1
|
Song Q, Yu Z, Lu W, Zhuo Z, Chang L, Mei H, Cui Y, Zhang D. PD-1/PD-L1 inhibitors related adverse events: A bibliometric analysis from 2014 to 2024. Hum Vaccin Immunother 2025; 21:2424611. [PMID: 39757956 DOI: 10.1080/21645515.2024.2424611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/14/2024] [Accepted: 10/29/2024] [Indexed: 01/07/2025] Open
Abstract
Programmed cell death-1 (PD-1) inhibitors and programmed cell death ligand 1 (PD-L1) inhibitors are considered effective alternatives for the primary treatment of recurrent metastatic cancers. However, they can induce various adverse events affecting multiple organ systems, potentially diminishing patients' quality of life, and even leading to treatment interruptions. Adverse events related to PD-1/PD-L1 inhibitors differ from those associated with CTLA-4 inhibitors and are more commonly observed in the treatment of solid tumors. This study aimed to address the knowledge gap regarding adverse events related to PD-1/PD-L1 inhibitors. A visual bibliometric network was constructed using VOSviewer, CiteSpace, R software, and the Web of Science Core Collection (WoSCC) to quantitatively analyze this research field. Future research directions were also explored. The USA ranked first in publication count and total citations. Over time, publication types transitioned from case reports to clinical trials. Research on for nivolumab was the most prevalent. The spectrum of cancers treated by PD-1/PD-L1 inhibitors expanded beyond melanoma and lung cancer to include renal cell carcinoma, esophageal cancer, and others. Common adverse events included pneumonitis, myasthenia gravis, and vitiligo. There was a significant increase in multi-phase clinical trials and studies related to biomarkers. This study offers valuable insights for potential collaborators and institutions, highlighting trends in the study of adverse events related to PD-1/PD-L1 inhibitors. The management of these adverse events has become more refined and standardized. Biomarker research and multi-phase clinical trials are likely to be key areas of focus in future studies.
Collapse
Affiliation(s)
- Qingya Song
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zongliang Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Wenping Lu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhili Zhuo
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lei Chang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Heting Mei
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yongjia Cui
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dongni Zhang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Craig-Meyer D, Hollenbaugh JA, Morgado S, McGee K, Perkins E, Yarzabek B, Lapinski P, Rowse A, Cooper C, Fortunato M, Cocco M, Cadwallader K, Munday J. Immunophenotypical characterization of immune checkpoint receptor expression in cynomolgus monkeys and human healthy volunteers in resting and in T-cell stimulatory conditions in vitro. J Immunotoxicol 2025; 22:2462106. [PMID: 39945090 DOI: 10.1080/1547691x.2025.2462106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 01/14/2025] [Accepted: 01/29/2025] [Indexed: 04/12/2025] Open
Abstract
Immunotherapeutics targeting immune checkpoint receptors or their ligands (i.e., immune checkpoint inhibitors), have been groundbreaking in the field of oncology, radically changing the approach to treatment and improving the clinical outcomes of an ever-expanding list of solid tumors and hematological malignancies. However, immune checkpoint inhibitors (ICI) are not devoid of side effects, collectively regarded as immune-related adverse events (irAE); they are not easily uncovered in preclinical immunotoxicological investigations and are often due to the very low expression of their targets in immunologically-unchallenged non-clinical species. We have characterized expression of a broad range of immune checkpoint receptors in peripheral blood mononuclear cell (PBMC) subpopulations from cynomolgus monkeys and healthy human volunteers, under resting and T-cell stimulatory conditions by multicolor flow cytometry to inform appropriate species selection for modeling potential irAE in immunotherapeutic preclinical research. Focusing on the response of the main lymphocyte populations to interleukin (IL)-2 alone, or in combination with anti-CD3 and anti-CD28 antibodies, checkpoints with shared similarities and key differences between the two species were identified. The results of this first study provide a database for the expression and response to stimulation for immune checkpoint receptors and can help guide future model selection in the design of preclinical studies involving immunotherapeutics directed against these targets.
Collapse
Affiliation(s)
| | | | - Sara Morgado
- Labcorp Early Development Laboratories Limited, Huntingdon, UK
| | - Karen McGee
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - Ethan Perkins
- Labcorp Early Development Laboratories Limited, Harrogate, UK
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, UK
| | | | | | - Amber Rowse
- Labcorp Early Development Laboratories Inc, Ann Arbor, MI
| | - Chris Cooper
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - Mara Fortunato
- Labcorp Early Development Laboratories Limited, Huntingdon, UK
| | - Mario Cocco
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | | | - James Munday
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| |
Collapse
|
3
|
Enssle S, Sax A, May P, El Khawanky N, Soliman N, Perl M, Enssle JC, Krey K, Ruland J, Pichlmair A, Bassermann F, Poeck H, Heidegger S. Gasdermin E links tumor cell-intrinsic nucleic acid signaling to proinflammatory cell death for successful checkpoint inhibitor cancer immunotherapy. Oncoimmunology 2025; 14:2504244. [PMID: 40366863 PMCID: PMC12080277 DOI: 10.1080/2162402x.2025.2504244] [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: 11/22/2024] [Revised: 05/05/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
Durable clinical responses to immune checkpoint inhibitors (ICI) are limited to a minority of patients, and molecular pathways that modulate their efficacy remain incompletely defined. We have recently shown that activation of the innate RNA-sensing receptor RIG-I and associated apoptotic tumor cell death can facilitate tumor immunosurveillance and -therapy, but the mechanism that drives its immunogenicity remained unclear. We here show that intratumoral activity of the pore-forming protein gasdermin E (GSDME) links active RIG-I signaling and apoptotic cell death in tumor cells to inflammatory pyroptosis. Activation of tumor-intrinsic RIG‑I triggered cleavage of GSDME, pore formation, loss of cell membrane integrity and leakage of cytosolic components from dying tumor cells. Tumor antigen cross-presentation by dendritic cells and subsequent expansion of cytotoxic T cells strongly relied on tumor-intrinsic GSDME activity. In preclinical murine cancer models, defective GSDME signaling rendered tumors resistant to ICI therapy. Epigenetic reprogramming with upregulation of Gdsme enhanced the susceptibility of tumor cells to inflammatory cell death and immunotherapy. In humans, transcriptome analysis of melanoma samples showed strong correlation between genetic activity of the RIG-I and pyroptosis pathways. In melanoma patients, high transcriptional activity of a pyroptosis gene set was associated with prolonged survival and beneficial response to ICI therapy. In summary, our data show that GSDME links RIG-I and apoptotic signaling to inflammatory cell death, thereby driving its immunogenicity and responsiveness to ICI. A deeper understanding of these pathways may allow for the development of novel combined modality approaches to improve ICI treatment responses in cancer patients.
Collapse
Affiliation(s)
- Stefan Enssle
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Anna Sax
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Peter May
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Nadia El Khawanky
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Nardine Soliman
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Markus Perl
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Julius C. Enssle
- Department of Medicine II, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Karsten Krey
- Institute of Virology, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Jürgen Ruland
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Pichlmair
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Virology, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Florian Bassermann
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hendrik Poeck
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
- Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
- Center for immunomedicine intransplantation and oncology (CITO), Regensburg, Germany
| | - Simon Heidegger
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
4
|
O'Connor MH, Rhodin KE, Tyler DS, Beasley GM. Management of In-transit Disease: Regional Therapies, Intralesional Therapies, and Systemic Therapy. Surg Oncol Clin N Am 2025; 34:393-410. [PMID: 40413006 PMCID: PMC12104569 DOI: 10.1016/j.soc.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
In-transit (IT) melanoma represents a distinct, heterogeneous pattern of disease that arises as superficial tumors along the track between the primary site and the draining regional lymph node basin. Many therapies have been explored for treatment of this disease with the goal of maximizing delivery of the therapeutic agent to the tumor while minimizing systemic toxicities. These include regional chemotherapies, intralesional injections, checkpoint inhibitors, immunomodulators, and vaccines in various combinations or as monotherapy. Here, we review the general managemnt of patients with ITmelanoma, the range of currently available treatment options, and recommendations for specific therapies for individual patients.
Collapse
Affiliation(s)
- Margaret H O'Connor
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Kristen E Rhodin
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Douglas S Tyler
- Department of Surgery, Texas Medical Branch, Galveston, TX, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
5
|
Ton Nu QC, Deka G, Park PH. CD8 + T cell-based immunotherapy: Promising frontier in human diseases. Biochem Pharmacol 2025; 237:116909. [PMID: 40179991 DOI: 10.1016/j.bcp.2025.116909] [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: 12/10/2024] [Revised: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
The abundant cell components of the adaptive immune system called T lymphocytes (T cells) play important roles in mediating immune responses to eliminate the invaders and create the memory of the germs to form a new immunity for the next encounter. Among them, cytotoxic T cells expressing cell-surface CD8 are the most critical effector cells that directly eradicate the target infected cells by recognizing antigens presented by major histocompatibility complex class I molecules to protect our body from pathological threats. In the continuous evolution of immunotherapy, various CD8+ T cell-based therapeutic strategies have been developed based on the role and molecular concept of CD8+ T cells. The emergence of such remarkable therapies provides promising hope for multiple human disease treatments such as autoimmunity, infectious disease, cancer, and other non-infectious diseases. In this review, we aim to discuss the current knowledge on the utilization of CD8+ T cell-based immunotherapy for the treatment of various diseases, the molecular basis involved, and its limitations. Additionally, we summarize the recent advances in the use of CD8+ T cell-based immunotherapy and provide a comprehensive overview of CD8+ T cells, including their structure, underlying mechanism of function, and markers associated with CD8+ T cell exhaustion. Building upon these foundations, we delineate the advancement of CD8+ T cell-based immunotherapies with fundamental operating principles followed by research studies, and challenges, as well as illustrate human diseases involved in this development.
Collapse
Affiliation(s)
- Quynh Chau Ton Nu
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Gitima Deka
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea; Research institute of cell culture, Yeungnam University, Gyeongsan, Republic of Korea.
| |
Collapse
|
6
|
Seldomridge AN, Weiser R, Holder AM. Systemic Therapy for Melanoma: What Surgeons Need to Know. Surg Oncol Clin N Am 2025; 34:359-374. [PMID: 40413004 DOI: 10.1016/j.soc.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Immune checkpoint inhibitors and targeted therapies (BRAF/MEK inhibitors) have transformed the care of patients with stage IV melanoma, now with 5-year overall survival rates around 50%. Surgeons should be acquainted with these drugs, the multidisciplinary considerations of their use, and the unique immune-related adverse events (irAEs) they can cause. In this review, we discuss systemic therapies for cutaneous melanoma, including the biology of immune checkpoint inhibition, treatment indications, and toxicities. We also explain how these irAEs and other toxicities can impact surgical planning and perioperative management.
