1
|
Le J, Sun Y, Deng G, Dian Y, Xie Y, Zeng F. Immune checkpoint inhibitors in cancer patients with autoimmune disease: Safety and efficacy. Hum Vaccin Immunother 2025; 21:2458948. [PMID: 39894761 PMCID: PMC11792813 DOI: 10.1080/21645515.2025.2458948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/09/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025] Open
Abstract
The utilization of immune-checkpoint inhibitors (ICIs) in cancer immunotherapy frequently leads to the occurrence of immune-related adverse events (irAEs), making it generally not recommended for patients with preexisting autoimmune diseases. Hence, we conducted a meta-analysis on safety and efficacy of ICIs in cancer patients with preexisting autoimmune diseases to provide further insights. PubMed, EMBASE, and Cochrane Library were systematically searched until December 20, 2024. The main summary measures used were pooled rate and risk ratio (RR) with 95% confidential interval (CI), which were analyzed using R statistic software. A total of 52 articles were included in the study. When cancer patients with preexisting autoimmune diseases received ICIs treatment, the overall incidence was 0.610 (95% CI: 0.531-0.686) for any grade irAEs, 0.295 (95% CI: 0.248-0.343) for flares, 0.325 (95% CI: 0.258-0.396) for de novo irAEs, 0.238 (95% CI: 0.174-0.309) for grade ≥3 irAEs, and 0.143 (95% CI: 0.109-0.180) for discontinuation due to immunotoxicity. Compared with those without autoimmune diseases, cancer patients with autoimmune diseases experienced a higher risk of any-grade irAEs (RR: 1.23, 95% CI: 1.12-1.35) and discontinuation due to immunotoxicity (1.40, 95% CI: 1.11-1.78). However, no statistically significant differences were observed in the incidence of grade ≥3 irAEs, objective response rate (ORR), disease control rate (DCR), overall survival (OS), and progression-free survival (PFS) between the two groups. During ICIs treatment, irAEs are common among cancer patients with autoimmune diseases, but severe irAEs is relatively low. ICIs are effective in this population, but should be strictly monitored when used to avoid immunotoxicity.
Collapse
Affiliation(s)
- Jiayuan Le
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Furong Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Furong Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Furong Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Furong Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanli Xie
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
Kuusisalo S, Iivanainen S, Koivunen JP. Association of anti-PD-(L)1 treatment duration to efficacy in advanced solid tumors: a single center retrospective study. Ann Med 2025; 57:2476729. [PMID: 40091413 PMCID: PMC11915729 DOI: 10.1080/07853890.2025.2476729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 11/28/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are a standard of care in multiple cancers. Only a minority benefits, thus, optimal use and treatment duration remain indistinct. While biomarkers albeit PD-L1 are scarce, declined performance status and cancer-related systemic inflammation detected by blood inflammatory markers such as C-reactive protein (CRP) have been linked to inferior prognosis. MATERIALS AND METHODS We investigated the association of limited anti-PD-(L)1 treatment duration to therapy efficacy in melanoma and non-small cell lung cancer (NSCLC) patients who received therapy in a non-curative setting in Oulu University Hospital 2014-2022. Baseline prognostic factors (e.g. ECOG, CRP, and PD-L1 for NSCLC) were collected. Progression-free (PFS), overall (OS), and IO-free survival were analyzed using the Kaplan-Meier and Cox regression methods. RESULTS 126 patients (NSCLC, n = 72; melanoma, n = 54) were included. Majority (n = 101) were treated in the first line. Objective response rate was 34.9%. The median (m) anti-PD-(L)1 treatment duration was 3.42 months (mo). The mPFS and mOS were 6.8 mo (CI 95% 4.4-9.3) and 19.1 mo (CI 95% 13.3-24.9). Of the baseline factors, ECOG and CRP retained their significance in multivariate analysis for PFS (HR 0.34, CI 95% 0.19-0.59; HR 0.34, CI 95% 0.22-054) and OS (HR 0.38, CI 95% 0.20-0.71; HR 0.29, CI 95% 0.17-0.49). No difference was observed in PFS (HR 1.40, CI 95% 0.68-2.90) or OS (HR 0.69, CI 95% 0.29-1.65) according to treatment duration (3-6mo vs. > 6 mo). Long median IO-free survival (10.2 months; CI 95%, 4.1-16.3) was detected. CONCLUSION We characterized an anti-PD-(L)1 treated advanced NSCLC and melanoma cohort in which treatment benefit occurs irrespective of treatment duration and long-term benefit is observed off-treatment.
Collapse
Affiliation(s)
- Saara Kuusisalo
- Department of Medical Oncology and Radiotherapy and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Sanna Iivanainen
- Department of Medical Oncology and Radiotherapy and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jussi P. Koivunen
- Department of Medical Oncology and Radiotherapy and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
3
|
Li G, Che X, Wang S, Liu D, Xie D, Jiang B, Zheng Z, Zheng X, Wu G. The role of cisplatin in modulating the tumor immune microenvironment and its combination therapy strategies: a new approach to enhance anti-tumor efficacy. Ann Med 2025; 57:2447403. [PMID: 39757995 PMCID: PMC11705547 DOI: 10.1080/07853890.2024.2447403] [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: 01/16/2024] [Revised: 05/27/2024] [Accepted: 11/23/2024] [Indexed: 01/07/2025] Open
Abstract
Cisplatin is a platinum-based drug that is frequently used to treat multiple tumors. The anti-tumor effect of cisplatin is closely related to the tumor immune microenvironment (TIME), which includes several immune cell types, such as the tumor-associated macrophages (TAMs), cytotoxic T-lymphocytes (CTLs), dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), and natural killer (NK) cells. The interaction between these immune cells can promote tumor survival and chemoresistance, and decrease the efficacy of cisplatin monotherapy. Therefore, various combination treatment strategies have been devised to enhance patient responsiveness to cisplatin therapy. Cisplatin can augment anti-tumor immune responses in combination with immune checkpoint blockers (such as PD-1/PD-L1 or CTLA4 inhibitors), lipid metabolism disruptors (like FASN inhibitors and SCD inhibitors) and nanoparticles (NPs), resulting in better outcomes. Exploring the interaction between cisplatin and the TIME will help identify potential therapeutic targets for improving the treatment outcomes in cancer patients.
Collapse
Affiliation(s)
- Guandu Li
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shijin Wang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Deqian Xie
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bowen Jiang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zunwen Zheng
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xu Zheng
- Department of Cell Biology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
4
|
Wiecken M, Machiraju D, Chakraborty S, Mayr EM, Lenoir B, Eurich R, Richter J, Pfarr N, Halama N, Hassel JC. The immune checkpoint LAG-3 is expressed by melanoma cells and correlates with clinical progression of the melanoma. Oncoimmunology 2025; 14:2430066. [PMID: 39716918 DOI: 10.1080/2162402x.2024.2430066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 12/25/2024] Open
Abstract
Immune checkpoint blockers have substantially improved prognosis of melanoma patients, nevertheless, resistance remains a significant problem. Here, intrinsic and extrinsic factors in the tumor microenvironment are discussed, including the expression of alternative immune checkpoints such as lymphocyte activation gene 3 (LAG-3) and T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3). While most studies focus on immune cell expression of these proteins, we investigated their melanoma cell intrinsic expression by immunohistochemistry in melanoma metastases of 60 patients treated with anti-programmed cell death protein 1 (PD-1) and/or anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) therapy, and correlated it with the expression of potential ligands, RNA sequencing data and clinical outcome. LAG-3 and TIM-3 were commonly expressed in melanoma cells. In the stage IV cohort, expression of LAG-3 was associated with M1 stage (p < 0.001) and previous exposure to immune checkpoint inhibitors (p = 0.029). Moreover, in the anti-PD-1 monotherapy treatment group patients with high LAG-3 expression by tumor cells tended to have a shorter progression-free survival (p = 0.088), whereas high expression of TIM-3 was associated with a significantly longer overall survival (p = 0.007). In conclusion, we provide a systematic analysis of melanoma cell intrinsic LAG-3 and TIM-3 expression, highlighting potential implications of their expression on patient survival.
Collapse
Affiliation(s)
- Melanie Wiecken
- Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Devayani Machiraju
- Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Shounak Chakraborty
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Eva-Maria Mayr
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Bénédicte Lenoir
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit "Applied Tumor Immunity"(TME unit), Heidelberg, Germany
| | - Rosa Eurich
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit "Applied Tumor Immunity"(TME unit), Heidelberg, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Division of Translational Immunotherapy, Heidelberg, Germany
| | - Jasmin Richter
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Nicole Pfarr
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Niels Halama
- German Cancer Research Center (DKFZ) Heidelberg, Division of Translational Immunotherapy, Heidelberg, Germany
- Department of Medical Oncology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica C Hassel
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
5
|
Hamza M, Wang S, Liu Y, Li K, Zhu M, Chen L. Unraveling the potential of bioengineered microbiome-based strategies to enhance cancer immunotherapy. Microbiol Res 2025; 296:128156. [PMID: 40158322 DOI: 10.1016/j.micres.2025.128156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
The human microbiome plays a pivotal role in the field of cancer immunotherapy. The microbial communities that inhabit the gastrointestinal tract, as well as the bacterial populations within tumors, have been identified as key modulators of therapeutic outcomes, affecting immune responses and reprogramming the tumor microenvironment. Advances in synthetic biology have made it possible to reprogram and engineer these microorganisms to improve antitumor activity, enhance T-cell function, and enable targeted delivery of therapies to neoplasms. This review discusses the role of the microbiome in modulating both innate and adaptive immune mechanisms-ranging from the initiation of cytokine production and antigen presentation to the regulation of immune checkpoints-and discusses how these mechanisms improve the efficacy of immune checkpoint inhibitors. We highlight significant advances with bioengineered strains like Escherichia coli Nissle 1917, Lactococcus lactis, Bifidobacterium, and Bacteroides, which have shown promising antitumor efficacy in preclinical models. These engineered microorganisms not only efficiently colonize tumor tissues but also help overcome resistance to standard therapies by reprogramming the local immune environment. Nevertheless, several challenges remain, such as the requirement for genetic stability, effective tumor colonization, and the control of potential safety issues. In the future, the ongoing development of genetic engineering tools and the optimization of bacterial delivery systems are crucial for the translation of microbiome-based therapies into the clinic. This review highlights the potential of bioengineered microbiota as an innovative, personalized approach in cancer immunotherapy, bringing hope for more effective and personalized treatment options for patients with advanced malignancies.
Collapse
Affiliation(s)
- Muhammad Hamza
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| | - Yike Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Kun Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Motao Zhu
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
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
|
Joshi UM, Hundal J, Mata JR, Schollenberger MD, Warrier G, Luke JJ, Lipson EJ, Funchain P. Beyond Checkpoint Inhibition: Keeping Therapeutic Options Open. Am Soc Clin Oncol Educ Book 2025; 45:e473856. [PMID: 40233298 DOI: 10.1200/edbk-25-473856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Combination immune checkpoint inhibitor therapy (ICI) with ipilimumab (anti-cytotoxic T-lymphocyte-associated protein 4) + nivolumab (anti-PD-1) in untreated, metastatic melanoma has achieved a ten-year melanoma-specific survival of 52%. However, approximately 40%-55% of patients with metastatic melanoma have primary resistance and do not initially respond to anti-PD-1, and an additional 25% of patients develop secondary resistance, exhibiting an initial response followed by disease progression. In PD-1-refractory melanoma, treatment options are limited. Addition of ipilimumab, relatlimab (anti-LAG3), or lenvatinib (VEGFR TKI) has minimal to modest efficacy. Switching to targeted BRAF/MEK inhibition improves survival for BRAF-mutant disease. MEK and KIT inhibitors have limited activity in NRAS- and KIT-mutant metastatic melanoma, respectively. Recently, personalized, autologous tumor-infiltrating lymphocyte therapy has become a US Food and Drug Administration-approved second-line option; lifileucel demonstrates durable response (approximately 30%) in heavily pretreated, metastatic melanoma. Emerging therapeutics that show promising clinical benefit in ongoing clinical trials include novel engineered oncolytic viral and human leukocyte antigen (HLA)-restricted immune-mediated T-cell therapies. As a therapy which is limited to patients who are HLA-A*02:01, T-cell receptor (TCR) engineered T cells (TCR-T) iterates on personalized adoptive cell transfer, and immune mobilizing monoclonal TCRs against cancer are CD3 bispecifics that bind glycoprotein 100 (tebentafusp, approved for metastatic uveal melanoma) or PRAME to activate T cells. Finally, in patients at high risk for immune-related adverse events (irAEs), ICI should still be considered. ICI may be given with modified immunosuppression in patients with autoimmune disease or previous organ transplantation. Cumulative data support safe administration in older patients and in ICI rechallenge for patients with previous irAE.
