inhibitors: Opportunities for the future. Curr Probl Cancer 2025; 57:101204. [PMID: 40339394 DOI: 10.1016/j.currproblcancer.2025.101204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025]
Abstract
With recent breakthroughs in immunotherapy, particularly with the approval of immune checkpoint inhibitors for various cancer indications, patients now have a wider array of treatment options, even for those with metastatic disease. Still, the survival benefit of immune-checkpoint inhibitors is modest, and there is concern about drug toxicity. In addition, there is ongoing exploration into combination therapy involving immune-checkpoint inhibitors, which come at the risk of increased toxicity. Unfortunately, due to the cost of the currently approved doses and dosing intervals, many patients in the community in the United States and low- and middle-income countries lack access to these transformative therapies. Further, the observation of resistance to immune-checkpoint inhibitors and limitations of currently approved doses and dosing intervals warrants changes in current practice. This review paper discusses both model-based and clinical studies in the current literature. Strategies for improving access to immune-checkpoint inhibitors and expanding their utilization, including weight-based dosing instead of fixed dosing, dose and dose interval adjustments, development of biomarkers and scoring systems for personalization of immune-checkpoint inhibitors, and alternative trial design, are discussed.
Collapse
Affiliation(s)
- Won Jin Jeon
- Division of Hematology & Oncology, Loma Linda University, Loma Linda, CA, USA
| | - So Young Son
- Loma Linda University School of Pharmacy, Loma Linda University, Loma Linda, CA, USA
| | - Faith Abodunrin
- Biological Sciences Division, University of Chicago, Chicago, IL, USA
| | | | | | - Rajat Thawani
- Division of Hematology & Oncology, Knight Cancer Institute Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
40
|
Tawbi HA, Hodi FS, Lipson EJ, Schadendorf D, Ascierto PA, Matamala L, Castillo Gutiérrez E, Rutkowski P, Gogas H, Lao CD, Janoski De Menezes J, Dalle S, Arance AM, Grob JJ, Ratto B, Rodriguez S, Mazzei A, Dolfi S, Long GV. Three-Year Overall Survival With Nivolumab Plus Relatlimab in Advanced Melanoma From RELATIVITY-047. J Clin Oncol 2025; 43:1546-1552. [PMID: 39671533 PMCID: PMC12054981 DOI: 10.1200/jco.24.01124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/23/2024] [Accepted: 10/14/2024] [Indexed: 12/15/2024] Open
Abstract
Nivolumab plus relatlimab demonstrated a statistically significant improvement in progression-free survival (PFS), along with a clinically meaningful, but not statistically significant improvement in overall survival (OS) and a numerically higher objective response rate (ORR) compared with nivolumab in the RELATIVITY-047 trial (ClinicalTrials.gov identifier: NCT03470922). We report updated descriptive efficacy and safety results from RELATIVITY-047 with a median follow-up of 33.8 months. Median PFS was 10.2 months (95% CI, 6.5 to 15.4) with nivolumab plus relatlimab and 4.6 months (95% CI, 3.5 to 6.5) with nivolumab (hazard ratio [HR], 0.79 [95% CI, 0.66 to 0.95]); median OS was 51.0 months (95% CI, 34.0 to not reached) and 34.1 (95% CI, 25.2 to 44.7) months, respectively (HR, 0.80 [95% CI, 0.66 to 0.99]). ORR was 43.7% (95% CI, 38.4 to 49.0) with nivolumab plus relatlimab and 33.7% (95% CI, 28.8 to 38.9) with nivolumab. Efficacy across the majority of prespecified subgroups favored the combination. No new or unexpected safety signals were identified. Overall, at 3-year follow-up, the benefit observed with nivolumab plus relatlimab compared with nivolumab in patients with advanced melanoma was sustained, with the OS HR 95% CI upper bound now <1. This benefit is accompanied by a safety profile consistent with previous reports.
