1
|
Lotti F, Melixetian M, Vlachou T, Nobile MS, Bacciu L, Malferrari M, Quaresima N, Rapino S, Marocchi F, Barberis M, Soriani C, Gallo B, Mollo V, Ferrarotto I, Bossi D, Ferrucci PF, Pelicci PG, Luzi L, Lanfrancone L. GPNMB marks a quiescent cell population in melanoma and promotes metastasis formation. EMBO Rep 2025:10.1038/s44319-025-00501-w. [PMID: 40528051 DOI: 10.1038/s44319-025-00501-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: 12/11/2024] [Revised: 05/26/2025] [Accepted: 06/02/2025] [Indexed: 06/20/2025] Open
Abstract
Melanoma exhibits high intratumoral heterogeneity, characterized by a diverse population of cells undergoing dynamic transitions between cellular states. These adaptive changes enable melanoma cells to survive in the harsh tumor microenvironment, acquire drug resistance, and metastasize. One such state, quiescence, has been linked to both relapse and drug resistance, but its underlying biology and molecular mechanisms remain poorly understood. Our study challenges the conventional understanding of melanoma quiescence. Contrary to the notion of a rare, unique subpopulation, we demonstrate that quiescence is a highly dynamic state accessible to most, if not all, melanoma cells. This state is exquisitely sensitive to microenvironmental cues. We identify GPNMB as a marker of quiescence, that is expressed in both primary and metastatic tumors. GPNMB-positive cells exhibit a pro-metastatic phenotype and are enriched in metastatic sites, suggesting a potential role for quiescence in tumor dissemination. Our findings position GPNMB as a valuable marker for isolating quiescent melanoma cells and as a potential therapeutic target to tackle metastasis.
Collapse
Affiliation(s)
- Fiorenza Lotti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Marine Melixetian
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Thalia Vlachou
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Purposeful, Tritis Septembriou 144, Athens, 11251, Greece
| | - Marco S Nobile
- Department of Environmental Science, Computer Science and Statistics, University of Ca' Foscari, Venice, Italy
| | - Leone Bacciu
- Department of Environmental Science, Computer Science and Statistics, University of Ca' Foscari, Venice, Italy
| | - Marco Malferrari
- Department of Chemistry "Giacomo Ciamician", Università di Bologna, Via Selmi 2, 40126, Bologna, Italy
| | - Nicolò Quaresima
- Department of Chemistry "Giacomo Ciamician", Università di Bologna, Via Selmi 2, 40126, Bologna, Italy
| | - Stefania Rapino
- Department of Chemistry "Giacomo Ciamician", Università di Bologna, Via Selmi 2, 40126, Bologna, Italy
| | - Federica Marocchi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Massimo Barberis
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Soriani
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Barbara Gallo
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Velia Mollo
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Ilaria Ferrarotto
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Daniela Bossi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Institute of Oncology Research, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Pier Francesco Ferrucci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology, Gruppo MultiMedica, Milano, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Lucilla Luzi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
2
|
Huang W, Pan Y, Zhong T, He S, Qi Y, Huang Y. Near-infrared 10B-BODIPY for precise guidance of tracer imaging and treatment in boron neutron capture therapy. Chem Commun (Camb) 2025; 61:9079-9082. [PMID: 40401390 DOI: 10.1039/d5cc01671a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
A new near-infrared fluorescence probe (PBA-BDP) was developed by coupling boron-10 (10B) to a phenylboronic acid (PBA)-functionalized BODIPY dye. This multifunctional probe not only delivers 10B for boron neutron capture therapy (BNCT) but also enhances tumor-specific targeting and provides dynamic insight into boron distribution during treatment.