Collapse
Affiliation(s)
- Urvashi Mitbander Joshi
- Division of Malignant Hematology and Medical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Jasmin Hundal
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | | | - Megan D Schollenberger
- Department of Oncology, Johns Hopkins University, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Govind Warrier
- Department of Oncology, Johns Hopkins University, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jason J Luke
- Division of Malignant Hematology and Medical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Evan J Lipson
- Department of Oncology, Johns Hopkins University, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Pauline Funchain
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Amrane K, Le Noac'h P, Hemon P, Abgral R, Le Meur C, Pradier O, Misery L, Legoupil D, Berthou C, Uguen A. MHC class II: a predictor of outcome in melanoma treated with immune checkpoint inhibitors. Melanoma Res 2025; 35:176-186. [PMID: 39945603 DOI: 10.1097/cmr.0000000000001022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
This study aimed to evaluate the predictive value of MHC class II (MHC-II) expression by melanoma cells in a large cohort of metastatic cutaneous melanoma patients treated with immune checkpoint inhibitors (ICIs). We conducted a single-center, retrospective study involving stage IV cutaneous melanoma patients who received ICI as first-line therapy. MHC-II expression in melanoma cells was quantified using dual-color anti-SOX10 and anti-MHC-II immunohistochemistry on tumor samples from 95 patients. The primary endpoint was event-free survival (EFS), with secondary endpoints including 1-year EFS, 1-year overall survival (OS), disease control rate (DCR), and the correlation between MHC-II expression and clinico-biological characteristics. The cohort had a median age of 67 years (range, 33-90), with a male-to-female ratio of 50 : 45. Thirty-three percent of patients received the ipilimumab-nivolumab combination. The median follow-up was 16.8 months. Disease progression occurred in 58 patients (61%), with a median time to progression of 4.8 months. Forty-six patients (48.4%) experienced an event within the first year, and 52 patients (54.7%) died during follow-up. MHC-II positivity was observed in ≥10% of melanoma cells in 6.3% of patients. MHC-II expression was significantly associated with 1-year EFS ( P = 0.037) and DCR ( P = 0.032), but not with EFS or 1-year OS. Age, phototype, and brain metastases were correlated with MHC-II expression status. Our findings suggest that MHC-II expression by melanoma cells may serve as a favorable predictive biomarker for survival in metastatic cutaneous melanoma patients treated with ICIs.
Collapse
Affiliation(s)
- Karim Amrane
- Department of Oncology, Regional Hospital of Morlaix, Morlaix
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
| | - Pierre Le Noac'h
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- Department of Pathology, University Hospital of Brest
| | - Patrice Hemon
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
| | - Ronan Abgral
- Department of Nuclear Medicine, University Hospital of Brest
- UMR Inserm 1304 GETBO, IFR 148, University of Western Brittany
| | - Coline Le Meur
- Department of Radiotherapy, University Hospital of Brest
| | - Olivier Pradier
- Department of Radiotherapy, University Hospital of Brest
- Inserm, UMR1101, LaTIM, University of Western Brittany
| | - Laurent Misery
- Department of Dermatology, University Hospital of Brest
- Laboratoire sur les Interactions Épithéliums-Neurones (LIEN-EA4685), Université de Bretagne Occidentale
| | - Delphine Legoupil
- Department of Dermatology, University Hospital of Brest
- Laboratoire sur les Interactions Épithéliums-Neurones (LIEN-EA4685), Université de Bretagne Occidentale
| | - Christian Berthou
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- Department of Hematology, University Hospital of Brest, Brest, France
| | - Arnaud Uguen
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- UMR Inserm 1304 GETBO, IFR 148, University of Western Brittany
| |
Collapse
|
11
|
Sperduto PW, Marqueen KE, Chang E, Li J, Davies MA, Ebner DK, Breen WG, Lamba N, Shih HA, Edwards D, Kim MM, Mahal A, Rahman R, Ankrah N, Boggs DH, Lewis C, Hyer D, Buatti JM, Johri F, Soliman H, Masucci L, Roberge D, Aneja S, Chiang V, Phuong C, Braunstein S, Dajani S, Sachdev S, Wan Z, Niedzwiecki D, Vaios E, Kirkpatrick JP, Pasetsky J, Wang TJ, Kutuk T, Kotecha R, Ross RB, Rusthoven CG, Nakano T, Tawbi HA, Mehta MP. Improved Survival and Prognostication in Melanoma Patients With Brain Metastases: An Update of the Melanoma Graded Prognostic Assessment. J Clin Oncol 2025; 43:1910-1919. [PMID: 40245362 PMCID: PMC12119226 DOI: 10.1200/jco-24-01351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/24/2025] [Accepted: 02/21/2025] [Indexed: 04/19/2025] Open
Abstract
PURPOSE Survival for patients with melanoma has recently improved. The propensity of melanoma to metastasize to the brain remains a common and serious feature of this disease. The purposes of this study were to evaluate prognostic factors for patients with newly diagnosed melanoma brain metastases (MBMs) in a large cohort treated with modern multimodal therapies, compare those results with those in prior eras, and update the Melanoma Graded Prognostic Assessment (GPA). METHODS Univariable and multivariable (MVA) analyses of prognostic factors and treatments associated with survival were performed on 1,796 patients with newly diagnosed MBM treated between January 01, 2015, and December 31, 2021, using a multi-institutional retrospective database. Multiple imputation was used to address missingness of potential predictors. Significant variables in combined MVA were used to update the Melanoma GPA. Comparisons were made with legacy cohorts. RESULTS Median survivals for cohorts A (1985-2007, n = 481), B (2006-2015, n = 823), and C (2015-2021, n = 1,796) were 6.7, 9.8, and 16.6 months and median follow-up times were 40.1, 43.6, and 48.8 months, respectively. In combined MVA, significant prognostic factors for survival were higher Karnofsky Performance Status, fewer MBMs, absence of extracranial metastases, lower serum lactate dehydrogenase, and no immunotherapy before MBM. These factors were incorporated into the updated Melanoma GPA. The combined median and 3-year survivals for patients with GPA 0-1, 1.5-2, and 2.5-4.0 were 5.4, 13.2, and 43.2 months and 12.4%, 28.8%, and 51.6%, respectively. CONCLUSION Prognostic factors have changed and survival has improved for patients with MBM but varies widely by GPA. The updated Melanoma GPA calculator (BrainMetGPA), available free online, can be used to estimate survival, individualize treatment, stratify clinical trials, guide surveillance, and augment clinical trial eligibility. Multidisciplinary treatment is essential. Trials are needed to elucidate the optimal sequencing of various therapeutic modalities.
Collapse
Affiliation(s)
| | | | - Enoch Chang
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jing Li
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | | | | | | | | - Nii Ankrah
- University of Alabama, Birmingham, Birmingham, AL
| | | | | | | | | | - Fasila Johri
- Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Canada
| | - Hany Soliman
- Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Canada
| | - Laura Masucci
- Centre Hospitalier de l’Universite de Montreal, Montreal, Canada
| | - David Roberge
- Centre Hospitalier de l’Universite de Montreal, Montreal, Canada
| | | | | | | | | | | | | | - Zihan Wan
- Duke Cancer Institute-Biostatistics Shared Resource, Durham, NC
| | | | | | | | | | | | - Tugce Kutuk
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | - Rupesh Kotecha
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | | | | | | | | | - Minesh P. Mehta
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| |
Collapse
|
12
|
Padmanaban V, Contreras CM. Role of Surgery for Metastatic Melanoma in the Era of Checkpoint Blockade. Surg Clin North Am 2025; 105:663-679. [PMID: 40412893 DOI: 10.1016/j.suc.2024.11.007] [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
Treatment paradigms have shifted with the introduction of checkpoint inhibitors, facilitating less extensive surgical procedures in some scenarios. Resection and adjuvant therapy are key components in patients with locally advanced or limited metastatic disease. Alternately, these patient populations may benefit from peri-operative systemic therapy based on recent clinical trial data. Resection is also integral to administering tumor infiltrating lymphocyte therapy in patients with treatment-refractory, unresectable melanoma. The role of surgery continues to evolve, and ranges the spectrum from resection with curative intent, to palliative interventions intended to effectively manage symptoms or disease-related complications.
Collapse
Affiliation(s)
- Vennila Padmanaban
- Division of Surgical Oncology, Department of Surgery, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Carlo M Contreras
- Division of Surgical Oncology, Department of Surgery, James Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
13
|
Kubo T, Kato H, Horiguchi S, Kozuki T, Asagi A, Yoshida M, Udono H, Kiura K, Hotta K. Phase-Ib dose-finding and pharmacokinetic trial of metformin combined with nivolumab for refractory/recurrent solid tumors. Int J Clin Oncol 2025:10.1007/s10147-025-02786-2. [PMID: 40434511 DOI: 10.1007/s10147-025-02786-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 05/06/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND Our previous findings showed that the addition of metformin to nivolumab resulted in remarkable tumor regression and increased the number of tumor-infiltrating T cells in mouse models. Therefore, we conducted a phase Ib study using combination therapy with nivolumab and metformin in patients with refractory/recurrent solid tumors. METHODS This study consisted of two parts: 1, evaluating the maximum tolerated dose (MTD), safety, pharmacokinetics in solid tumors, and 2, principally investigating the safety at the recommended dose limited to thoracic and pancreatic cancers. Metformin and nivolumab were administered orally at doses of 750-2,250 mg/day and biweekly at a fixed intravenous dose of 3 mg/kg, respectively. Dose-limiting toxicity was evaluated within the first 4 weeks. Both metformin and nivolumab were continued until disease progression or discontinued because of toxicity. RESULTS In total, 17 and 24 patients were enrolled in parts 1 and 2, respectively. One patient experienced increased pancreatic enzyme levels (grade 4) and lactic acidosis (grade 3). No Grade 5 adverse events were observed. MTD was not reached up to 2,250 mg/day of metformin, 2,250 mg/day was selected for part 2. An objective response was observed in 4 of 41 patients. One-year progression-free and overall survival rates were 9.8% and 56.8%, respectively. Two patients remained alive without disease progression for more than three years. CONCLUSIONS Nivolumab and metformin combination therapy was well-tolerated and showed preliminary signals of efficacy in a subset of patients. Further verification of the underlying mechanism in cases where treatment is effective is required. TRIAL REGISTRATION NUMBERS UMIN registration number 000028405 https://upload.umin.ac.jp/cgi-open-bin/ctr_e/ctr_view.cgi?recptno=R000031915 .
Collapse
Affiliation(s)
- Toshio Kubo
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
- Center for Clinical Oncology, Okayama University Hospital, Okayama, Japan
| | - Hironari Kato
- Department of Gastroenterology, Okayama University Hospital, Okayama, Japan
| | - Shigeru Horiguchi
- Department of Gastroenterology, Okayama University Hospital, Okayama, Japan
| | - Toshiyuki Kozuki
- Department of Thoracic Oncology and Medicine, NHO Shikoku Cancer Center, Matsuyama, Japan
| | - Akinori Asagi
- Department of Gastrointestinal Medical Oncology, NHO Shikoku Cancer Center, Matsuyama, Japan
| | - Michihiro Yoshida
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Heiichiro Udono
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Katsuyuki Kiura
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Katsuyuki Hotta
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan.
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan.
| |
Collapse
|
14
|
Miglino N, Toussaint NC, Ring A, Bonilla X, Tusup M, Gosztonyi B, Mehra T, Gut G, Jacob F, Chevrier S, Lehmann KV, Casanova R, Jacobs A, Sivapatham S, Boos L, Rahimzadeh P, Schuerch M, Sobottka B, Chicherova N, Yu S, Wegmann R, Mena J, Milani ES, Goetze S, Esposito C, Sarabia Del Castillo J, Frei AL, Nowak M, Irmisch A, Kuipers J, Baciu-Drăgan MA, Ferreira PF, Singer F, Bertolini A, Prummer M, Lischetti U, Tumor Profiler Consortium, Aebersold R, Bacac M, Maass G, Moch H, Weller M, Theocharides APA, Manz MG, Beerenwinkel N, Beisel C, Pelkmans L, Snijder B, Wollscheid B, Heinzelmann V, Bodenmiller B, Levesque MP, Koelzer VH, Rätsch G, Dummer R, Wicki A. Feasibility of multiomics tumor profiling for guiding treatment of melanoma. Nat Med 2025:10.1038/s41591-025-03715-6. [PMID: 40425842 DOI: 10.1038/s41591-025-03715-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 04/09/2025] [Indexed: 05/29/2025]
Abstract
There is limited evidence supporting the feasibility of using omics and functional technologies to inform treatment decisions. Here we present results from a cohort of 116 melanoma patients in the prospective, multicentric observational Tumor Profiler (TuPro) precision oncology project. Nine independent technologies, mostly at single-cell level, were used to analyze 126 patient samples, generating up to 500 Gb of data per sample (40,000 potential markers) within 4 weeks. Among established and experimental markers, the molecular tumor board selected 54 to inform its treatment recommendations. In 75% of cases, TuPro-based data were judged to be useful in informing recommendations. Patients received either standard of care (SOC) treatments or highly individualized, polybiomarker-driven treatments (beyond SOC). The objective response rate in difficult-to-treat palliative, beyond SOC patients (n = 37) was 38%, with a disease control rate of 54%. Progression-free survival of patients with TuPro-informed therapy decisions was 6.04 months, (95% confidence interval, 3.75-12.06) and 5.35 months (95% confidence interval, 2.89-12.06) in ≥third therapy lines. The proof-of-concept TuPro project demonstrated the feasibility and relevance of omics-based tumor profiling to support data-guided clinical decision-making. ClinicalTrials.gov identifier: NCT06463509 .