Collapse
Affiliation(s)
| | | | - Evan J. Lipson
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Dirk Schadendorf
- University of Essen and the German Cancer Consortium, Essen, Germany
| | - Paolo A. Ascierto
- Istituto Nazionale dei Tumori IRCCS “Fondazione G. Pascale,” Naples, Italy
| | - Luis Matamala
- Instituto Oncológico Fundación Arturo López Pérez and Department of Oncology, Instituto Nacional dfoel Cáncer, Santiago, Chile
| | | | - Piotr Rutkowski
- Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Helen Gogas
- National and Kapodistrian University of Athens, Athens, Greece
| | - Christopher D. Lao
- Michigan Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | | | - Stéphane Dalle
- Hospices Civils de Lyon, Cancer Research Center of Lyon, Pierre-Bénite, France
| | | | | | | | | | | | | | - Georgina V. Long
- Melanoma Institute Australia, The University of Sydney, Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| |
Collapse
|
41
|
Calvo-Barreiro L, Secor M, Damjanovic J, Abdel-Rahman SA, Lin YS, Gabr M. Computational Design of a Bicyclic Peptide Inhibitor Targeting the ICOS/ICOS-L Protein-Protein Interaction. Chem Biol Drug Des 2025; 105:e70117. [PMID: 40317592 PMCID: PMC12121478 DOI: 10.1111/cbdd.70117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/19/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025]
Abstract
The interaction between the inducible T-cell costimulatory molecule (ICOS) and its ligand (ICOS-L) is a critical pathway in T-cell activation and immune regulation. We computationally designed a bicyclic peptide (CP5) that inhibits the ICOS/ICOS-L protein-protein interaction (PPI). Using the structural insights derived from the ICOS/ICOS-L co-crystal structure (PDB: 6X4G) and bias-exchange metadynamics simulations (BE-META), we first designed monocyclic peptide candidates containing the β-strand (residues 51-55 51YVYWQ55) of ICOS-L that interact with ICOS. Using Rosetta's flex ddG calculations and further disulfide-bond restraint, we arrived at CP5 (cyclo-RVY[CQPGWC]WVLpG) as a potential ICOS/ICOS-L inhibitor. Using dynamic light scattering (DLS), we examined the interaction between CP5 and ICOS. Importantly, we validated the ICOS/ICOS-L inhibitory activity of CP5 using both TR-FRET assay and ELISA. Notably, CP5 demonstrated satisfactory in vitro pharmacokinetic properties, such as metabolic stability and lipophilicity, positioning it as a promising candidate for further drug development. Our findings provide a foundation for future drug discovery efforts aiming to develop cyclic peptides that specifically target the ICOS/ICOS-L interaction.
Collapse
Affiliation(s)
- Laura Calvo-Barreiro
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, New York, USA
| | - Maxim Secor
- Department of Chemistry, Tufts University, Medford, Massachusetts, USA
| | - Jovan Damjanovic
- Department of Chemistry, Tufts University, Medford, Massachusetts, USA
| | - Somaya A Abdel-Rahman
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, New York, USA
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Yu-Shan Lin
- Department of Chemistry, Tufts University, Medford, Massachusetts, USA
| | - Moustafa Gabr
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
42
|
Zhao Y, Wucherpfennig KW. Unlocking LAG3: Ubiquitin's unexpected role. Cell 2025; 188:2307-2309. [PMID: 40315815 DOI: 10.1016/j.cell.2025.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 05/04/2025]
Abstract
The inhibitory receptor LAG3 is the target of the FDA-approved mAb relatlimab, but its mechanism of signaling is not well understood. In this issue of Cell, Jiang et al. demonstrate that ubiquitination of its cytoplasmic domain is essential for the inhibitory function of LAG3. Co-expression of LAG3 and the CBL E3 ligases represents a biomarker of clinical response to LAG3 inhibition in human melanoma.