Collapse
Affiliation(s)
- Wenyong Huang
- Faculty of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| | - Yong Pan
- Faculty of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| | - Tianyuan Zhong
- Faculty of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| | - Shasha He
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen 361005, China
| | - Yanxin Qi
- Faculty of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| |
Collapse
|
3
|
Sun M, Ni C, Li A, Liu J, Guo H, Xu F, Li K, Cao X, Shi X, Guo R. A biomimetic nanoplatform mediates hypoxia-adenosine axis disruption and PD-L1 knockout for enhanced MRI-guided chemodynamic-immunotherapy. Acta Biomater 2025:S1742-7061(25)00435-0. [PMID: 40516842 DOI: 10.1016/j.actbio.2025.06.021] [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: 04/08/2025] [Revised: 05/21/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
Malignant melanoma is an extremely aggressive and fatal form of skin cancer due to the limited efficacy of conventional therapies. While immune checkpoint blockade therapy and chemodynamic therapy (CDT) have emerged as promising strategies for melanoma treatment, their effectiveness is compromised by the immunosuppressive and complex tumor microenvironment (TME). Here, cancer cell membrane-camouflaged nanoplatforms (PPMC@CM) were developed to co-deliver the CRISPR/Cas9-PD-L1 system and manganese dioxide nanoparticles (MnO2 NPs) for magnetic resonance imaging (MRI)-guided CDT and enhanced immunotherapy. The formed PPMC@CM could efficiently accumulate at tumor sites by homologous targeting, generate O2 to relieve hypoxia, and deplete glutathione (GSH) to enhance Mn2+-mediated Fenton-like reactions for enhanced CDT. Meanwhile, CRISPR/Cas9-mediated PD-L1 knockout effectively suppressed the PD-L1 expression, while hypoxia relief attenuated the immunosuppressive hypoxia-CD39/CD73-adenosine (ADO) pathway, thereby boosting the PD-L1-mediated immunotherapy. In vivo experimental results demonstrated that PPMC@CM nanoplatform could efficiently inhibit the growth and metastasis of melanoma by enhanced CDT and amplified immunotherapy, and provide targeted MRI of tumors. This work presents a novelty strategy to design biomimetic theranostic nanoplatform for melanoma by the combination of CDT and improved immunotherapy with CRISPR/Cas9-PD-L1 system and hypoxia-ADO axis inhibition. STATEMENT OF SIGNIFICANCE: Malignant melanoma is a highly aggressive and treatment-refractory skin cancer, where conventional therapies exhibit limited efficacy and immune checkpoint blockade (ICB) is often compromised by the immunosuppressive tumor microenvironment (TME). To address these challenges, we developed a biomimetic nanoplatform (PPMC@CM) to codeliver MnO₂ nanoparticles and the CRISPR/Cas9-PD-L1 gene-editing system for MRI-guided chemodynamic therapy and enhanced immunotherapy. The PPMC@CM nanoplatform could efficiently accumulate at tumor sites by homologous targeting and relieve hypoxia to suppress the hypoxia-CD39/CD73-adenosine immunosuppressive axis. Additionally, the CRISPR/Cas9-mediated PD-L1 knockout significantly suppresses PD-L1 expression, thereby boosting ICB efficacy. Moreover, PPMC@CM could deplete glutathione in the TME to amplify Mn2+-mediated Fenton-like reactions for enhanced chemodynamic therapy. This research represents a promising theranostic nanoplatform for melanoma by combining chemodynamic therapy and immunotherapy.
Collapse
Affiliation(s)
- Mengyu Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China; College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, China
| | - Cheng Ni
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Aiyu Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Junjie Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China; College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, China
| | - Honghua Guo
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201620, China
| | - Fanyong Xu
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201620, China
| | - Kangan Li
- Mini-invasive Interventional Therapy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xueyan Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Rui Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| |
Collapse
|
4
|
Han R, Balsiger T, Dobrzyński M, Dürr L, Hell T, Smieško M, Solis PN, Hamburger M, Pertz O, Teufel R, Garo E. Coumarins and betulinic acid analogues from Mammea americana and their inhibitory activities on oncogenic MAPK/ERK and PI3K/AKT pathways in human melanoma cells. PHYTOCHEMISTRY 2025; 238:114580. [PMID: 40516889 DOI: 10.1016/j.phytochem.2025.114580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 06/11/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
Melanoma, a highly aggressive form of skin cancer, is primarily driven by two key oncogenic signaling pathways: MAPK/ERK and PI3K/AKT. In a discovery program aiming to identify natural products that inhibit one or both pathways, an in-house library of 2'576 plant extracts was screened using a high-content screening assay with melanoma cells expressing ERK/AKT activity biosensors to quantify inhibition at the single-cell level. The ethyl acetate extract from the leaves of Mammea americana was found to inhibit both pathways in the patient-derived cell line MM121224 and was selected for HPLC-based activity profiling. Scale-up isolation of the compounds eluting in the active window of the chromatogram afforded ten previously undescribed betulinic acid analogues (1-10), along with nine known coumarins (11-19). The isolated compounds were individually evaluated for their inhibitory activity on ERK and AKT in MM121224 cells. Interestingly, none of the betulinic acid derivatives 1-10 showed activity in the assay. In contrast, the isolated coumarins were shown to inhibit both pathways, with the most potent theraphin B (11) exhibiting an IC50 value of 37.0 ± 0.4 μM against the AKT pathway, while also demonstrating weaker activity against the ERK pathway.