Collapse
Affiliation(s)
- Nicola Miglino
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Nora C Toussaint
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Swiss Data Science Center SDSC, Zurich, Switzerland
| | - Alexander Ring
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Ximena Bonilla
- Department of Computer Science, Institute of Machine Learning, ETH Zurich, Zurich, Switzerland
| | - Marina Tusup
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Benedict Gosztonyi
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Tarun Mehra
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Gabriele Gut
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Francis Jacob
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Stephane Chevrier
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Kjong-Van Lehmann
- Department of Computer Science, Institute of Machine Learning, ETH Zurich, Zurich, Switzerland
- Department of Biology, RWTH Aachen, Aachen, Germany
| | - Ruben Casanova
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Andrea Jacobs
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Sujana Sivapatham
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Laura Boos
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Parisa Rahimzadeh
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Manuel Schuerch
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Bettina Sobottka
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Natalia Chicherova
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Shuqing Yu
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Swiss Data Science Center SDSC, Zurich, Switzerland
| | - Rebekka Wegmann
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Julien Mena
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Emanuela S Milani
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Sandra Goetze
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), ETH Zurich, Zurich, Switzerland
| | - Cinzia Esposito
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | | - Anja L Frei
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Marta Nowak
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Anja Irmisch
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Jack Kuipers
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Monica-Andreea Baciu-Drăgan
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Pedro F Ferreira
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Franziska Singer
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Anne Bertolini
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Michael Prummer
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Ulrike Lischetti
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | | | - Rudolf Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Marina Bacac
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Gerd Maass
- Roche Diagnostics GmbH, MWG, Penzberg, Germany
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Alexandre P A Theocharides
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Niko Beerenwinkel
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Lucas Pelkmans
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Berend Snijder
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Bernd Wollscheid
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Viola Heinzelmann
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Bernd Bodenmiller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital, Zurich, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Gunnar Rätsch
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Computer Science, Institute of Machine Learning, ETH Zurich, Zurich, Switzerland
- Biomedical Informatics, University Hospital Zurich, Zurich, Switzerland
- AI Center at ETH Zurich, ETH Zurich, Zurich, Switzerland
- Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Andreas Wicki
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland.
| |
Collapse
Collaborators
Melike Ak, Faisal S Al-Quaddoomi, Silvana I Albert, Jonas Albinus, Ilaria Alborelli, Sonali Andani, Per-Olof Attinger, Monica-Andreea Baciu-Drăgan, Daniel Baumhoer, Beatrice Beck-Schimmer, Lara Bernasconi, Lars Bosshard, Byron Calgua, Stéphane Chevrier, Ricardo Coelho, Maya D'Costa, Esther Danenberg, Natalie R Davidson, Stefanie Engler, Martin Erkens, Katja Eschbach, André Fedier, Joanna Ficek-Pascual, Bruno Frey, Linda Grob, Detlef Günther, Pirmin Haeuptle, Viola Heinzelmann-Schwarz, Sylvia Herter, Rene Holtackers, Tamara Huesser, Alexander Immer, Tim M Jaeger, Alva R James, Philip M Jermann, André Kahles, Abdullah Kahraman, Werner Kuebler, Christian P Kunze, Christian Kurzeder, Mitchell Levesque, Flavio C Lombardo, Sebastian Lugert, Philipp Markolin, Martin Mehnert, Julian M Metzler, Simone Muenst, Riccardo Murri, Charlotte K Y Ng, Stefan Nicolet, Monica Nunez Lopéz, Patrick Ga Pedrioli, Salvatore Piscuoglio, Laurie Prélot, Natalie Rimmer, Mathilde Ritter, Christian Rommel, María L Rosano-González, Natascha Santacroce, Ramona Schlenker, Petra C Schwalie, Severin Schwan, Tobias Schär, Gabriela Senti, Wenguang Shao, Vipin T Sreedharan, Stefan Stark, Daniel J Stekhoven, Tanmay Tanna, Tinu M Thomas, Markus Tolnay, Vinko Tosevski, Mustafa A Tuncel, Audrey Van Drogen, Marcus Vetter, Tatjana Vlajnic, Sandra Weber, Walter P Weber, Fabian Wendt, Norbert Wey, Mattheus He Wildschut, Johanna Ziegler, Marc Zimmermann, Martin Zoche, Gregor Zuend,
Collapse
|
15
|
He L, He J, Jiang T, Gong R, Wan X, Duan M, Chen Z, Cheng Y. Inhibition of UCH-L1 enhances immunotherapy efficacy in triple-negative breast cancer by stabilizing PD-L1. Eur J Pharmacol 2025; 1000:177743. [PMID: 40389130 DOI: 10.1016/j.ejphar.2025.177743] [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: 01/28/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Recent research indicates that programmed death 1 (PD-1) and programmed death-ligand 1 (PD-L1) inhibitors show promise in treating triple-negative breast cancer (TNBC), but their efficacy is lower than anticipated, especially when used alone. Therefore, enhancing the anti-tumor immune response strategy for TNBC is crucial. Ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), involved in tumor cell regulation and a potential therapeutic target, has an undefined role in TNBC immunotherapy. In this study, we explored the inverse correlation between UCH-L1 and PD-L1 in TNBC patient tissues. Through in vitro experiments, we found that UCH-L1 negatively regulates PD-L1 by stabilizing the E3 ubiquitin ligase ariadne-1 homolog (ARIH1), which promotes PD-L1 ubiquitination and degradation. Further analysis in Balb/c mice xenograft tumors showed that UCH-L1 correlates with GZMB+/CD8+ T cell infiltration in TNBC, suggesting potential synergistic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. Overall, in TNBC, UCH-L1 stabilizes ARIH1, leading to low PD-L1 expression, which may explain the limited effectiveness of immunotherapy in TNBC patients. Our mouse experiments showed improved therapeutic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. These findings offer a new avenue for immunotherapy in TNBC patients.
Collapse
Affiliation(s)
- Linhao He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Jiaying He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ting Jiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Rong Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Xiaoya Wan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Mingwu Duan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Zonglin Chen
- Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China; Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China; NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, 410008, China; Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Changsha, 410011, China.
| |
Collapse
|
16
|
Pires de Oliveira Neto C, Nascimento GC, Damianse SDSP, Faria MDS. Recent advances in craniopharyngioma pathophysiology and emerging therapeutic approaches. Front Endocrinol (Lausanne) 2025; 16:1562942. [PMID: 40433410 PMCID: PMC12106005 DOI: 10.3389/fendo.2025.1562942] [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: 01/18/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Craniopharyngiomas are rare intracranial tumors originating from the Rathke's pouch, affecting the sellar and parasellar regions. Despite their benign nature, they cause significant morbidity and mortality due to their proximity to vital structures such as the optic pathways and the hypothalamic-pituitary axis, resulting in endocrine, visual, neurological impairment, and hypothalamic syndrome. Classified into adamantinomatous (ACP) and papillary (PCP), these tumors differ in epidemiology, histology, and pathophysiology. ACP, the most common type, presents a bimodal peak incidence between 5 and 15 years of age and 45 and 60 years of age, while PCP is more restricted to adults. Traditional treatments such as surgery and radiotherapy face significant challenges, including high recurrence rates. Intracystic chemotherapy is used in monocystic ACP but with limited efficacy and adverse effects related to toxicity. Recent advances in molecular biology have introduced targeted therapies, such as BRAF and MEK inhibitors, which show potential benefits in craniopharyngioma patients, particularly in the PCP. For ACP, however, therapeutic outcomes remain limited despite advances in molecular understanding, including mutations in the CTNNB1 gene and growth factors. Increasing investigation into the inflammatory microenvironment and immune response of these tumors presents new therapeutic possibilities and promising alternatives for tumor control, such as the use of anti-IL-6R, anti-VEGF agents and immune checkpoints inhibitors. This review aims to synthesize advancements in the pathophysiology of craniopharyngiomas and explore emerging therapeutic implications, focusing on precision medicine approaches for the management of this challenging disease.
Collapse
Affiliation(s)
- Clariano Pires de Oliveira Neto
- Post-Graduate Program in Health Sciences (PPGCS), Federal University of Maranhão (UFMA), São Luis, Brazil
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
- Clinical Research Center (CEPEC), University Hospital of the Federal University of Maranhao (HUUFMA), São Luís, Brazil
| | - Gilvan Cortês Nascimento
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
- Clinical Research Center (CEPEC), University Hospital of the Federal University of Maranhao (HUUFMA), São Luís, Brazil
- Department of Medicine I, Federal University of Maranhao (UFMA), São Luis, Brazil
| | - Sabrina da Silva Pereira Damianse
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
- Clinical Research Center (CEPEC), University Hospital of the Federal University of Maranhao (HUUFMA), São Luís, Brazil
| | - Manuel dos Santos Faria
- Post-Graduate Program in Health Sciences (PPGCS), Federal University of Maranhão (UFMA), São Luis, Brazil
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Clinical Research Center (CEPEC), University Hospital of the Federal University of Maranhao (HUUFMA), São Luís, Brazil
- Department of Medicine I, Federal University of Maranhao (UFMA), São Luis, Brazil
| |
Collapse
|
17
|
Weiss SA, Djureinovic D, Wei W, Tran T, Austin M, Markowitz J, Eroglu Z, Khushalani NI, Hegde U, Cohen J, Sznol M, Anderson G, Johnson B, Piteo C, Mahajan A, Adeniran A, Jilaveanu L, Goldberg S, Chiang V, Forsyth P, Kluger HM. Phase II Trial of Pembrolizumab in Combination With Bevacizumab for Untreated Melanoma Brain Metastases. J Clin Oncol 2025; 43:1685-1694. [PMID: 40048689 PMCID: PMC12058415 DOI: 10.1200/jco-24-02219] [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: 10/06/2024] [Revised: 12/10/2024] [Accepted: 01/17/2025] [Indexed: 03/15/2025] Open
Abstract
PURPOSE Anti-vascular endothelial growth factor therapy enhances PD-1 inhibitor activity in preclinical models and has been used to treat perilesional cerebral edema and radiation necrosis. METHODS We conducted a two-institution phase II trial of bevacizumab and pembrolizumab in patients with untreated melanoma brain metastasis (MBM) (ClinicalTrials.gov identifier: NCT02681549). Patients were anti-PD-(L)-1-naïve, and had ≥one asymptomatic, nonhemorrhagic 5-20 mm MBM, not requiring immediate local therapy or steroids. RESULTS Thirty-seven patients received four doses of bevacizumab and pembrolizumab every 3 weeks followed by up to 2 years of pembrolizumab. The brain metastasis response rate (primary end point) was 54.1% (95% CI, 36.9 to 70.5). The extracranial response rate was 56.3% (95% CI, 37.7 to 73.6). Median intracranial progression-free survival was 2.2 years (95% CI, 0.41 to not reached [NR]). Median overall survival (OS) was 4.3 years (95% CI, 1.6 to NR). Four-year OS rate was 51.6%. Grade 3 treatment-related adverse event rates from bevacizumab and pembrolizumab were 10.8% and 18.9%, respectively. Higher pretreatment vessel density in metastatic tumors and smaller on-therapy increases in circulating angiopoietin-2 were associated with response. CONCLUSION Pembrolizumab with bevacizumab was well tolerated and demonstrated substantial activity in patients with untreated MBM with promising OS, justifying further evaluation of this regimen.
Collapse
Affiliation(s)
- Sarah A. Weiss
- Rutgers Cancer Institute (Medical Oncology), New Brunswick, NJ
| | | | - Wei Wei
- Yale University School of Medicine (Biostatistics), New Haven, CT
| | - Thuy Tran
- Yale University School of Medicine (Medical Oncology), New Haven, CT
| | - Matthew Austin
- Yale University School of Medicine (Medical Oncology), New Haven, CT
| | - Joseph Markowitz
- Moffitt Cancer Center (Department of Cutaneous Oncology), Tampa, FL
| | - Zeynep Eroglu
- Moffitt Cancer Center (Department of Cutaneous Oncology), Tampa, FL
| | | | - Upendra Hegde
- University of Connecticut School of Medicine (Medical Oncology), Farmington, CT
| | - Justine Cohen
- Dana Farber Cancer Institute (Medical Oncology), Boston, MA
| | - Mario Sznol
- Yale University School of Medicine (Medical Oncology), New Haven, CT
| | - Gail Anderson
- Yale University School of Medicine (Medical Oncology), New Haven, CT
| | - Barbara Johnson
- Yale University School of Medicine (Medical Oncology), New Haven, CT
| | - Cecily Piteo
- Moffitt Cancer Center (Department of Cutaneous Oncology), Tampa, FL
| | - Amit Mahajan
- Yale University School of Medicine (Radiology), New Haven, CT
| | | | - Lucia Jilaveanu
- Yale University School of Medicine (Medical Oncology), New Haven, CT
| | - Sarah Goldberg
- Yale University School of Medicine (Medical Oncology), New Haven, CT
| | - Veronica Chiang
- Yale University School of Medicine (Neurosurgery), New Haven, CT
| | - Peter Forsyth
- Moffitt Cancer Center (Departments of Neuro-Oncology and Cell Biology), Tampa, FL
| | - Harriet M. Kluger
- Yale University School of Medicine (Medical Oncology), New Haven, CT
| |
Collapse
|
18
|
Yamamoto Y, Kitadani J, Hayata K, Goda T, Tominaga S, Nakai T, Nagano S, Iwamoto R, Kawai M. Promising Treatment Strategy for Primary Malignant Melanoma of the Esophagus by Radical Esophagectomy and Nivolumab as Adjuvant Therapy: A Case Report. Surg Case Rep 2025; 11:25-0027. [PMID: 40356806 PMCID: PMC12068939 DOI: 10.70352/scrj.cr.25-0027] [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: 01/21/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
INTRODUCTION Primary malignant melanoma of the esophagus (PMME) is a rare malignant tumor of the esophagus with very poor prognosis due to high rates of recurrence and metastasis even after radical resection. Recently, however, immune checkpoint inhibitors such as anti-programmed cell death-1 antibodies have been suggested to improve the prognosis of malignant melanoma. This report describes the use of postoperative nivolumab as adjuvant therapy after surgical resection of PMME, with recurrence-free follow-up for more than 1 year. CASE PRESENTATION A 69-year-old man had chest discomfort and tightness in his throat. Upper gastrointestinal endoscopy revealed multiple melanosis and an elevated lesion in the middle esophagus. After histological examination, he was diagnosed as having PMME, so he underwent thoracoscopic subtotal esophagectomy, three-field lymphadenectomy, pedunculated jejunum reconstruction with super-charge and super-drainage, and feeding jejunostomy due to the past history of gastrectomy. The adjuvant therapy using nivolumab (every 2 weeks, 240 mg) for 1 year was completed with no serious side effects, and there was no recurrence for more than 1 year postoperatively. CONCLUSIONS Although cases of PMME treated with adjuvant nivolumab have rarely been reported, the present case suggests that this approach may represent a promising treatment option, similar to cutaneous melanoma.