Collapse
Affiliation(s)
- Ashlee N Seldomridge
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Roi Weiser
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Ashley M Holder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
7
|
Zhang N, Zhu J, Hu X, Zhou Y, Wang Q, Cai S, Xie Q, Qiu L, Lv G, Lin J. Development and evaluation of biphenyl-based small-molecule radiotracers for PET imaging of PD-L1 in tumor. Bioorg Med Chem Lett 2025; 122:130187. [PMID: 40086606 DOI: 10.1016/j.bmcl.2025.130187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Accurate identification of programmed cell death ligand 1 (PD-L1) expression is crucial for anti-tumor immunotherapy. However, the heterogeneity of PD-L1 expression in tumors makes it challenging to detect by immunohistochemistry. In this study, we developed two novel PD-L1 small-molecule PET tracers, [18F]LGT-1 and [18F]LGT-2, to enable the non-invasive and precise measurement of PD-L1 expression in tumors through PET imaging. The radiochemical yields for [18F]LGT-1 and [18F]LGT-2 were 12.54±2.73% and 10.54±2.21%, respectively, with both tracers exhibiting approximately 98% radiochemical purity and molar activities of 12.23±2.84 GBq/μmol and 11.41±1.47 GBq/μmol. Both tracers demonstrated good stability in PBS (pH 7.4) and mouse serum after 2 hours of incubation. In cellular uptake assays, [18F]LGT-1 achieved a maximum uptake of 5.47±0.03 %AD at 4 hours, which could be significantly inhibited by the non-radioactive compound LGT-1. In contrast, [18F]LGT-2 exhibited high non-specific uptake in tumor cells. PET imaging revealed that [18F]LGT-1 quickly accumulated in tumors within 5 minutes, achieving an uptake of 1.48±0.15 %ID/mL, and maintained a stable level for 60 minutes, while [18F]LGT-2 showed minimal tumor uptake. Additionally, [18F]LGT-1 had significantly lower liver uptake compared to [18F]LGT-2. Despite the high uptake in non-target tissues for [18F]LGT-1, which complicates its application, this study provides new insights for developing novel PD-L1 small-molecule tracers, with further optimization of the tracers currently in progress.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Junyi Zhu
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Xin Hu
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yuxuan Zhou
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Qianhui Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Shuyue Cai
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Quan Xie
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Ling Qiu
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Gaochao Lv
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China.
| | - Jianguo Lin
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China.
| |
Collapse
|
8
|
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
|
9
|
Boyd R, Charakidis M, Burgess CP, Dugdale S, Castillon Jawoyn Katherine Region Nt C, Sutandar D, Wright A. Improved cancer survival in the Northern Territory: identifying progress and disparities for Aboriginal peoples, 1991-2020. Med J Aust 2025; 222:517-523. [PMID: 40259612 DOI: 10.5694/mja2.52656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/06/2024] [Indexed: 04/23/2025]
Abstract
OBJECTIVES To investigate cancer survival for Aboriginal and non-Aboriginal peoples in the Northern Territory during the period 1991-2020, across the 15 most prevalent primary cancer sites. STUDY DESIGN Retrospective cohort study of Northern Territory Cancer Registry notifications. SETTING, PARTICIPANTS NT residents diagnosed with an invasive cancer from 1 January 1991 to 31 December 2020. MAIN OUTCOME MEASURES Five-year survival for all cancers and for 15 primary cancer sites by 10-year periods of diagnosis, and excess hazard ratios comparing excess mortality following cancer diagnosis for Aboriginal peoples compared with non-Aboriginal peoples. RESULTS Of 17 759 cancer registrations analysed, 3350 (18.9%) had Indigenous status recorded as Aboriginal. Five-year survival improved significantly from 1991-2000 to 2011-2020 for all populations, including Aboriginal (males, 20.5% to 37.1%; females, 32.3% to 47.2%) and non-Aboriginal (males, 50.0% to 65.9%; females, 64.5% to 75.4%). The gap in 5-year cancer survival for Aboriginal peoples closed by 12.4% (4.0 percentage difference) for females, but only by 2.4% (0.7 percentage difference) for males. For all 15 cancer sites, 5-year survival improved, but Aboriginal peoples experienced excess mortality, ranging from an excess hazard ratio of 1.3 for mortality following diagnosis of liver cancer to 6.1 for prostate cancer during the period 2011-2020. CONCLUSIONS Cancer survival has improved for Aboriginal and non-Aboriginal peoples in the NT. However, the gap in survival outcomes for Aboriginal peoples persists. Further research is required on pathways to close the gap, including: improving access to care, understanding social and cultural factors, reducing diagnostic and treatment delays, and promoting greater equity of Aboriginal participation in clinical trials. Quality improvement approaches led by Aboriginal peoples should be prioritised to tailor culturally appropriate preventive strategies.
Collapse
Affiliation(s)
- Rowena Boyd
- Northern Territory Department of Health, Darwin, NT
| | - Michail Charakidis
- Northern Territory Department of Health, Darwin, NT
- Royal Darwin Hospital, Darwin, NT
- Charles Darwin University, Darwin, NT
| | | | | | | | | | | |
Collapse
|
10
|
Bhatt KA, Vaynrub AJ, Cham J, Iyer SG, Izar B. Diagnosis and management of concurrent metastatic melanoma and chronic myelomonocytic leukemia. Melanoma Res 2025; 35:192-196. [PMID: 39903257 DOI: 10.1097/cmr.0000000000001025] [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: 02/06/2025]
Abstract
While the association between chronic lymphocytic leukemia (CLL) and a higher incidence of melanoma is well documented, the diagnosis of concurrent high-risk chronic myelomonocytic leukemia (CMML) and metastatic melanoma (MM) has not previously been described. Moreover, the treatment of MM and CMML differ greatly in the mechanism of action of their corresponding antineoplastic therapies: treatment of MM frequently involves immune checkpoint inhibitors (ICI), while patients with CMML receive myelosuppressive agents. Simultaneous management of these malignancies can be nuanced due to the potential impact of one treatment's constituents on the activity of the other and the broad and nonoverlapping array of potential adverse effects of these agents. Here, we describe the clinical course of a patient who was diagnosed with concurrent MM and CMML and our approach to the challenging balance of delivering ICI concurrently with the hypomethylating agent azacitidine and the BCL-2 inhibitor venetoclax.
Collapse
Affiliation(s)
- Kishan A Bhatt
- Columbia University Vagelos College of Physicians and Surgeons
| | - Anna J Vaynrub
- Columbia University Vagelos College of Physicians and Surgeons
| | - Jason Cham
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center
| | - Sunil G Iyer
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center
| | - Benjamin Izar
- Columbia University Vagelos College of Physicians and Surgeons
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center
- Columbia Center for Translational Immunology
- Department of Systems Biology, Program for Mathematical Genomics, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
11
|
Haugh AM, Fisher DE, Lawrence DP. Tumor-Infiltrating Lymphocyte Therapy for Melanoma Has Arrived (at Last). J Invest Dermatol 2025; 145:1257-1259. [PMID: 40146097 DOI: 10.1016/j.jid.2024.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 03/28/2025]
Affiliation(s)
- Alexandra M Haugh
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David E Fisher
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Donald P Lawrence
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
12
|
Schuiveling M, Ter Maat LS, Van Duin IAJ, Verheijden RJ, Troenokarso MF, Moeskops P, Verhoeff JJC, Elias SG, van Amsterdam WAC, Burgers F, Van den Berkmortel FWPJ, Boers-Sonderen MJ, Boomsma MF, De Groot JW, Haanen JBAG, Hospers GAP, Piersma D, Vreugdenhil G, Westgeest HM, Kapiteijn E, Labots M, Veldhuis WB, Van Diest PJ, De Jong PA, Pluim JPW, Leiner T, Veta M, Suijkerbuijk KPM. Body composition and checkpoint inhibitor treatment outcomes in advanced melanoma: a multicenter cohort study. J Natl Cancer Inst 2025; 117:1245-1252. [PMID: 39980388 PMCID: PMC12145918 DOI: 10.1093/jnci/djaf039] [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: 05/15/2024] [Revised: 12/18/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND The association of body composition with checkpoint inhibitor outcomes in melanoma is a matter of ongoing debate. In this study, we aim to investigate body mass index (BMI) alongside computed tomography (CT)-derived body composition metrics in the largest cohort to date. METHODS Patients treated with first-line anti-PD1 ± anti-CTLA4 for advanced melanoma were retrospectively identified from 11 melanoma centers in The Netherlands. From baseline CT scans, 5 body composition metrics were extracted: subcutaneous adipose tissue index, visceral adipose tissue index, skeletal muscle index, density, and gauge. These metrics were correlated in univariable and multivariable Cox proportional hazards analysis with progression-free survival, overall survival, and melanoma-specific survival (PFS, OS, and MSS). RESULTS A total of 1471 eligible patients were included. Median PFS and OS were 9.1 and 38.1 months, respectively. Worse PFS was observed in underweight patients (multivariable hazard ratio [HR] = 1.86, 95% CI = 1.14 to 3.06). Furthermore, prolonged OS was observed in patients with higher skeletal muscle density (multivariable HR = 0.88, 95% CI = 0.81 to 0.97) and gauge (multivariable HR = 0.61, 95% CI = 0.82 to 0.998), whereas higher visceral adipose tissue index was associated with worse OS (multivariable HR = 1.12, 95% CI = 1.04 to 1.22). No association with survival outcomes was found for overweight, obesity, or subcutaneous adipose tissue. CONCLUSION Our findings suggest that underweight BMI is associated with worse PFS, whereas higher skeletal muscle density and lower visceral adipose tissue index were associated with improved OS. These associations were independent of known prognostic factors, including sex, age, performance status, and extent of disease. No significant association between higher BMI and survival outcomes was observed.
Collapse
Affiliation(s)
- Mark Schuiveling
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Laurens S Ter Maat
- Image Sciences Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Isabella A J Van Duin
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rik J Verheijden
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Max F Troenokarso
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Pim Moeskops
- Quantib (currently DeepHealth), Rotterdam, The Netherlands
| | - Joost J C Verhoeff
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sjoerd G Elias
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wouter A C van Amsterdam
- Department of Data Science and Biostatistics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Femke Burgers
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Marye J Boers-Sonderen
- Department of Medical Oncology, Radboudumc, Radboud University, Nijmegen, The Netherlands
| | | | | | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
- Melanoma Clinic, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Geke A P Hospers
- Department of Medical Oncology, UMC Groningen, University of Groningen, Groningen, The Netherlands
| | - Djura Piersma
- Department of Medical Oncology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Gerard Vreugdenhil
- Department of Medical Oncology, Maxima Medical Center, Veldhoven, The Netherlands
| | - Hans M Westgeest
- Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - Mariette Labots
- Department of Medical Oncology, UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Wouter B Veldhuis
- Quantib (currently DeepHealth), Rotterdam, The Netherlands
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Paul J Van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Pim A De Jong
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Josien P W Pluim
- Image Sciences Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Medical Image Analysis, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Tim Leiner
- Quantib (currently DeepHealth), Rotterdam, The Netherlands
- Department of Radiology, Mayo Clinical, Rochester, MN, United States
| | - Mitko Veta
- Medical Image Analysis, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
13
|
Wang S, Yang C, Zhang Y, Hu Y, Xiao L, Ding W, Qiu B, Li F. Co-encapsulated Ce6 + CpG and biopeptide-modified liposomes for enhanced transdermal photo-immunotherapy of superficial tumors. Mater Today Bio 2025; 32:101669. [PMID: 40177379 PMCID: PMC11964550 DOI: 10.1016/j.mtbio.2025.101669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 04/05/2025] Open
Abstract
Cancer immunotherapy encounters challenges of a low treatment response rate due to the tumor immunosuppressive microenvironment and immune-related adverse events caused by off-target immunotherapy agents delivered through systemic administration in clinical practice. Photodynamic therapy (PDT) offers a viable approach to improve the immunotherapy efficacy through inducing immunogenic tumor cell death and is particularly advantageous in superficial tumor treatment. Therefore, leveraging integrated nanomaterials for photo-immunotherapy appears to be an ideal strategy to improve therapeutic outcome. In this study, we develop a transdermal-enhancing peptide (TD)-modified cationic liposome that simultaneously encapsulated with photosensitizer chlorine 6 (Ce6) and immunoadjuvant CpG, denoted as Ce6/CpG@Lip-TD, to mediate photo-immunotherapy of superficial tumors via the skin. The functionalization of TD peptide and positively charged surface endow the liposomes enhanced skin penetration capability. The combination of Ce6 and CpG within the liposomes synergistically potentiates the photo-immunotherapy effect when exposed to laser irradiation. In both melanoma and breast cancer murine models, Ce6/CpG@Lip-TD demonstrated substantial tumor-suppressing properties, along with an augmented systemic immune response against distal tumors. As a topical therapeutic agent, Ce6/CpG@Lip-TD circumvents the regulatory challenges associated with the systemic delivery of nanomaterials and significantly reduces systemic side effects, holding great promise for rapid translation into clinical applications.