Collapse
Affiliation(s)
- Ye Zhao
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Department of Neurology, Brigham & Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
43
|
Jiang Y, Dai A, Huang Y, Li H, Cui J, Yang H, Si L, Jiao T, Ren Z, Zhang Z, Mou S, Zhu H, Guo W, Huang Q, Li Y, Xue M, Jiang J, Wang F, Li L, Zhong Q, Wang K, Liu B, Wang J, Fan G, Guo J, Chen L, Workman CJ, Shen Z, Kong Y, Vignali DAA, Xu C, Wang H. Ligand-induced ubiquitination unleashes LAG3 immune checkpoint function by hindering membrane sequestration of signaling motifs. Cell 2025; 188:2354-2371.e18. [PMID: 40101708 DOI: 10.1016/j.cell.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 08/16/2024] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
Lymphocyte activation gene 3 (LAG3) has emerged as a promising cancer immunotherapy target, but the mechanism underlying LAG3 activation upon ligand engagement remains elusive. Here, LAG3 was found to undergo robust non-K48-linked polyubiquitination upon ligand engagement, which promotes LAG3's inhibitory function instead of causing degradation. This ubiquitination could be triggered by the engagement of major histocompatibility complex class II (MHC class II) and membrane-bound (but not soluble) fibrinogen-like protein 1 (FGL1). LAG3 ubiquitination, mediated redundantly by the E3 ligases c-Cbl and Cbl-b, disrupted the membrane binding of the juxtamembrane basic residue-rich sequence, thereby stabilizing the LAG3 cytoplasmic tail in a membrane-dissociated conformation enabling signaling. Furthermore, LAG3 ubiquitination is crucial for the LAG3-mediated suppression of antitumor immunity in vivo. Consistently, LAG3 therapeutic antibodies repress LAG3 ubiquitination, correlating with their checkpoint blockade effects. Moreover, patient cohort analyses suggest that LAG3/CBL coexpression could serve as a biomarker for response to LAG3 blockade. Collectively, our study reveals an immune-checkpoint-triggering mechanism with translational potential in cancer immunotherapy.
Collapse
Affiliation(s)
- Yong Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Anran Dai
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuwei Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Lingang Laboratory, Shanghai 200031, China
| | - Hua Li
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jian Cui
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Haochen Yang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Tao Jiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Zhengxu Ren
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Si Mou
- BeiGene, Ltd, Beijing 102206, China
| | | | - Wenhui Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Qiang Huang
- School of Medicine, Shanghai University, Shanghai 200444, China; State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an 710032, China
| | - Yilin Li
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Manman Xue
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jingwei Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Fei Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Li Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qinying Zhong
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kun Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Baichuan Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Lingang Laboratory, Shanghai 200031, China
| | - Jinjiao Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Liang Chen
- School of Medicine, Shanghai University, Shanghai 200444, China; State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an 710032, China
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | | | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China.
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Chenqi Xu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Shanghai Academy of Natural Sciences (SANS), Shanghai 200031, China.
| | - Haopeng Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Academy of Natural Sciences (SANS), Shanghai 200031, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
44
|
Qureshi Z, Zaheer Z, Asghar Z, Bakhtiar M, Fatima E, Altaf F. Cardiovascular Risk Profile of Nivolumab Anti-cancer Therapy: A Review of Current Literature. Am J Clin Oncol 2025; 48:235-241. [PMID: 40008416 DOI: 10.1097/coc.0000000000001166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
OBJECTIVES Immune checkpoint inhibitors (ICI) upregulate host antitumor immunity, proving efficacy across diverse tumor types. Currently approved ICI treatment primarily targets the programmed cell death receptor 1 (PD-1) and its ligand PD-L1, and cytotoxic T lymphocyte-antigen 4 (CTLA-4). Nivolumab is a monoclonal antibody that targets the human PD-1 receptor and is an entirely human immunoglobulin G4 (IgG4), approved by the FDA for various cancers like advanced melanoma, metastatic renal cell carcinoma, Hodgkin lymphoma, and advanced lung carcinoma. This review will summarise and discuss the recent literature on cardiotoxicity associated with nivolumab therapy. METHODS We searched online databases like PubMed, Scopus, Google Scholar, and Embase for articles related to Nivolumab. RESULTS Cardiotoxicity with ICI use is most commonly represented as myocarditis. Patients present with complaints of shortness of breath, palpitations, edema, and fatigue. Takotsubo cardiomyopathy, or broken heart syndrome, is characterized by systolic dysfunction of the left ventricle, mimicking a myocardial infarction but without associated coronary ischemia and with minimal elevation of cardiac enzymes. In the CHECKMATE-037 trial, ventricular arrhythmias occurred in <10% of those who received nivolumab. In a retrospective analysis of patients treated with ICI (predominantly nivolumab monotherapy) for lung cancer, 11% of the patients developed major adverse cardiac events, including myocarditis, non-ST-segment elevated myocardial infarction, supraventricular tachycardia, and pericardial disorders. CONCLUSION Close collaboration between cardiology and oncology specialists is crucial for early detection and effective management of cardiac complications, enhancing the safety of nivolumab anticancer therapy.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | | | - Zoha Asghar