Collapse
Affiliation(s)
- Yusuke Yamamoto
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Junya Kitadani
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Keiji Hayata
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Taro Goda
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Shinta Tominaga
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Tomoki Nakai
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Shotaro Nagano
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Ryuta Iwamoto
- Department of Human Pathology, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Manabu Kawai
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| |
Collapse
|
19
|
Sánchez-Camacho A, Torres-Zurita A, Gallego-López L, Hernández-Pacheco R, Silva-Romeiro S, Álamo de la Gala MDC, Peral-Gutiérrez de Ceballos E, de la Cruz-Merino L. Management of immune-related myocarditis, myositis and myasthenia gravis (MMM) overlap syndrome: a single institution case series and literature review. Front Immunol 2025; 16:1597259. [PMID: 40406130 PMCID: PMC12095175 DOI: 10.3389/fimmu.2025.1597259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/17/2025] [Indexed: 05/26/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of various malignancies, particularly melanoma. However, immune-related adverse events (irAEs) pose significant challenges, particularly in cases of severe toxicity syndromes. One such life-threatening irAE is the myocarditis, myositis, and myasthenia gravis (MMM) overlap syndrome, which occurs in less than 1% of patients but has in-hospital mortality rates ranging from 40 to 60%. Due to its rarity and complexity, early recognition and a multidisciplinary approach are critical to improving outcomes. Methods We present a single-institution case series of four patients diagnosed with MMM overlap syndrome following ICI therapy. Clinical presentation, laboratory findings, imaging, and electrophysiological tests were analyzed to confirm the diagnosis. Therapeutic interventions-including corticosteroids, intravenous immunoglobulins (IVIG), plasma exchange (PLEX), tocilizumab, and rituximab- were evaluated in terms of efficacy and clinical outcomes. Results The onset of MMM syndrome varied from 2 to 4 weeks after initiating ICI therapy. Patients presented with rapidly progressive symptoms, including ptosis, bulbar dysfunction, respiratory distress, myopathy, and cardiac conduction abnormalities. Immunosuppressive therapy with high-dose corticosteroids was initiated in all cases. Additional immunomodulatory treatments (IVIG, tocilizumab, PLEX, and rituximab) were administered based on clinical deterioration and autoimmune profile. Two patients achieved complete recovery, one remains on maintenance immunosuppression, and one died due to respiratory failure despite aggressive treatment. Conclusion MMM overlap syndrome is a severe and often fatal irAE associated with ICI therapy. Early identification, aggressive immunosuppressive treatment, and individualized therapeutic strategies are essential to optimize patient outcomes. Further research is needed to refine diagnostic criteria, identify predictive biomarkers, and establish standardized treatment protocols.
Collapse
Affiliation(s)
| | - Alberto Torres-Zurita
- Department of Medical Oncology, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Laura Gallego-López
- Autoimmune Disease Unit, Department of Internal Medicine, Hospital Universitario Virgen Macarena, Seville, Spain
| | | | - Silvia Silva-Romeiro
- Department of Medical Oncology, Hospital Universitario Virgen Macarena, Seville, Spain
- Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen Macarena, CSIC, University of Seville, Seville, Spain
| | | | | | - Luis de la Cruz-Merino
- Department of Medical Oncology, Hospital Universitario Virgen Macarena, Seville, Spain
- Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen Macarena, CSIC, University of Seville, Seville, Spain
- Department of Medicine, University of Seville, Seville, Spain
| |
Collapse
|
20
|
Canali B, Apolone G, Ascierto PA, De Braud F, Grossi F, Perrone F, Fiorentino F, Di Costanzo A, Candelora L, Patanè G, Zapparelli G, Mezzanotte C, Didoni G, Riccaboni M. Effect of immuno-oncology on clinical and economic outcomes for a selection of cancers in Italy. Expert Rev Pharmacoecon Outcomes Res 2025:1-11. [PMID: 40329477 DOI: 10.1080/14737167.2025.2493130] [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: 10/09/2024] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVES This study assesses the impact of immune-oncology (IO) drugs' availability on cancer incidence-adjusted mortality rates from melanoma, lung, and renal cancers at population level in Italy between 2008 and 2019. METHODS We conducted a retrospective study on cross-sectional time-series aggregated data collected from publicly available sources and IQVIA proprietary databases. Three fixed-effects regression models were used to estimate how IO availability affects incidence-adjusted mortality for each cancer type. Estimated deaths were compared with deaths in a scenario with no IO drugs availability. Finally, the number of averted deaths was valued using the human capital approach. RESULTS A 1% increase in IO availability reduces incidence-adjusted mortality rates for melanoma, lung, and renal cancers by 0.125% (95% CI: 0.138-0.112; p < 0.01), 0.011% (95% CI: 0.013-0.009; p < 0.01) and 0.005% (95% CI: 0.006-0.003; p < 0.01) between the introduction of the drug in the therapeutic area and 2019. This reduction resulted in total savings of € 49.0 million, € 61.3 million, and € 10.9 million in indirect costs due to premature mortality, respectively. CONCLUSIONS IO drugs introduction in Italy between 2008 and 2019 was associated with a significant decrease in deaths from each cancer and, consequently, in savings in indirect costs related to premature mortality.
Collapse
Affiliation(s)
- Beatrice Canali
- Real World Solutions, IQVIA Solutions Italy S.r.l, Milan, Italy
| | - Giovanni Apolone
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Filippo De Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesco Grossi
- Department of Medicine and Technological Innovation, Università degli Studi dell'Insubria, Varese, Italy
- Medical Oncology Division, ASST Sette Laghi, Varese, Italy
| | - Francesco Perrone
- Clinical Trial Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | | | | | - Laura Candelora
- Real World Solutions, IQVIA Solutions Italy S.r.l, Milan, Italy
| | | | | | - Claudia Mezzanotte
- Pricing, Access Strategy & Health Economics, Bristol Myers Squibb, Rome, Italy
| | - Guido Didoni
- Pricing, Access Strategy & Health Economics, Bristol Myers Squibb, Rome, Italy
| | | |
Collapse
|
21
|
Li G, Wang J, Fang Q, Dai L, Du W. Oncologic outcomes following neoadjuvant immunochemotherapy in locally advanced oral squamous cell carcinoma. Front Immunol 2025; 16:1571285. [PMID: 40406105 PMCID: PMC12095181 DOI: 10.3389/fimmu.2025.1571285] [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: 02/05/2025] [Accepted: 04/07/2025] [Indexed: 05/26/2025] Open
Abstract
Background To assess the oncologic outcomes in patients with oral squamous cell carcinoma (SCC) who underwent treatment with radiotherapy (RT) or chemoradiation therapy (CRT) following neoadjuvant immunochemotherapy and surgery. Methods Data from patients who underwent neoadjuvant immunochemotherapy, surgery, and adjuvant therapy were collected prospectively and analyzed retrospectively. The primary outcomes assessed were 3-year overall survival and locoregional control. Secondary endpoints included the objective response rate (ORR), rates of pathologic complete response (pCR) and major pathologic response (MPR), as well as safety. Results A total of 137 patients were included in the analysis. Neoadjuvant therapy yielded an ORR of 81.7%, with pCR and MPR achieved in 47 and 73 patients, respectively. Grade III and IV adverse events were rare, comprising only 1.6% of all events. The addition of adjuvant chemotherapy to RT did not show a significant reduction in the risk of locoregional recurrence. However, with regards to overall survival, the hazard ratios were 0.85 (95% CI: 0.73-0.96) for the MPR group and 0.66 (95% CI: 0.37-0.89) for the pCR group, both significantly higher than that in patients with incomplete pathologic response. The addition of adjuvant chemotherapy to RT was associated with a 5% reduction in the risk of mortality (95% CI: 1%-14%), the protective effect of CRT was the most obvious in patients with MPR. Conclusion Neoadjuvant immunochemotherapy demonstrated high safety and efficacy in oral SCC. CRT was superior to RT in terms of overall survival especially in patients with MPR when administered following neoadjuvant immunochemotherapy and surgery.
Collapse
Affiliation(s)
| | | | | | | | - Wei Du
- Department of Head Neck and Thyroid, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
22
|
Jiang D, Yoou MS, Cho S, Choi Y. Molecular Dynamics-Guided Repositioning of FDA-Approved Drugs for PD-L1 Inhibition with In Vitro Anticancer Potential. Int J Mol Sci 2025; 26:4497. [PMID: 40429641 PMCID: PMC12110937 DOI: 10.3390/ijms26104497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Programmed death-ligand 1 (PD-L1) is a crucial immune checkpoint protein that tumors often exploit to evade immune surveillance. This study systematically screened a library of 1031 FDA-approved drugs using a high-throughput molecular dynamics approach to identify potential inhibitors targeting PD-L1. From this screening, five promising compounds-vorapaxar, delafloxacin, tenofovir disoproxil, pivmecillinam, and fursultiamine-showed significant binding affinities to PD-L1 and demonstrated cytotoxic activity against A549 lung tumor cells. These candidates were further evaluated through extended molecular dynamics simulations lasting up to 150 ns to assess their structural stability, residue fluctuations, and binding free energy. Among the identified compounds, pivmecillinam demonstrated the most favorable results, exhibiting stable binding interactions and a binding free energy of -18.01 kcal/mol, comparable to that of the known PD-L1 inhibitor BMS-1. These findings suggest that pivmecillinam has promising immunomodulatory potential and could serve as a candidate for further development in cancer immunotherapy. Overall, this study underscores the value of integrating high-throughput MD and experimental approaches for drug repositioning to identify novel therapeutic agents.
Collapse
Affiliation(s)
- Dejun Jiang
- Department of Environmental Engineering, Hoseo University, Asan 31499, Republic of Korea;
| | - Myoung-Schook Yoou
- Eulji Medi-Bio Research Institute, Eulji University, Daejeon 34824, Republic of Korea;
| | - Sungjoon Cho
- Department of Bio-Applied Toxicology, Hoseo University, Asan 31499, Republic of Korea;
| | - Youngjin Choi
- Department of Bio-Applied Toxicology, Hoseo University, Asan 31499, Republic of Korea;
- Department of Food Science & Technology, Hoseo University, Asan 31499, Republic of Korea
| |
Collapse
|
23
|
Lee D, Ahn J, Choi J. PathNetDRP: a novel biomarker discovery framework using pathway and protein-protein interaction networks for immune checkpoint inhibitor response prediction. BMC Bioinformatics 2025; 26:119. [PMID: 40325361 PMCID: PMC12051301 DOI: 10.1186/s12859-025-06125-0] [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: 12/17/2024] [Accepted: 03/31/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Predicting immune checkpoint inhibitor (ICI) response remains a significant challenge in cancer immunotherapy. Many existing approaches rely on differential gene expression analysis or predefined immune signatures, which may fail to capture the complex regulatory mechanisms underlying immune response. Network-based models attempt to integrate biological interactions, but they often lack a quantitative framework to assess how individual genes contribute within pathways, limiting the specificity and interpretability of biomarkers. Given these limitations, we developed PathNetDRP, a framework that integrates biological pathways, protein-protein interaction networks, and machine learning to identify functionally relevant biomarkers for ICI response prediction. RESULTS We introduce PathNetDRP, a novel biomarker discovery approach that applies the PageRank algorithm to prioritize ICI-associated genes, maps them to relevant biological pathways, and calculates PathNetGene scores to quantify their contribution to immune response. Unlike conventional methods that focus solely on gene expression differences, PathNetDRP systematically incorporates biological context to improve biomarker selection. Validation across multiple independent cancer cohorts showed that PathNetDRP achieved strong predictive performance, with cross-validation the area under the receiver operating characteristic curves increasing from 0.780 to 0.940. Interestingly, PathNetDRP did not merely improve predictive accuracy; it also provided insights into key immune-related pathways, reinforcing its potential for identifying clinically relevant biomarkers. CONCLUSION The biomarkers identified by PathNetDRP demonstrated robust predictive performance across cross-validation and independent validation datasets, suggesting their potential utility in clinical applications. Furthermore, enrichment analysis highlighted key immune-related pathways, providing a deeper understanding of their role in ICI response regulation. While these findings underscore the promise of PathNetDRP, future work will explore the integration of additional predictive features, such as tumor mutational burden and microsatellite instability, to further refine its applicability.
Collapse
Affiliation(s)
- Dohee Lee
- Department of Computer Science and Engineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon, 22012, Republic of Korea
| | - Jaegyoon Ahn
- Department of Computer Science and Engineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon, 22012, Republic of Korea.
| | - Jonghwan Choi
- Division of Software, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea.
| |
Collapse
|
24
|
Wang SJ, Xiu J, Butcher KM, DeClerck BK, Kim GH, Moser J, Gibney GT, Hernandez‐Aya LF, Lutzky J, Abdulla F, Margolin KA, Possik PA, Robles‐Espinoza CD, Ito F, In GK. Comprehensive Profiling of Acral Lentiginous Melanoma Reveals Downregulated Immune Activation Compared to Cutaneous Melanoma. Pigment Cell Melanoma Res 2025; 38:e70027. [PMID: 40405404 PMCID: PMC12099029 DOI: 10.1111/pcmr.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2025] [Revised: 04/27/2025] [Accepted: 05/13/2025] [Indexed: 05/24/2025]
Abstract
Acral lentiginous melanoma (ALM) is a rare and insufficiently understood subtype of melanoma lacking in effective treatment options. Recent work has demonstrated that the response of ALM to immune checkpoint blockade is inferior to that of cutaneous melanoma (CM). Here we performed bulk genomic and transcriptomic sequencing of tumor tissue from 28 ALM and 5692 CM cases. Similar to prior studies, ALM was associated with a significantly lower incidence of point mutations, including in the TERT promoter and BRAF, but increased numbers of gene amplifications, notably of CCND1, HMGA2, and MDM2. Reactome pathway analysis revealed enhancement of keratinization and PI3K/AKT signaling pathways. Overall immunogenicity was decreased in ALM, which possessed lower IFNγ (p < 0.001) and T-cell inflammatory (p = 0.03) pathway scores than CM. Despite higher computationally inferred levels of myeloid dendritic cells (p = 0.006), neoantigen load independent of predicted HLA binding affinity was lower (p < 0.01) in ALM versus CM. Assessment of classical and nonclassical HLA mRNA levels revealed upregulation of HLA-G, suggesting alternative ALM immune evasion pathways in the setting of lower PD-L1 expression (p = 0.005). Additional research is needed to better understand and therapeutically target signaling networks in the ALM tumor microenvironment.
