1
|
Lereim RR, Dunn C, Aamdal E, Chauhan SK, Straume O, Guren TK, Kyte JA. Plasma protein dynamics during ipilimumab treatment in metastatic melanoma: associations with tumor response, adverse events and survival. Oncoimmunology 2025; 14:2440967. [PMID: 39703053 DOI: 10.1080/2162402x.2024.2440967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024] Open
Abstract
The immune checkpoint inhibitor ipilimumab provides long term survival in some metastatic melanoma patients, but the majority has no benefit, and may experience serious side effects. Here, we investigated the dynamics of plasma cytokine concentrations and their potential utility for predicting treatment response, adverse events and overall survival (OS) in patients with metastatic melanoma undergoing ipilimumab monotherapy. A cohort of 148 patients was examined, with plasma samples collected prior to treatment initiation and at the end of the first and second treatment cycles. Concentrations of 48 plasma proteins were measured using a multiplex immunoassay. The results revealed a general increase in cytokine levels following the first ipilimumab dose, consistent with immune activation. Patients not responding to treatment exhibited significantly elevated baseline levels of G-CSF, IL-2RA, MIP-1a, and SCF, compared to tumor responders (p < 0.05). Furthermore, high levels of IL-2RA, IFNγ, PDGF-bb and MIG were linked to inferior OS, while high concentrations of MIF and RANTES were associated with improved OS (p < 0.05). A multivariate model containing CRP, LDH, ECOG, IL-2RA and PDGF-bb identified a subgroup of patients with poor OS. Patients who experienced severe immune-related adverse events within three months of treatment initiation had higher baseline concentrations of several cytokines, indicating a potential association between preexisting inflammation and adverse events. These findings indicate that the first dose of ipilimumab induces a systemic response with increased levels of circulating cytokines and suggest candidate biomarkers for clinical response, immune-mediated toxicity and survival. Further studies in independent patient cohorts are required to confirm the findings.
Collapse
Affiliation(s)
| | - Claire Dunn
- Department of Cancer Immunology, Oslo University Hospital, Oslo, Norway
| | - Elin Aamdal
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Oddbjørn Straume
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Tormod Kyrre Guren
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Jon Amund Kyte
- Department of Cancer Immunology, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| |
Collapse
|
2
|
Long Z, Yi Z, Yan W, Wang H. Trends in the immunotherapy for glioblastoma: A two-decade bibliometric analysis. Hum Vaccin Immunother 2025; 21:2466299. [PMID: 39950580 PMCID: PMC11834472 DOI: 10.1080/21645515.2025.2466299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/27/2025] [Accepted: 02/09/2025] [Indexed: 02/20/2025] Open
Abstract
Glioblastoma is a life-threatening primary malignant brain tumor with an unfavorable prognosis. Contributing factors to its poor outcome include tumor heterogeneity, low mutational burden, and immunosuppression within the tumor microenvironment. Recognizing these challenges, immunotherapeutic strategies have emerged as a promising avenue for glioblastoma treatment. Although several dynamic research and scientific trend have increasingly taken pace in the immunotherapeutic approaches to glioblastoma, systematic bibliometric studies on such trends are few. On this note, this study explores a bibliometric analysis of the research hotspots and trends in glioblastoma immunotherapy. We conducted a search in the Web of Science Core Collection database for articles on glioblastoma immunotherapy published between 2004 and 2024. Using VOSviewer and CiteSpace software, we analyzed collected articles to explore aspects such as country of origin, journal of publication, affiliated institute, authorship, keywords, and citation patterns. As of May 1, 2024, we retrieved 3,729 papers on Glioblastoma Immunotherapy. In the field of glioblastoma immunotherapy, the United States stands out as the leading contributor, with 1,708 publications and a substantial 90,590 citations. Following closely, China has made significant contributions through 926 publications, earning 17,533 citations, while Germany adds to the body of knowledge with 349 publications and 16,355 citations. Furthermore, Authoritative journals in this field include Clinical Cancer Research and Neuro-Oncology. The top five keywords during this period were temozolomide, radiotherapy, dendritic cell, cytotoxic T lymphocyte, and vaccination. Moreover, Hotspots in the field include immune checkpoint inhibitors and chimeric antigen receptor T cell therapy.
Collapse
Affiliation(s)
- Zhi Long
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Yan
- The First Department of General Surgery, Hunan Provincial People’s Hospital, Hunan Normal University, Changsha, China
| | - Hongxin Wang
- Department of Neurosurgery, The Affiliated Changsha Central Hospital, Hengyang Medical School,University of South China, Changsha, China
| |
Collapse
|
3
|
Kuusisalo S, Iivanainen S, Koivunen JP. Association of anti-PD-(L)1 treatment duration to efficacy in advanced solid tumors: a single center retrospective study. Ann Med 2025; 57:2476729. [PMID: 40091413 PMCID: PMC11915729 DOI: 10.1080/07853890.2025.2476729] [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: 02/05/2024] [Revised: 11/28/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are a standard of care in multiple cancers. Only a minority benefits, thus, optimal use and treatment duration remain indistinct. While biomarkers albeit PD-L1 are scarce, declined performance status and cancer-related systemic inflammation detected by blood inflammatory markers such as C-reactive protein (CRP) have been linked to inferior prognosis. MATERIALS AND METHODS We investigated the association of limited anti-PD-(L)1 treatment duration to therapy efficacy in melanoma and non-small cell lung cancer (NSCLC) patients who received therapy in a non-curative setting in Oulu University Hospital 2014-2022. Baseline prognostic factors (e.g. ECOG, CRP, and PD-L1 for NSCLC) were collected. Progression-free (PFS), overall (OS), and IO-free survival were analyzed using the Kaplan-Meier and Cox regression methods. RESULTS 126 patients (NSCLC, n = 72; melanoma, n = 54) were included. Majority (n = 101) were treated in the first line. Objective response rate was 34.9%. The median (m) anti-PD-(L)1 treatment duration was 3.42 months (mo). The mPFS and mOS were 6.8 mo (CI 95% 4.4-9.3) and 19.1 mo (CI 95% 13.3-24.9). Of the baseline factors, ECOG and CRP retained their significance in multivariate analysis for PFS (HR 0.34, CI 95% 0.19-0.59; HR 0.34, CI 95% 0.22-054) and OS (HR 0.38, CI 95% 0.20-0.71; HR 0.29, CI 95% 0.17-0.49). No difference was observed in PFS (HR 1.40, CI 95% 0.68-2.90) or OS (HR 0.69, CI 95% 0.29-1.65) according to treatment duration (3-6mo vs. > 6 mo). Long median IO-free survival (10.2 months; CI 95%, 4.1-16.3) was detected. CONCLUSION We characterized an anti-PD-(L)1 treated advanced NSCLC and melanoma cohort in which treatment benefit occurs irrespective of treatment duration and long-term benefit is observed off-treatment.
Collapse
Affiliation(s)
- Saara Kuusisalo
- Department of Medical Oncology and Radiotherapy and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Sanna Iivanainen
- Department of Medical Oncology and Radiotherapy and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jussi P. Koivunen
- Department of Medical Oncology and Radiotherapy and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
4
|
Bick F, Blanchetot C, Lambrecht BN, Schuijs MJ. Targeting γc family cytokines with biologics: current status and future prospects. MAbs 2025; 17:2468312. [PMID: 39967341 PMCID: PMC11845063 DOI: 10.1080/19420862.2025.2468312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
Over the recent decades the market potential of biologics has substantially expanded, and many of the top-selling drugs worldwide are now monoclonal antibodies or antibody-like molecules. The common gamma chain (γc) cytokines, Interleukin (IL-)2, IL-4, IL-7, IL-9, IL-15, and IL-21, play pivotal roles in regulating immune responses, from innate to adaptive immunity. Dysregulation of cell signaling by these cytokines is strongly associated with a range of immunological disorders, which includes cancer as well as autoimmune and inflammatory diseases. Given the essential role of γc cytokines in maintaining immune homeostasis, the development of therapeutic interventions targeting these molecules poses unique challenges. Here, we provide an overview of current biologics targeting either single or multiple γc cytokines or their respective receptor subunits across a spectrum of diseases, primarily focusing on antibodies, antibody-like constructs, and antibody-cytokine fusions. We summarize therapeutic biologics currently in clinical trials, highlighting how they may offer advantages over existing therapies and standard of care, and discuss recent advances in this field. Finally, we explore future directions and the potential of novel therapeutic intervention strategies targeting this cytokine family.
Collapse
Affiliation(s)
- Fabian Bick
- Argenx BV, Zwijnaarde, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | | | - Bart N. Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Martijn J. Schuijs
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
5
|
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
|
6
|
Huang R, Wu Y, Shen F, Chen S, Yang X, Lin Y, Fang Y, Shen J. Manganese-coordinated nanoparticles loaded with CHK1 inhibitor dually activate cGAS-STING pathway and enhance efficacy of immune checkpoint therapy. Biomaterials 2025; 319:123199. [PMID: 40009899 DOI: 10.1016/j.biomaterials.2025.123199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/26/2025] [Accepted: 02/16/2025] [Indexed: 02/28/2025]
Abstract
Notable advancements have been made in utilizing immune checkpoint blockade (ICB) for the treatment of various cancers. However, the overall response rates and therapeutic effectiveness remain unsatisfactory. One cause is the inadequate immune environment characterized by poor T cell infiltration in tumors. To address these limitations, enhancing immune infiltration is crucial for optimizing the therapeutic efficacy of ICB. Activating the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway is essential for initiating immune response and has become a potential target for developing combination therapies with ICB. In this study, we designed and fabricated manganese-containing nanoparticles loaded with the CHK1 inhibitor PF477736, which were subsequently encapsulated with macrophage membrane (PF/MMSN@MPM). This innovative design achieved excellent tumor targeting and demonstrated potent antitumor effects. The combination therapy dually amplified the cGAS-STING pathway, causing a cascade of enhanced therapeutic effects against tumors. Furthermore, single-cell mass cytometry (CyTOF) analysis revealed that PF/MMSN@MPM enhanced the activation and infiltration of immune cells. Moreover, the combination of PF/MMSN@MPM with anti-PD-1 (αPD-1) exhibited a stronger therapeutic effect compared to αPD-1 alone. PF/MMSN@MPM precisely and synergistically activated the cGAS-STING pathway, significantly improving therapeutic efficacy of ICB, and offering promising potential for tumor therapy.
Collapse
Affiliation(s)
- Rui Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yijia Wu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Feiyang Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuai Chen
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyu Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yao Lin
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yan Fang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jianfeng Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
7
|
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
|
8
|
Ma C, Yu X, Zhang X, Su L, Jiang O, Cui R. Combination of radiotherapy and ICIs in advanced hepatocellular carcinoma: A systematic review of current evidence and future prospects (Review). Oncol Lett 2025; 30:342. [PMID: 40438865 PMCID: PMC12117537 DOI: 10.3892/ol.2025.15088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/24/2025] [Indexed: 06/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a global health concern because of its rising prevalence and high fatality rates. Conventional treatments for advanced HCC (aHCC) have limited success, emphasizing the need for novel treatment options. Radiotherapy (RT) treatments, such as stereotactic body radiation and proton therapy, improve local tumor management via precision targeting. Moreover, immune checkpoint inhibitors (ICIs) that target the programmed cell death protein 1(PD-1)/PD ligand 1 (PD-L1) and cytotoxic T lymphocyte associated protein 4 (CTLA-4) pathways have promise for systemic antitumor effectiveness. The combination of RT and ICIs takes advantage of their complementary mechanisms: RT kills immunogenic cells and controls the tumor microenvironment to increase antigen presentation, whereas ICIs enhance and maintain antitumor immune responses. This combination enhances tumor regression and immune response in aHCC, improving response rate and progression-free survival with manageable safety. The present review aimed to summarize the rationale for combining RT + ICIs in patients with aHCC and clinical outcomes, as well as ways to enhance this combination technique. The combination of these models is a promising technique for improving outcomes for patients with aHCC and warrants further investigation.
Collapse
Affiliation(s)
- Cheng Ma
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Xinlin Yu
- Department of Oncology, The Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Xialin Zhang
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Lihong Su
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Ou Jiang
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Ran Cui
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| |
Collapse
|
9
|
Janes LA, Haykal T, Angeles CV. Vaccines in Melanoma: Past, Present, and Future. Surg Oncol Clin N Am 2025; 34:411-421. [PMID: 40413007 DOI: 10.1016/j.soc.2025.01.004] [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
While immune checkpoint inhibitor (ICI) immunotherapy has significantly improved the outcomes for advanced melanoma patients, more than 50% of patients do not respond. Cancer vaccines can induce a robust anti-tumor immune response, which may provide the necessary immunomodulation required for ICI efficacy. To date, there has only been modest clinical benefit with melanoma vaccines. Recent advancements with improved vaccine technology including the use of mRNA vaccines have promised to lead better clinical outcomes. This review will discuss the historical use of vaccines for melanoma, approaches to vaccine development, the current ongoing trials, and future directions.