Collapse
Affiliation(s)
- Shaozhen Wang
- Medical Imaging Center, Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Chen Yang
- Medical Imaging Center, Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yuanyuan Zhang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
| | - Yi Hu
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230027, China
| | - Lan Xiao
- Department of Gynecology Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Weiping Ding
- Medical Imaging Center, Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Bensheng Qiu
- Medical Imaging Center, Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Fenfen Li
- Medical Imaging Center, Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
14
|
Dong H, Li S, Peng Y, Zhang X, Zheng J, Xue C, Zheng Y, Yu Y, Lu X, Hu Z, Cui H. Durvalumab‑induced type 1 diabetes mellitus in lung adenocarcinoma: A case report and literature review. Oncol Lett 2025; 29:277. [PMID: 40247987 PMCID: PMC12005073 DOI: 10.3892/ol.2025.15023] [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: 11/15/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
Immune checkpoint inhibitor-induced type 1 diabetes mellitus (ICI-T1DM) is a rare adverse reaction associated with durvalumab. Among the adverse reactions to durvalumab, the incidence of new-onset diabetes is relatively rare, occurring in ~0.2% of cases. The present study reports the case of a 62-year-old woman who developed ICI-T1DM following two cycles of durvalumab, presenting with thirst, polydipsia and polyuria. Laboratory examinations (glycated hemoglobin and glutamic acid decarboxylase antibody), along with consultations from an endocrinologist, led to the patient being diagnosed with ICI-T1DM. Immunotherapy was discontinued, and insulin replacement therapy was initiated. Blood glucose levels were closely monitored using a subcutaneous meter. The onset of diabetic ketoacidosis (DKA) was prevented due to timely treatment. In conclusion, medical oncologists need to be aware that durvalumab, an immunotherapy agent, can induce ICI-T1DM. Therefore, regular monitoring of blood glucose levels and collaborative consultations with endocrinologists are essential for an accurate diagnosis when elevated blood sugar levels are detected. The prompt diagnosis of ICI-T1DM is crucial to prevent the occurrence of DKA.
Collapse
Affiliation(s)
- Huijing Dong
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Shengfu Li
- Department of Tuberculosis, Tai Yuan Fourth Peoples (Tuberculosis) Hospital, Taiyuan, Shanxi 030053, P.R. China
| | - Yanmei Peng
- Department of Oncology, Fangshan Hospital Beijing University of Chinese Medicine, Beijing 102400, P.R. China
| | - Xu Zhang
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Jiabin Zheng
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Chongxiang Xue
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Yumin Zheng
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Yixuan Yu
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Xingyu Lu
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Zixin Hu
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Huijuan Cui
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| |
Collapse
|
15
|
Gianmarco M, Carolina P, Gregorio M, Michela V, Monica P, Claire GG, Michele M, Giulia M, Roberta M, Cinzia A, Lorena B, Marcello T, Fabiana P, Roberta M. Circulating tumor DNA monitoring in advanced mutated melanoma (LIQUID-MEL). THE JOURNAL OF LIQUID BIOPSY 2025; 8:100295. [PMID: 40276578 PMCID: PMC12019447 DOI: 10.1016/j.jlb.2025.100295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025]
Abstract
Introduction Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of metastatic melanoma, but a percentage of patients did not show benefit. Circulating tumor DNA (ctDNA) has emerged as a potential non-invasive tool for monitoring disease evolution and treatment response. The present study aimed to evaluate the clinical utility of ctDNA dynamics in patients with metastatic melanoma receiving ICIs, while exploring its role in the oncological course. Materials and methods The LIQUID-MEL study is a prospective, single-centre pilot study including patients with BRAF/NRAS-mutant metastatic melanoma. ctDNA was quantified using digital droplet PCR (ddPCR) at four different time points. Uni- and multivariable Cox regression models were used to assess the correlation between shedding and progression-free survival (PFS), and overall survival (OS). Results Overall, 23 patients were included. At baseline, ctDNA was detectable in 5/23 (21.7 %) cases. Baseline ctDNA shedding was associated with shorter PFS (3.88 months vs. 0.69 months, p=0.012). A strong numerical trend was observed also in OS (12.66 months vs. 2.53 months, p=0.287). Shedding at baseline did not demonstrate independent prognostic or predictive value in the uni- and multivariable analysis. The longitudinal analysis revealed intriguing patterns of ctDNA shedding in individual patients. Conclusion ctDNA detectability and its dynamic changes during treatment may have potential clinical utility in patients with metastatic melanoma, offering a valuable non-invasive tool for monitoring disease and treatment response. The small sample size limited the statistical power of the analysis. Further studies with larger cohorts are needed to validate its role in routine clinical practice.
Collapse
Affiliation(s)
| | - Palazzi Carolina
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Monica Gregorio
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Verzè Michela
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Pluchino Monica
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Maffezzoli Michele
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Portsmouth Hospital University NHS Trust, Portsmouth, United Kingdom
| | - Mazzaschi Giulia
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Manuguerra Roberta
- Pathology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Azzoni Cinzia
- Pathology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Bottarelli Lorena
- Pathology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Tiseo Marcello
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Perrone Fabiana
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Minari Roberta
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| |
Collapse
|
16
|
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
|
17
|
Liu D, Liu L, Zhao X, Zhang X, Chen X, Che X, Wu G. A comprehensive review on targeting diverse immune cells for anticancer therapy: Beyond immune checkpoint inhibitors. Crit Rev Oncol Hematol 2025; 210:104702. [PMID: 40122356 DOI: 10.1016/j.critrevonc.2025.104702] [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: 02/10/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, primary resistance and acquired resistance continue to limit their efficacy for many patients. To address resistance and enhance the anti-tumor activity within the tumor immune microenvironment (TIME), numerous therapeutic strategies targeting both innate and adaptive immune cells have emerged. These include combination therapies with ICIs, chimeric antigen receptor T-cell (CAR-T), chimeric antigen receptor macrophages (CAR-Ms) or chimeric antigen receptor natural killer cell (CAR-NK) therapy, colony stimulating factor 1 receptor (CSF1R) inhibitors, dendritic cell (DC) vaccines, toll-like receptor (TLR) agonists, cytokine therapies, and chemokine inhibition. These approaches underscore the significant potential of the TIME in cancer treatment. This article provides a comprehensive and up-to-date review of the mechanisms of action of various innate and adaptive immune cells within the TIME, as well as the therapeutic strategies targeting each immune cell type, aiming to deepen the understanding of their therapeutic potential.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinming Zhao
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoman Zhang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaochi Chen
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
18
|
Foiani G, Melchiotti E, Capello K, Porcellato I, Brachelente C, Iussich S, Giacobino D, Morello E, Martano M, Buracco P, Vascellari M. PD-L1, PD-1, and CTLA-4 mRNA In Situ Expression by Canine Oral Melanoma Cells and Immune Cells of the Tumour Microenvironment. Vet Comp Oncol 2025; 23:141-151. [PMID: 39789732 DOI: 10.1111/vco.13039] [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: 09/24/2024] [Revised: 12/10/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025]
Abstract
Canine oral melanoma (OM) exhibits poor prognosis and limited treatment options. The success of immune checkpoint inhibitors (ICIs) in human melanoma has driven interest in similar therapeutic approaches in the dog, although the immunosuppressive mechanisms adopted by canine OM remain unclear. This study aimed to evaluate the expression of the immune checkpoints PD-1/PD-L1 and CTLA-4 by RNAscope in situ hybridization (ISH) in canine OM, to investigate their expression pattern and explore their potential role in melanoma progression. Twenty-four formalin-fixed, paraffin-embedded canine OM were included in the study. PD-L1 expression by tumour cells was detected in 100% melanomas (score 1-3), especially at the host-tumour interface. PD-1 and CTLA-4 expression by tumour cells was detected in 13/24 (54%, score 1-2) and 18/24 (75%, score 1) melanomas, respectively. Dual ISH-immunohistochemistry with Melanoma Triple Cocktail, CD3, CD20 and Iba1 demonstrated the expression of tested immune checkpoints in neoplastic and immune cells. Notably, PD-1 and CTLA-4 were predominantly expressed by tumour-infiltrating T lymphocytes, while PD-L1 was primarily expressed by tumour-associated macrophages. PD-1 expression in neoplastic cells was significantly correlated with mitotic count (p < 0.05), while no associations were found between immune checkpoint expression and disease-free interval or overall survival. Whole tumour PD-L1 and PD-1 expression, assessed by image analysis, correlated to PD-L1 scores in neoplastic cells and the grade of tumour-infiltrating lymphocytes, respectively. Collectively, PD-L1, PD-1 and CTLA-4 likely contribute to immunosuppression in canine OM. Further studies are warranted to investigate whether ISH can serve as a biomarker for selecting patients suitable for ICI treatment.