Collapse
Affiliation(s)
- Stephanie J. Wang
- Department of MedicineUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
| | | | - Katherine M. Butcher
- Division of OncologyUniversity of Southern California Keck School of Medicine, Norris Comprehensive Cancer CenterLos AngelesCaliforniaUSA
| | - Brittney K. DeClerck
- Department of DermatologyUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
- Department of PathologyUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
| | - Gene H. Kim
- Department of DermatologyUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
- Department of PathologyUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
| | - Justin Moser
- HonorHealth Research and Innovation InstituteScottsdaleArizonaUSA
| | | | | | - Jose Lutzky
- University of Miami Sylvester Comprehensive Cancer CenterMiamiFloridaUSA
| | | | - Kim A. Margolin
- Department of Medical OncologySaint John's Cancer Institute, Providence Saint John's Health CancerSanta MonicaCaliforniaUSA
| | - Patrícia Abrão Possik
- Division of Basic and Experimental ResearchBrazilian National Cancer InstituteRio de JaneiroBrazil
| | | | - Fumito Ito
- Department of SurgeryUniversity of Southern California Keck School of Medicine, Norris Comprehensive Cancer CenterLos AngelesCaliforniaUSA
| | - Gino K. In
- Department of MedicineUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
- Division of OncologyUniversity of Southern California Keck School of Medicine, Norris Comprehensive Cancer CenterLos AngelesCaliforniaUSA
- Department of DermatologyUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
| |
Collapse
|
25
|
Wang Y, Hu X, Wang J, Zhang Y, Guo P, Lv Y, Ma G, Wei W, Wang S. Versatile PLGA-Based Drug Delivery Systems for Tumor Immunotherapy. SMALL METHODS 2025; 9:e2401623. [PMID: 39924767 DOI: 10.1002/smtd.202401623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/07/2025] [Indexed: 02/11/2025]
Abstract
Tumor immunotherapy, which utilizes the immune system to fight cancer, represents a revolutionary method for cancer treatment. Poly (lactic-co-glycolic acid) (PLGA) copolymer has emerged as a promising material for tumor immunotherapy due to its biocompatibility, biodegradability, and versatility in drug delivery. By tuning the size, shape, and surface properties of PLGA-based systems, researchers have improved their ability to align with the requirements for diverse tumor immunotherapy modalities. In this review, the basic properties of the PLGA materials are first introduced and further the principal forms of the PLGA systems for controlled release are summarized and delivery applications are targeted. In addition, recent advances in the use of PLGA delivery systems are highlighted to enhance antitumor immune responses in terms of tumor vaccines, immunogenic cell death-mediated immune responses, tumor microenvironment modulation, and combination immunotherapies. Finally, prospects for the future research and clinical translation of PLGA materials are proposed.
Collapse
Affiliation(s)
- Yishu Wang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiaoming Hu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinghui Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, P. R. China
| | - Yu Zhang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Peilin Guo
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanlin Lv
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Wei Wei
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Shuang Wang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
26
|
Qureshi Z, Zaheer Z, Asghar Z, Bakhtiar M, Fatima E, Altaf F. Cardiovascular Risk Profile of Nivolumab Anti-cancer Therapy: A Review of Current Literature. Am J Clin Oncol 2025; 48:235-241. [PMID: 40008416 DOI: 10.1097/coc.0000000000001166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
OBJECTIVES Immune checkpoint inhibitors (ICI) upregulate host antitumor immunity, proving efficacy across diverse tumor types. Currently approved ICI treatment primarily targets the programmed cell death receptor 1 (PD-1) and its ligand PD-L1, and cytotoxic T lymphocyte-antigen 4 (CTLA-4). Nivolumab is a monoclonal antibody that targets the human PD-1 receptor and is an entirely human immunoglobulin G4 (IgG4), approved by the FDA for various cancers like advanced melanoma, metastatic renal cell carcinoma, Hodgkin lymphoma, and advanced lung carcinoma. This review will summarise and discuss the recent literature on cardiotoxicity associated with nivolumab therapy. METHODS We searched online databases like PubMed, Scopus, Google Scholar, and Embase for articles related to Nivolumab. RESULTS Cardiotoxicity with ICI use is most commonly represented as myocarditis. Patients present with complaints of shortness of breath, palpitations, edema, and fatigue. Takotsubo cardiomyopathy, or broken heart syndrome, is characterized by systolic dysfunction of the left ventricle, mimicking a myocardial infarction but without associated coronary ischemia and with minimal elevation of cardiac enzymes. In the CHECKMATE-037 trial, ventricular arrhythmias occurred in <10% of those who received nivolumab. In a retrospective analysis of patients treated with ICI (predominantly nivolumab monotherapy) for lung cancer, 11% of the patients developed major adverse cardiac events, including myocarditis, non-ST-segment elevated myocardial infarction, supraventricular tachycardia, and pericardial disorders. CONCLUSION Close collaboration between cardiology and oncology specialists is crucial for early detection and effective management of cardiac complications, enhancing the safety of nivolumab anticancer therapy.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | | | - Zoha Asghar
- Department of Medicine, Ziauddin University, Karachi
| | | | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| |
Collapse
|
27
|
Yan A, Madigan L, Korman A, Shearer S, Dulmage B, Patel T, Milani-Nejad N, Chung C, Fisher K, Kaffenberger B. Morbilliform Eruptions: Differentiating Low-Risk Drug Eruptions, Severe Cutaneous Adverse Reactions, Viral Eruptions, and Acute Graft-Versus-Host Disease. Am J Clin Dermatol 2025; 26:379-393. [PMID: 39888589 PMCID: PMC12085335 DOI: 10.1007/s40257-025-00924-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Morbilliform eruptions, which are a clinical reaction pattern characterized by erythematous macules and papules coalescing into patches that cover most of the skin surface, are one of the most common cutaneous findings in the inpatient setting. In the hospital setting, most causes are benign and due to low-risk drug exanthems; however, morbilliform eruptions may also be a sign of high-risk diseases, including Stevens-Johnson syndrome/toxic epidermal necrolysis, drug reaction with eosinophilia and systemic symptoms/drug-induced hypersensitivity syndrome, acute generalized exanthematous pustulosis, and graft-versus-host disease. Proper identification of the etiology and risk stratification of a morbilliform eruption is critical to ensure proper management and optimize patient outcomes. In this review, we discuss the key features that differentiate high-risk from low-risk morbilliform eruptions, as well as specific characteristics that differentiate the different high-risk eruptions. Additionally, we offer a clinical algorithm that may be applied in the management of a patient who presents with a morbilliform rash.
Collapse
Affiliation(s)
- Allison Yan
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Lauren Madigan
- Department of Dermatology, The University of Utah, Salt Lake City, UT, USA
| | - Abraham Korman
- Department of Dermatology, The Ohio State University, 1328 Dublin Rd, Suite 100, Columbus, OH, 43212, USA
| | | | - Brittany Dulmage
- Department of Dermatology, The Ohio State University, 1328 Dublin Rd, Suite 100, Columbus, OH, 43212, USA
| | - Tejesh Patel
- Department of Dermatology, The University of Tennessee, Memphis, TN, USA
| | - Nima Milani-Nejad
- Department of Dermatology, University of California Los Angeles, Los Angeles, CA, USA
| | - Catherine Chung
- Department of Dermatology, The Ohio State University, 1328 Dublin Rd, Suite 100, Columbus, OH, 43212, USA
| | - Kristopher Fisher
- Department of Dermatology, The Ohio State University, 1328 Dublin Rd, Suite 100, Columbus, OH, 43212, USA
| | - Benjamin Kaffenberger
- Department of Dermatology, The Ohio State University, 1328 Dublin Rd, Suite 100, Columbus, OH, 43212, USA.
| |
Collapse
|
28
|
Steinberg FT, Simon M, Wawer-Matos Reimer PA, Rokohl AC, Heindl LM. [New systemic treatment approaches for conjunctival melanoma]. DIE OPHTHALMOLOGIE 2025; 122:349-356. [PMID: 40067450 DOI: 10.1007/s00347-025-02207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/30/2024] [Accepted: 02/05/2025] [Indexed: 05/08/2025]
Abstract
Conjunctival melanoma is a rare disease that nevertheless has a high tumor-associated mortality rate. A resection in sano with adjuvant local treatment currently represents the therapeutic gold standard and systemic treatment is used for metastasized conjunctival melanoma and/or very advanced nonresectable local findings. New knowledge on molecular changes in conjunctival melanoma shows a clear similarity to those of cutaneous melanoma. Therefore, many findings on new systemic forms of treatment for cutaneous melanoma can be transferred to conjunctival melanoma. In the clinical application BRAF/MEK inhibitors and immune checkpoint inhibitors are already in use and good response rates have been shown in small retrospective studies and case reports. Due to the rarity of conjunctival melanoma, there are no larger prospective studies comparing different systemic therapeutic agents. In a nonrandomized retrospective comparison, a better overall survival was shown for a combination of BRAF/MEK inhibitors (progression-free 1‑year survival probability of 54.7%; overall survival of 29.1 months) compared to a combination of PD1/CTLA4 antibodies (progression-free 1‑year survival probability of 42%; overall survival of 18 months). The current recommendation is to perform genomic profiling for every conjunctival melanoma, particularly to investigate a BRAF mutation. If a BRAF mutation is present, BRAF/MEK inhibitor treatment should preferably be initiated. Treatment with immune checkpoint inhibitors can be used in the case of BRAF-negative mutation status or treatment failure with BRAF/MEK inhibitors. Monotherapy with the CTLA4 antibody ipilimumab is not recommended due to its inferiority to PD1 antibodies. New knowledge in the genomic profiling of conjunctival melanoma could enable further targeted treatment options in the future.
Collapse
Affiliation(s)
| | - Michael Simon
- Zentrum für Augenheilkunde, Universitätsklinikum Köln, Köln, Deutschland
| | | | - Alexander C Rokohl
- Zentrum für Augenheilkunde, Universitätsklinikum Köln, Köln, Deutschland
| | - Ludwig M Heindl
- Zentrum für Augenheilkunde, Universitätsklinikum Köln, Köln, Deutschland
| |
Collapse
|
29
|
Kong WY, Soderholm A, Brooks AJ, Gonzalez Cruz JL, Wells JW. Harnessing cytokine immunocomplexes and cytokine fusion proteins for cancer Therapy: Mechanisms and clinical potential. Cancer Treat Rev 2025; 136:102937. [PMID: 40233680 DOI: 10.1016/j.ctrv.2025.102937] [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] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
Cytokines are pivotal regulators of cellular functions and immune responses, making them highly promising targets for cancer immunotherapy. Despite their widespread clinical application, the effectiveness of cytokine immunotherapy is often hampered by their pleiotropic effects, short half-lives, uneven biodistribution, and severe side effects at high dosages. Recent advancements in cytokine biology have led to the development of cytokine-antibody immunocomplexes and cytokine fusion proteins, offering a new paradigm in cancer treatments. These innovations foster the ability of cytokines to selectively activate specific cancer-targeting immune cell populations, such as CD8 T cells and NK cells, effectively inhibiting tumour progression. Furthermore, both therapeutic approaches can mitigate systemic toxicities and prolong the biological activity of cytokines in the body. This review delves into the recent advancements of cytokine immunocomplexes and cytokine fusion proteins, with a particular focus on interleukin-2 (IL-2), IL-7 and IL-15, which are in clinical/preclinical development. Moreover, we discuss the therapeutic benefits of these approaches observed in recent preclinical and clinical studies, along with the challenges that must be addressed to fully unlock their potential in cancer immunotherapy.
Collapse
Affiliation(s)
- Wei Yang Kong
- Frazer Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Amelia Soderholm
- Frazer Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Andrew J Brooks
- Frazer Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Queensland, Australia; School of Science & Technology, University of New England, Armidale, New South Wales, Australia
| | - Jazmina L Gonzalez Cruz
- Frazer Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - James W Wells
- Frazer Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Queensland, Australia; Dermatology Research Centre, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
30
|
Fujimoto A, Sakata S, Kataoka K, Kogure Y, Chiba K, Okada A, Shiraishi Y, Baba S, Maruyama D, Ogawa S, Takeuchi K. High-accuracy Detection of PD-L1 3'-UTR Disruption by Immunohistochemistry and Fluorescence in Situ Hybridization on Formalin-fixed Paraffin-embedded Sections. Am J Surg Pathol 2025; 49:490-498. [PMID: 40026197 DOI: 10.1097/pas.0000000000002372] [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: 03/04/2025]
Abstract
Programmed death-ligand 1 (PD-L1/CD274) structural variation (SV) disrupting the 3'-untranslated region has been highlighted as being associated with PD-L1 overexpression. In the present study, we evaluated lymphoma tissue samples to investigate the applicability of immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) for detecting the PD-L1 SV involving the 3'-untranslated region. In total, 1052 lymphoma samples were screened using IHC, and 99 IHC screening-positive samples were evaluated with FISH (non-Hodgkin lymphoma [NHL, n=58] and Hodgkin lymphoma [HL, n=41]). Of these, 92 samples showed strong PD-L1 expression with 2 PD-L1 antibodies (E1J2J and SP142) (concordant PD-L1 IHC), whereas 7 samples showed strong PD-L1 expression only with E1J2J (discordant PD-L1 IHC). Abnormal FISH findings for PD-L1 were detected in all evaluated samples (51 NHLs and 41 HLs). A structural abnormality pattern was observed in 17 of the 51 evaluated NHL samples (33%). In contrast, all 41 HL samples showed a copy number abnormality pattern, with 1 exhibiting a structural abnormality pattern. Target-capture sequencing of the PD-L1 gene was performed on 73 of the 99 IHC screening-positive samples, comprising 41 NHLs and 32 HLs. PD-L1 SVs were detected in 16 (39%) of the 41 NHL samples and in only one of the 32 HL samples (3%). Samples exhibiting discordant PD-L1 IHC and/or FISH structural abnormality patterns were shown to harbor PD-L1 SV by target-capture sequencing, with positive and negative predictive values of 94% and 96%, respectively. Our approach is an alternative to target-capture sequencing for evaluating PD-L1 gene abnormalities.