Collapse
Affiliation(s)
- Lindsay A Janes
- Department of Surgery, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Tarek Haykal
- Department of Medicine, University of Michigan, Rogel Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Christina V Angeles
- Departments of Surgery and Dermatology, University of Michigan, Rogel Cancer Center, 1500 East Medical Center Drive, Cancer Center 6219, Ann Arbor, MI 48109, USA.
| |
Collapse
|
10
|
Marciniak M, Stachowicz-Suhs M, Wagner M. The role of innate immune cells in modulating vascular dynamics in skin malignancies. Biochim Biophys Acta Rev Cancer 2025; 1880:189331. [PMID: 40280501 DOI: 10.1016/j.bbcan.2025.189331] [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: 01/28/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
A developing tumor relies heavily on blood vessels to supply oxygen and nutrients. As a result, angiogenesis, the formation of new blood vessels, supports tumor growth and progression. Similarly, lymphangiogenesis, the formation of new lymphatic vessels, plays a critical role in metastatic dissemination by providing pathways for malignant cells to spread. The tumor microenvironment is crucial for establishing and maintaining these vascular networks, with innate immune cells playing a key regulatory role. Notably, immune cells are specifically enriched in barrier tissues, such as the skin, emphasizing their importance in skin malignancies. Therefore, understanding their role in regulating angiogenesis and lymphangiogenesis is essential for developing novel therapeutic strategies. This review article explores how innate immune cells influence tumor vasculature and highlights the therapeutic potential that may arise from this knowledge.
Collapse
Affiliation(s)
- Mateusz Marciniak
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland; Department of Biochemistry and Immunochemistry, Wrocław Medical University, Wrocław, Poland
| | - Martyna Stachowicz-Suhs
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland
| | - Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland.
| |
Collapse
|
11
|
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
|
12
|
Wang X, Tian Y, Wu X, Zhu Y, Chen H, Wang Z, Liu Z, Tan J, Pan Z, Cao J, Li Z, Zhang X, Shi Z, Wang J, Liu T. Targeting PERP promotes anti-tumor immunity in HNSCC by regulating tumor immune microenvironment and metabolic homeostasis. Mol Cancer 2025; 24:168. [PMID: 40483487 PMCID: PMC12144827 DOI: 10.1186/s12943-025-02360-4] [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: 02/24/2025] [Accepted: 05/20/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND PERP may have the potential to function as an oncogene. However, the precise function, prognostic value, and predictive significance remain shrouded in ambiguity. METHODS We conducted an in-depth analysis using pan-cancer RNA sequencing data and various online web tools to investigate the correlation between PERP and crucial clinical outcomes such as prognosis, tumor microenvironment, and tumor metabolism. In addition, we explored the tumor-promoting role of PERP and its potential mechanisms through models such as immunofluorescence staining, flow cytometry, cell proliferation assays, wound healing assays, cell migration assays, mass spectrometry analysis and isotope tracing. Further in vivo models confirmed the functional consistency of PERP across pan-cancer. Finally, we analyzed the potential of PERP as a predictive factor for immunotherapy sensitivity in a clinical cohort. RESULTS PERP exhibits elevated expression in the majority of cancer types and impedes immune cell infiltration as well as immune checkpoint reactivity in pan-cancer. We confirmed that PERP can promote tumor progression by tumor cell proliferation, scratch and transwell experiments. Meanwhile, the absence of PERP restricts the flux of 13C6-glucose into glycolysis and the tricarboxylic acid (TCA) cycle. Importantly, the deficiency of PERP enhances the in vivo anti-tumor efficacy of PD1 monoclonal antibodies. In addition, low PERP expression is highly correlated with the response of head and neck squamous cell carcinoma (HNSCC) patients to immunotherapy. CONCLUSIONS PERP represents a promising predictive/diagnostic biomarker and therapeutic target for HNSCC patients.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150000, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Yuxi Tian
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- National Medical Metabolomics International Collaborative Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xiaohong Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Yewen Zhu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Huihong Chen
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Zeyao Wang
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150000, China
| | - Zihan Liu
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150000, China
| | - Jiaqi Tan
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Zhaoyu Pan
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Jiaoyan Cao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Zhenjiang Li
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Xin Zhang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Zhongjie Shi
- Department of Pediatrics, Wayne State University, Detroit, MI, 48201, USA
| | - Juncheng Wang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
- National Medical Metabolomics International Collaborative Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| | - Tong Liu
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150000, China.
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China.
| |
Collapse
|
13
|
Hoeijmakers LL, Rozeman EA, Lopez-Yurda M, Grijpink-Ongering LG, Heeres BC, van de Wiel BA, Flohil C, Sari A, Heijmink SWTPJ, van den Broek D, Broeks A, de Groot JWB, Vollebergh MA, Wilgenhof S, van Thienen JV, Haanen JBAG, Blank CU. Durable responses upon short-term addition of targeted therapy to anti-PD1 in advanced melanoma patients: 5-year progression-free and overall survival update of the IMPemBra trial. Eur J Cancer 2025; 222:115431. [PMID: 40279684 DOI: 10.1016/j.ejca.2025.115431] [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: 01/23/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/27/2025]
Abstract
BACKGROUND The addition of targeted therapy (TT) to immune checkpoint inhibitors has been shown to transiently increase immune infiltration in melanoma. This formed the rationale for the IMPemBra trial, which showed a numerical increase in progression-free survival (PFS) in patients treated with short-term/intermittent TT and anti-PD1 compared to anti-PD1 alone. In this report, the final toxicity-analysis, 5-year PFS and exploratory analysis of overall survival (OS) will be reported, together with an analysis of subsequent therapies. PATIENTS AND METHODS 32 treatment-naïve patients with a BRAFV600E/K-mutated advanced melanoma were treated with 2 cycles of pembrolizumab 200 mg every 3 weeks, followed by randomization to continue pembrolizumab monotherapy for six weeks in cohort-1 versus pembrolizumab plus intermittent dabrafenib 150 mg BID + trametinib 2 mg QD 2×1-week (cohort 2), 2×2-weeks (cohort 3), or 1×6-weeks (cohort 4). After week 12, all patients continued pembrolizumab monotherapy for a maximum of 2 years. RESULTS With a median follow-up of 73 months, final grade 3-4 immune-related adverse events are 12 % (cohort 1), 12 % (cohort 2), 38 % (cohort 3) and 63 % (cohort 4). Estimated 5-year PFS and OS rates were 25 % and 50 % for pembrolizumab monotherapy (cohort-1) and 46 % and 71 % for pembrolizumab + intermittent TT (cohorts 2-4). Estimated 5-year PFS and OS were 63 % and 63 % (cohort 2), 38 % and 75 % (cohort 3), and 38 % and 75 % (cohort 4), respectively. The subsequent therapies were balanced between cohorts. Patients treated with short-term/intermittent schemes achieved durable responses upon subsequent TT again. CONCLUSION This survival update from the IMPemBra trial demonstrates that combination of short-term TT and checkpoint inhibition can induce long-lasting responses, warranting further analyses in larger cohorts, and in a randomized design.
Collapse
Affiliation(s)
- L L Hoeijmakers
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - E A Rozeman
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M Lopez-Yurda
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - B C Heeres
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - B A van de Wiel
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - C Flohil
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A Sari
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - S W T P J Heijmink
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - D van den Broek
- Department of Laboratory medicine, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A Broeks
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - M A Vollebergh
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - S Wilgenhof
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - J V van Thienen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - J B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, Leiden University Medical Center, the Netherlands
| | - C U Blank
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, Leiden University Medical Center, the Netherlands; Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
14
|
Saamarthy K, Daams R, Sime W, Persson C, Chygorin E, Ahlqvist K, Evans-Axelsson S, Strand D, Massoumi R. An optimised Bcl-3 inhibitor for melanoma treatment. Br J Pharmacol 2025; 182:2426-2446. [PMID: 39943627 DOI: 10.1111/bph.17467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND AND PURPOSE Malignant melanoma is the most lethal form of skin cancer, characterised by a poor survival rate. One of the key factors driving the aggressive growth of melanoma cells is the elevated expression of the proto-oncogene Bcl-3. This study aims to optimise, evaluate and characterise a second-generation Bcl-3 inhibitor, using melanoma as a model to demonstrate its potential therapeutic efficacy. EXPERIMENTAL APPROACH We synthesised and screened a series of structural analogues and selected A27, the most promising candidate for further investigation. We assessed whether A27 disrupted the interaction between Bcl-3 and its binding partner, p50, and examined the subsequent effects on cyclin D1 expression. Additionally, we evaluated the impact of A27 on melanoma cell proliferation and migration in vitro, as well as its therapeutic efficacy in various in vivo melanoma models. KEY RESULTS Nuclear magnetic resonance (NMR) confirmed that A27 directly binds to Bcl-3, effectively inhibiting its function. By disrupting the Bcl-3/p50 interaction, A27 led to a significant down-regulation of cyclin D1 expression. In cellular assays, A27 markedly reduced proliferation and migration of melanoma cells. In vivo, treatment with A27 resulted in a substantial reduction in melanoma tumour growth, with no observed toxicity in treated animals. CONCLUSIONS AND IMPLICATIONS At present, no other Bcl-3 inhibitors exist for clinical application in the field of oncology, and as a result, our novel findings provide a unique opportunity to develop a highly specific drug against malignant melanoma to meet an urgent clinical need.
Collapse
Affiliation(s)
- Karunakar Saamarthy
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Renée Daams
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Wondossen Sime
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Cecilia Persson
- Swedish NMR Center, Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Eduard Chygorin
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Kristofer Ahlqvist
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Susan Evans-Axelsson
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Daniel Strand
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Ramin Massoumi
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Fraterman I, Cerquin LE, de Ligt KM, van der Loo I, Wilgenhof S, van de Poll-Franse LV, Beets-Tan RGH, Tissier RLM, Trebeschi S. Muscle atrophy and organ enlargement associated with quality of life during systemic therapy for melanoma: findings from an AI-based body composition analysis. Eur Radiol 2025; 35:3476-3485. [PMID: 39702633 DOI: 10.1007/s00330-024-11289-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/26/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024]
Abstract
INTRODUCTION Health-related quality of life (HRQoL) is emerging as an endpoint, adjunct to survival, in cancer treatment. For this reason, the European Organization for Research and Treatment of Cancer (EORTC) has developed standardized quality-of-life questionnaires to collect patient-reported outcome measurement (PROM), which so far have been widely used in clinical trials to evaluate the impact of new drugs on cancer patients. However, while these questionnaires comprehensively describe patient functions, little is known about their association with patient characteristics. This study aims to bridge this gap and investigate the association between patient body composition and HRQoL. MATERIALS AND METHODS A retrospective cohort of melanoma patients treated with systemic therapy who completed HRQoL questionnaires and had regular imaging follow-ups was included. The primary endpoint was the association between n = 116 AI-measured longitudinal volumes of thoracic and abdominal organs, subcutaneous and visceral fat, skeleton and muscles (estimated by TotalSegmentator), and physical functioning (PF), role functioning (RF) and fatigue (FA) (estimated by the EORTC-QLQ-C30). RESULTS The n = 358 patients were included. Our findings show larger liver, spleen, and gallbladder volumes associated with decreased PF and RF and an increase in FA (p < 0.05). Furthermore, larger muscle volumes were associated with an increase in PF and RF and a decrease in FA (p < 0.01). DISCUSSION Our findings show significant associations between AI-measured body and organ analysis and HRQoL in patients with melanoma on systemic treatment. Future research is needed to understand the underlying cause and determine the possible predictive ability of these imaging features. KEY POINTS Question Are changes in body composition associated with changes in HRQoL in melanoma patients undergoing systemic therapy? Findings AI-based body composition analysis shows that larger muscle volumes are linked to improved HRQoL, while organ enlargement is associated with a decline in it. Clinical relevance Our findings indicate new imaging biomarkers that can help monitor patients and evaluate treatment responses. These biomarkers link patient function to physical changes during treatment, offering insights for creating response evaluation criteria that also consider improvements in quality of life.