Collapse
Affiliation(s)
- Greta Foiani
- Histopathology Laboratory, Istituto Zooprofilattico Sperimentale delle Venezie, Padua, Italy
| | - Erica Melchiotti
- Histopathology Laboratory, Istituto Zooprofilattico Sperimentale delle Venezie, Padua, Italy
| | - Katia Capello
- Epidemiology and Biostatistics Unit, Istituto Zooprofilattico Sperimentale delle Venezie, Padua, Italy
| | - Ilaria Porcellato
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Chiara Brachelente
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Selina Iussich
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Davide Giacobino
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Emanuela Morello
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Marina Martano
- Department of Veterinary Medical Sciences, University of Parma, Parma, Italy
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Marta Vascellari
- Histopathology Laboratory, Istituto Zooprofilattico Sperimentale delle Venezie, Padua, Italy
| |
Collapse
|
19
|
Kött J, Gebhardt C. [Biomarkers in adjuvant and neoadjuvant treatment of melanoma]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2025; 76:361-364. [PMID: 40335648 DOI: 10.1007/s00105-025-05506-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/07/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Personalized treatment of melanoma is becoming increasingly more important. Biomarkers offer the possibility of controlling treatment more precisely and reducing side effects. OBJECTIVE The aim of this text is to provide an overview of current tissue-based, blood-based and radiological biomarkers and their clinical application in melanomas. MATERIAL AND METHODS A literature research and analysis of current studies on biomarkers in adjuvant and neoadjuvant treatment of melanomas were carried out and relevant congress contributions were additionally included. RESULTS Tissue-based programmed cell death 1 ligand 1 (PD-L1) expression, interferon gamma (IFNγ) signature, gene expression profiles (GEP) and tumor mutational burden (TMB) are of prognostic and predictive relevance. Blood-based circulating tumor DNA (ctDNA) in the sense of a liquid biopsy should be emphasized as a personalized biomarker for longitudinal tracking during treatment or aftercare. Positron emission tomography computed tomography (PET-CT) and body composition enable an improved assessment of treatment efficiency. There are currently no data from prospective validation studies on these biomarkers; initial data from the NivoMela study are awaited. CONCLUSION The combination of tissue-based, blood-based and radiological biomarkers in terms of multiparametric approaches is promising but further prospective validation is needed for broad clinical use. These are currently not comprehensively implemented in the clinical routine in centers or in remuneration procedures.
Collapse
Affiliation(s)
- Julian Kött
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Deutschland
- Fleur Hiege Centrum für Hautkrebsforschung, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Deutschland
| | - Christoffer Gebhardt
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Deutschland.
- Fleur Hiege Centrum für Hautkrebsforschung, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Deutschland.
- Universitäres Hauttumorzentrum Hamburg, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Deutschland.
| |
Collapse
|
20
|
Olsson-Brown A, Jain A, Frazer R, Farrugia D, Carser J, Houghton J, Lewis RD, D'Mello S, Emanuel G. Clinical Management and Outcomes of Immune-Related Adverse Events During Treatment with Immune Checkpoint Inhibitor Therapies in Melanoma and Renal Cell Carcinoma: A UK Real-World Evidence Study. Oncol Ther 2025:10.1007/s40487-025-00349-z. [PMID: 40448748 DOI: 10.1007/s40487-025-00349-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 05/15/2025] [Indexed: 06/02/2025] Open
Abstract
INTRODUCTION Immune checkpoint inhibitor (ICI) therapy, although effective in treating patients with a variety of advanced malignancies, can result in potentially severe or even fatal immune-related adverse events (irAEs). This study aimed to generate real-world evidence on irAE characteristics, clinical management and clinical outcomes among patients with advanced (unresectable or metastatic) malignant melanoma (a/mMM) or advanced renal cell carcinoma (aRCC) treated with nivolumab (NIVO) and/or ipilimumab (IPI) in the UK. METHODS This was a multi-centre, retrospective cohort study of adult patients with a/mMM or aRCC, who received NIVO and/or IPI at one of five specialist treatment centres in the UK between 1 January 2016 and 31 March 2020. The incidence and grading of irAEs were described, as well as time to irAE onset, the management of irAEs and overall survival (OS). RESULTS In total, 199 patients were included in the study: 162 with a/mMM and 37 with aRCC. The majority of patients in both a/mMM (75.3%) and aRCC (62.2%) cohorts reported any irAE, while 45.9% and 30.4% in the a/mMM and aRCC cohorts reported grade 3 or 4 irAEs, respectively. Colitis/diarrhoea, skin reactions and hepatitis were most frequently reported, and the predominant treatment prescribed for any irAE was corticosteroids only. Analysis indicated a positive association between the development of an irAE and longer OS. CONCLUSIONS Findings from this study support current literature, provide further insights into the characteristics and clinical management of irAEs and support an association between the development of an irAE and improved OS in these patient groups.
Collapse
Affiliation(s)
| | - Ankit Jain
- The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | | | - David Farrugia
- Gloucestershire Hospitals NHS Foundation Trust, Gloucester, UK
| | | | - John Houghton
- Health Economics and Outcomes Research Ltd, Cardiff, UK
| | - Ruth D Lewis
- Health Economics and Outcomes Research Ltd, Cardiff, UK
| | - Simon D'Mello
- Bristol Myers Squibb, Sanderson Road, Denham, Uxbridge, UB8 1DH, UK.
| | | |
Collapse
|
21
|
Ito T, Tanaka Y, Tanegashima K, Nishio K, Hashimoto H, Ichiki T, Ohno F, Kaku-Ito Y, Nakahara T. KS-NailMel-1: a novel cell line of nail apparatus melanoma. Hum Cell 2025; 38:112. [PMID: 40437181 PMCID: PMC12119781 DOI: 10.1007/s13577-025-01242-7] [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: 01/16/2025] [Accepted: 05/17/2025] [Indexed: 06/01/2025]
Abstract
Nail apparatus melanoma (NAM) is a specific type of cutaneous melanoma that develops in the nail apparatus of the hands and feet. The prognosis for metastatic NAM is poor due to a lack of fully effective systemic therapies. However, the difficulty in obtaining a NAM model has hindered basic research aimed at discovering effective treatment strategies. In this study, we established a NAM cell line, named KS-NailMel-1, from a primary tumor located on the nail apparatus of the left ring finger of a 68-year-old Japanese female. The cells were successfully maintained for over 9 months, exhibiting a doubling time of 38.6 ± 1.94 h. KS-NailMel-1 displayed consistent growth, spheroid formation, and invasiveness, and was confirmed to be identical to the original tumor through short tandem repeat analyses, whole-exome sequencing, and immunohistochemistry. Western blotting of the cells demonstrated the protein expression of NECTIN4, which has recently attracted attention as a potential therapeutic target for melanoma. The KS-NailMel-1 cell line represents a valuable resource for basic and preclinical research on NAM, deepening our understanding of the tumor characteristics and facilitating the development of treatment strategies for this rare form of cancer.
Collapse
Affiliation(s)
- Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Yuka Tanaka
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Keiko Tanegashima
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kiichiro Nishio
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroki Hashimoto
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Toshio Ichiki
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Fumitaka Ohno
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yumiko Kaku-Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
22
|
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
|
23
|
Noringriis IM, Donia M, Madsen K, Schmidt H, Haslund CA, Bastholt L, Svane IM, Ellebaek E. Long-term clinical outcome of patients with metastatic melanoma and initial stable disease during anti-PD-1 checkpoint inhibitor immunotherapy with pembrolizumab. Br J Cancer 2025:10.1038/s41416-025-03048-8. [PMID: 40419744 DOI: 10.1038/s41416-025-03048-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/22/2025] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND A substantial number of patients with metastatic melanoma (MM) treated with anti-PD-1 monotherapy have initial stable disease (SD), yet the real-world prognosis of these patients remains unclear. METHODS In this nationwide cohort study, we analysed real-world outcomes of patients with MM treated with pembrolizumab in Denmark. Focusing on patients with initial SD, we assessed best overall response (BOR), progression-free survival (PFS), and overall survival (OS) and identified predictors of survival in multivariable analyses. RESULTS Out of 1048 included patients, 233 (22.2%) had initial SD with a median PFS and OS of 14.7 and 50.1 months. Subsequent partial response (PR) or complete response (CR) was developed by 44 (18.9%) and 52 (22.3%) patients showing significantly improved PFS compared to patients with continued SD (PR: HR 0.52, 95% CI 0.34-0.81, p = 0.003; CR: HR 0.15, 95% CI 0.07-0.32, p < 0.001) and survival rates comparable to patients with initial PR and CR, respectively. Furthermore, 49 (21.0%) patients showed continued disease control (median follow-up of 82.3 months). For 51.0% of these patients, the last dose of pembrolizumab was administered during SD with a median treatment duration of 12.4 months. CONCLUSIONS Of patients with initial SD, 40% developed a subsequent objective response with improved long-term prognosis comparable to patients with initial response. More than 20% exhibited continued disease control.
Collapse
Affiliation(s)
- Inge Mansfield Noringriis
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Marco Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Kasper Madsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.
| | - Eva Ellebaek
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.
| |
Collapse
|
24
|
Fojo T, Bates S. Skimming the Median and the Problem of Exaggerated Survival Gains. JAMA Oncol 2025:2834385. [PMID: 40402485 DOI: 10.1001/jamaoncol.2025.0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
This Viewpoint describes a phenomenon termed skimming the median and discusses 3 examples to emphasize why an entire dataset should be considered to reach precise conclusions.
Collapse
Affiliation(s)
- Tito Fojo
- Columbia University, New York, New York
- The James J. Peters Veterans Administration Medical Center, New York, New York
| | - Susan Bates
- Columbia University, New York, New York
- The James J. Peters Veterans Administration Medical Center, New York, New York
| |
Collapse
|
25
|
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
|
26
|
Lin J, Liu T, Chen J, Lin Y, Chen X, Zhuo Y, Li Y, Jiang Y, Yang L, Tu C, Liu B, Zou J, Chen L, Sun Y. Efficacy and safety of cadonilimab combined with chemotherapy as the first-line treatment for primary advanced or recurrent endometrial cancer: a prospective single-arm open-label phase II clinical trial. BMJ Open 2025; 15:e094649. [PMID: 40389320 PMCID: PMC12090854 DOI: 10.1136/bmjopen-2024-094649] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 05/06/2025] [Indexed: 05/21/2025] Open
Abstract
INTRODUCTION Recently, immunotherapy has significantly transformed the treatment landscape of endometrial cancer (EC). Results from KEYNOTE-158, RUBY and AtTEnd showed programmed cell death 1 (PD-1) or programmed cell death-ligand 1 inhibitors with promising efficacy in primary advanced or recurrent EC. However, few studies focused on the role of dual immune checkpoints in primary advanced or recurrent EC. Cadonilimab is an immune checkpoint inhibitor targeting the PD-1 and T-lymphocyte antigen-4, which is expected to show substantial clinical efficacy in EC. Combining cadonilimab with standard chemotherapy may have synergistic effects, making this combination a promising first-line treatment for primary advanced or recurrent EC. Furthermore, incorporating molecular classification for guidance on the use of cadonilimab may hold valuable clinical benefits. METHODS AND ANALYSIS In this multicentre, open-label, phase II study, patients with histologically confirmed EC were eligible. Forty-five patients will be recruited. Seventeen patients will be enrolled in stage I, and at least seven cases of complete response (CR) and partial response (PR) should be observed before entering stage II. All patients will receive cadonilimab at a dosage of 10 mg/kg along with carboplatin (area under the curve (AUC)=4-5) plus paclitaxel (175 mg/m2) every 3 weeks (Q3W) for 6-8 cycles. Subsequently, patients with CR, PR or stable disease will receive maintenance of cadonilimab at 10 mg/kg Q3W for 24 months or until progressive disease or adverse events are reported. The objective response rate is the primary endpoint. The secondary endpoints include the disease control rate, duration of response, progression-free survival, overall survival and safety. Additionally, exploratory endpoints involve biomarkers that may predict the efficacy of cadonilimab and chemotherapy, as well as their relationship with molecular classifications. The interim analysis will be conducted after 17 patients have been enrolled. ETHICS AND DISSEMINATION The study protocol meets the approval of the ethical committee of Fujian Cancer Hospital (K2023-173-04) and all other participating hospitals. Study findings will be disseminated in peer-reviewed publications. TRIAL REGISTRATION NUMBER NCT06066216.