- Department of Medicine, Ziauddin University, Karachi
| | | | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| |
Collapse
|
45
|
Wang J, Chen Q, Shan Q, Liang T, Forde P, Zheng L. Clinical development of immuno-oncology therapeutics. Cancer Lett 2025; 617:217616. [PMID: 40054657 PMCID: PMC11930610 DOI: 10.1016/j.canlet.2025.217616] [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: 11/05/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Immuno-oncology (IO) is one of the fastest growing therapeutic areas within oncology. IO agents work indirectly via the host's adaptive and innate immune system to recognize and eradicate tumor cells. Despite checkpoint inhibitors being only introduced to the market since 2011, they have become the second most approved product category. Current Food and Drug Administration (FDA)-approved classes of IO agents include: immune checkpoint inhibitors (ICIs), chimeric antigen receptor T-cell therapy (CAR-T), bi-specific T-cell engager (BiTE) antibody therapy, T-cell receptor (TCR) engineered T cell therapy, tumor-infiltrating lymphocyte (TIL) therapy, cytokine therapy, cancer vaccine therapy, and oncolytic virus therapy. Cancer immunotherapy has made progress in multiple cancer types including melanoma, non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), and urothelial carcinoma; however, several cancers remain refractory to immunotherapy. Future directions of IO include exploration in the neoadjuvant/perioperative setting, combination strategies, and optimizing patient selection through improved biomarkers.
Collapse
Affiliation(s)
- Jianxin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qi Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qiang Shan
- Department of General Surgery, Haining People's Hospital, Haining, 314400, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Patrick Forde
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
46
|
Giudice GC, Beckermann KE, Siqueira Do Amaral P, Rini BI. Immunotherapy Strategies After Immune Checkpoint Inhibitor Exposure in Renal Cell Carcinoma: A Review. JAMA Oncol 2025; 11:554-561. [PMID: 40146173 DOI: 10.1001/jamaoncol.2025.0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Importance Immune checkpoint inhibitors have transformed the treatment landscape for metastatic renal cell carcinoma; however, the failure of first-line therapeutic strategies remains a considerable challenge. Currently, clinicians face various issues, such as managing cases in patients who progress during treatment or relapse after adjuvant immunotherapy. Observations This review evaluates different strategies for treating patients with advanced kidney cancer previously exposed to immunotherapy. Evidence from other malignant neoplasms suggests potential effectiveness for rechallenging with immune checkpoint inhibitors. The most important available data are presented, including retrospective, prospective, and randomized clinical trials, to explore the role of immunotherapy in patients with renal cell carcinoma who have experienced prior failure of immune checkpoint inhibitors. Conclusions and Relevance Although retrospective data suggest modest effectiveness of an immunotherapy rechallenge treatment, larger phase 3 trials failed to demonstrate substantial benefit in progression-free survival and overall survival. Currently, no randomized evidence supports the use of agents targeting conventional immune checkpoints in patients with renal cell carcinoma who have previously received immunotherapy.
Collapse
Affiliation(s)
- Giulia Claire Giudice
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Kathryn E Beckermann
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Paulo Siqueira Do Amaral
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Brian I Rini
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| |
Collapse
|
47
|
Long GV, Shklovskaya E, Satgunaseelan L, Mao Y, da Silva IP, Perry KA, Diefenbach RJ, Gide TN, Shivalingam B, Buckland ME, Gonzalez M, Caixeiro N, Vergara IA, Bai X, Rawson RV, Hsiao E, Palendira U, Phan TG, Menzies AM, Carlino MS, Quek C, Grimmond SM, Vissers JHA, Yeo D, Rasko JEJ, Khasraw M, Neyns B, Reardon DA, Ashley DM, Wheeler H, Back M, Scolyer RA, Drummond J, Wilmott JS, Rizos H. Neoadjuvant triplet immune checkpoint blockade in newly diagnosed glioblastoma. Nat Med 2025; 31:1557-1566. [PMID: 40016450 DOI: 10.1038/s41591-025-03512-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/14/2025] [Indexed: 03/01/2025]
Abstract
Glioblastoma (GBM) is an aggressive primary adult brain tumor that rapidly recurs after standard-of-care treatments, including surgery, chemotherapy and radiotherapy. While immune checkpoint inhibitor therapies have transformed outcomes in many tumor types, particularly when used neoadjuvantly or as a first-line treatment, including in melanoma brain metastases, they have shown limited efficacy in patients with resected or recurrent GBM. The lack of efficacy has been attributed to the scarcity of tumor-infiltrating lymphocytes (TILs), an immunosuppressive tumor microenvironment and low tumor mutation burden typical of GBM tumors, plus exclusion of large molecules from the brain parenchyma. We hypothesized that upfront neoadjuvant combination immunotherapy, administered with disease in situ, could induce a stronger immune response than treatment given after resection or after recurrence. Here, we present a case of newly diagnosed IDH-wild-type, MGMT promoter unmethylated GBM, treated with a single dose of neoadjuvant triplet immunotherapy (anti-programmed cell death protein 1 plus anti-cytotoxic T-lymphocyte protein 4 plus anti-lymphocyte-activation gene 3) followed by maximal safe resection 12 days later. The anti-programmed cell death protein 1 drug was bound to TILs in the resected GBM and there was marked TIL infiltration and activation compared with the baseline biopsy. After 17 months, there is no definitive sign of recurrence. If used first line, before safe maximal resection, checkpoint inhibitors are capable of immune activation in GBM and may induce a response. A clinical trial of first-line neoadjuvant combination checkpoint inhibitor therapy in newly diagnosed GBM is planned (GIANT; trial registration no. NCT06816927 ).