Collapse
Affiliation(s)
- Itske Fraterman
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Laura Estacio Cerquin
- Department of Radiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- GROW-Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Kelly M de Ligt
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Iris van der Loo
- Department of Radiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- GROW-Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Sofie Wilgenhof
- Department of Medical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Lonneke V van de Poll-Franse
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organization, Utrecht, The Netherlands
- Department of Medical and Clinical Psychology, Center of Research On Psychological and Somatic Disorders (CoRPS), Tilburg University, Tilburg, The Netherlands
| | - Regina G H Beets-Tan
- GROW-Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, The Netherlands
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Renaud L M Tissier
- Biostatistics Center, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Stefano Trebeschi
- Department of Radiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
- GROW-Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
16
|
Janßen S, Moufarrej A, Springer R, Häberle L, Jäger P, Antke C, Homey B, Lindhof HH. Diffuse large B-cell lymphoma mimicking metastatic melanoma: the importance of biopsies in the era of immune checkpoint inhibitors. Melanoma Res 2025; 35:197-200. [PMID: 40013997 DOI: 10.1097/cmr.0000000000001028] [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: 02/28/2025]
Abstract
Positron emission tomography (PET)/computed tomography (CT) imaging is an established tool in diagnosing and staging for various malignancies, however, during immune checkpoint inhibitor (ICI) therapy not only inflammatory changes may mimic disease progression, but also secondary malignancies should be considered in the setting of unusual clinical and radiographic findings. Here, we present the case of a 64-year-old man with a lymphogenic metastatic malignant melanoma treated with ipilimumab/nivolumab, in whom PET/CT indicated tumor progression of an intra-abdominal mass. Biopsy revealed an unusual reactive T-cell expansion without clonal expansion, pathologically consistent with ICI-induced immune response. As the patient's general condition worsened, we switched to targeted therapy, which had to be discontinued due to increasing fatigue. Follow-up PET/CT at 6 months showed further intra-abdominal progression. Subsequent histopathology of the extirpated mesenteric lymph node conglomerate now revealed diffuse large B-cell lymphoma. Our case highlights the importance of repeated histologic examinations of radiologic pathologies to distinguish secondary malignancies from ICI-induced inflammatory reactions or progressive disease.
Collapse
Affiliation(s)
| | | | | | | | - Paul Jäger
- Department of Hematology, Oncology, and Clinical Immunology
| | - Christina Antke
- Department of Nuclear Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | | | | |
Collapse
|
17
|
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
|
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
|
Rajak P. Immune checkpoint inhibitors: From friend to foe. Toxicol Rep 2025; 14:102033. [PMID: 40353246 PMCID: PMC12063143 DOI: 10.1016/j.toxrep.2025.102033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 04/18/2025] [Accepted: 04/20/2025] [Indexed: 05/14/2025] Open
Abstract
Immune checkpoints are crucial in regulating the activation of cell-mediated and humoral immune responses. However, cancer cells hijack this mechanism to evade the immune surveillance and anti-cancer response. Typically, receptors like PD-1 and CTLA4, expressed on immune cells, prevent the activation and differentiation of T cells. They also inhibit the development of autoimmune reactions. However, ligands such as PD-L1 for the receptor PD-1 are also expressed on the surface of cancer cells that help prevent the activation of anti-cancer immune responses by blocking the signalling pathways mediated by PD-1 and CTLA4. Immune checkpoint inhibitors (ICIs) have promising therapeutic efficacy for treating several cancers by activating T cells and their differentiation into effector cells against tumours. Nonetheless, hyperactivated immune cells usually contribute to detrimental issues, also known as immune-related adverse effects (IrAE). IrAEs have been observed in multiple organs, leading to neurological issues, colitis, endocrine dysfunction, renal issues, hepatitis, pneumonitis, and dermatitis. The interplay between hyperactivated T cells and Treg cells helps in orchestrating the development of autoimmunity. Moreover, the crosstalk between proinflammatory interleukins and the development of autoantibodies also mediates the multiorgan effects of ICIs in cancer patients. IrAEs are generally managed by terminating the ICI therapy, reducing the ICI dose, and by using corticosteroids to subvert inflammation. Therefore, the present review aims to delineate the impacts of ICIs on the development of autoimmune diseases and inflammatory outcomes in cancer patients. In addition, mechanistic insight involving immune cells, cytokines, and autoantibodies for ICI-mediated IrAEs will also be discussed with updated findings in this field.
Collapse
Affiliation(s)
- Prem Rajak
- Toxicology Research Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| |
Collapse
|
20
|
Tay DL, Perkins R, Muehlberger JR, Casucci T, Campbell T, McFarland MM, Ellington L. The Role of Palliative Nursing in the Context of Immunotherapies: A Scoping Review of Patient- and Family-Centered Care Considerations. J Hosp Palliat Nurs 2025; 27:E118-E128. [PMID: 40063380 DOI: 10.1097/njh.0000000000001111] [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: 05/08/2025]
Abstract
Immunotherapies are increasingly used to treat advanced cancers and can extend survival in some patients. The potential for longer survival may inflate prognosis expectations among patients and caregivers and delay palliative care. This review sought to identify issues impacting patients and caregivers that influence specialized palliative care use in the context of immunotherapies. A scoping review was conducted with guidance from the JBI Manual for Evidence Synthesis. Studies were identified using MEDLINE, EMBASE, CINAHL Complete, AgeLine, Cochrane Library, and APA PsycINFO, without any database filters or limiters. Inclusion criteria included palliative care utilization among adult cancer patients receiving immunotherapy and their family caregivers in any geographical or specialty setting. The protocol was registered in the Open Science Framework. Using Covidence, 11 898 studies were screened by reviewers, with 10 studies meeting inclusion criteria. Themes of patient and family considerations that affect the use of specialized palliative care included (1) understanding of disease/treatment, (2) hope/optimism, (3) communication barriers, and (4) emotional distress and uncertainty. This review is one of the first to identify issues impacting patients receiving immunotherapies and their families. Findings have implications for nurse communication, education, and psychosocial support with advanced cancer patients and families receiving immunotherapies.
Collapse
|
21
|
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
|
22
|
He Y, Wang X. A comprehensive investigation of associations between cell death pathways and molecular and clinical features in pan-cancer. Clin Transl Oncol 2025; 27:2731-2749. [PMID: 39487950 DOI: 10.1007/s12094-024-03769-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Regulated cell death (RCD) pathways play significant roles in tumorigenesis. However, systematic investigation into correlations between RCD and various molecular and clinical features, particularly anti-tumor immunity and immunotherapy response in pan-cancer remains lacking. METHODS Using the single-sample gene set enrichment analysis, we quantified the activities of six RCD pathways (apoptosis, autophagy, ferroptosis, cuproptosis, necroptosis, and pyroptosis) in each cancer specimen. Then, we explored associations of these six RCD pathways with tumor immunity, genomic instability, tumor phenotypes and clinical features, and responses to immunotherapy and targeted therapies in pan-cancer by statistical analyses. RESULTS Our results showed that the RCD (except autophagy) activities were oncogenic signatures, as evidenced by their hyperactivation in late stage or metastatic cancer patients, positive correlations with tumor proliferation, stemness, genomic instability and intratumor heterogeneity, and correlation with worse survival outcomes in cancer. In contrast, autophagy was a tumor suppressive signature as its associations with molecular and clinical features in cancer shows an opposite pattern compared to the other RCD pathways. Furthermore, heightened RCD (except cuproptosis) activities were correlated with increased sensitivity to immune checkpoint inhibitors. Additionally, elevated activities of pyroptosis, autophagy, cuproptosis and necroptosis were associated with increased drug sensitivity in a broad spectrum of anti-tumor targeted therapies, while the elevated activity of ferroptosis was correlated with decreased sensitivity to numerous targeted therapies. CONCLUSION RCD (except autophagy) activities correlate with unfavorable cancer prognosis, while the autophagy activity correlate with favorable clinical outcomes. RCD (except cuproptosis) activities are positive biomarkers for anti-tumor immunity and immunotherapy response.
Collapse
Affiliation(s)
- Yin He
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Intelligent Pharmacy Interdisciplinary Research Center, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Intelligent Pharmacy Interdisciplinary Research Center, China Pharmaceutical University, Nanjing, 211198, China.
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
23
|
Green N, Kurt M, Moshyk A, Larkin J, Baio G. A Bayesian Hierarchical Mixture Cure Modelling Framework to Utilize Multiple Survival Datasets for Long-Term Survivorship Estimates: A Case Study From Previously Untreated Metastatic Melanoma. Stat Med 2025; 44:e70132. [PMID: 40444748 PMCID: PMC12124107 DOI: 10.1002/sim.70132] [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: 01/29/2024] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 06/02/2025]
Abstract
Time to an event of interest over a lifetime is a central measure of the clinical benefit of an intervention used in a health technology assessment (HTA). Within the same trial, multiple end-points may also be considered. For example, overall and progression-free survival time for different drugs in oncology studies. A common challenge is when an intervention is only effective for some proportion of the population who are not clinically identifiable. Therefore, latent group membership as well as separate survival models for identified groups need to be estimated. However, follow-up in trials may be relatively short leading to substantial censoring. We present a general Bayesian hierarchical framework that can handle this complexity by exploiting the similarity of cure fractions between end-points; accounting for the correlation between them and improving the extrapolation beyond the observed data. Assuming exchangeability between cure fractions facilitates the borrowing of information between end-points. We undertake a comprehensive simulation study to evaluate the model performance under different scenarios. We also show the benefits of using our approach with a motivating example, the CheckMate 067 phase 3 trial consisting of patients with metastatic melanoma treated with first line therapy.
Collapse
Affiliation(s)
| | - Murat Kurt
- Worldwide Health Economics and Outcomes Research, Bristol Myers SquibbLawrenceNew JerseyUSA
| | - Andriy Moshyk
- Worldwide Health Economics and Outcomes Research, Bristol Myers SquibbLawrenceNew JerseyUSA
| | | | | |
Collapse
|
24
|
Enzler T, Frankel TL. Pancreatic cancer precursor lesions - Can immunotherapy prevent progression into pancreatic ductal adenocarcinoma? Cancer Lett 2025; 619:217662. [PMID: 40127814 DOI: 10.1016/j.canlet.2025.217662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers, with a 5-year survival rate of only 12.5 %. Early detection of PDAC or addressing risk factors for PDAC development are ways to improve outcomes. PDAC can arise from precursor lesions, including pancreatic intraepithelial neoplasia (PanIN), intraductal papillary mucinous neoplasm (IPMN), and less frequent, mucinous cystic neoplasm (MCN), and other rare precursor variants. High-risk precursor lesions harbor a substantial chance of evolving into PDAC. Such lesions can often be found in resected PDAC specimens adjacent to the cancer. Unfortunately, recognizing precursor lesions that need to be resected is often tricky, and resections frequently end in major surgical interventions. Thus, better ways to handle precursor lesions are desperately needed. We mapped the immune microenvironments (IMEs) of PanINs, IPMNs, and MCNs on a cellular level using multiplex immunofluorescence and computational imaging technology and compared the findings to PDACs and normal pancreatic tissues. We found distinct and potentially targetable mechanisms of immunosuppression between the two main precursor lesions, PanIN and IMPN. Immunosuppression in IPMNs seems partly mediated by programmed cell death protein 1 ligand (PD-L1) expression on antigen-presenting cells (APCs). By contrast, elevated numbers of regulatory T cells (Tregs) seem to be key players in the immunosuppression of PanINs. Thus, treating high-risk IPMNs with anti-PD-1 and high-risk PanINs with agents targeting Tregs, such as anti-lymphocyte associated protein 4 (anti-CTLA-4) antibodies, could reverse their immunosuppressive state. Reversal of immunosuppression will restore immunosurveillance and eventually prevent progression into PDAC. We also review relevant published and ongoing non-surgical treatment approaches for high-risk IPMNs and PanINs.
Collapse
Affiliation(s)
- Thomas Enzler
- Department of Medicine, University of Michigan, Ann Arbor, MI, 40109, USA.
| | - Timothy L Frankel
- Department of Surgery, University of Michigan, Ann Arbor, MI, 40109, USA
| |
Collapse
|
25
|
Shi Y, Li X, Li Z, Sun J, Gao T, Wei G, Guo Q. Nano-formulations in disease therapy: designs, advances, challenges, and future directions. J Nanobiotechnology 2025; 23:396. [PMID: 40448105 DOI: 10.1186/s12951-025-03442-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 05/05/2025] [Indexed: 06/02/2025] Open
Abstract
Nano-formulations, as an innovative drug delivery system, offer distinct advantages in enhancing drug administration methods, improving bioavailability, promoting biodegradability, and enabling targeted delivery. By exploiting the unique size advantages of nano-formulations, therapeutic agents, including drugs, genes, and proteins, can be precisely reorganized at the microscale level. This modification not only facilitates the precise release of these agents but also significantly enhances their efficacy while minimizing adverse effects, thereby creating novel opportunities for treatment of a wide range of diseases. In this review, we discuss recent advancements, challenges, and future perspectives in nano-formulations for therapeutic applications. For this aim, we firstly introduce the development, design, synthesis, and action mechanisms of nano-formulations. Then, we summarize their applications in disease diagnosis and treatment, especially in fields of oncology, pulmonology, cardiology, endocrinology, dermatology, and ophthalmology. Furthermore, we address the challenges associated with the medical applications of nanomaterials, and provide an outlook on future directions based on these considerations. This review offers a comprehensive examination of the current applications and potential significance of nano-formulations in disease diagnosis and treatment, thereby contributing to the advancement of modern medical therapies.