Collapse
Affiliation(s)
- Jie Lin
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Tongyu Liu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Jian Chen
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Yingtao Lin
- Clinical Medical Research Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Xiaoxiang Chen
- Department of Gynecology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Yanhong Zhuo
- Department of Radiation Oncology, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Yuzhi Li
- Department of Gynecology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Yu Jiang
- Department of Obstetrics and Gynecology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Linlin Yang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Chunhua Tu
- Department of Obstetrics and Gynecology, The First Affilated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Bin Liu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Jianping Zou
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Lijun Chen
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Yang Sun
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| |
Collapse
|
27
|
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
|
28
|
Guetter S, König C, Koerkel-Qu H, Markiewicz A, Scheitler S, Katzer M, Berneburg M, Renner P, Cucuruz B, Guttenberger L, Naimer V, Weidele K, Treitschke S, Werno C, Jaser H, Bargmann T, Braun A, Weber F, Evert K, Rochwarger A, Schürch CM, Limm K, Oefner PJ, Rachel R, Baumann F, Warfsmann J, Schmidleithner L, Guetter K, Mohammadi P, Ulmer A, Haferkamp S, Klein CA, Werner-Klein M. MCSP + metastasis founder cells activate immunosuppression early in human melanoma metastatic colonization. NATURE CANCER 2025:10.1038/s43018-025-00963-w. [PMID: 40379833 DOI: 10.1038/s43018-025-00963-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 04/01/2025] [Indexed: 05/19/2025]
Abstract
To investigate the early, poorly understood events driving metastatic progression, we searched for the earliest detectable disseminated cancer cells (DCCs), also often referred to as disseminated tumor cells (DTCs), in sentinel lymph node (SLN) biopsies of 492 patients with stage I-III melanoma. Using micromanipulator-assisted isolation of rare DCCs, single-cell mRNA and DNA sequencing, codetection by indexing immunofluorescence imaging and survival analysis, we identified melanoma-associated chondroitin sulfate proteoglycan (MCSP)+ melanoma cells as metastasis founder cells (MFCs). We found that DCCs entering SLNs predominantly exhibited a transitory phenotype that, upon interferon-γ exposure triggered by CD8 T cells, dedifferentiated into a neural-crest-like phenotype. This was accompanied by increased production of small extracellular vesicles (sEVs) carrying the immunomodulatory proteins CD155 and CD276 but rarely programmed cell death protein 1 ligand 1. The sEVs suppressed CD8 T cell proliferation and function, facilitating colony formation. Targeting MCSP+ MFCs or their immune escape mechanisms could be key to curing melanoma early by preventing manifestation of metastasis.
Collapse
Affiliation(s)
- Severin Guetter
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Courtney König
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Huiqin Koerkel-Qu
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Aleksandra Markiewicz
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Sebastian Scheitler
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Marie Katzer
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Philipp Renner
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Beatrix Cucuruz
- Department of Vascular Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Leonhard Guttenberger
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Veronika Naimer
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Kathrin Weidele
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Steffi Treitschke
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Christian Werno
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Hanna Jaser
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Tonia Bargmann
- Preclinical Pharmacology and Toxicology, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM member of Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH) in the German Center for Lung Research (DZL), Hannover, Germany
| | - Armin Braun
- Preclinical Pharmacology and Toxicology, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM member of Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH) in the German Center for Lung Research (DZL), Hannover, Germany
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Florian Weber
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Katja Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Alexander Rochwarger
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Christian M Schürch
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Katharina Limm
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Reinhard Rachel
- Center for Electron Microscopy, University of Regensburg, Regensburg, Germany
| | - Felix Baumann
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Germany
| | - Jens Warfsmann
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Lisa Schmidleithner
- Leibniz Institute for Immunotherapy, University Hospital Regensburg, Regensburg, Germany
| | - Kathrin Guetter
- Leibniz Institute for Immunotherapy, University Hospital Regensburg, Regensburg, Germany
| | - Parvaneh Mohammadi
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Anja Ulmer
- Department of Dermatology, University of Tübingen, Tübingen, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Christoph A Klein
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany.
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany.
| | - Melanie Werner-Klein
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany.
- Leibniz Institute for Immunotherapy, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
29
|
Prasai A, Baral N, Elajami MK, Vallabhaneni E, Mitchell JD, Iqbal U, Bahmad HF, Welty FK, Elajami TK. Trends of all-cause, melanoma-specific, and cardiovascular mortality in melanoma patients from 2005 to 2020. Cancer Epidemiol 2025; 97:102848. [PMID: 40381435 DOI: 10.1016/j.canep.2025.102848] [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: 02/15/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Recent advances in melanoma treatment, including immunotherapy and targeted therapy, have significantly improved survival among melanoma patients after 2010. However, these changes may have influenced mortality trends, including those related to cardiovascular (CV) events. OBJECTIVE In this study, we assess mortality trends including CV mortality in melanoma patients. METHODS Using data from the Surveillance, Epidemiology, and End Results (SEER) 17 registry, we examined melanoma incidence and CV, melanoma-specific, and all-cause mortalities from 2005 to 2020. We utilized Joinpoint software to estimate the annual percentage change (APC). RESULTS Our study included 299,993 melanoma patients (173,889 males; 126,104 females). The incidence of melanoma increased by 0.6 % annually (95 % CI: 0.3-1.5, p < 0.05) from 2005 to 2019, followed by a 6.3 % decrease (95 % CI: -9.9 to -0.7, p < 0.05) in 2020. All-cause mortality increased annually by 8.3 % (95 % CI: 6.7-11.3, p < 0.05) from 2005 to 2010, then by 3.3 % (95 % CI: 2.8-3.7, p < 0.05) after 2010. Melanoma-specific mortality increased by 2.0 % annually (95 % CI: 0.8-3.5, p < 0.05) from 2005 until 2013, after which it declined by 5.1 % (95 % CI: -7.3 to -3.8, p < 0.05). CV mortality increased by 7.1 % annually (95 % CI: 4.6-9.4, p < 0.05) from 2005 to 2020. Trends were similar across sexes, with a non-significant higher APC in CV mortality noted among females from 2017 to 2020. CONCLUSIONS Our study shows despite the significant decrease in all-cause and melanoma-specific mortalities after 2010, likely reflecting the benefits of modern therapies, CV mortality continued to rise. These findings underscore the need for long-term surveillance and CV risk management in melanoma patients.
Collapse
Affiliation(s)
- Astha Prasai
- Department of Internal Medicine, Rochester Regional Health, Rochester, NY, USA
| | - Nischit Baral
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, USA
| | - Mohamad K Elajami
- Department of Internal Medicine, Hartford Hospital, Hartford, CT, USA
| | - Esha Vallabhaneni
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, USA
| | - Joshua D Mitchell
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Uzma Iqbal
- Department of Internal Medicine, Rochester Regional Health, Rochester, NY, USA; Department of Cardiology, Sands, Constellation Heart Institute, Rochester Regional Health, Rochester, NY, USA
| | - Hisham F Bahmad
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Francine K Welty
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tarec K Elajami
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, USA.
| |
Collapse
|
30
|
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
|
31
|
Giri VK, McDermott DF, Zaemes J. The emerging role of lymphocyte-activation gene 3 targeting in the treatment of solid malignancies. Cancer 2025; 131:e35892. [PMID: 40344213 DOI: 10.1002/cncr.35892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
PD-(L)1-based immune checkpoint inhibitor therapies have profoundly impacted the treatment of many solid malignancies. Although the addition of CTLA-4 checkpoint inhibitors can enhance anticancer activity, it also significantly increases the rate of immune-related adverse events. Therefore, there has been much interest in identifying additional immune checkpoints to improve the outcomes seen with PD-1-based therapy while minimizing additional side effects. One such target, lymphocyte-activation gene 3 (LAG-3), has long been recognized as an important inhibitor of T-cell function via modulation of the T-cell receptor pathway. Several drugs targeting LAG-3 have been developed, including most prominently the monoclonal antibody relatlimab. To date, the most significant demonstration of efficacy in targeting LAG-3 has been the use of relatlimab with the PD-1 inhibitor nivolumab in the treatment of advanced melanoma. The combination of nivolumab plus relatlimab is more efficacious compared to PD-1 inhibition alone, as has been previously seen with the combination of CTLA-4 inhibitor ipilimumab with nivolumab. However, nivolumab plus relatlimab offers a potentially more favorable toxicity profile. Here, the authors review the mechanism of the LAG-3 pathway and its rationale as a target for anticancer therapy as well as currently available data regarding the use of LAG-3 agents in treating melanoma and other solid tumors. Other investigational agents that target LAG-3 via novel mechanisms are also reviewed.
Collapse
Affiliation(s)
- Vinay K Giri
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - David F McDermott
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jacob Zaemes
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
32
|
Nazarova VV, Orlova KV, Magomedova ZR, Romanov DS, Maskalenka R, Yarovoy AA, Yarovaya VA, Trofimova OP, Shubina IZ, Demidov LV. Combined stereotactic radiation therapy and immunotherapy for metastatic uveal melanoma. Front Oncol 2025; 15:1567504. [PMID: 40406270 PMCID: PMC12094999 DOI: 10.3389/fonc.2025.1567504] [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: 01/27/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Aim Uveal melanoma (UM) is a rare primary intraocular malignant tumor with an extremely poor prognosis. Our study evaluated the feasibility to improve metastatic UM treatment outcomes with a combined approach of immunotherapy and radiation therapy. Methods The retrospective study enrolled 24 patients with metastatic uveal melanoma who had combined treatment with stereotactic radiation therapy (RT) and immune checkpoint inhibitor therapy. 35% of patients received combination immunotherapy, and the others received mono-immunotherapy with anti-PD-1 drugs. All patients underwent stereotactic RT for metastases in the liver (75% patients), bones (8%), soft tissues (8%), brain (4%), and lungs (4%). Results Overall response rate (ORR) was 39.1%. Complete response (CR) was achieved in 8.7% patients and partial response (PR) - in 30.4% patients, median progression free survival (PFS) was 11.6 months [95% confidence interval (CI), 5.4-14.4], and median overall survival (OS) was 27.6 months [95% CI, 16.9 - 49.1]. Conclusions The study has demonstrated a safe combination of stereotactic radiation therapy and immune checkpoint inhibitor immunotherapy in patients with metastatic uveal melanoma. The combination shows a potential treatment option for this patient cohort since no other effective therapies are available at present.