Collapse
Affiliation(s)
- Ayumi Fujimoto
- Pathology Project for Molecular Targets, Cancer Institute
- Division of Pathology, Cancer Institute
| | - Seiji Sakata
- Pathology Project for Molecular Targets, Cancer Institute
- Division of Pathology, Cancer Institute
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research
| | - Keisuke Kataoka
- Division of Molecular Oncology, National Cancer Center Research Institute
- Division of Hematology, Department of Medicine, Keio University School of Medicine
| | - Yasunori Kogure
- Division of Molecular Oncology, National Cancer Center Research Institute
| | - Kenichi Chiba
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute
| | - Ai Okada
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute
| | - Yuichi Shiraishi
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute
| | - Satoko Baba
- Pathology Project for Molecular Targets, Cancer Institute
- Division of Pathology, Cancer Institute
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research
| | - Dai Maruyama
- Department of Hematology-Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kengo Takeuchi
- Pathology Project for Molecular Targets, Cancer Institute
- Division of Pathology, Cancer Institute
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research
| |
Collapse
|
31
|
Triggiano G, Pezzicoli G, Tucci M. Immunotherapy in Advanced Cutaneous Melanoma: From the Optimal Treatment Duration to the Impact on Survival in Case of Early Discontinuation Due to Immune-Related Adverse Events. Biomolecules 2025; 15:651. [PMID: 40427544 PMCID: PMC12109418 DOI: 10.3390/biom15050651] [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: 02/17/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have improved the prognosis of patients with cutaneous melanoma. Immunotherapy (IT) is generally well tolerated, but an increasing area of investigation concerns the optimal treatment duration of anti-programmed cell death-1 (anti-PD1) regimens to limit the immune-related adverse events in patients who obtained a clinical response. Another point of interest is the impact of the early discontinuation of ICIs on the maintenance of response in terms of survival in patients developing grade 3-4 adverse events that mostly occur in those receiving the combo-IT. Currently, we are still far from having final conclusions on these topics and, thus, the present review aims to describe the recent data about the optimal treatment duration and the maintenance of response in the case of early discontinuation. In this context, we include data on the real life of patients from our Medical Oncology Center who discontinued anti-PD1 after at least a stable disease or those interrupting the combo-IT due to adverse events.
Collapse
Affiliation(s)
- Giacomo Triggiano
- Medical Oncology Unit, Policlinico of Bari, 70124 Bari, Italy; (G.T.)
| | - Gaetano Pezzicoli
- Medical Oncology Unit, Policlinico of Bari, 70124 Bari, Italy; (G.T.)
| | - Marco Tucci
- Medical Oncology Unit, Policlinico of Bari, 70124 Bari, Italy; (G.T.)
- Department of Interdisciplinary Medicine (DIM), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| |
Collapse
|
32
|
Zhang J, Yang H, Li L, Peng C, Li J. Noninvasive Transdermal Delivery of STING Agonists Reshapes the Immune Microenvironment of Melanoma and Potentiates Checkpoint Blockade Therapy Efficacy. ACS APPLIED BIO MATERIALS 2025; 8:3156-3166. [PMID: 40191891 DOI: 10.1021/acsabm.4c02004] [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: 04/22/2025]
Abstract
The emergence of immunotherapy as a revolutionary therapeutic modality has fostered confidence and underscored its potent efficacy in tumor therapy. However, enhancing the therapeutic efficacy of immunotherapy by precise and judicious administration poses a significant challenge. In this context, we have developed a disulfide-bearing transdermal nanovaccine by integrating a thiol-reactive agent lipoic acid (LA) into a metal-coordinated cyclic dinucleotide nanoassembly, designated as LA-Mn-cGAMP (LMC) nanovaccines. Upon topical application to the skin with melanoma, the dithiolane moiety of LA enables thiol-disulfide dynamic exchange in the skin, hence facilitating penetration into both the skin and subcutaneous tumor tissues via the thiol-mediated uptake (TMU) mechanism. Our findings demonstrate that transdermal administration of LMC significantly enhances STING activation, mitigates the immunosuppressive tumor microenvironment (TME), and retards melanoma progression. Moreover, the remodeled TME amplifies the efficacy of immune checkpoint inhibitors. This advancement offers an administration strategy for existing STING agonist therapy, potentially improving the biosafety of immunotherapy.
Collapse
Affiliation(s)
- Junjie Zhang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
| | - Hui Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
| | - Liang Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, China
| | - Changkun Peng
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
| | - Jingying Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, China
| |
Collapse
|
33
|
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
|
34
|
Pauken KE, Alhalabi O, Goswami S, Sharma P. Neoadjuvant immune checkpoint therapy: Enabling insights into fundamental human immunology and clinical benefit. Cancer Cell 2025; 43:623-640. [PMID: 40118048 DOI: 10.1016/j.ccell.2025.03.005] [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: 01/21/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/23/2025]
Abstract
While immune checkpoint therapy (ICT) has revolutionized cancer treatment, most patients with advanced disease fail to achieve durable benefit. To address this challenge, it is essential to integrate mechanistic research with clinical studies to: (1) understand response mechanisms, (2) identify patient-specific resistance pathways, (3) develop biomarkers for patient selection, and (4) design novel therapies to overcome resistance. We propose that incorporating "direct-in-patient" studies into clinical trials is crucial for bridging the gap between fundamental science and clinical oncology. In this review, we first highlight recent clinical success of ICT in the neoadjuvant setting, where treatment is given in earlier disease stages to improve outcomes. We then explore how neoadjuvant clinical trials could be utilized to drive mechanistic laboratory-based investigations. Finally, we discuss novel scientific concepts that will potentially aid in overcoming resistance to ICT, which will require future clinical trials to understand their impact on human immune responses.
Collapse
Affiliation(s)
- Kristen E Pauken
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Omar Alhalabi
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sangeeta Goswami
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; James P Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Padmanee Sharma
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; James P Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
35
|
Fager A, Samuelsson M, Olofsson Bagge R, Pivodic A, Bjursten S, Levin M, Jespersen H, Ny L. Immune checkpoint inhibitor therapy is associated with a decreased risk of developing melanoma brain metastases. BJC REPORTS 2025; 3:22. [PMID: 40217072 PMCID: PMC11992042 DOI: 10.1038/s44276-025-00137-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 02/27/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Despite recent advancements in metastatic melanoma treatment, the emergence of melanoma brain metastases (MBM) continues to pose a challenge. This study aimed to explore factors associated with MBM development. METHODS This retrospective study included patients diagnosed with advanced melanoma (unresectable stages III and IV [M1a-c]) between 2013 and 2019 at Sahlgrenska University Hospital, Gothenburg, Sweden. Differences in baseline and primary tumor characteristics, mutational status, biomarker levels, and first-line treatment between patients who developed MBM (BM+) and patients who did not develop MBM (BM-) were analyzed using univariable and multivariable Cox proportional hazard regression. RESULT Of 395 patients, 91 subsequently developed MBM. Patients who received immune checkpoint inhibitors (ICI) as first-line treatment had a reduced risk of MBM development (p ≤ 0.001). None of the eleven patients who received CTLA-4 inhibitors as monotherapy or in combination with PD-1 inhibitors as first-line treatment developed brain metastases. Elevated plasma levels of S100B (p = 0.021) and higher metastatic stage (p = 0.047) were also associated with an increased risk of MBM development. CONCLUSION ICI treatment is associated with a decreased risk of MBM development, suggesting a protective role. Elevated S100B levels and stage IV disease at advanced melanoma diagnosis might indicate an increased risk of MBM development.
Collapse
Affiliation(s)
- Anna Fager
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Matilda Samuelsson
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Sara Bjursten
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Max Levin
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Jespersen
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Lars Ny
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
36
|
Rimassa L, Chan SL, Sangro B, Lau G, Kudo M, Reig M, Breder V, Ryu MH, Ostapenko Y, Sukeepaisarnjaroen W, Varela M, Tougeron D, Crysler OV, Bouattour M, Van Dao T, Tam VC, Faccio A, Furuse J, Jeng LB, Kang YK, Kelley RK, Paskow MJ, Ran D, Xynos I, Kurland JF, Negro A, Abou-Alfa GK. Five-year overall survival update from the HIMALAYA study of tremelimumab plus durvalumab in unresectable HCC. J Hepatol 2025:S0168-8278(25)00226-0. [PMID: 40222621 DOI: 10.1016/j.jhep.2025.03.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND & AIMS In the phase III HIMALAYA study (NCT03298451), STRIDE (Single Tremelimumab Regular Interval Durvalumab) significantly improved overall survival (OS) versus sorafenib in unresectable HCC (uHCC) and demonstrated long-term survival benefits. We report an updated exploratory analysis of OS with 5 years of follow-up, including survival by multiple tumour response measures. METHODS Participants were randomised to STRIDE (tremelimumab plus durvalumab), durvalumab or sorafenib. OS, depth of response and serious adverse events (AEs) were assessed. Extended long-term survivors (eLTS; ≥48 months beyond randomisation) were described. Updated data cut-off: 01 March 2024. RESULTS Median (95% CI) follow-up durations were 62.49 (59.47-64.79) months (STRIDE) and 59.86 (58.32-61.54) months (sorafenib). The OS HR (95% CI) for STRIDE versus sorafenib was 0.76 (0.65-0.89). OS rates at 60 months for STRIDE versus sorafenib were 19.6% versus 9.4% overall, 28.7% versus 12.7% in participants achieving disease control per RECIST v1.1 and 50.7% versus 26.3% in participants achieving >25% tumour shrinkage. No late-onset treatment-related serious AEs were reported for STRIDE. There were more eLTS with STRIDE (83/393, 21.1%) than sorafenib (45/389, 11.6%), and extended long-term survival occurred across all clinically relevant subgroups. CONCLUSIONS At 5 years, STRIDE sustained an OS benefit versus sorafenib and maintained a manageable safety profile. OS benefit with STRIDE was improved in participants with disease control. Data suggest that any degree of tumour shrinkage with STRIDE can be associated with improved OS, indicating that conventional response measures may not fully capture STRIDE benefits. Nevertheless, participants experiencing deep responses appear to have the greatest benefit. STRIDE continues to set new benchmarks in uHCC with 1 in 5 patients alive at 5 years. IMPACT AND IMPLICATIONS The phase III HIMALAYA study showed that STRIDE (Single Tremelimumab Regular Interval Durvalumab) improved overall survival (OS) versus sorafenib in participants with unresectable HCC (uHCC), including after 4 years of follow-up. Understanding the efficacy and safety of STRIDE over the longer term is important for healthcare providers; here, we demonstrate that STRIDE sustained an OS benefit versus sorafenib and maintained a manageable safety profile after 5 years of follow-up. OS benefit with STRIDE was improved in participants with disease control and any degree of tumour shrinkage, indicating that conventional response measures may not fully capture the benefits of STRIDE. These findings are important as they set new benchmarks in uHCC and may help guide clinical decisions in the future.
Collapse
Affiliation(s)
- Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| | - Stephen L Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir Yue-Kong Pao Center for Cancer, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bruno Sangro
- Liver Unit and HPB Oncology Area, Clínica Universidad de Navarra and CIBEREHD, Pamplona - Madrid, Spain
| | - George Lau
- Humanity and Health Clinical Trial Center, Humanity and Health Medical Group, Hong Kong SAR, China
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Maria Reig
- Barcelona Clinic Liver Cancer (BCLC), Liver Unit, Hospital Clinic de Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain
| | - Valeriy Breder
- Department of Chemotherapy, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russian Federation
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yuriy Ostapenko
- Department of Minimally Invasive and Endoscopic Surgery, Interventional Radiology, National Cancer Institute, Kyiv, Ukraine
| | | | - Maria Varela
- Liver Unit, Hospital Universitario Central de Asturias, IUOPA, ISPA, FINBA, Universidad de Oviedo, Oviedo, Spain
| | - David Tougeron
- Department of Gastroenterology and Hepatology, Poitiers University Hospital, Poitiers, France
| | - Oxana V Crysler
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Tu Van Dao
- Cancer Research and Clinical Trials Center, Department of Optimal Therapy, National Cancer Hospital, Hanoi, Vietnam
| | - Vincent C Tam
- Department of Oncology, Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Adilson Faccio
- Department of Oncology, CEON - Centro Especializado em Oncologia, Ribeirao Preto, Brazil
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Long-Bin Jeng
- Department of Surgery, China Medical University and Hospital, Taichung, Taiwan, Republic of China
| | - Yoon Koo Kang
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Robin K Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Michael J Paskow
- Global Medical Affairs, AstraZeneca, Gaithersburg, Maryland, USA
| | - Di Ran
- Statistics, AstraZeneca, Gaithersburg, Maryland, USA
| | - Ioannis Xynos
- Oncology R&D, Late-Stage Development, AstraZeneca, Cambridge, UK
| | - John F Kurland
- Oncology R&D, Late-Stage Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Alejandra Negro
- Oncology R&D, Late-Stage Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Ghassan K Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Cornell University, New York, New York, USA; Weill Medical College, Cornell University, New York, New York, USA; Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
37
|
Słaby J, Wnuk M, Błoniarz D, Stec P, Szmatoła T, Kaznowska E, Reich A, Moros M, Lewińska A. ITGA1, the alpha 1 subunit of integrin receptor, is a novel marker of drug-resistant senescent melanoma cells in vitro. Arch Toxicol 2025:10.1007/s00204-025-04028-w. [PMID: 40202610 DOI: 10.1007/s00204-025-04028-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025]
Abstract
Chemotherapy-induced senescence may promote drug resistance and treatment failure. Precise detection and elimination of senescent cancer cells is considered as a novel promising anticancer strategy. However, data on senescence-associated skin cancer cell surface markers as potential therapeutic targets are limited. In the present study, we have established two models of drug-induced senescence in vitro using DNA damaging chemotherapeutics, namely etoposide (0.75-5 µM) and cisplatin (1.25-5 µM), and ten skin cancer cell lines, both melanoma (n = 8, A375, G-361, MM370, SH-4, SK-MEL-1, MeWo, MM127, RPMI-7951) and non-melanoma (n = 2, A431, MCC13), to investigate the levels of 97 cell surface markers. Initial gene expression analysis revealed the increasing tendency in the levels of seven transcripts (ITGA1, ITGA3, VAMP3, STX4, ARMCX3, ULBP2, and PLAUR) and five transcripts (ITGA1, ITGA3, STX4, ARMCX3, and PLAUR) in five etoposide and cisplatin-induced senescent melanoma cell lines, respectively, compared to corresponding proliferating cells. Elevated pools of integrin α1 (ITGA1) were confirmed at mRNA and protein levels in eight drug-induced senescent melanoma cell lines. Similar pattern of changes in integrin α1 levels was not observed in drug-induced senescent non-melanoma skin cancer cells. Analysis using clinical melanoma samples also showed that the levels of ITGA1 and ITGA3 were correlated with the presence of melanoma cells in a section. We document that integrin α1 can be considered as a novel marker of drug-induced senescent melanoma cells. Thus, we postulate that new integrin α1-based targeted therapies can be designed and tested against drug-induced senescent melanoma cells.