Collapse
Affiliation(s)
- YunYan Shi
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Xiao Li
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Zhiyuan Li
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Jialin Sun
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Tong Gao
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Gang Wei
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China.
| | - Qie Guo
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China.
| |
Collapse
|
26
|
Collins LG, Elliott TM, Webb A, Reid IR, Sinclair C, Comans T, Karnon J, Foeglein A, van Gorp K, Fanning V, Neale RE. Estimated health and economic effects associated with over- and under-exposure to solar ultraviolet radiation in Australia and New Zealand using the SUNEX simulation model. Photochem Photobiol Sci 2025:10.1007/s43630-025-00726-7. [PMID: 40434562 DOI: 10.1007/s43630-025-00726-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 04/19/2025] [Indexed: 05/29/2025]
Abstract
Multiple health problems are associated with either over- or under-exposure to ultraviolet (UV) radiation. Using an agent-based microsimulation model, we examined the joint health and economic effects of conditions arising from over-exposure to sunlight (i.e., melanoma, keratinocyte skin carcinoma (KC) and cataract) and under-exposure to sunlight via vitamin D deficiency (i.e., fragility fractures and multiple sclerosis). We developed an agent-based model to estimate and compare incident cases, disease-specific deaths, healthcare costs and losses in quality-adjusted life years (QALYs) attributable to over- or under-exposure to UV radiation. Simulations were performed over a 20-year period for populations in 14 locations across Australia and New Zealand. Conditions caused by over-exposure to UV radiation were predicted to result in 6.0 and 1.2 million new cases compared with 0.12 and 0.08 million cases from under-exposure in Australia and New Zealand, respectively. However, the number of deaths due to under-exposure (Australia: 58,503; New Zealand: 20,104) were higher than those arising from over-exposure (Australia: 49,320; New Zealand: 7136), but this was dependent on the definition of vitamin D deficiency used. The expected healthcare costs from over-exposure to UV radiation were AU$12.4 billion in Australia and NZ$5.2 billion in New Zealand, three-fold higher than costs for conditions attributable to under-exposure in both countries. Despite the enormous burden of skin cancers, highlighting the importance of sun protection, avoidable deaths and healthcare costs of fragility fractures due to a lack of UV radiation requires a reduction in vitamin D deficiency in Australians and New Zealanders.
Collapse
Affiliation(s)
- Louisa G Collins
- Viertel Cancer Research Centre, Cancer Council Queensland, Brisbane, QLD, Q4006, Australia.
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Q4006, Australia.
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Q4029, Australia.
- School of Nursing and Cancer and Palliative Care Outcomes Centre, Queensland University of Technology (QUT), Brisbane, QLD, Q4059, Australia.
| | - Thomas M Elliott
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Q4006, Australia
| | - Ann Webb
- Department of Earth and Environmental Sciences, University of Manchester, Manchester, M13 9PL, UK
| | - Ian R Reid
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Craig Sinclair
- Cancer Council Victoria, Melbourne, VIC, 3002, Australia
| | - Tracy Comans
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Q4029, Australia
| | - Jonathan Karnon
- College of Medicine and Public Health, Flinders University, Adelaide, SA, SA5042, Australia
| | - Anna Foeglein
- Heisenberg Analytics, Brisbane, QLD, 4068, Australia
| | | | - Vanessa Fanning
- Lived Experience Expert Panel, Multiple Sclerosis Australia, Sydney, NSW, Australia
| | - Rachel E Neale
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Q4006, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Q4029, Australia
| |
Collapse
|
27
|
Wu Y, Zhou Y, Xia S, Meng Z. The real-world safety of Nivolumab: a pharmacovigilance analysis based on the FDA adverse event reporting system. Front Immunol 2025; 16:1605958. [PMID: 40491923 PMCID: PMC12146392 DOI: 10.3389/fimmu.2025.1605958] [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: 04/04/2025] [Accepted: 05/08/2025] [Indexed: 06/11/2025] Open
Abstract
Background Nivolumab, a human immunoglobulin IgG4 monoclonal antibody targeting PD-1 receptor, received initial FDA approval in 2014 for treating unresectable or metastatic malignant melanoma (MM), followed by approval for metastatic squamous and non-squamous non-small cell lung cancer (NSCLC) in 2015. With expanding clinical applications of nivolumab, comprehensive evaluation of its safety profile in real-world healthcare settings becomes increasingly crucial. Methods We compiled a real-world safety dataset of nivolumab from the FDA Adverse Event Reporting System (FAERS) database, encompassing reports from Q4-2014 through Q2 2024. To evaluate the association between nivolumab and adverse events (AEs), we employed four distinct disproportionality analysis methods: Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Multi-item Gamma Poisson Shrinker (MGPS) and Bayesian Confidence Propagation Neural Network (BCPNN). Additionally, we utilized Weibull distribution modeling to characterize the temporal risk patterns of identified adverse events. Results Our analysis identified 64,627 AEs reports associated with nivolumab. The most frequently reported AEs included fatigue, dyspnea, musculoskeletal pain, decreased appetite, cough, nausea, and constipation. Notably, we detected several potential safety signals not currently listed in the prescribing information: Malignant neoplasm progression, weight decreased, sepsis myocarditis, encephalitis and hypotension. Conclusions Our large-scale pharmacovigilance study identified significant safety signals associated with nivolumab, including previously unrecognized adverse drug reactions. The identification of these safety signals underscores the importance of ongoing post-marketing surveillance for immune checkpoint inhibitors. Future studies should investigate the mechanisms underlying these associations and develop targeted monitoring protocols.
Collapse
Affiliation(s)
- Yutong Wu
- Department of Neurology, Second Affiliated Hospital of Army Medical University, Chong Qing, China
| | - Yue Zhou
- Department of Neurology, Second Affiliated Hospital of Army Medical University, Chong Qing, China
| | - Shiyue Xia
- Basic Medical College, Army Medical University, Chong Qing, China
| | - Zhaoyou Meng
- Department of Neurology, Second Affiliated Hospital of Army Medical University, Chong Qing, China
| |
Collapse
|
28
|
Yau T, Galle PR, Decaens T, Sangro B, Qin S, da Fonseca LG, Karachiwala H, Blanc JF, Park JW, Gane E, Pinter M, Peña AM, Ikeda M, Tai D, Santoro A, Pizarro G, Chiu CF, Schenker M, He A, Chon HJ, Wojcik-Tomaszewska J, Verset G, Wang QQ, Stromko C, Neely J, Singh P, Jimenez Exposito MJ, Kudo M, CheckMate 9DW investigators. Nivolumab plus ipilimumab versus lenvatinib or sorafenib as first-line treatment for unresectable hepatocellular carcinoma (CheckMate 9DW): an open-label, randomised, phase 3 trial. Lancet 2025; 405:1851-1864. [PMID: 40349714 DOI: 10.1016/s0140-6736(25)00403-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Patients with unresectable hepatocellular carcinoma have a poor prognosis, and treatments with long-term benefits are needed. We report results from the preplanned interim analysis of the CheckMate 9DW trial assessing nivolumab plus ipilimumab versus lenvatinib or sorafenib for unresectable hepatocellular carcinoma in the first-line setting. METHODS This open-label, randomised, phase 3 trial enrolled patients aged 18 years or older with unresectable hepatocellular carcinoma without previous systemic therapy at 163 hospitals and cancer centres across 25 countries in Asia, Australia, Europe, North America, and South America. Patients had at least one measurable untreated lesion per Response Evaluation Criteria in Solid Tumours (RECIST) version 1.1, a Child-Pugh score of 5 or 6, and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned (1:1) via an interactive response technology system to receive nivolumab (1 mg/kg) plus ipilimumab (3 mg/kg) intravenously every 3 weeks for up to four doses, followed by nivolumab 480 mg every 4 weeks or investigator's choice of either oral lenvatinib (8 mg or 12 mg mg daily depending on bodyweight) or oral sorafenib (400 mg twice daily). Randomisation was stratified by aetiology; the presence of macrovascular invasion, extrahepatic spread, or both; and baseline alpha-fetoprotein concentration. The primary endpoint was overall survival, which was assessed in all randomly assigned patients; safety was an exploratory endpoint and was assessed in all randomly assigned patients who received at least one dose of study medication. This trial is registered with ClinicalTrials.gov, NCT04039607 (ongoing). FINDINGS Between Jan 6, 2020, and Nov 8, 2021, 668 patients were randomly assigned to nivolumab plus ipilimumab (n=335) or lenvatinib or sorafenib (n=333). Early crossing of the overall survival Kaplan-Meier curves reflected a higher number of deaths during the first 6 months after randomisation with nivolumab plus ipilimumab (hazard ratio 1·65 [95% CI 1·12-2·43]) but was followed by a sustained separation of the curves thereafter in favour of nivolumab plus ipilimumab (0·61 [0·48-0·77]). After a median follow-up of 35·2 months (IQR 31·1-39·9), overall survival was significantly improved with nivolumab plus ipilimumab versus lenvatinib or sorafenib (median 23·7 months [95% CI 18·8-29·4] vs 20·6 months [17·5-22·5]; hazard ratio 0·79 [0·65-0·96]; two-sided stratified log-rank p=0·018); respective overall survival rates were 49% (95% CI 44-55) versus 39% (34-45) at 24 months and 38% (32-43) versus 24% (19-30) at 36 months. Overall, 137 (41%) of 332 patients receiving nivolumab plus ipilimumab and 138 (42%) of 325 patients receiving lenvatinib or sorafenib had grade 3-4 treatment-related adverse events. 12 deaths were attributed to treatment with nivolumab plus ipilimumab and three were attributed to treatment with lenvatinib or sorafenib. INTERPRETATION Nivolumab plus ipilimumab showed a significant overall survival benefit versus lenvatinib or sorafenib and manageable safety in patients with previously untreated unresectable hepatocellular carcinoma. These results support nivolumab plus ipilimumab as a first-line treatment in this setting. FUNDING Bristol Myers Squibb.
Collapse
Affiliation(s)
- Thomas Yau
- Centre of Cancer Medicine and University Department of Medicine, The University of Hong Kong, Hong Kong
| | - Peter R Galle
- University Medical Center, I Medical Department, Mainz, Germany.
| | - Thomas Decaens
- University of Grenoble Alpes, CHU Grenoble Alpes, Institute for Advanced Biosciences, CNRS UMR 5309-INSERM U1209, Grenoble, France
| | - Bruno Sangro
- Clinica Universidad de Navarra and CIBEREHD, Pamplona-Madrid, Spain
| | - Shukui Qin
- Nanjing Tianyinshan Hospital of China Pharmaceutical University, Nanjing, China
| | - Leonardo G da Fonseca
- Instituto do Cancer do Estado de São Paulo, ICESP, University of São Paulo School of Medicine, São Paulo, Brazil
| | | | | | - Joong-Won Park
- National Cancer Center and Myongji Hospital, Goyang, South Korea
| | - Edward Gane
- University of Auckland, Auckland, New Zealand
| | | | - Ana Matilla Peña
- Hospital General Universitario Gregorio Marañón, CIBEREHD, Madrid, Spain
| | - Masafumi Ikeda
- National Cancer Center Hospital East, Kashiwa Chiba, Japan
| | | | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, and IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | | | | | - Aiwu He
- MedStar Georgetown University Hospital, Washington, DC, USA
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, South Korea
| | | | - Gontran Verset
- HUB-Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Qi Qi Wang
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Arya SP, Timilsina H, Thennakoon SKS, Postema RM, Jahan R, Reynolds AM, Tan X. A novel peptide targeting PD-1: implications for protein-protein interaction studies and immunotherapy. Chem Commun (Camb) 2025; 61:7680-7683. [PMID: 40304419 DOI: 10.1039/d5cc01198a] [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: 05/02/2025]
Abstract
Chemical ligand binding to PD-1 is a powerful approach for studying protein-protein interactions and advancing immunotherapy. In this study, we introduce a novel II peptide with strong binding affinity for PD-1. Molecular docking analysis reveals key interactions between the II peptide and PD-1, supporting inhibition ELISA data and indicating that the II peptide overlaps with the PD-1/PD-L1 interaction interface.