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia.
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia.
- Mater Hospital, North Sydney, New South Wales, Australia.
- Royal North Shore Hospital, St Leonards, New South Wales, Australia.
| | - Elena Shklovskaya
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Macquarie University, Macquarie Park, New South Wales, Australia
| | - Laveniya Satgunaseelan
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Yizhe Mao
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Inês Pires da Silva
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Kristen A Perry
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Russell J Diefenbach
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Macquarie University, Macquarie Park, New South Wales, Australia
| | - Tuba N Gide
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Brindha Shivalingam
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | - Michael E Buckland
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Maria Gonzalez
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
| | - Nicole Caixeiro
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Ismael A Vergara
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Xinyu Bai
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Robert V Rawson
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- NSW Health Pathology, Sydney, New South Wales, Australia
| | - Edward Hsiao
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Umaimainthan Palendira
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Tri Giang Phan
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, North Sydney, New South Wales, Australia
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Blacktown Hospital, Blacktown, New South Wales, Australia
- Westmead Hospital, Westmead, New South Wales, Australia
| | - Camelia Quek
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Sean M Grimmond
- Collaborative Centre for Genomic Cancer Medicine, University of Melbourne, Melbourne, Victoria, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Joseph H A Vissers
- Collaborative Centre for Genomic Cancer Medicine, University of Melbourne, Melbourne, Victoria, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Dannel Yeo
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Centenary Institute, Camperdown, New South Wales, Australia
| | - John E J Rasko
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Centenary Institute, Camperdown, New South Wales, Australia
| | | | - Bart Neyns
- Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Helen Wheeler
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Michael Back
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- NSW Health Pathology, Sydney, New South Wales, Australia
| | - James Drummond
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
- North Shore Radiology & Nuclear Medicine, St Leonards, New South Wales, Australia
- Brain Imaging Laboratory, The Brain Cancer Group, St Leonards, New South Wales, Australia
| | - James S Wilmott
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Helen Rizos
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Macquarie University, Macquarie Park, New South Wales, Australia
| |
Collapse
|
48
|
Maul LV, Ramelyte E, Dummer R, Mangana J. Management of metastatic melanoma with combinations including PD-1 inhibitors. Expert Opin Biol Ther 2025; 25:1-12. [PMID: 40159098 DOI: 10.1080/14712598.2025.2485315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Melanoma is among the most immunogenic malignancies. The advent of immune checkpoint inhibitors (ICIs) has revolutionized the landscape of melanoma treatment. Long-term durable cancer control is possible in nearly 50% of non-resectable, metastatic melanoma patients with anti-CTLA4 and anti-PD-1 antibodies. AREAS COVERED This review provides a critical overview of the current data and future research directions on the management of metastatic melanoma with ICIs. We reviewed the efficacy and safety of combinations with PD-1 inhibitors through PubMed database research (Nov 2024-Mar 2025). EXPERT OPINION A decade after ipilimumab's approval, challenges remain. To cure more patients, the development of combinations is warranted. Combinations with a limited number of ipilimumab applications improve the overall survival outcome by approximately 10%, with a dramatic increase in adverse events including fatal events. Anti-LAG3/nivolumab is a promising alternative, offering similar efficacy to ipilimumab/nivolumab with better tolerability. In our opinion, ipilimumab/nivolumab combination should be the first-line therapy for high-risk patients (high LDH, brain or liver metastasis), while nivolumab/relatlimab or PD-1 monotherapy may be preferable for lower-risk cases. However, treatment decisions are increasingly complex, since most patients nowadays are pretreated in the (neo)-adjuvant setting. The key limitation today is the lack of biomarkers to guide individualized treatment strategies.