Collapse
Affiliation(s)
- Valeria V. Nazarova
- FSBI “N.N. Blokhin National Medical Research Center of Oncology”, Ministry of Health of the Russian Federation, Moscow, Russia
- Federal State Autonomous Educational Institution (FSAEI) of Higher Education «N.I. Pirogov Russian National Research Medical University» of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Kristina V. Orlova
- FSBI “N.N. Blokhin National Medical Research Center of Oncology”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Zakhra R. Magomedova
- FSBI “N.N. Blokhin National Medical Research Center of Oncology”, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | | | - Andrey A. Yarovoy
- Svyatoslav Nikolaevich (S.N.) Fedorov National Medical Research Center (NMRC) “Mezhotraslevoy Nauchno-Technichesky Komplex (MNTK) “Eye Microsurgery”, Moscow, Russia
| | - Vera A. Yarovaya
- Svyatoslav Nikolaevich (S.N.) Fedorov National Medical Research Center (NMRC) “Mezhotraslevoy Nauchno-Technichesky Komplex (MNTK) “Eye Microsurgery”, Moscow, Russia
| | - Oxana P. Trofimova
- FSBI “N.N. Blokhin National Medical Research Center of Oncology”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Irina Zh. Shubina
- FSBI “N.N. Blokhin National Medical Research Center of Oncology”, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Lev V. Demidov
- FSBI “N.N. Blokhin National Medical Research Center of Oncology”, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
33
|
von Bubnoff D, Schmitt C, Goldinger SM, Schadendorf D, Kähler KC, Hafner C, Kramer N, Fröhlich W, Dummer R, Berking C, Schliep S, Kirchberger MC, Heinzerling L. Prognostic and predictive value of IDO expression in metastatic melanoma treated with Ipilimumab. PLoS One 2025; 20:e0321937. [PMID: 40334245 PMCID: PMC12058187 DOI: 10.1371/journal.pone.0321937] [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: 06/27/2024] [Accepted: 03/11/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND The tumor microenvironment is crucial for prognosis and response to immunotherapy in several tumor entities. METHODS In a multicenter retrospective study, a total of 86 tumor samples from patients with metastatic melanoma were evaluated for baseline expression of indoleamine 2,3-dioxygenase (IDO) and programmed death ligand 1 (PD-L1). Expression patterns of IDO and PD-L1 on tumor cells and antigen-presenting cells (APCs) as determined by immunohistochemical (IHC) staining of paraffin-embedded tissue sections were correlated with response to ipilimumab and overall survival (OS). Statistical analysis was performed using the Spearman correlation, the Mann-Whitney test and Kaplan-Meier estimator. RESULTS IDO expression in tumor cells or APCs was not predictive for treatment response. The median OS was 26 months in IDO-positive and IDO-negative patients, regardless of IDO expression in tumor cells or APCs. A correlation of IHC expression scores of IDO and PD-L1 could not be documented. CONCLUSION The exact role of IDO in creating an immunosuppressive tumor environment and its reversal needs to be further elucidated.
Collapse
Affiliation(s)
| | - Christina Schmitt
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| | - Simone M. Goldinger
- Department of Dermatology, University Hospital of Zurich, Zürich, Switzerland
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Katharina C. Kähler
- Department of Dermatology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Christian Hafner
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Nora Kramer
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| | - Waltraud Fröhlich
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Zürich, Switzerland
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Stefan Schliep
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Michael C. Kirchberger
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Lucie Heinzerling
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| |
Collapse
|
34
|
Peeters E, van Genugten EAJ, Heskamp S, de Vries IJM, van Herpen C, Koenen HJPM, Kneilling M, van der Post RS, van Dop WA, Westdorp H, Aarntzen E. Exploring molecular imaging to investigate immune checkpoint inhibitor-related toxicity. J Immunother Cancer 2025; 13:e011009. [PMID: 40341021 PMCID: PMC12060888 DOI: 10.1136/jitc-2024-011009] [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: 12/09/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
Immune checkpoint inhibitors (ICI) boost the endogenous anticancer immunity, evoking long-lasting anticancer responses in a subset of patients with solid tumors. Simultaneously, ICI are also associated with serious toxicities, impacting treatment duration and the quality of life. The proposed processes underlying ICI-related toxicity include T-cell activation and recruitment to non-tumor tissues, involvement of other immune cells and fibroblasts and the host' microbiome composition. However, the exact mechanisms of these processes remain incompletely understood, hindering clinicians' ability to predict and identify ICI-related toxicity in the early stages of treatment. Molecular imaging may play a role as a non-invasive biomarker, providing a tool to study ICI-related toxicity. This review discusses the applications of molecular imaging to answer questions regarding the mechanisms, detection, and prediction of ICI-related toxicity. Potential targets and the current state of development of suitable imaging techniques are discussed.
Collapse
Affiliation(s)
- Eva Peeters
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Sandra Heskamp
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carla van Herpen
- Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tubingen, Baden-Württemberg, Germany
- Department of Dermatology, University of Tübingen, Tubingen, Baden-Württemberg, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls Universität Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Rachel S van der Post
- Department of Pathology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Willemijn A van Dop
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Harm Westdorp
- Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik Aarntzen
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Nuclear Medicine, Eberhard Karls Universität Tübingen, Tübingen, Baden-Württemberg, Germany
- Department of Nuclear Medicine and Molecular Imaging, University Medical Centre Groningen, Groningen, Groningen, The Netherlands
| |
Collapse
|
35
|
Zhang ZF, Zhang Y, Chen YW, Cao GS, Zheng XD, Sun R, Peng H, Tian ZG, Sun HY. CD200R blockade enhances anti-tumor immunity by unleashing NK and CD8 + T cells in tumor. Acta Pharmacol Sin 2025:10.1038/s41401-025-01556-0. [PMID: 40329005 DOI: 10.1038/s41401-025-01556-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/30/2025] [Indexed: 05/08/2025]
Abstract
Immune checkpoint inhibitors have revolutionized cancer therapy, but a large proportion of patients do not respond well to current checkpoint immunotherapies. CD200R (also known as OX2R) is a transmembrane glycoprotein of the immunoglobulin superfamily that is mainly expressed on myeloid and lymphoid-derived immunocompetent cells such as myeloid cells, natural killer (NK), and CD8+ T cells. In this study, we investigated the therapeutic potential and cellular mechanisms of targeting CD200R in tumor immunotherapy. We established 4 subcutaneous tumor mouse models using MC38 (colon cancer), MCA205 (fibrosarcoma), LLC (lung cancer), and EO771 (mammary cancer) cell lines. We found that CD200R was highly expressed on tumor-infiltrating NK and CD8+ T cells with exhausted phenotypes in the four subcutaneous tumor mouse models. Either genetic ablation or antibody blockade of CD200R retarded tumor growth and prolonged the survival of tumor-bearing mice by preventing or reversing exhaustion of both NK cells and CD8+ T cells. The combined therapy of CD200R antibody with anti-PD-1/anti-PD-L1 synergistically inhibited tumor growth. By depletion of NK or/and CD8+ T cells, we demonstrated that both cell types contributed to the anti-tumor efficacy of CD200R blockade in tumor-bearing mice. Further, the blockade of human CD200R significantly enhanced human NK cell function and inhibited human tumor growth in PBMC-reconstituted xenograft mice. Our results demonstrate that CD200R is a potential immune checkpoint molecule that can suppress the tumoricidal activities of NK and CD8+ T cells, and could thus be exploited as a therapeutic target in the future.
Collapse
Affiliation(s)
- Zheng-Feng Zhang
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Yu Zhang
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ya-Wen Chen
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Guo-Shuai Cao
- Hefei TG ImmunoPharma Corporation Limited, Hefei, 230027, China
| | - Xiao-Dong Zheng
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Rui Sun
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hui Peng
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Zhi-Gang Tian
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Hao-Yu Sun
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
36
|
Paun RA, Li L, Mouncef A, Radzioch D, Tabrizian M. Liposome-Polymer Nanoparticles Loaded with Copper Diethyldithiocarbamate and 6-Bromo-Indirubin-3'-Oxime Enable the Treatment of Refractive Melanoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2409012. [PMID: 40317886 DOI: 10.1002/smll.202409012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/26/2025] [Indexed: 05/07/2025]
Abstract
Despite significant advances in cancer immunotherapy, many patients fail to respond to current treatments, outlining the need to develop novel therapeutic modalities. Therapeutic resistance in cancer cells is mediated by significant genomic instability due to their oncogenic transformation and evolutionary pressures inside the tumor microenvironment (TME). However, these cellular and molecular adaptations can result in a significant increase in the baseline endoplasmic reticulum (ER) stress in TME-resident cells. This can be taken advantage of as a therapeutic strategy by using the metal chelate copper diethyldithiocarbamate (CuET), a potent inhibitor of the p97-UFD1-NPL4 protein complex to induce cytotoxicity and exacerbate ER stress in cancer cells. Here, CuET is combined with the anti-inflammatory drug 6-bromo-indirubin-3'-oxime (BIO), a potent GSK3 inhibitor, to modulate the aberrant inflammatory response inside the TME. However, both CuET and BIO are highly hydrophobic and exhibit poor bioavailability, requiring the development of an appropriate carrier. Herein, it is demonstrated that CuET and BIO can be efficiently loaded into liposomes that are stabilized by poly(vinylpyrrolidone). The liposome-loaded drug combination resulted in a significant decrease of 47% and 76% in the tumor burden of syngeneic B16F10 and YUMM1.7 mouse models, respectively, without any major acute toxicity.
Collapse
Affiliation(s)
- Radu A Paun
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, 3775 University Street, Montreal, QC, H3A 2B6, Canada
- Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Ling Li
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, 3775 University Street, Montreal, QC, H3A 2B6, Canada
| | - Adam Mouncef
- Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Danuta Radzioch
- Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
- Department of Human Genetics, Faculty of Medicine and Health Sciences, 3640 University Street, Montreal, QC, H3A 0C7, Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, 3775 University Street, Montreal, QC, H3A 2B6, Canada
- Faculty of Dentistry and Oral Health Sciences, McGill University, McGill University, 2001 McGill College Ave, Montreal, QC, H3A 1G1, Canada
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
Chang CW, Yang ST, Liu CH, Chang WH, Lee WL, Wang PH. Endometrial cancer: Part II. Multimodality treatment of uterine high-grade serous carcinoma (clinical course I). Taiwan J Obstet Gynecol 2025; 64:413-424. [PMID: 40368509 DOI: 10.1016/j.tjog.2024.03.034] [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] [Accepted: 03/03/2024] [Indexed: 05/16/2025] Open
Abstract
Endometrial cancer (EC) is the most frequently diagnosed gynecological malignancy with rapid growth of incidence in the high-income countries and Taiwan. Since the integration of modern molecular pathology into traditional clinico-pathology for the diagnosis and classification of EC, the treatment is apparently switched to more precise molecular-guided or -targeted therapy, having been reviewed in the part I (2022). The current review is the part II describing the previous history (clinical course I) of the complex clinical course occurred in a 66-year-old woman with uterine high-grade serous carcinoma (HGSC, post-curettage diagnosis), who was treated with robotic staging surgery without additional postoperative adjuvant therapy due to absence of any residual malignancy in February 2019 (Rationales and controversial issues 1-5). Recurrences at the multiple sites, including vaginal cuff, liver, adrenal and lung metastases occurred in December 2020. Suboptimal cytoreductive surgery and following 6 cycles of paclitaxel-carboplatin regimen between January and April 2021 achieved nearly complete remission (Rationales 6,7 and controversial issues 6,7 for the first recurrence). However, this equivocal clinical situation made the following treatment in confusion. Using positron emission tomography/magnetic resonance image (MRI) or computed tomography (CT) not only serving as a valuable tool for detecting occult metastatic lesions but also giving an answer for uncertain clinical diagnosis provided the additional therapy (Rationales 8-10 and Controversial issues 7-9). This case highlights an aggressive nature of uterine HGSC, even though the initial diagnosis was an early-stage disease (no residual tumor) and the hospital shopping makes the clinical course much complex. This part II (Clinical course I) has explored the first journey from the initial diagnosis and initial treatment at the hospital "A" and "B" to the subsequent management of the recurrent disease at the hospitals "C,D,E,F", including the rationale and controversial issues in the clinical course I.