Collapse
Affiliation(s)
- Satya Prakash Arya
- Department of chemistry and Center for Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, USA.
| | - Hari Timilsina
- Department of chemistry and Center for Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, USA.
| | | | - Rick Mason Postema
- Department of chemistry and Center for Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, USA.
| | - Raunak Jahan
- Department of chemistry and Center for Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, USA.
| | - Andrew Michael Reynolds
- Department of chemistry and Center for Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, USA.
| | - Xiaohong Tan
- Department of chemistry and Center for Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, USA.
| |
Collapse
|
30
|
N V, B S, A T, M DB. Asymptomatic hypercalcemia, caused by sarcoid-like granulomatosis, induced by checkpoint inhibition. Acta Clin Belg 2025:1-7. [PMID: 40389384 DOI: 10.1080/17843286.2025.2506467] [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: 01/27/2025] [Accepted: 05/07/2025] [Indexed: 05/21/2025]
Abstract
OBJECTIVES To illustrate the diagnostic challenges, as well as the importance of early recognition of rare immunotherapy-induced complications, presenting a case and literature of sarcoid-like granulomatous reaction. METHODS This report presents a case of asymptomatic hypercalcemia, revealing a sarcoid-like granulomatosis in a patient with metastatic melanoma, treated with an immune checkpoint inhibitor (ICI). In the discussion, an overview of the existing literature is provided through a PubMed search. RESULTS Thorough investigations are essential to rule out disease progression and other possible explanations. Ultimately, biopsy with extensive staining led to the diagnosis of sarcoid-like granulomatosis. As there is no consensus in treatment, we suggest a case-by-case assessment, if possible by discussion within the multidisciplinary treatment team, to decide discontinuation of the causal ICI-therapy or the use of systemic steroids as supportive therapy. CONCLUSION This case demonstrates the importance of a broad differential diagnosis when identifying an asymptomatic hypercalcemia as well as new CT-graphic lesions, since the diagnosis of sarcoid-like granulomatosis can avoid not only unnecessary changes in treatment plans, avoiding toxicity, but also be a sign of good prognosis.
Collapse
Affiliation(s)
- Vandemaele N
- Department of Medical Oncology, Ghent University Hospital, Ghent, Belgium
| | - Stragier B
- Department of Medical Oncology, AZ Delta Roeselare, Roeselare, Belgium
| | - Tamsin A
- Department of Pathology, AZ Delta Roeselare, Roeselare, Belgium
| | - De Bock M
- Department of Medical Oncology, AZ Delta Roeselare, Roeselare, Belgium
| |
Collapse
|
31
|
Nardin C, Vernerey D, Aubin F. Ipilimumab + nivolumab in combination vs. nivolumab monotherapy as first-line treatment in patients with metastatic melanoma: conclusions from a real-world French national cohort. Br J Dermatol 2025; 192:971-972. [PMID: 40036378 DOI: 10.1093/bjd/ljaf080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Affiliation(s)
- Charlée Nardin
- Hôpital Jean Minjoz, Besançon, Bourgogne-Franche-Comté, France
| | - Dewi Vernerey
- Methodology and Quality of Life Unit in Oncology, Besançon University Hospital Center, Besançon, Bourgogne-Franche-Comté, France
| | - François Aubin
- Methodology and Quality of Life Unit in Oncology, Besançon University Hospital Center, Besançon, Bourgogne-Franche-Comté, France
| |
Collapse
|
32
|
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
|
33
|
Patel RP, Lim LRJ, Saleh R, Schenk D, Lee MK, Lelliott E, Rao AD, Arabi S, Smith L, Trigos AS, Haynes N, McArthur GA, Sheppard KE. Sensitivity to immune checkpoint inhibitors in BRAF/MEK inhibitor refractory melanoma. J Immunother Cancer 2025; 13:e011551. [PMID: 40379272 PMCID: PMC12083385 DOI: 10.1136/jitc-2025-011551] [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: 01/13/2025] [Accepted: 04/28/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Resistance to BRAF and MEK inhibitors (BRAFi/MEKi) in metastatic melanoma frequently results in cross-resistance to immune checkpoint inhibitors (ICI), limiting effective treatment options. However, a subset of BRAFi/MEKi-resistant patients remains responsive to second-line ICI, suggesting heterogeneous underlying resistance mechanisms. This study aimed to explore the tumor immune microenvironment in BRAFi/MEKi-resistant melanoma to uncover factors influencing sensitivity to second-line ICI therapy. METHOD To investigate mechanisms underlying resistance and responsiveness to second-line ICIs, BRAFi/MEKi-resistant melanoma mouse models were used. Flow cytometry was employed to analyze immune cell populations within the tumor microenvironment, focusing on changes in CD8+T effector cells and other key immune subsets. RNA sequencing was performed to profile transcriptomic changes in resistant tumors, providing insights into the signaling pathways associated with resistance. Clinical samples from BRAFi/MEKi-resistant patients were further evaluated for correlations between immune profiles and key signaling pathways to support findings from the preclinical models. RESULTS Using BRAFi/MEKi-resistant melanoma mouse models, we observed distinct alterations in the tumor-immune microenvironment. Tumors exhibiting resistance showed a significant increase in CD8+T effector cells following BRAFi/MEKi treatment, suggesting an immune-stimulatory response. Mechanistic analysis identified the activation of the EGFR-STAT signaling pathway as a key driver of intrinsic resistance in these models. Notably, these tumors retained sensitivity to second-line ICI therapy, contrasting with NRAS-driven BRAFi/MEKi-resistant tumors, which demonstrated cross-resistance to ICIs. Supporting these findings, clinical samples from BRAFi/MEKi-resistant patients revealed a correlation between elevated EGFR activation and higher immune scores, indicating potential sensitivity to ICI therapy in this subset of patients. CONCLUSION EGFR overexpression emerges as a potential predictive biomarker for responsiveness to second-line ICIs in BRAFi/MEKi-resistant melanoma. These findings underscore the need for stratified therapeutic approaches and highlight EGFR as a target for improving outcomes in ICI therapy.
Collapse
Affiliation(s)
- Riyaben P Patel
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Lydia Rui Jia Lim
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Reem Saleh
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Darius Schenk
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael K Lee
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Emily Lelliott
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- La Trobe University, Melbourne, Victoria, Australia
- The Walter and Eliza Hall Institute of Medical Research Melbourne, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Aparna D Rao
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Shaghayegh Arabi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Lorey Smith
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Anna S Trigos
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Nicole Haynes
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Grant A McArthur
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Karen E Sheppard
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
34
|
Baldassarre G, L de la Serna I, Vallette FM. Death-ision: the link between cellular resilience and cancer resistance to treatments. Mol Cancer 2025; 24:144. [PMID: 40375296 PMCID: PMC12080166 DOI: 10.1186/s12943-025-02339-1] [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: 02/18/2025] [Accepted: 04/22/2025] [Indexed: 05/18/2025] Open
Abstract
One of the key challenges in defeating advanced tumors is the ability of cancer cells to evade the selective pressure imposed by chemotherapy, targeted therapies, immunotherapy and cellular therapies. Both genetic and epigenetic alterations contribute to the development of resistance, allowing cancer cells to survive initially effective treatments. In this narration, we explore how genetic and epigenetic regulatory mechanisms influence the state of tumor cells and their responsiveness to different therapeutic strategies. We further propose that an altered balance between cell growth and cell death is a fundamental driver of drug resistance. Cell death programs exist in various forms, shaped by cell type, triggering factors, and microenvironmental conditions. These processes are governed by temporal and spatial constraints and appear to be more heterogeneous than previously understood. To capture the intricate interplay between death-inducing signals and survival mechanisms, we introduce the concept of Death-ision. This framework highlights the dynamic nature of cell death regulation, determining whether specific cancer cell clones evade or succumb to therapy. Building on this understanding offers promising strategies to counteract resistant clones and enhance therapeutic efficacy. For instance, combining DNMT inhibitors with immune checkpoint blockade may counteract YAP1-driven resistance or the use of transcriptional CDK inhibitors could prevent or overcome chemotherapy resistance. Death-ision aims to provide a deeper understanding of the diversity and evolution of cell death programs, not only at diagnosis but also throughout disease progression and treatment adaptation.
Collapse
Affiliation(s)
- Gustavo Baldassarre
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, 33081, Italy.
| | - Ivana L de la Serna
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA.
| | - François M Vallette
- Centre de Recherche en Cancérologie et Immunologie Intégrées Nantes Angers (CRCI2 NA), INSERM UMR1307/CNRS UMR 6075/Nantes Université/Univ. Angers. Nantes, 44007, Nantes, France.
- Institut de Cancérologie de L'Ouest (ICO), 44085, Saint-Herblain, France.
| |
Collapse
|
35
|
Kase S, Yamashita Y, Takeuchi S, Ishida S. Case Report: Laser speckle flowgraphy in a patient with uveitis due to immune-related adverse events by immune checkpoint inhibitors. Front Oncol 2025; 15:1492011. [PMID: 40444081 PMCID: PMC12119479 DOI: 10.3389/fonc.2025.1492011] [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: 09/06/2024] [Accepted: 04/21/2025] [Indexed: 06/02/2025] Open
Abstract
Purpose It remains unknown whether choroidal circulation could be altered at the onset of immune checkpoint inhibitor (ICI) uveitis compared with that before ICI treatment. Herein we report a patient with Vogt-Koyanagi-Harada (VKH) disease-like uveitis in the unaffected eye as an immune-related adverse effect (irAE) due to ICIs for metastatic choroidal melanoma who had received enucleation. Moreover, choroidal circulation and choroidal thickness were measured before and after treatment. Methods A 58-year-old man had a medical history of enucleation in his left eye due to choroidal melanoma 6 years ago. Metastatic lesions in the gastrointestinal tracts and lung were found, and then he received ICIs three times. About 1 month later, he suffered from blurred vision and metamorphopsia in his right eye. Choroidal circulation was evaluated by mean blur rate (MBR), a relative value showing choroidal blood velocity on laser speckle flowgraphy. Central choroidal thickness (CCT) was measured on optical coherence tomography. Results Since ophthalmic findings revealed VKH-like uveitis, oral prednisolone of 30 mg was given for 2 weeks, which were then tapered. MBR was reduced and CCT increased at the onset of ICI uveitis compared with its baseline and resolution after corticosteroid treatment. Conclusions Choroidal circulation was disrupted, possibly due to ICI-induced autoinflammatory reaction to the choroid, which was managed by corticosteroid treatment. The combination of MBR and CCT could be a useful biomarker for managing the patients with VKH-like uveitis by ICIs.
Collapse
Affiliation(s)
- Satoru Kase
- Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yui Yamashita
- Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Takeuchi
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Susumu Ishida
- Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
36
|
Bungaro C, Guida M, Apollonio B. Spatial proteomics of the tumor microenvironment in melanoma: current insights and future directions. Front Immunol 2025; 16:1568456. [PMID: 40443654 PMCID: PMC12119572 DOI: 10.3389/fimmu.2025.1568456] [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: 01/29/2025] [Accepted: 04/25/2025] [Indexed: 06/02/2025] Open
Abstract
Over the past years, cancer research has transitioned from a 'cancer cell-centered' focus to a more integrative view of tumors as dynamic ecosystems. This paradigm shift emphasizes the tumor microenvironment (TME) as a complex network of interacting cellular and acellular components, where tumor cells orchestrate a supportive environment that facilitates progression, metastasis, and immune evasion. Understanding the spatial organization of these components within the TME is crucial, as the positioning and interactions between cancerous and non-cancerous cells significantly influence tumor behavior and therapy response. Spatial proteomics has emerged as a powerful tool for TME analysis, enabling the detection and quantification of proteins within intact tissue architecture at subcellular resolution. This approach provides insights into cellular interactions, signaling pathways, and functional states, facilitating the discovery of novel biomarkers and therapeutic targets linked to specific tissue regions and cellular contexts. Translating spatial proteomics into clinical practice requires overcoming challenges related to technology refinement, standardization of workflows, and adaptation to routine pathology settings. Melanoma is an aggressive, highly immunogenic malignancy with variable response rates to existing immunotherapies. Given that over half of patients treated with immune checkpoint inhibitors (ICIs) fail to respond or experience disease progression, the identification of novel biomarkers and therapeutic targets to enhance current therapies is urgently required. Spatial imaging technologies are increasingly being utilized to dissect the complex interplay between stroma, melanoma, and immune cell types within the TME to address this need. This review examines key spatial proteomics methods, their applications in melanoma biology, and associated image analysis pipelines. We highlight the current limitations, and future directions, emphasizing the potential for clinical translation to guide personalized treatment strategies, inform prognosis, and predict therapeutic response.