Collapse
Affiliation(s)
- Lara Valeska Maul
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
49
|
Yochum ZA, Braun DA. Immunotherapy for Renal Cell Carcinoma-What More is to Come? Target Oncol 2025; 20:467-483. [PMID: 40208564 DOI: 10.1007/s11523-025-01143-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2025] [Indexed: 04/11/2025]
Abstract
The treatment of renal cell carcinoma (RCC), a malignancy that is typically chemoresistant, has drastically evolved with the introduction of vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR TKIs) and immune checkpoint inhibitors (ICIs). The introduction of ICI-based regimens has significantly improved outcomes for patients with metastatic RCC. Currently, first-line therapy for patients with metastatic RCC involves multiple ICI-based regimens, either dual ICIs (with anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA- 4) and anti-programmed cell death- 1 (PD- 1) therapies) or anti-PD- 1 therapy in combination with VEGFR TKIs. Despite improving patient outcomes with ICI-based regimens, durable responses remain uncommon, highlighting the need for innovative treatment strategies. In this review, we highlight the current standard of care ICI-based regimens followed by ongoing clinical trials with novel combinations of existing FDA-approved agents and targets. We also discuss novel immunotherapies currently in clinical trials, which aim to improve antitumor T cell immunity either by improving T cell activation or T cell navigation to the tumor microenvironment. The incorporation of these novel therapies offers the potential to improve RCC patient outcomes, particularly by enhancing the durability of treatment responses.
Collapse
Affiliation(s)
- Zachary A Yochum
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Center of Molecular and Cellular Oncology, Yale Cancer Center, New Haven, CT, USA
| | - David A Braun
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- Center of Molecular and Cellular Oncology, Yale Cancer Center, New Haven, CT, USA.
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA.
- Department of Urology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
50
|
Lodde G, Albrecht LJ, Schadendorf D. [Treatment of metastatic melanoma: update 2025]. Dtsch Med Wochenschr 2025; 150:562-569. [PMID: 40262755 DOI: 10.1055/a-2500-0927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Immune checkpoint inhibition and targeted therapy with BRAF/MEK inhibition for BRAF-mutated melanoma have significantly improved progression-free and overall survival in patients with metastatic melanoma. Current research focuses on novel treatment strategies for PD-1 resistance, neoadjuvant approaches, and cellular therapies. 10-year follow-up data of randomized clinical trials show that both combined CTLA-4 and PD-1 immune checkpoint inhibition and PD-1 immune checkpoint inhibition alone can achieve long-term survival in metastatic melanoma. Potential surrogate markers of long-term response include a progression-free survival at 3 years after start of treatment and a reduction in tumour burden of at least 80%. The management of PD-1 resistance remains a challenge. Advances in molecular pathology have led to the identification of new therapeutic targets. Several cellular therapies are currently being evaluated in clinical trials as alternatives for melanoma patients refractory to immune checkpoint inhibition or targeted BRAF/MEK inhibition. In BRAF-mutant melanoma, combined BRAF/MEK inhibition is an alternative to immune checkpoint inhibition. Real-world data and clinical trial results on treatment sequencing suggest that immune checkpoint inhibition may improve survival in the first line setting, particularly in the absence of prior adjuvant systemic therapy. Adjuvant treatment leads to improved progression-free survival in melanoma patients while overall survival data are still pending. Neoadjuvant treatment seems to be a promising alternative to conventional adjuvant therapy for specific subgroups of melanoma patients. Participation in clinical trials offers patients the best opportunity to benefit from the latest treatment options.
Collapse
|