Collapse
Affiliation(s)
- Che-Wei Chang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Szu-Ting Yang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Hao Liu
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Hsun Chang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Female Cancer Foundation, Taipei, Taiwan
| | - Wen-Ling Lee
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Female Cancer Foundation, Taipei, Taiwan; Department of Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan.
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Female Cancer Foundation, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
40
|
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
|
41
|
Ren F, Pang X, Liu N, Zhu L. Multiomics evaluation and machine learning optimize molecular classification, prediction of prognosis and immunotherapy response for ovarian cancer. Pathol Res Pract 2025; 269:155925. [PMID: 40168774 DOI: 10.1016/j.prp.2025.155925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/18/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Ovarian cancer (OC), owing to its substantial heterogeneity and high invasiveness, has historically been devoid of precise, individualized treatment options. This study aimed to establish integrated consensus subtypes of OC using different multiomics integration methodologies. METHODS We integrated five distinct multiomics datasets from multicentric cohorts to identify high-resolution molecular subgroups using a combination of 10 and 101 clustering and machine learning algorithms, respectively, to develop a robust consensus multiomics-related machine learning signature (CMMS). RESULTS Two cancer subtypes with prognostic significance were identified using multiomics clustering analysis. 10 essential genes were identified in the CMMS. Patients in the high CMMS group exhibited a poorer prognosis, with a "cold tumor" phenotype and an immunosuppressive state with reduced immune cell infiltration. In contrast, patients in the low CMMS group exhibited a more favorable prognosis, with immune activation and a "hot tumor" phenotype characterized by increased tumor mutation burden, tumor neoantigen burden, PD-L1 expression, and enriched M1 macrophages. Eight independent immunotherapy datasets were validated to further corroborate our findings regarding patients in the low CMMS group who responded better to immunotherapy. CONCLUSIONS CMMS detection has significant utility in the prognosis of patients at an early stage and identification of potential candidates for immunotherapy.
Collapse
Affiliation(s)
- Fang Ren
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Xiaoao Pang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Ning Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Liancheng Zhu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
42
|
Sam I, Benhamouda N, Biard L, Da Meda L, Desseaux K, Baroudjan B, Nakouri I, Renaud M, Sadoux A, Alkatrib M, Deleuze JF, Battistella M, Shen Y, Resche-Rigon M, Mourah S, Lebbe C, Tartour E. Soluble CD27 differentially predicts resistance to anti-PD1 alone but not with anti-CTLA-4 in melanoma. EMBO Mol Med 2025; 17:909-922. [PMID: 40148586 DOI: 10.1038/s44321-025-00203-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/25/2025] [Accepted: 02/10/2025] [Indexed: 03/29/2025] Open
Abstract
Metastatic melanoma can be treated with anti-PD-1 monotherapy or in combination with anti-CTLA-4 or anti-Lag3. However, combination therapy is associated with a high risk of toxicity. Recently, we reported that high plasma soluble CD27 (sCD27) levels reflect the intratumoral interaction of CD70-CD27 and dysfunctional T cells in the tumor microenvironment of renal cell carcinoma. In this study, we first characterized the intratumoral expression of CD70 and CD27 in melanoma tumors and their interaction in vivo. We then reported a significant association between baseline sCD27 and anti-PD-1 resistance as assessed by progression-free survival, overall survival, or 12-month complete response in two prospective cohorts of melanoma patients. Multivariate analysis confirmed that sCD27 was independently associated with clinical outcomes. Notably, sCD27 did not predict clinical response to combination therapy in either cohort. This differential predictive value of sCD27 for the two therapeutic options was later confirmed by propensity score analysis. Our results suggest that high plasma sCD27 levels predict poorer efficacy of anti-PD1 monotherapy in metastatic melanoma, justifying therapeutic escalation with a combination of anti-PD1 and anti-CTLA-4.
Collapse
Affiliation(s)
- Ikuan Sam
- Universite Paris Cite, INSERM, PARCC, Paris, France
- Department of Immunology, APHP, Hôpital Europeen Georges Pompidou (HEGP)-Hôpital Necker, Paris, France
| | - Nadine Benhamouda
- Universite Paris Cite, INSERM, PARCC, Paris, France
- Department of Immunology, APHP, Hôpital Europeen Georges Pompidou (HEGP)-Hôpital Necker, Paris, France
| | - Lucie Biard
- APHP, Department of Biostatistics and Medical Information, APHP, Saint-Louis Hospital, Paris, INSERM, UMR-1153, ECSTRRA Team, Paris, France
| | - Laetitia Da Meda
- Universite Paris Cité, APHP Dermato-Oncology, Cancer Institute AP-HP, Nord Paris Cité, INSERM U976, Saint Louis Hospital Paris, Paris, France
| | - Kristell Desseaux
- APHP, Department of Biostatistics and Medical Information, APHP, Saint-Louis Hospital, Paris, INSERM, UMR-1153, ECSTRRA Team, Paris, France
| | - Barouyr Baroudjan
- Universite Paris Cité, APHP Dermato-Oncology, Cancer Institute AP-HP, Nord Paris Cité, INSERM U976, Saint Louis Hospital Paris, Paris, France
| | - Ines Nakouri
- Universite Paris Cité, APHP Dermato-Oncology, Cancer Institute AP-HP, Nord Paris Cité, INSERM U976, Saint Louis Hospital Paris, Paris, France
| | - Marion Renaud
- Universite Paris Cité, APHP Dermato-Oncology, Cancer Institute AP-HP, Nord Paris Cité, INSERM U976, Saint Louis Hospital Paris, Paris, France
| | - Aurélie Sadoux
- Department of Pharmacology and Tumor Genomics, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Jean-François Deleuze
- Fondation Jean Dausset-CEPH (Centre d'Etude du Polymorphisme Humain), CEPH-Biobank, Paris, France
| | - Maxime Battistella
- Department of Pathology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Yimin Shen
- Fondation Jean Dausset-CEPH (Centre d'Etude du Polymorphisme Humain), CEPH-Biobank, Paris, France
| | - Matthieu Resche-Rigon
- APHP, Department of Biostatistics and Medical Information, APHP, Saint-Louis Hospital, Paris, INSERM, UMR-1153, ECSTRRA Team, Paris, France
| | - Samia Mourah
- Department of Pharmacology and Tumor Genomics, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Cité, INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), Paris, France
| | - Celeste Lebbe
- Universite Paris Cité, APHP Dermato-Oncology, Cancer Institute AP-HP, Nord Paris Cité, INSERM U976, Saint Louis Hospital Paris, Paris, France.
| | - Eric Tartour
- Universite Paris Cite, INSERM, PARCC, Paris, France.
- Department of Immunology, APHP, Hôpital Europeen Georges Pompidou (HEGP)-Hôpital Necker, Paris, France.
| |
Collapse
|
43
|
Maul LV, Ramelyte E, Dummer R, Mangana J. Management of metastatic melanoma with combinations including PD-1 inhibitors. Expert Opin Biol Ther 2025; 25:1-12. [PMID: 40159098 DOI: 10.1080/14712598.2025.2485315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Melanoma is among the most immunogenic malignancies. The advent of immune checkpoint inhibitors (ICIs) has revolutionized the landscape of melanoma treatment. Long-term durable cancer control is possible in nearly 50% of non-resectable, metastatic melanoma patients with anti-CTLA4 and anti-PD-1 antibodies. AREAS COVERED This review provides a critical overview of the current data and future research directions on the management of metastatic melanoma with ICIs. We reviewed the efficacy and safety of combinations with PD-1 inhibitors through PubMed database research (Nov 2024-Mar 2025). EXPERT OPINION A decade after ipilimumab's approval, challenges remain. To cure more patients, the development of combinations is warranted. Combinations with a limited number of ipilimumab applications improve the overall survival outcome by approximately 10%, with a dramatic increase in adverse events including fatal events. Anti-LAG3/nivolumab is a promising alternative, offering similar efficacy to ipilimumab/nivolumab with better tolerability. In our opinion, ipilimumab/nivolumab combination should be the first-line therapy for high-risk patients (high LDH, brain or liver metastasis), while nivolumab/relatlimab or PD-1 monotherapy may be preferable for lower-risk cases. However, treatment decisions are increasingly complex, since most patients nowadays are pretreated in the (neo)-adjuvant setting. The key limitation today is the lack of biomarkers to guide individualized treatment strategies.
Collapse
Affiliation(s)
- Lara Valeska Maul
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
Lowe MC, Delman KA, Ollila DW. The Landmark Series: Melanoma and Adjuvant Therapy. Ann Surg Oncol 2025; 32:3212-3215. [PMID: 40067605 DOI: 10.1245/s10434-025-17102-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 02/17/2025] [Indexed: 04/24/2025]
Affiliation(s)
- Michael C Lowe
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA.
| | - Keith A Delman
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - David W Ollila
- Department of Surgery, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
45
|
Chang CW, Yang ST, Chang WH, Lee WL, Wang PH. Endometrial cancer: Part II. Multimodality treatment of uterine high-grade serous carcinoma (clinical course II). Taiwan J Obstet Gynecol 2025; 64:425-433. [PMID: 40368510 DOI: 10.1016/j.tjog.2025.04.001] [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] [Accepted: 03/19/2025] [Indexed: 05/16/2025] Open
Abstract
The part II (clinical course I) of endometrial cancer (EC) describing the complex clinical course (initial treatment and therapy for the first recurrence at both local-regional and distant metastatic sites) of a 66-year-old woman with uterine high-grade serous carcinoma (HGSC, post-curettage diagnosis) after minimally invasive surgery (MIS) treatment-confirmed absence of residual tumor. In the clinical course I, we discussed the recent trend in using MIS for treating early-stage EC patients, regardless whether histologic types are classified, as well as the rationale of absent following postoperative adjuvant therapy due to no residual tumor of gross specimen after complete staging surgery. The patient had multiple recurrences at both loco-regional and distant sites after 22-month disease-free survival (DFS). After salvage therapy, including incomplete cytoreductive surgery for symptom control, chemotherapy, radiotherapy and final systemic therapy, the patient had a durable response period (10-month treatment plus 9 months of the 2nd progression-free survival [PFS]). The current report is the subsequent clinical course of this patient (clinical course II), which further managed this patient at the second recurrence based on multiple modality treatment to achieve the durable response and significantly improve the quality of life.
Collapse
Affiliation(s)
- Che-Wei Chang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Szu-Ting Yang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Hsun Chang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Female Cancer Foundation, Taipei, Taiwan
| | - Wen-Ling Lee
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Female Cancer Foundation, Taipei, Taiwan; Department of Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan.