Collapse
Affiliation(s)
| | | | - Benedetta Apollonio
- Rare Tumors and Melanoma Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| |
Collapse
|
37
|
Park J, Joung JG, Lim MC, Lee J, Kim BG, Kim JW, Shin SJ, Kim S, Park E, Choi CH, Kim HS, Park SY, Lee JY. Neoadjuvant Chemotherapy with Dual Immune Checkpoint Inhibitors for Advanced-Stage Ovarian Cancer: Final Analysis of TRU-D Phase II Nonrandomized Clinical Trial. Clin Cancer Res 2025; 31:1865-1876. [PMID: 40043003 DOI: 10.1158/1078-0432.ccr-24-3753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/12/2025] [Accepted: 03/03/2025] [Indexed: 05/16/2025]
Abstract
PURPOSE This open-label, investigator-initiated, phase II study was conducted to evaluate the safety, survival, and neoadjuvant outcomes of neoadjuvant chemotherapy (NAC) combined with dual immune checkpoint inhibitors in advanced-stage epithelial ovarian cancer (EOC). PATIENTS AND METHODS Between June 2019 and July 2021, 45 patients with unresectable stage III to IV EOC were enrolled. The patients received three cycles of NAC combined with durvalumab and tremelimumab. All patients underwent interval debulking surgery and received three cycles of durvalumab and adjuvant chemotherapy, followed by 12 cycles of durvalumab as maintenance therapy. The primary endpoint was the 12-month progression-free survival (PFS) rate; the secondary endpoints were the objective response rate after NAC, a chemotherapy response score, pathologic complete response, overall survival, and safety. The preplanned exploratory analyses assessed the lymphocyte infiltration, PD-L1 expression, and genomic profiles of pretreatment tumors. RESULTS The 12-month PFS rate was 65.9% [95% confidence interval (CI), 52.8-not estimated (NE)], whereas the 24- and 30-month PFS rates were 38.6% (95% CI, 26.7-NE) and 36.4% (95% CI, 24.7-NE), respectively. After NAC, the objective response rate was 86.7%, whereas 14 patients (31.1%) had a chemotherapy response score of three, and five (11.1%) achieved pathologic complete response. The 30-month overall survival rate was 87.7%. The most common grade ≥3 adverse event was neutropenia (26.7%). In an exploratory analysis, patients with pre-NAC tumors showing PD-L1 (combined positive score) ≥1, high Mutation Signature 3, and a high extracellular matrix signature demonstrated improved PFS outcomes. CONCLUSIONS NAC combined with dual immune checkpoint inhibitors is feasible for advanced-stage EOC and shows promising activity with a durable clinical response.
Collapse
Affiliation(s)
- Junsik Park
- Department of Obstetrics and Gynecology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, South Korea
| | - Je-Gun Joung
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Myong Cheol Lim
- Center for Gynecologic Cancer, National Cancer Center, Goyang, South Korea
| | - Jungbok Lee
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Byoung-Gie Kim
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| | - Jae-Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - So Jin Shin
- Department of Obstetrics and Gynecology, Keimyung University Dongsan Medical Center, Daegu, South Korea
| | - Sunghoon Kim
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunhyang Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Chel Hun Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| | - Hee Seung Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Yoon Park
- Center for Gynecologic Cancer, National Cancer Center, Goyang, South Korea
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
38
|
Huang R, Wang Y, Teng H, Xu M, He K, Shen Y, Guo G, Feng X, Li T, Zhou B, Bajenoff M, Lawrence T, Liang Y, Lu L, Zhang L. Tyrosinase in melanoma inhibits anti-tumor activity of PD-1 deficient T cells. BMC Biol 2025; 23:135. [PMID: 40375241 PMCID: PMC12083179 DOI: 10.1186/s12915-025-02237-4] [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: 08/23/2024] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Melanoma is one of the most commonly diagnosed malignancies and serves as a model for studying immunotherapy. The B16 melanoma model, resembling human cold tumors that lack T cell infiltration and show minimal response to PD-1 blockade, is widely used for studying melanoma and its resistance to immunotherapy. Therefore, understanding the molecular basis that prevents T cell-mediated anti-tumor activity in B16 melanoma is of great significance. RESULTS In this study, we generated tyrosinase knockout B16 melanoma cells using CRISPR/Cas9 and discovered that tyrosinase in melanoma significantly inhibits the anti-tumor activity of T cells. Tyrosinase deficiency leads to a 3.80-fold increase in T-cell infiltration and enhances T-cell activation within the tumor. Single-cell RNA sequencing reveals an altered cold tumor immunophenotype in tyrosinase-deficient B16 melanoma. In wild-type mice, T cells in tyrosinase-deficient tumors express elevated levels of PD-1 and Foxp3. However, strikingly, in PD-1 deficient mice, the loss of tyrosinase in B16 melanoma unleashes the anti-tumor activity of PD-1 deficient T cells. This enhanced anti-tumor activity is explained by significantly increased tumor T cell infiltration accompanied by reduced frequencies of regulatory T cells in PD-1 knockout mice. CONCLUSIONS These findings suggest that targeting tyrosinase could convert cold tumors into an immune-responsive state in vivo using murine models. Inhibiting tyrosinase could enhance the effectiveness of PD-1 blockade, offering a new approach for melanoma patients who fail in current PD-1 inhibitor treatment.
Collapse
Affiliation(s)
- Rong Huang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
| | - Yingbin Wang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Haitao Teng
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Mengjun Xu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Kexin He
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Yingzhuo Shen
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Guo Guo
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
- Centre d'immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, Marseille, France
| | - Xinyu Feng
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Tianhan Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Binhui Zhou
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Marc Bajenoff
- Centre d'immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, Marseille, France
| | - Toby Lawrence
- Centre d'immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, Marseille, France
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Yinming Liang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
- Center of Disease Model and Immunology, Hunan Academy of Chinese Medicine, Changsha, China.
| | - Liaoxun Lu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
| | - Lichen Zhang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
- Center of Disease Model and Immunology, Hunan Academy of Chinese Medicine, Changsha, China.
| |
Collapse
|
39
|
Nahon P. Establishing five-year overall survival as a new standard for trials in advanced HCC. J Hepatol 2025:S0168-8278(25)02199-3. [PMID: 40373978 DOI: 10.1016/j.jhep.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Affiliation(s)
- Pierre Nahon
- AP-HP, Hôpitaux Universitaires Paris Seine Saint-Denis, APHP, Liver Unit, Bobigny; Université Sorbonne Paris Nord, F-93000 Bobigny; Inserm, UMR-1138 "Functional Genomics of Solid Tumors", Centre de recherche des Cordeliers, Université de Paris, Paris.
| |
Collapse
|
40
|
Borys K, Lodde G, Livingstone E, Weishaupt C, Römer C, Künnemann MD, Helfen A, Zimmer L, Galetzka W, Haubold J, Friedrich CM, Umutlu L, Heindel W, Schadendorf D, Hosch R, Nensa F. Fully volumetric body composition analysis for prognostic overall survival stratification in melanoma patients. J Transl Med 2025; 23:532. [PMID: 40355935 PMCID: PMC12067685 DOI: 10.1186/s12967-025-06507-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Accurate assessment of expected survival in melanoma patients is crucial for treatment decisions. This study explores deep learning-based body composition analysis to predict overall survival (OS) using baseline Computed Tomography (CT) scans and identify fully volumetric, prognostic body composition features. METHODS A deep learning network segmented baseline abdomen and thorax CTs from a cohort of 495 patients. The Sarcopenia Index (SI), Myosteatosis Fat Index (MFI), and Visceral Fat Index (VFI) were derived and statistically assessed for prognosticating OS. External validation was performed with 428 patients. RESULTS SI was significantly associated with OS on both CT regions: abdomen (P ≤ 0.0001, HR: 0.36) and thorax (P ≤ 0.0001, HR: 0.27), with lower SI associated with prolonged survival. MFI was also associated with OS on abdomen (P ≤ 0.0001, HR: 1.16) and thorax CTs (P ≤ 0.0001, HR: 1.08), where higher MFI was linked to worse outcomes. Lastly, VFI was associated with OS on abdomen CTs (P ≤ 0.001, HR: 1.90), with higher VFI linked to poor outcomes. External validation replicated these results. CONCLUSIONS SI, MFI, and VFI showed substantial potential as prognostic factors for OS in malignant melanoma patients. This approach leveraged existing CT scans without additional procedural or financial burdens, highlighting the seamless integration of DL-based body composition analysis into standard oncologic staging routines.
Collapse
Affiliation(s)
- Katarzyna Borys
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Girardetstraße 2, 245131, Essen, Germany.
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany.
| | - Georg Lodde
- Institute of Dermatology, University Hospital Essen, Essen, Germany
| | | | - Carsten Weishaupt
- Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Christian Römer
- Clinic for Radiology, University Hospital Münster, Münster, Germany
| | | | - Anne Helfen
- Clinic for Radiology, University Hospital Münster, Münster, Germany
| | - Lisa Zimmer
- Institute of Dermatology, University Hospital Essen, Essen, Germany
| | - Wolfgang Galetzka
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Johannes Haubold
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Girardetstraße 2, 245131, Essen, Germany
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Christoph M Friedrich
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Computer Science, University of Applied Sciences and Arts Dortmund, Dortmund, Germany
| | - Lale Umutlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Walter Heindel
- Clinic for Radiology, University Hospital Münster, Münster, Germany
| | - Dirk Schadendorf
- Institute of Dermatology, University Hospital Essen, Essen, Germany
| | - René Hosch
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Girardetstraße 2, 245131, Essen, Germany
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Felix Nensa
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Girardetstraße 2, 245131, Essen, Germany
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| |
Collapse
|
41
|
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
|
42
|
Lu Q, Li J, Chen W, Wang Z, Wang D, Liu C, Sun Y, Jiang H, Zhang C, Chang Y, Zhou J, Wu X, Gao Y, Ning S. NetLnc: A Network-Based Computational Framework to Identify Immune Checkpoint-Related lncRNAs for Immunotherapy Response in Melanoma. Int J Mol Sci 2025; 26:4557. [PMID: 40429702 PMCID: PMC12110832 DOI: 10.3390/ijms26104557] [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: 03/17/2025] [Revised: 04/27/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) could alter the tumor immune microenvironment and regulate the expression of immune checkpoints (ICPs) by regulating target genes in tumors. However, only a few lncRNAs have precise functions in immunity and potential for predicting ICP inhibitors (ICI) response. Here, we developed a computational multi-step framework that leverages interaction network-based analysis to identify cancer- and immune-context ICP-related lncRNAs (NetLnc). Based on bulk and single-cell RNA sequencing data, these lncRNAs were significantly correlated with immune cell infiltration and immune expression signature. Specific hub ICP-related lncRNAs such as BANCR, MIAT, and SNHG15 could predict three- and five-year prognosis of melanoma in independent datasets. We also validated that some NetLnc-based predictions could better effectively predict ICI response compared to single molecules using three kinds of machine learning algorithms following independent datasets. Taken together, this study presents the use of a network-based framework to efficiently select ICP-related lncRNAs, which contributes to a comprehensive understanding of lncRNA functions and accelerates the discovery of lncRNA-based biomarkers in ICI treatment.
Collapse
Affiliation(s)
- Qianyi Lu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Jian Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Wenli Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Zhuoru Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Di Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Chenyu Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yue Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Han Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Caiyu Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yetong Chang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Jiajun Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Xiaohong Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin 150081, China;
| | - Yue Gao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
43
|
Wang Q, Yuan F, Zuo X, Li M. Breakthroughs and challenges of organoid models for assessing cancer immunotherapy: a cutting-edge tool for advancing personalised treatments. Cell Death Discov 2025; 11:222. [PMID: 40335487 PMCID: PMC12059183 DOI: 10.1038/s41420-025-02505-w] [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: 11/08/2024] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Organoid models are powerful tools for evaluating cancer immunotherapy that provide a more accurate representation of the tumour microenvironment (TME) and immune responses than traditional models. This review focuses on the latest advancements in organoid technologies, including immune cell co-culture, 3D bioprinting, and microfluidic systems, which enhance the modelling of TME and facilitate the assessment of immune therapies such as immune checkpoint inhibitors (ICIs), CAR-T therapies, and oncolytic viruses. Although these models have great potential in personalised cancer treatment, challenges persist in immune cell diversity, long-term culture stability, and reproducibility. Future developments integrating artificial intelligence (AI), multi-omics, and high-throughput platforms are expected to improve the predictive power of organoid models and accelerate the clinical translation of immunotherapy.
Collapse
Affiliation(s)
- Qian Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210009, Jiangsu, PR China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, 210009, Jiangsu, PR China
| | - Fangwei Yuan
- Department of Thoracic Surgery, Lian Shui County People's Hospital, Huaian, 223400, Jiangsu, PR China
| | - Xianglin Zuo
- Biobank of Jiangsu Cancer Hospital (Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University), Nanjing, 210000, Jiangsu, PR China.
| | - Ming Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210009, Jiangsu, PR China.