Collapse
Affiliation(s)
- Julia Słaby
- Doctoral School, University of Rzeszow, Rejtana 16C, 35-959, Rzeszow, Poland
| | - Maciej Wnuk
- Department of Biotechnology, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland
| | - Dominika Błoniarz
- Department of Biotechnology, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland
| | - Paulina Stec
- Department of Biotechnology, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland
| | - Tomasz Szmatoła
- Department of Basic Sciences, University of Agriculture in Krakow, al. Mickiewicza 24/28, 30-059, Kraków, Poland
| | - Ewa Kaznowska
- Department of Medical Sciences, University of Rzeszow, Warzywna 1a, 35-310, Rzeszów, Poland
| | - Adam Reich
- Department of Medical Sciences, University of Rzeszow, Warzywna 1a, 35-310, Rzeszów, Poland
| | - María Moros
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/ Pedro Cerbuna 12, 50009, Saragossa, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029, Madrid, Spain
| | - Anna Lewińska
- Department of Biotechnology, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland.
| |
Collapse
|
38
|
Tarin M, Oryani MA, Javid H, Karimi-Shahri M. Exosomal PD-L1 in non-small cell lung Cancer: Implications for immune evasion and resistance to immunotherapy. Int Immunopharmacol 2025; 155:114519. [PMID: 40199140 DOI: 10.1016/j.intimp.2025.114519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025]
Abstract
Exosomes, characterized by their bilayer lipid structure, are crucial in mediating intercellular signaling and contributing to various physiological processes. Tumor cells produce distinct exosomes facilitating cancer progression, angiogenesis, and metastasis by conveying signaling molecules. A notable feature of these tumor-derived exosomes is the presence of programmed death-ligand 1 (PD-L1) on their surface. The PD-L1/programmed cell death receptor-1 (PD-1) signaling axis serves as a critical immune checkpoint, enabling tumors to evade immune detection and antitumor activity. The advancement of immunotherapy targeting the PD-1/PD-L1 pathway has significantly impacted the treatment landscape for non-small cell lung cancer (NSCLC). Despite its promise, evidence indicates that many patients experience limited responses or develop resistance to PD-1/PD-L1 inhibitors. Recent studies suggest that exosomal PD-L1 contributes to this resistance by modulating immune responses and tumor adaptability. This study reviews the PD-1/PD-L1 pathway's characteristics, current clinical findings on PD-L1 inhibitors in NSCLC, and exosome-specific attributes, with a particular focus on exosomal PD-L1. Furthermore, it examines the growing body of research investigating the role of exosomal PD-L1 in cancer progression and response to immunotherapy, underscoring its potential as a target for overcoming resistance in NSCLC treatment.
Collapse
Affiliation(s)
- Mojtaba Tarin
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mahsa Akbari Oryani
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
39
|
van Dijk B, Schoenaker IJH, van der Veldt AAM, de Groot JWB. Exhaled breath analysis with the use of an electronic nose to predict response to immune checkpoint inhibitors in patients with metastatic melanoma: melaNose trial. Front Immunol 2025; 16:1564463. [PMID: 40248699 PMCID: PMC12003352 DOI: 10.3389/fimmu.2025.1564463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
Introduction Immune checkpoint inhibitors (ICIs) have significantly improved the overall survival for patients with different solid tumors. However, there is an urgent need for predictive biomarkers to identify patients with metastatic melanoma who do not benefit from treatment with ICIs, to prevent unnecessary immune related adverse events (irAEs). Electronic noses (eNoses) showed promising results in the detection of cancer as well as the prediction of response outcome in patients with cancer. In this feasibility study, we aimed to investigate whether the breath pattern measured using eNose can be used as a simple biomarker to predict clinical benefit to first-line treatment with ICIs in patients with metastatic melanoma. Methods In this prospective, observational single-center feasibility study, patients with metastatic melanoma performed a breath test using Aeonose™ before start of first-line treatment with ICIs. The detected exhaled breath pattern of volatile organic compounds (VOC) was used for machine learning in a training set to develop a model to identify patients who do not benefit from treatment with ICIs. Lack of clinical benefit was defined as progressive disease according to best tumor response using RECIST v1.1. Primary outcome measures were sensitivity, specificity and accuracy. Results The eNose showed a distinct breath pattern between patients with and without clinical benefit from ICIs. To identify patients who do not benefit from first-line ICIs treatment, breath pattern analysis using the eNose resulted in a sensitivity of 88%, specificity of 79%, and accuracy of 85%. Conclusion Exhaled breath analysis using eNose can identify patients with metastatic melanoma who will not benefit from first-line treatment with ICIs and guide treatment strategies. When validated in an external cohort, eNose could be a useful tool to select these patients for alternative treatment strategies in clinical practice.
Collapse
Affiliation(s)
- Brigit van Dijk
- Department of Medical Oncology, Erasmus Medical Center Medical Center (MC), Rotterdam, Netherlands
| | | | - Astrid A. M. van der Veldt
- Department of Medical Oncology, Erasmus Medical Center Medical Center (MC), Rotterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center (MC), Rotterdam, Netherlands
| | | |
Collapse
|
40
|
Lonberg N. The Problem with Syngeneic Mouse Tumor Models. Cancer Immunol Res 2025; 13:456-462. [PMID: 39996612 DOI: 10.1158/2326-6066.cir-24-1046] [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: 10/31/2024] [Revised: 12/16/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025]
Abstract
The advent of syngeneic mouse tumor models provided the scientific foundation for cancer immunotherapies now in widespread use. However, in many respects, these models do not faithfully recapitulate the interactions between cancer cells and the immune systems of human patients who have solid tumors because they represent a very early stage in the immune response to the newly transplanted cancer cells compared with the relatively mature stage found in human patients at the time of treatment. The lack of translatability of syngeneic models is probably responsible for many failed clinical trials conducted at considerable expense, involving far too many patients with cancer who received no benefit. Better mouse models would substantially accelerate the pace of discovery of new immunotherapies. Until these models emerge, a better understanding of the differences between the existing syngeneic models and human cancers may provide a more efficient path for moving experimental drugs into clinical development. To accomplish this, we must consider mice transplanted with syngeneic tumor cells to be in vivo assays, potentially useful for understanding the mechanism of action of immunotherapies rather than disease models.
Collapse
|
41
|
Reitmajer M, Nanz L, Müller N, Leiter U, Amaral T, Aebischer V, Flatz L, Forschner A. Comparative real-world outcomes of stage III melanoma patients treated with talimogene laherparepvec or interleukin 2. Ther Adv Med Oncol 2025; 17:17588359251324035. [PMID: 40171522 PMCID: PMC11960150 DOI: 10.1177/17588359251324035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/12/2025] [Indexed: 04/03/2025] Open
Abstract
Background Talimogene laherparepvec (T-VEC) and interleukin-2 (IL-2) are both used in the intralesional treatment of melanoma skin metastases. T-VEC received regulatory approval from the European Medicines Agency and the U.S. Food and Drug Administration in 2015, while IL-2 has been used off-label for this purpose for many years. Despite their use in clinical practice, there is a lack of comparative data on the efficacy and safety of these treatments. Objectives This retrospective study aimed to compare the efficacy and safety of intralesional T-VEC and IL-2 in non-resectable stage III patients with melanoma treated at a single center between January 2016 and September 2024. Methods We identified eligible patients using the Central Malignant Melanoma Registry and the local University Hospital Pharmacy database. Overall survival (OS) and progression-free survival (PFS) were calculated. Furthermore, best response rates and occurrence of adverse events (AEs) were compared between the T-VEC and the IL-2 group. Concomitant systemic treatment was allowed. Results A total of 62 patients were included, with 37 receiving T-VEC and 25 receiving IL-2 as first-line therapy. Ten patients received both therapies subsequently. The median PFS for the cohort was 5.0 months, and the median OS was 34.0 months. No significant differences in PFS (p = 0.790), OS (p = 0.894), or best response rates (p = 0.468) were found between groups. Common AEs included local injection site reactions and fever, with no severe events leading to discontinuation by a physician. Conclusion No significant differences in PFS, OS, or best response rates were observed between IL-2 and T-VEC treatments. The choice of therapy may be influenced by factors such as availability, physician preference, and patient-specific considerations.
Collapse
Affiliation(s)
- Markus Reitmajer
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstraße 25, Tuebingen 72076, Germany
| | - Lena Nanz
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Nina Müller
- University Pharmacy, University Hospital Tuebingen, Tuebingen, Germany
| | - Ulrike Leiter
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Teresa Amaral
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Valentin Aebischer
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Lukas Flatz
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Andrea Forschner
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
42
|
Fina E, Vitale E, De Summa S, Gadaleta-Caldarola G, Tommasi S, Massafra R, Brunetti O, Rizzo A. Liquid biopsy for guiding breast cancer immunotherapy. Immunotherapy 2025; 17:369-383. [PMID: 40083311 PMCID: PMC12045575 DOI: 10.1080/1750743x.2025.2479426] [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: 10/27/2024] [Accepted: 03/11/2025] [Indexed: 03/16/2025] Open
Abstract
Liquid biopsy is a laboratory test used to detect and analyze circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and other tumor-derived components, in a blood sample. In the context of breast cancer (BC), liquid biopsies hold significant promise for guiding the use of immune checkpoint inhibitors and immune-based combinations, offering real-time insights into tumor dynamics, treatment response, and resistance mechanisms. This review explores the role of liquid biopsy in BC immunotherapy, focusing on its applications, benefits, issues, and current and future research directions.
Collapse
Affiliation(s)
- Emanuela Fina
- Thoracic Surgery Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elsa Vitale
- Scientific Directorate, IRCCS Istituto Tumori “Giovanni Paolo II, Bari, Italy
| | - Simona De Summa
- Unità di Diagnostica Molecolare e Farmacogenetica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | | | - Stefania Tommasi
- Unità di Diagnostica Molecolare e Farmacogenetica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Raffaella Massafra
- Scientific Directorate, IRCCS Istituto Tumori “Giovanni Paolo II, Bari, Italy
| | - Oronzo Brunetti
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II, Bari, Italy
| | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II, Bari, Italy
| |
Collapse
|
43
|
Wu Z, Sun W, He B, Wang C. Clinical characteristics, treatment, and outcomes of nivolumab-induced uveitis. Immunopharmacol Immunotoxicol 2025; 47:222-227. [PMID: 39885372 DOI: 10.1080/08923973.2025.2461056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/26/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Nivolumab has been linked to occurrences of uveitis, yet the clinical features associated with these episodes remain unclear. This study aimed to explore the clinical characteristics of uveitis induced by nivolumab and to offer guidance for its prevention, diagnosis, and treatment. METHODS We conducted a retrospective analysis by gathering case reports related to nivolumab-induced uveitis from both Chinese and English databases, covering the period from inception until 30 September 2024. RESULTS A total of 38 patients with uveitis were included, with a median age of 63 years (range 35 and 92). The onset of uveitis occurred between 1 week and 24 months post-administration, with a median onset time of 1.4 months. Blurred vision was the primary complaint among patients. Sixteen patients (42.1%) exhibited uveitis resembling Vogt-Koyanagi-Harada (VKH) disease. Bilateral uveitis was the most prevalent form (89.2%), followed by unilateral uveitis (8.1%). Anterior uveitis was the most frequently observed type (52.6%), succeeded by posterior uveitis (23.7%), panuveitis (21.1%), and intermediate uveitis (2.6%). Uveitis showed significant improvement or resolution following treatment with topical or systemic corticosteroids, with a median improvement time of 4 weeks post-therapy. CONCLUSIONS Uveitis is a relatively uncommon adverse effect of nivolumab, typically manifesting within 5 months of treatment. Prompt recognition of nivolumab-induced uveitis and appropriate management are crucial, as most cases are treatable.