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Female Cancer Foundation, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
46
|
Ren D, Tran TB, Chow WA, Barnachea S, Wei K, Lee BA. Complete remission of locoregionally metastatic melanoma after one single dose of pembrolizumab: A case report. JAAD Case Rep 2025; 59:122-126. [PMID: 40290795 PMCID: PMC12022409 DOI: 10.1016/j.jdcr.2025.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Affiliation(s)
- Dong Ren
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, California
| | - Thuy B. Tran
- Division of Surgical Oncology, University of California, Irvine, Irvine, California
| | - Warren A. Chow
- Division of Hematology/Oncology, University of California, Irvine, Irvine, California
| | - Shelby Barnachea
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, California
| | - Katherine Wei
- Department of Radiology, University of California, Irvine, Irvine, California
| | - Bonnie A. Lee
- Departments of Dermatology and Pathology, University of California, Irvine, Irvine, California
| |
Collapse
|
47
|
Deshmukh MG, Brooks VT, Roy SF, Milette S, Bosenberg M, Micevic G. DNA methylation in melanoma immunotherapy: mechanisms and therapeutic opportunities. Clin Epigenetics 2025; 17:71. [PMID: 40307913 PMCID: PMC12044936 DOI: 10.1186/s13148-025-01865-5] [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: 10/24/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Abnormal DNA methylation is a hallmark of cancer and a nearly universal feature of melanoma. DNA methylation plays well-appreciated melanoma cell-intrinsic roles, including silencing tumor-suppressor genes, regulating genomic stability, deregulating expression of oncogenes to potentiate proliferative signaling and tumor migration. With the recent success of immunological therapies for melanoma, important roles for DNA methylation are also emerging at the interface between melanoma and immune cells with the potential to regulate the anti-tumor immune response. These newly recognized roles for DNA methylation in controlling melanoma cell immunogenicity, expression of MHC and immune checkpoint molecules as well as T cell phenotypes in the tumor microenvironment raise the possibility of using DNA methylation to develop improved therapies and methylation-based biomarkers. In addition to reviewing the "immune dimension" of DNA methylation, we summarize recent developments with potential clinical applications in melanoma, such as targeted DNA methylation editing, single-cell methylation approaches, and measurement of circulating methylated DNA. An improved understanding of the immune roles of DNA methylation presents an exciting opportunity for continued improvement of care and outcomes for patients with melanoma.
Collapse
Affiliation(s)
- Maya G Deshmukh
- Medical Scientist Training Program (MD-PhD), Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Veronica T Brooks
- Department of Dermatology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Simon F Roy
- Department of Dermatology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Simon Milette
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Marcus Bosenberg
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale Center for Immuno-Oncology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Goran Micevic
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, 06520, USA.
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520, USA.
- Department of Dermatology, Yale School of Medicine, New Haven, CT, 06520, USA.
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
48
|
Awad MM, Forde PM, Girard N, Spicer J, Wang C, Lu S, Mitsudomi T, Felip E, Broderick SR, Swanson SJ, Brahmer J, Kerr K, Saylors GB, Chen KN, Gharpure V, Neely J, Balli D, Hu N, Provencio Pulla M. Neoadjuvant Nivolumab Plus Ipilimumab Versus Chemotherapy in Resectable Lung Cancer. J Clin Oncol 2025; 43:1453-1462. [PMID: 39778121 PMCID: PMC12005868 DOI: 10.1200/jco-24-02239] [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: 10/09/2024] [Accepted: 10/31/2024] [Indexed: 01/11/2025] Open
Abstract
PURPOSE Neoadjuvant immune checkpoint blockade with nivolumab plus ipilimumab improves overall survival (OS) in non-small cell lung cancer (NSCLC); however, randomized data for resectable lung cancer are limited. We report results from the exploratory concurrently randomized nivolumab plus ipilimumab and chemotherapy arms of the international phase III CheckMate 816 trial. METHODS Adults with stage IB-IIIA (American Joint Committee on Cancer seventh edition) resectable NSCLC received three cycles of nivolumab once every 2 weeks plus one cycle of ipilimumab or three cycles of chemotherapy (on day 1 or days 1 and 8 of each 3-week cycle) followed by surgery. Analyses included event-free survival (EFS), OS, pathologic response, surgical outcomes, biomarker analyses, and safety. RESULTS A total of 221 patients were concurrently randomly assigned to nivolumab plus ipilimumab (n = 113) or chemotherapy (n = 108). At a median follow-up of 49.2 months, the median EFS was 54.8 months (95% CI, 24.4 to not reached [NR]) with nivolumab plus ipilimumab versus 20.9 months (95% CI, 14.2 to NR) with chemotherapy (HR, 0.77 [95% CI, 0.51 to 1.15]); 3-year EFS rates were 56% versus 44%. Higher rates of EFS events were initially seen, with later benefit favoring nivolumab plus ipilimumab; 3-year OS rates were 73% versus 61% (HR, 0.73 [95% CI, 0.47 to 1.14]); pathologic complete response rates were 20.4% versus 4.6%, respectively. In the respective arms, 83 (74%) and 82 patients (76%) underwent definitive surgery. Grade 3-4 treatment-related adverse events occurred in 14% and 36% of patients, respectively. CONCLUSION Neoadjuvant nivolumab plus ipilimumab showed potential long-term clinical benefit versus chemotherapy, despite early crossing of EFS curves in the preoperative phase and a lower rate of high-grade toxicity.
Collapse
Affiliation(s)
| | - Patrick M. Forde
- The Bloomberg–Kimmel Institute for Cancer Immunotherapy, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD
| | - Nicolas Girard
- Institut du Thorax Curie-Montsouris, Institut Curie, Paris, France
| | | | - Changli Wang
- Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | | | - Enriqueta Felip
- Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology, Universitat Autònoma Barcelona, Barcelona, Spain
| | - Stephen R. Broderick
- The Bloomberg–Kimmel Institute for Cancer Immunotherapy, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD
| | | | - Julie Brahmer
- The Bloomberg–Kimmel Institute for Cancer Immunotherapy, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD
| | - Keith Kerr
- Aberdeen Royal Infirmary, Aberdeen, United Kingdom
| | | | - Ke-Neng Chen
- Peking University School of Oncology, Beijing Cancer Hospital, Beijing, China
| | | | | | | | - Nan Hu
- Bristol Myers Squibb, Princeton, NJ
| | | |
Collapse
|
49
|
Bonzano E, Barruscotti S, Chiellino S, Montagna B, Bonzano C, Imarisio I, Colombo S, Guerrini F, Saddi J, La Mattina S, Tomasini CF, Spena G, Pedrazzoli P, Lancia A. Current Treatment Paradigms for Advanced Melanoma with Brain Metastases. Int J Mol Sci 2025; 26:3828. [PMID: 40332507 PMCID: PMC12027546 DOI: 10.3390/ijms26083828] [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: 02/15/2025] [Revised: 04/12/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
The therapeutic management of melanoma brain metastases has undergone a profound revolution during recent decades. Optimal integration of systemic therapies with local treatments seems to represent the strategy to pursue in order to maximize clinical outcomes, stressing the need for real multidisciplinary care in this setting of patients. However, the current approach in the clinics does not necessarily reflect what the current guidelines state, and several pending issues are present, from the ideal therapeutic sequence between stereotactic radiosurgery (SRS) and drug administration to the current role of surgery and whole brain radiotherapy (WBRT), all of which need to be addressed. This narrative review aims to provide practical help for navigating the current controversies, with an eye towards possible future advancements in the field, which could help to obtain a comprehensive molecular characterization of the tumor and a more personalized patient-centered therapeutic approach.
Collapse
Affiliation(s)
- Elisabetta Bonzano
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (S.L.M.)
| | | | - Silvia Chiellino
- Unit of Oncology, Department of Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (B.M.)
| | - Benedetta Montagna
- Unit of Oncology, Department of Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (B.M.)
| | - Chiara Bonzano
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, IRCCS Ospedale Policlinico San Martino, University Eye Clinic, 16132 Genoa, Italy
| | - Ilaria Imarisio
- Unit of Oncology, Department of Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (B.M.)
| | - Sara Colombo
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (S.L.M.)
| | - Francesco Guerrini
- Unit of Neurosurgery, Department of Head & Neck Surgery, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (F.G.)
| | - Jessica Saddi
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (S.L.M.)
| | - Salvatore La Mattina
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (S.L.M.)
| | | | - Giannantonio Spena
- Unit of Neurosurgery, Department of Head & Neck Surgery, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (F.G.)
| | - Paolo Pedrazzoli
- Unit of Oncology, Department of Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (B.M.)
| | - Andrea Lancia
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy (S.L.M.)
| |
Collapse
|
50
|
Babiker R, Wali AF, El-Tanani M, Rabbani SA, Rangraze I, Satyam SM, Patni MA, El-Tanani Y. Comparative Efficacy of Immune Checkpoint Inhibitors and Therapeutic Vaccines in Solid Tumors: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Vaccines (Basel) 2025; 13:423. [PMID: 40333324 PMCID: PMC12030876 DOI: 10.3390/vaccines13040423] [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: 02/24/2025] [Revised: 04/02/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
Background: Immune checkpoint inhibitors (ICIs) and therapeutic vaccines have emerged as promising immunotherapeutic strategies for solid tumors. However, their comparative efficacy in improving overall survival (OS) remains unclear. This systematic review and meta-analysis aimed to evaluate the efficacy of ICIs and therapeutic vaccines in improving OS in patients with solid tumors. Methods: A comprehensive search was conducted across PubMed, Cochrane Library, Embase, and Clinical Trials.gov for randomized controlled trials (RCTs) published between 1 January 2010 and 31 December 2024. Studies comparing ICIs or therapeutic vaccines against control treatments (placebo, standard of care, or active comparators) in adults with solid tumors were included. The primary outcome was OS, and data were pooled using RevMan (web). Risk of bias was assessed using the Cochrane Risk of Bias tool. Results: Thirteen RCTs involving 10,991 participants were included. A total of 5722 of them were treated with therapeutic vaccines or checkpoint inhibitors. Therapeutic vaccines demonstrated insignificant improvement in OS, with a pooled mean difference of 1.89 months (95% CI: -0.54-4.31; P = 0.13), although with homogeneity (I2 = 0%). ICIs showed a statistically significant OS benefit, with a pooled mean difference of 1.32 months (95% CI: 0.62-2.02; P = 0.0002) and low heterogeneity (I2 = 12%). Conclusions: Therapeutic vaccines provide a larger but less consistent benefit, whereas ICIs offer modest but more consistent survival advantage. These findings support the need for personalized immunotherapy approaches as well as further research to identify predictive biomarkers and optimize treatment strategies by acquiring deep insights into the TME dynamic and behaviors.
Collapse
Affiliation(s)
- Rasha Babiker
- Department of Physiology, RAK College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| | - Adil Farooq Wali
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates; (A.F.W.); (S.A.R.)
| | - Mohamed El-Tanani
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates; (A.F.W.); (S.A.R.)
| | - Syed Arman Rabbani
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates; (A.F.W.); (S.A.R.)
| | - Imran Rangraze
- Department of Internal Medicine, RAK College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates;
| | - Shakta Mani Satyam
- Department of Pharmacology, RAK College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates;
| | - Mohamed Anas Patni
- Department of Community Medicine, RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras al Khaimah P.O. Box 11172, United Arab Emirates;
| | | |
Collapse
|