- The Fourth Clinical College of Nanjing Medical University, Nanjing, 210009, Jiangsu, PR China.
| |
Collapse
|
44
|
Chu X, Pu N, Yang X, Xie Y, Liu L, Jin Y. Subtypes of tumor-associated neutrophils and their roles in cancer immunotherapy. Crit Rev Oncol Hematol 2025; 212:104763. [PMID: 40334802 DOI: 10.1016/j.critrevonc.2025.104763] [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: 03/10/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025] Open
Abstract
Neutrophils are essential components of the innate immune system. Tumor-associated neutrophils (TANs) are shaped by tumor microenvironment (TME), leading to significant heterogeneity in biological characteristics and functions. Recent advances in single-cell sequencing have revealed a wide array of TAN subtypes, while a comprehensive classification system is still lacking. This review aims to summarize the alterations observed in TAN subgroups following cancer immunotherapy, and identify the distinctions and commonalities between pro-tumor and anti-tumor subgroups. Current progress of preclinical and clinical studies is also highlighted, involving novel therapies targeting TANs.
Collapse
Affiliation(s)
- Xinyun Chu
- Department of Hepatobiliary & Pancreatic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650000, China; Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xue Yang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuqi Xie
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Yun Jin
- Department of Hepatobiliary & Pancreatic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650000, China.
| |
Collapse
|
45
|
Reinhard S, Utikal JS, Zaremba A, Lodde G, von Wasielewski I, Klespe KC, Meier F, Haferkamp S, Kähler KC, Herbst R, Gebhardt C, Sindrilaru A, Dippel E, Angela Y, Mohr P, Pfoehler C, Forschner A, Kaatz M, Schell B, Gesierich A, Loquai C, Hassel JC, Ulrich J, Meiss F, Schley G, Heinzerling LM, Sachse M, Welzel J, Weishaupt C, Sunderkötter C, Michl C, Lindhof HH, Kreuter A, Heppt MV, Wenk S, Mauch C, Berking C, Nedwed AS, Gutzmer R, Leiter U, Schadendorf D, Ugurel S, Weichenthal M, Haist M, Fleischer MI, Lang B, Grabbe S, Stege H. First-line checkpoint inhibitor therapy in metastatic acral lentiginous melanoma compared to other types of cutaneous melanoma: A multicenter study from the prospective skin cancer registry ADOREG. Eur J Cancer 2025; 220:115356. [PMID: 40121837 DOI: 10.1016/j.ejca.2025.115356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Melanoma is the main cause of skin cancer-related death. Treatment with immune checkpoint inhibitors (CPI) has improved the prognosis in recent years. However, subtypes of melanoma differ in their response. Acral lentiginous melanoma (ALM) has a worse prognosis compared to cutaneous melanoma other than ALM (CM) and is therefore of particular relevance. AIMS To evaluate the efficacy of CPI in first-line treatment of patients with advanced ALM compared CM. METHODS Retrospective analysis of patients with metastatic ALM (n = 45) or CM (n = 328) who received first-line CPI therapy from the multicenter prospective skin cancer registry ADOREG. Study endpoints were best overall response (BOR), progression-free survival (PFS) and overall survival (OS). RESULTS ALM patients had significantly higher rates of ulcerated tumors, loco regional metastases and fewer BRAF-mutated tumors compared to CM patients. Combined CPI was administered in 48.9 % ALM patients and 39.3 % of CM patients, while the remaining patients received PD-1 monotherapy. OS trended to be shorter in patients with ALM (18.1 vs. 43.8 months, p = 0.10) with no significant differences in PFS (7.0 vs. 11.5 months, p = 0.21). In patients with CM, median OS with combined CPI was not reached, whereas the median OS after PD-1 monotherapy was 37.8 months (p = 0.22). Conversely, in patients with ALM, OS with combined CPI was 17.8 months, compared to 26 months with PD-1 monotherapy (p = 0.15). There were no significant differences in BOR between patients with ALM or CM. CONCLUSION Analysis of this real-world cohort of patients with metastatic melanoma showed a trend towards poorer survival outcomes upon first-line treatment with CPI in ALM compared to cutaneous melanoma of other subtypes.
Collapse
Affiliation(s)
- Sören Reinhard
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | - Jochen Sven Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Anne Zaremba
- Department of Dermatology, Venereology and Allergology, University Hospital Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Georg Lodde
- Department of Dermatology, Venereology and Allergology, University Hospital Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Imke von Wasielewski
- Skin Cancer Center Hannover, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Kai Christian Klespe
- Skin Cancer Center Hannover, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Katharina C Kähler
- Department of Dermatology, Skin Cancer Center, University Hospital Schleswig-Holstein - Campus Kiel, Kiel, Germany
| | - Rudolf Herbst
- Department of Dermatology, HELIOS Hospital Erfurt, Erfurt, Germany
| | - Christoffer Gebhardt
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anca Sindrilaru
- Department of Dermatology, University Hospital of Ulm, Ulm, Germany
| | - Edgar Dippel
- Department of Dermatology, Ludwigshafen City Hospital, Ludwigshafen, Germany
| | - Yenny Angela
- Department of Dermatology, Muehlenkreiskliniken Minden and Ruhr University Bochum, Minden, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe Kliniken Buxtehude, Buxtehude, Germany
| | - Claudia Pfoehler
- Department of Dermatology, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Andrea Forschner
- Center for Dermatooncology, Department of Dermatology, Eberhard-Karls University of Tübingen, Tubingen, Germany
| | - Martin Kaatz
- Department of Dermatology, DRK Hospital Chemnitz-Rabenstein, Rabenstein, Germany
| | - Beatrice Schell
- Department of Dermatology, Wald-Klinikum Gera, Gera 07546, Germany
| | - Anja Gesierich
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Carmen Loquai
- Department of Dermatology, Klinikum Bremen-Ost, Gesundheit Nord gGmbH, Bremen, 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
| | - Jens Ulrich
- Department of Dermatology and Allergy, Harzklinikum Dorothea Christiane Erxleben GmbH, Quedlinburg, Germany
| | - Frank Meiss
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gaston Schley
- Department of Dermatology, HELIOS Hospital Schwerin, Schwerin, Germany
| | - Lucie M Heinzerling
- Department of Dermatology and Allergy, University Hospital, Ludwig-Maximilian Universität Munich, München, Germany
| | - Michael Sachse
- Department of Dermatology, Hospital Bremerhaven Reinkenheide, Bremerhaven, Germany
| | - Julia Welzel
- Department of Dermatology, University Hospital Augsburg, Augsburg, Germany
| | - Carsten Weishaupt
- Department of Dermatology, University Hospital of Muenster, Muenster, Germany
| | - Cord Sunderkötter
- Department of Dermatology and Venereology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Christiane Michl
- Department of Dermatology and Venereology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | | | - Alexander Kreuter
- Department of Dermatology, Venerology and Allergology, Helios St. Elisabeth Klinik Oberhausen, University Witten-Herdecke, Oberhausen, Germany
| | - Markus V Heppt
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN) and CCC Alliance WERA, Bavarian Cancer Research Center (BZKF), Erlangen 91052, Germany
| | - Saskia Wenk
- Department of Dermatology, Medical Center Klinikum Darmstadt, Teaching Hospital Goethe-University Frankfurt, Darmstadt, Germany
| | - Cornelia Mauch
- Department of Dermatology, Ruhr-Universität Bochum, Bochum, Germany
| | - Carola Berking
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN) and CCC Alliance WERA, Bavarian Cancer Research Center (BZKF), Erlangen 91052, Germany
| | - Annekathrin Silvia Nedwed
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center Mainz, Mainz, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Muehlenkreiskliniken Minden and Ruhr University Bochum, Minden, Germany
| | - Ulrike Leiter
- Center for Dermatooncology, Department of Dermatology, Eberhard-Karls University of Tübingen, Tubingen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Michael Weichenthal
- Department of Dermatology, Skin Cancer Center, University Hospital Schleswig-Holstein - Campus Kiel, Kiel, Germany
| | - Maximilian Haist
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | | | - Berenice Lang
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | - Henner Stege
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany.
| |
Collapse
|
46
|
Ata S, Köşeci T, Benli BA, Bayhan AZ, Kesen O, Solmaz AA, Demir H, Çil T, Bozkurt Duman B. The prognostic value of halp score in predicting the efficacy of nivolumab treatment in metastatic malignant melanoma patients: A real-life, retrospective, single center analysis. Medicine (Baltimore) 2025; 104:e42261. [PMID: 40324274 PMCID: PMC12055156 DOI: 10.1097/md.0000000000042261] [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: 07/29/2024] [Revised: 12/18/2024] [Accepted: 04/10/2025] [Indexed: 05/07/2025] Open
Abstract
Patients with metastatic malignant melanoma have a survival rate of less than one year. Nivolumab, a monoclonal antibody against programmed cell death 1 (PD-1) receptor, has improved survival in patients without BRAF mutations. The HALP score, calculated from hemoglobin, albumin, lymphocyte, and platelet levels, provides information about a patient immune and nutritional status. High HALP scores have been associated with a better prognosis in various cancers. This study aimed to investigate the effect of high HALP scores on response to nivolumab treatment in patients with metastatic malignant melanoma. A retrospective study was conducted on 44 patients with metastatic malignant melanoma treated with nivolumab at Adana City Training and Research Hospital between 2014 and 2021. Patients who received dabrafenib-trametinib before nivolumab treatment were excluded. The HALP scores were calculated using laboratory parameters before the first nivolumab treatment. Statistical analyses were performed using SPSS version 25.0. The study included 22 female and 22 male patients with a mean age of 61.4 ± 15.6 years. Of the patients, 10 (27.2%) had a positive BRAF mutation, whereas 34 (77.3%) did not. The HALP score cutoff value was determined as 30.1. Patients with high HALP scores had significantly longer progression-free survival (PFS) and overall survival (OS) compared to those with low HALP scores (PFS: median 5.8 vs 3.1 months, P = .041; OS: median 54.9 vs 14.4 months, P = .005). In this study, we found that high HALP scores were significantly associated with longer PFS and OS in metastatic malignant melanoma patients receiving nivolumab treatment. HALP score was associated with both PFS and OS in patients with metastatic malignant melanoma treated with nivolumab. This immuno-nutritional parameter may be useful in various cancers; however, further prospective studies with larger patient cohorts are needed for clinical application.
Collapse
Affiliation(s)
- Serdar Ata
- Department of Medical Oncology, University of Health Sciences, Adana City Training and Research Hospital, Adana, Turkey
| | - Tolga Köşeci
- Department of Medical Oncology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Burcu Arslan Benli
- Department of Medical Oncology, University of Health Sciences, Adana City Training and Research Hospital, Adana, Turkey
| | - Ahmet Ziya Bayhan
- Department of Medical Oncology, University of Health Sciences, Adana City Training and Research Hospital, Adana, Turkey
| | - Oğuzhan Kesen
- Department of Medical Oncology, University of Health Sciences, Adana City Training and Research Hospital, Adana, Turkey
| | - Ali Alper Solmaz
- Department of Medical Oncology, University of Health Sciences, Adana City Training and Research Hospital, Adana, Turkey
| | - Hakan Demir
- Department of Medical Oncology, University of Health Sciences, Adana City Training and Research Hospital, Adana, Turkey
| | - Timuçin Çil
- Department of Medical Oncology, University of Health Sciences, Adana City Training and Research Hospital, Adana, Turkey
| | - Berna Bozkurt Duman
- Department of Medical Oncology, University of Health Sciences, Adana City Training and Research Hospital, Adana, Turkey
| |
Collapse
|
47
|
Jansen P, Galetzka W, Thielmann CM, Murali R, Zaremba A, Standl F, Lodde G, Möller I, Sucker A, Paschen A, Hadaschik E, Ugurel S, Zimmer L, Livingstone E, Schadendorf D, Stang A, Griewank KG. pTERT mutational status is associated with survival in stage IV melanoma patients receiving first-line immune therapy. Eur J Cancer 2025; 220:115337. [PMID: 40056560 DOI: 10.1016/j.ejca.2025.115337] [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: 12/19/2024] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND TERT promoter mutations are the most prevalent mutations in melanoma. In this study, we investigated clinical characteristics and survival after first line therapies in a cohort of melanoma patients with known TERT promoter (pTERT) mutation status. METHODS Sequencing data from 2013 to 2021 covering 29 genes and the pTERT status was assessed and 774 melanomas patients with known pTERT status and clinical data were analyzed. Progression free survival (PFS) and overall survival (OS) of 374 melanoma patients in AJCC-stage IV who received first-line immune checkpoint inhibitors (ICI, anti-CTLA4 /anti-PD1 combination therapy or anti-PD1 monotherapy) or targeted therapy (TT) were assessed applying Cox uni-/ multivariable analyses and Kaplan-Meier curves. RESULTS The cohort included 573 cutaneous, 69 mucosal, 37 acral and 95 MUP (melanomas of unknown primary) melanoma patients with a median observational time from first diagnosis to patient death or censoring of 38.5 months. TERT promoter mutations were identified in 476 melanomas (61.5 %). Survival analysis of 374 patients with stage IV disease undergoing first-line systemic therapy (ICI or TT) suggested prolonged PFS and OS for patients with pTERT mutation positive tumors (pTERT(+)). Particularly, pTERT(+) patients receiving anti-CTLA4/anti-PD1 therapy showed mPFS of 14.8 months (95 % CI: 7.1-40.3) and mOS of 105.2 months (95 % CI: 27.6-not reached) compared to pTERT(-) patients with mPFS of 5.5 months (95 % CI: 2.7-10.0) and mOS of 14.7 months (95 % CI: 11.7-24.1). CONCLUSIONS Our findings suggest that presence of a pTERT mutation in melanomas might favor PFS and OS after first line ICI with the greatest improvement after receiving anti-CTLA4 / anti-PD1. If validated in larger prospective studies, pTERT mutation status may be a valuable prognostic marker for stage IV melanoma patients.