Collapse
Affiliation(s)
- Zhaoquan Wu
- College of Pharmacy, Changsha Medical University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan, China
| | - Wei Sun
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Binsheng He
- College of Pharmacy, Changsha Medical University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan, China
| | - Chunjiang Wang
- College of Pharmacy, Changsha Medical University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan, China
| |
Collapse
|
44
|
Ziogas DC, Theocharopoulos C, Aravantinou K, Boukouris AE, Stefanou D, Anastasopoulou A, Lialios PP, Lyrarakis G, Gogas H. Clinical benefit of immune checkpoint inhibitors in elderly cancer patients: Current evidence from immunosenescence pathophysiology to clinical trial results. Crit Rev Oncol Hematol 2025; 208:104635. [PMID: 39889861 DOI: 10.1016/j.critrevonc.2025.104635] [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: 06/27/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
The age-related decline in immunity appears to be associated not only with cancer development but also with differential responses to immune checkpoint inhibitors (ICIs). Despite their increasing utility across various malignancies and therapeutic settings, limited data -derived primarily from subgroup analyses of randomized controlled trials (RCTs), pooled meta-analyses, and retrospective studies- are available on the effects of aging on their efficacy and toxicity. Immunosenescence, characterized by the progressive decline of the function of the immune system, and inflammaging, a state of persistent low-grade sterile inflammation, may influence ICI outcomes. Additionally, the incidence, severity, and subtypes of immune-related adverse events (irAEs) may differ between older and younger individuals due to loss of immunotolerance. In the current review, starting from a a comprehensive discussion of the pathophysiology of immunosenescence, we proceed to critically review age-related retrospective and randomized evidence supporting FDA-approved ICIs. We highlight similarities or differences across age groups and the clinical benefit of ICIs in elderly versus younger cancer patients. The optimal integration of ICIs in geriatric oncology necessitates greater inclusion of this patient demographic in RCTs along with real-world data in order to acquire robust data which will guide evidence-based treatment decisions for this population.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Charalampos Theocharopoulos
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Katerina Aravantinou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Aristeidis E Boukouris
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Dimitra Stefanou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Amalia Anastasopoulou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Panagiotis-Petros Lialios
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - George Lyrarakis
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| |
Collapse
|
45
|
Ferrari M, Facchini BA, Ascierto PA, Sparano F. Melanoma neoadjuvant treatment: review and update of recent trials. Expert Rev Anticancer Ther 2025; 25:383-392. [PMID: 40043281 DOI: 10.1080/14737140.2025.2474182] [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: 10/16/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
INTRODUCTION Neoadjuvant immunotherapy is emerging as an effective approach for resectable stage III/IV melanoma, showing improvements in disease response and survival outcomes. AREAS COVERED This review summarizes findings from neoadjuvant treatment trials in melanoma patients. Using the PubMed search engine and including the keywords 'neoadjuvant,' 'immunotherapy,' and 'melanoma,' we selected 18 trials that showed efficacy in patients with melanoma, mainly testing checkpoint inhibitors alone or in combination. Across all trials examined, treatments showed objective disease responses, which frequently translated into improved disease-free survival. EXPERT OPINION Additional phase III studies comparing neoadjuvant and adjuvant therapies are needed to establish the optimal standard of care. The variety of regimens and dosing schedules investigated highlights the need for further research to determine the most appropriate treatments in this clinical setting. Advances in the study of biomarkers that can identify specific subgroups of patients will guide future research in this field.
Collapse
Affiliation(s)
- Marco Ferrari
- Azienda USL Toscana centro, U.O. Oncologia Medica, Ponte a Niccheri, Bagno a Ripoli, FI, Italy
| | - Bianca Arianna Facchini
- Istituto Nazionale Tumori IRCCS Fondazione Giovanni Pascale - Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Napoli, Napoli, Italy
| | - Paolo Antonio Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Giovanni Pascale - Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Napoli, Napoli, Italy
| | - Francesca Sparano
- Istituto Nazionale Tumori IRCCS Fondazione Giovanni Pascale - Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Napoli, Napoli, Italy
| |
Collapse
|
46
|
Shibano M, Takahashi M, Nakatsukasa H, Ishigami Y, Toyokawa T, Taira K, Kawaguchi T, Nakamura Y, Kaneda H. Proton pump inhibitors reduce nivolumab efficacy in unresectable advanced or recurrent gastric cancer. Immunotherapy 2025; 17:331-338. [PMID: 40228034 PMCID: PMC12045562 DOI: 10.1080/1750743x.2025.2491300] [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: 02/08/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Proton pump inhibitors (PPI) have been shown to decrease the efficacy of immune checkpoint inhibitors in patients with various cancer types. However, there are few reports on their effect on patients with gastric cancer (GC). Therefore, we investigated the efficacy of nivolumab in patients with GC receiving PPI. METHODS This retrospective study analyzed data of patients who received nivolumab monotherapy for unresectable advanced or recurrent GC at Osaka Metropolitan University Hospital between September 2017 and December 2021. The primary and secondary endpoints were progression-free survival (PFS) and overall survival (OS), respectively. PPI use was defined as within 30 days before and after initiation of nivolumab monotherapy. RESULTS Seventy-seven eligible patients were included in this analysis. PPIs were used in 33 patients, while 36 patients had a previous gastrectomy. Multivariate analysis revealed that only PPI use was an independent predictor of PFS (hazard ratio [HR] 1.93, 95% confidence interval [CI] 1.03-3.64, p = 0.042). Contrastingly, PPI use was not an independent predictor of OS. CONCLUSION PPIs may reduce the efficacy of nivolumab, and their use should be carefully considered in patients receiving nivolumab.
Collapse
Affiliation(s)
- Masahito Shibano
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Masaya Takahashi
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
- Department of Quality and Safety Management, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Hitomi Nakatsukasa
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Yusuke Ishigami
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Takahiro Toyokawa
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Koichi Taira
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Tomoya Kawaguchi
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yasutaka Nakamura
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Hiroyasu Kaneda
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
47
|
Gray-Gaillard EF, Shah AA, Bingham Iii CO, Elisseeff JH, Murray J, Brahmer J, Forde P, Anagnostou V, Mammen J, Cappelli LC. Higher levels of VEGF-A and TNFα in patients with immune checkpoint inhibitor-induced inflammatory arthritis. Arthritis Res Ther 2025; 27:74. [PMID: 40170117 PMCID: PMC11959780 DOI: 10.1186/s13075-025-03546-3] [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: 11/25/2024] [Accepted: 03/23/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI), a type of cancer immunotherapy, can cause side effects including inflammatory arthritis (ICI-IA). Previous studies of ICI-IA do not include a thorough characterization of associated immune responses to provide potential targets for treatment. We aimed to identify cytokines uniquely increased in ICI-IA and determine correlations with IA severity and persistence. METHODS We evaluated patients diagnosed with ICI-IA by a rheumatologist (n = 80); control serum was obtained from ICI-treated cancer patients without any diagnosed irAEs (n = 17) or diagnosed with an unrelated irAE (n = 19). Serum was assayed to quantify 9 cytokine levels (IFN-γ, IL-4, IL-6, IL-10, IL-12p70, IL-1α, TNF-α, IL-17a, VEGF-A) using MSD U-PLEX assay. Mann-Whitney U tests were performed to evaluate differences in cytokine levels between control and ICI-IA groups. The Kruskal-Wallis test and multivariable ordinal logistic regression were used to determine difference in cytokine levels between patients of differing disease activity. RESULTS VEGF-A and TNFα were significantly elevated in patients with ICI-IA compared to ICI-controls; results persisted when restricting analyses to patients not treated with immunosuppressants at the time of sampling. ICI-IA patients were stratified by IA severity using CDAI score; there was significantly higher VEGF-A in those with higher disease activity. Ordinal logistic regression showed higher levels of IL-6 and VEGF-A were associated with higher disease activity. CONCLUSION Elevated levels of VEGF-A and TNFα are associated with ICI-IA. There was also higher IL-6 and VEGF-A among those with higher disease activity when controlling for confounding. These cytokines could be used as biomarkers of ICI-IA severity and present therapeutic targets.
Collapse
Affiliation(s)
- Elise F Gray-Gaillard
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Ami A Shah
- Division of Rheumatology, Johns Hopkins School of Medicine, 5501 Hopkins Bayview Circle Suite 1B1, Baltimore, MD, 21224, USA
| | - Clifton O Bingham Iii
- Division of Rheumatology, Johns Hopkins School of Medicine, 5501 Hopkins Bayview Circle Suite 1B1, Baltimore, MD, 21224, USA
| | - Jennifer H Elisseeff
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Joseph Murray
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Julie Brahmer
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Patrick Forde
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Valsamo Anagnostou
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jennifer Mammen
- Division of Endocrinology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Laura C Cappelli
- Division of Rheumatology, Johns Hopkins School of Medicine, 5501 Hopkins Bayview Circle Suite 1B1, Baltimore, MD, 21224, USA.
| |
Collapse
|
48
|
Su X, Luo Y, Wang Y, Qu P, Liu J, Han S, Ma C, Deng S, Liang Q, Qi X, Cheng P, Hou L. A select inhibitor of MORC2 encapsulated by chimeric membranecoated DNA nanocage target alleviation TNBC progression. Mater Today Bio 2025; 31:101497. [PMID: 39906202 PMCID: PMC11791359 DOI: 10.1016/j.mtbio.2025.101497] [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: 11/14/2024] [Revised: 01/02/2025] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most malignant type of breast cancer and lacks effective targeted therapeutic drugs, resulting in a high recurrence rate and worse outcome. In this study, bioinformatic analysis and a series of experiments demonstrated that MOCR2 was highly expressed in TNBC and closely associated with poor prognosis, indicating that MOCR2 may be a potential therapeutic target for TNBC. Subsequently, Angoline was identified as an inhibitor of MORC2 protein by high-throughput screening and can significantly kill the TNBC cells by blocking cell cycle and inducing apoptosis. Furthermore, the biomimetic nanodrug delivery system (PMD) was designed by encapsulating tetrahedral DNA nanostructures with biomimetic cell membrane, and it can efficiently evade the phagocytosis of immune system and target TNBC tissue. Additionally, PMD can markedly enhance the killing effect of Angoline on TNBC tumors. Therefore, PMD-enveloped Angoline provide a highly effective targeted therapeutic regimen for TNBC and may improve the outcome for patients with TNBC.
Collapse
Affiliation(s)
- Xiaohan Su
- Breast Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Breast Surgery, Mianyang 404 hospital, Mianyang, China
| | - Yunbo Luo
- Breast Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yali Wang
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Peng Qu
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jun Liu
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shiqi Han
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Cui Ma
- Department of Mathematics, Army Medical University, Chongqing, China
| | - Shishan Deng
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qi Liang
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaowei Qi
- Department of Breast Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu, China
| | - Lingmi Hou
- Breast Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
49
|
Braverman EL, Vignali DAA, Workman CJ. Multiplex targeting of immune-modulating genes in cancer. Nat Immunol 2025; 26:531-533. [PMID: 40108420 DOI: 10.1038/s41590-025-02115-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Affiliation(s)
- Erica L Braverman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
50
|
Tsai KK, Chang AY, Abouzeid CM, Aras MA, Fang Q, Bibby D, Haghighat L, Hilton JF, Daud AI, Schiller NB. Longitudinal Evaluation of Immune Checkpoint Inhibitor-Induced Fatigue Syndrome by Rest, Stress, and Speckle-Tracking Strain Echocardiography. Echocardiography 2025; 42:e70158. [PMID: 40198654 DOI: 10.1111/echo.70158] [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: 01/06/2025] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI) anti-tumor therapy is commonly associated with reports of significant fatigue. Whether ICI-induced fatigue is linked to subclinical cardiac toxicity is not well understood. METHODS We performed a prospective observational study to monitor cardiac function following initiation of ICI in cancer patients. Rest and staged exercise stress transthoracic echocardiograms (TTE) were captured at baseline, 3 and 6 months after ICI initiation. RESULTS Thirteen patients completed baseline testing, of whom 10 completed 3 month testing and 8 completed 6 month testing. Patients had elevated fatigue as per Common Terminology Criteria for Adverse Events (CTCAE) criteria and FACIT-Fatigue subscale scores (47.5 [42.5-52] vs. 47 [38-48], p = 0.001). Among resting echocardiographic parameters, left ventricular (LV) outflow tract velocity time integral (VTI) was reduced at 6 months (22.6 cm [21.2-24.5] vs. 19.8 cm [17.2-21.2], p = 0.001), albeit still within normal range. Measures of biventricular size and systolic function, along with LV global longitudinal strain (GLS) and LA global strain, were not significantly changed. Among exercise parameters, peak heart rate was reduced at 6 months (143.4 bpm [128.5-153.5] vs. 123.5 [110.2-130.8], p = 0.048); of those who completed 6 month testing, two (29%) were unable to achieve the previous peak stage of exercise. CONCLUSIONS In patients starting ICI, statistically-significant reductions in cardiac systolic function (LVOT VTI) and peak HR were observed, with nearly one-third unable to achieve peak exercise at 6 months. This finding, combined with reported fatigue and lack of changes to biventricular systolic functional measures, suggests induced functional cardiac limitations are due to deconditioning, rather than cardiotoxicity.
Collapse
Affiliation(s)
- Katy K Tsai
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, California, USA
| | - Andrew Y Chang
- Department of Medicine, Section of Cardiovascular Medicine, Yale University, New Haven, Connecticut, USA
| | - Christiane M Abouzeid
- Department of Medicine, Division of Cardiology, University of Southern California, Los Angeles, California, USA
| | - Mandar A Aras
- Department of Medicine, Division of Cardiology, University of California, San Francisco, San Francisco, California, USA
| | - Qizhi Fang
- Department of Medicine, Division of Cardiology, University of California, San Francisco, San Francisco, California, USA
| | - Dwight Bibby
- Department of Medicine, Division of Cardiology, University of California, San Francisco, San Francisco, California, USA
| | - Leila Haghighat
- Department of Medicine, Division of Cardiology, University of California, San Francisco, San Francisco, California, USA
- Sutter Health, Palo Alto Medical Foundation, Burlingame, California, USA
| | - Joan F Hilton
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Adil I Daud
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, California, USA
| | - Nelson B Schiller
- Department of Medicine, Division of Cardiology, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|