Collapse
Affiliation(s)
- Philipp Jansen
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Wolfgang Galetzka
- Institute for medical informatics, biometry and epidemiology, University Hospital Essen, Essen, Germany
| | - Carl M Thielmann
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Rajmohan Murali
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Anne Zaremba
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Fabian Standl
- Graduate Center of Medicine and Health, Technical University Munich, Munich, Germany
| | - Georg Lodde
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Inga Möller
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Eva Hadaschik
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Andreas Stang
- Institute for medical informatics, biometry and epidemiology, University Hospital Essen, Essen, Germany
| | - Klaus G Griewank
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany.
| |
Collapse
|
48
|
Weichenthal M, Ellebaek E, Mangana J, Asher N, Gavrilova I, Kandolf L, Ugurel S, Hausschild A, Meier F, Leiter U, Livingstone E, Gebhardt C, Gutzmer R, Ruhlmann CH, Mahncke-Guldbrandt L, Haslund CA, Kopec S, Teterycz P, Bender M, Poudroux W, Muñoz-Couselo E, Berciano-Guerrero MA, Shalamanova G, DePalo DK, Brozić JM, Chiarion-Sileni V, Arance A, Ziogas D, Robert C, van de Velde AO, Gassama AA, Shapira R, BenBetzalel G, Grynberg S, Ramelyte E, Bertoldo F, DelPrete V, Gaudy-Marqueste C, Mohr P, Dummer R, Ascierto PA, Gogas H, Espinosa E, Lebbé C, Rutkowski P, Haanen J, Schadendorf D, Svane IM. Immune checkpoint inhibition in metastatic or non-resectable melanoma after failure of adjuvant anti-PD-1 treatment. A EUMelaReg real-world evidence study. Eur J Cancer 2025; 220:115339. [PMID: 40090216 DOI: 10.1016/j.ejca.2025.115339] [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: 01/22/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Adjuvant immune checkpoint inhibition (ICI) with anti-PD-1 antibodies in high-risk resected melanoma has been shown to improve recurrence-free survival. It is unclear whether prior adjuvant anti-PD-1 therapy is associated with altered response to subsequent ICI treatment in the metastatic setting. METHODS Using data from the European Melanoma Registry (EUMelaReg), we analyzed the efficiency of first-line (1L) ICI in non-resectable or metastatic melanoma after failure from prior adjuvant anti-PD-1 treatment. Both single-agent anti-PD-1 and combined anti-PD-1/CTLA-4 (Ipi/Nivo) 1L regimes were included in the analysis. We identified 389 patients receiving 1L ICI with prior adjuvant anti-PD-1 treatment. The control population was selected from a pool of 3390 PD-1-naive cases by 1:1 matching for the type of 1L ICI and various prognostic factors. As outcome measure, overall remission rates (ORR) were calculated and progression-free survival (PFS) was evaluated by Kaplan-Meier and Cox regression analysis. RESULTS Out of 389 patients, 303 (77.9 %) received Ipi/Nivo and 86 (22.1 %) anti-PD-1 in 1L. ORR was significantly lower in pre-treated patients (31.4 %) as compared to anti-PD-1 naive patients (48.8 %; p < 0.0001). Kaplan-Meier analysis showed significantly shorter median PFS for pre-treated patients. This applied to both anti-PD-1 and Ipi/Nivo treatment. Patients with early recurrence from adjuvant treatment (during or up to 12 weeks after end of treatment) showed lower ORR (28.5 %) and shorter PFS (3.1 months) than those who recurred later (37.7 % and 6.1 months, respectively). CONCLUSIONS Patients with metastatic melanoma, previously exposed to anti-PD-1 ICI in the adjuvant setting showed significantly lower ORR and shorter PFS to 1L ICI with either Ipi/Nivo or single-agent anti-PD-1 retreatment.
Collapse
Affiliation(s)
- Michael Weichenthal
- Skin Cancer Center Kiel, University Hospital Schleswig-Holstein, Kiel, Germany.
| | - Eva Ellebaek
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Nethanel Asher
- Skin Cancer and Melanoma Center at Davidoff Cancer Center, Rabin Medical Center, Israel
| | - Iva Gavrilova
- Oncodermatology Department, National Oncology Center, Sofia, Bulgaria
| | - Lidija Kandolf
- Department of Dermatology, Faculty of Medicine, Military Medical Academy, Belgrade, Serbia
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany
| | - Axel Hausschild
- Skin Cancer Center Kiel, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Leiter
- Universitäts-Hautklinik Tübingen, Tübingen, Germany.
| | - Elisabeth Livingstone
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Skin Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Johannes Wesling Medical Center, Ruhr University Bochum Campus Minden, Minden, Germany
| | | | | | | | - Sylwia Kopec
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Paweł Teterycz
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Department of Computational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Marc Bender
- Skin Cancer Center, Division of Molecular Cell Biology, Elbe Kliniken Stade-Buxtehude, Buxtehude, Germany
| | | | - Eva Muñoz-Couselo
- Medical oncology department, Vall d'Hebron Hospital, Barcelona-Spain and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - Miguel-Angel Berciano-Guerrero
- Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria University Hospitals, IBIMA Plataforma BIONAND, Málaga, Spain
| | | | - Danielle K DePalo
- Department of Cutaneous Oncology, Surgical Oncology Research Fellow, Moffitt Cancer Center, Tampa, FL, United States
| | - Jasmina Marić Brozić
- Department of Oncology, UHC Sestre milosrdnice, School of Medicine Zagreb, Zagreb, Croatia
| | | | - Anna Arance
- Department of Medical Oncology and IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Dimitrios Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Caroline Robert
- Gustave Roussy Cancer Campus and Université Paris Saclay, Villejuif, France
| | | | - Awa Aminata Gassama
- Scientific Administration, Biometrics Department, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ronnie Shapira
- The Ella Lemelbaum Institute for Immuno-Oncology and Melanoma, Ramat-Gan, Israel
| | - Guy BenBetzalel
- The Ella Lemelbaum Institute for Immuno-Oncology and Melanoma, Ramat-Gan, Israel
| | - Shirly Grynberg
- The Ella Lemelbaum Institute for Immuno-Oncology and Melanoma, Ramat-Gan, Israel
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.
| | - Fabio Bertoldo
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Valerio DelPrete
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Caroline Gaudy-Marqueste
- Aix-Marseille Univ, APHM, Hôpital Timone, Dermatology and Skin Cancer Department. Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, Marseilles, France
| | - Peter Mohr
- Department of Dermatology, Elbekliniken Buxtehude, Buxtehude, Germany
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Paolo A Ascierto
- Department of Skin Cancers, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Enrique Espinosa
- Service of Oncology, Hospital U. La Paz, U. Autónoma de Madrid, Madrid-CIBERONC, Spain
| | - Celeste Lebbé
- Université Paris Cite, AP-HP Dermato-oncology and CIC, Cancer Institute APHP, INSERM U976, Saint Louis Hospital, Paris, France.
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - John Haanen
- Netherlands Cancer Institut Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Dirk Schadendorf
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
49
|
Prod'homme C, Sena N, Forestier E, Le Berre R, Desmedt E, Mortier L, Touzet L. Patient views on continued immune checkpoint inhibition following progression in advanced melanoma: A qualitative study. Bull Cancer 2025; 112:469-477. [PMID: 39800639 DOI: 10.1016/j.bulcan.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Immune checkpoint inhibition has revolutionized the management of metastatic melanoma, including in the final stages of disease progression: because it is well tolerated, some teams do not discontinue it in hopes of slowing disease progression. The risks are that treatment may be continued unnecessarily, causing side effects, and reduce access to specialist palliative care, in addition to increasing the cost of treatment. METHOD We explored the experiences of 10 patients in a university hospital with metastatic melanoma under continued immune checkpoint inhibitors combined with specialist palliative care. Our goal was to gain a better understanding of the advantages and disadvantages perceived by patients. The comprehensive interviews were analysed using a method inspired by grounded theory that met the COREQ international recommendation criteria. RESULTS Receiving the information of disease progression, continued treatment and onset of palliative care impacts patients' lives: from this point onwards, death becomes a reality and takes on the image of a sword of Damocles. The experience is anxiety-provoking because of the uncertainty of tomorrow, and painful because of the physical suffering and successive bereavements. However, far from causing depression and despair, joint oncology-palliative management is well accepted and helps to improve patients' daily lives and well-being. CONCLUSION Our findings highlight patients' ambivalence. On the one hand, they recount their experiences of suffering in connection with the disease and the confrontation with death, and on the other hand, their need to continue to live and to hope. Joint care provided by oncology and palliative care teams, symbolically representing hope and death, may mirror patients' psychological mindset and provide just the support they need.
Collapse
Affiliation(s)
- Chloé Prod'homme
- Palliative Care Unit, ULR 2694 METRICS, CHU de Lille, Université de Lille, 59000 Lille, France.
| | - Nicolas Sena
- Palliative Care Unit, ULR 2694 METRICS, CHU de Lille, Université de Lille, 59000 Lille, France
| | - Emmanuelle Forestier
- Palliative Care Unit, ULR 2694 METRICS, CHU de Lille, Université de Lille, 59000 Lille, France
| | - Rozenn Le Berre
- Experiment, Transhumanism, Human Interactions, Care and Society (ETHICS) - EA 7446, Lille Catholic University, Lille, France
| | - Eve Desmedt
- Department of Dermatology, CHU de Lille, Université de Lille, 59000 Lille, France
| | - Laurent Mortier
- Department of Dermatology, CHU de Lille, Université de Lille, 59000 Lille, France
| | - Licia Touzet
- Palliative Care Unit, ULR 2694 METRICS, CHU de Lille, Université de Lille, 59000 Lille, France
| |
Collapse
|
50
|
Alghabban A, Corke L, Katzberg H, Bril V, Barnett-Tapia C, Mason W, Alothman R, Albiruni Ryan AR, Hogg D, Sridhar S, Dhani N, Spreafico A, Eng L, Sacher A, Bradbury P, Liu G, Leighl N, Shepherd FA. Myasthenia Gravis in Patients Treated With Immune Checkpoint Inhibitors. JTO Clin Res Rep 2025; 6:100772. [PMID: 40242666 PMCID: PMC12002810 DOI: 10.1016/j.jtocrr.2024.100772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/07/2024] [Accepted: 11/16/2024] [Indexed: 04/18/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have improved outcomes significantly for patients across multiple tumor types, and now are being used in combination with other therapies and in earlier settings where treatment intent is curative. Immune-related adverse events occur commonly and there are clear guidelines regarding management. Neurological toxicities such as myasthenia gravis (MG) with or without myositis are rare but are associated with high morbidity and mortality. Methods This single-centre study presents a series of patients treated with ICIs who subsequently developed immune-related MG. Presenting symptoms, treatments and outcomes were abstracted from retrospective chart review. Results We identified 16 patients (9 thoracic malignancies, 7 other tumor sites) who were diagnosed with MG after one or more cycles of ICI. Eleven had overlapping myositis. The median time from the first ICI treatment to the onset of symptoms was 49 days (range 17-361). All patients received steroids (prednisone 1-2 mg/kg); six required other immunosuppressive agents, and five underwent plasma exchange. Only two patients had complete resolution, eight improved with residual symptoms, two experienced initial improvement followed by deterioration, and four worsened despite treatment. Six patients died as a result of myasthenia-related complications (38%), three from progressive cancer (19%) and seven remain alive at the time of review (44%). Conclusion ICI-related MG is a rare and potentially fatal adverse event. Diagnosis and management remain a challenge, especially with negative serological markers and in the presence of overlapping syndromes with high mortality rates. Prompt recognition and multimodality treatment are key. Clinicians should have a low threshold for diagnosis and early management.
Collapse
Affiliation(s)
- Abdulrahman Alghabban
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Lucy Corke
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Hans Katzberg
- Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Vera Bril
- Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Carolina Barnett-Tapia
- Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Warren Mason
- Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Raed Alothman
- Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Abdul Razak Albiruni Ryan
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - David Hogg
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Srikala Sridhar
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Neesha Dhani
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Anna Spreafico
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Lawson Eng
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Penelope Bradbury
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Natasha Leighl
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Frances A. Shepherd
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|