Collapse
Affiliation(s)
- Ruzhe Han
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Tamara Balsiger
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Maciej Dobrzyński
- Institute of Cell Biology, University of Bern, Bern, 3012, Switzerland
| | - Lara Dürr
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Tanja Hell
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Martin Smieško
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Pablo N Solis
- Centro de Investigaciones Farmacognosticas de la Flora Panameña - Dr. Mahabir P. Gupta, Faculty of Pharmacy, University of Panama, Panama City, 3366-4, Republic of Panama
| | - Matthias Hamburger
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Olivier Pertz
- Institute of Cell Biology, University of Bern, Bern, 3012, Switzerland
| | - Robin Teufel
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Eliane Garo
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland.
| |
Collapse
|
5
|
Zhang M, Qiu H, Han Z, Ma Y, Hou J, Yuan J, Jia H, Zhou M, Lu H, Wu Y. Topical transdermal administration of lenalidomide nanosuspensions-based hydrogels against melanoma: In vitro and in vivo studies. Int J Pharm X 2025; 9:100316. [PMID: 39898009 PMCID: PMC11787432 DOI: 10.1016/j.ijpx.2025.100316] [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: 08/27/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
Percutaneous neoadjuvant therapy has proven effective in diminishing tumor size and the surgical intervention area, which couldeffectively mitigate the risk of tumor recurrence and enhance immunotherapy efficacy. Lenalidomide, an approved medication orally used to treat myeloma, was loaded into nanosuspensions-based hydrogels (Len-NBHs) for transdermal administration as a percutaneous neoadjuvant therapy. This study was designed to investigate the inhibitory effect and mechanism of Len-NBHs on melanoma. Network pharmacology and transcriptomic analyses identified key targets and signaling pathways. The effects of lenalidomide on melanoma were further verified through Western blotting, immunohistochemistry, immunofluorescence, and quantitative real-time polymerase chain reaction,using both in vitro cell experiments and in vivo melanoma mouse models. Lenalidomide could induce melanoma cells apoptosis, disrupt cell cycle progression, impede cell migration and invasion, and modify tumor microenvironment (TME). Mechanistically, lenalidomide reversed the abnormal activation of the PI3K-AKT signaling pathway and the overexpression of CD93, while also recruiting CD8+ T cells, CD4+ T cells, and dendritic cells to infiltrate the tumor site. Transdermal administration of Len-NBHs represents a promising adjuvant therapy for the treatment of malignant melanoma. Preoperative administration of Len-NBHs can inhibit the outward spread of melanoma, reduce tumor size, thereby decreasing the surgical excision area and improving patient survival rates and prognosis.
Collapse
Affiliation(s)
| | | | - Zheyi Han
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Yazhong Ma
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Jingjing Hou
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Jingwei Yuan
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Haiyan Jia
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Menglu Zhou
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Hongjie Lu
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Yan Wu
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| |
Collapse
|
6
|
Meng Y, Zhou Q, Dian Y, Zeng F, Deng G, Chen X. Ferroptosis: A Targetable Vulnerability for Melanoma Treatment. J Invest Dermatol 2025; 145:1323-1344. [PMID: 39797894 DOI: 10.1016/j.jid.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] [Received: 08/08/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 01/13/2025]
Abstract
Melanoma is a devastating form of skin cancer characterized by a high mutational burden, limited treatment success, and dismal prognosis. Although immunotherapy and targeted therapies have significantly revolutionized melanoma treatment, the majority of patients fail to achieve durable responses, highlighting the urgent need for novel therapeutic strategies. Ferroptosis, an iron-dependent form of regulated cell death driven by the overwhelming accumulation of lipid peroxides, has emerged as a promising therapeutic approach in preclinical melanoma models. A deeper understanding of the ferroptosis landscape in melanoma based on its biology characteristics, including phenotypic plasticity, metabolic state, genomic alterations, and epigenetic changes, as well as the complex role and mechanisms of ferroptosis in immune cells could provide a foundation for developing effective treatments. In this review, we outline the molecular mechanisms of ferroptosis, decipher the role of melanoma biology in ferroptosis regulation, reveal the therapeutic potential of ferroptosis in melanoma, and discuss the pressing questions that should guide future investigations into ferroptosis in melanoma.
Collapse
Affiliation(s)
- Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| |
Collapse
|
7
|
Zolghadri S, Bakhshi Kashi M, Vahidfar N, Farzanefar S, Karimian A, Mohammadi Ashnani MH, Yousefnia H. First-in-human dosimetry and safety evaluation of 68Ga-αMSH derivative for PET imaging of melanoma. Sci Rep 2025; 15:17748. [PMID: 40404705 PMCID: PMC12098654 DOI: 10.1038/s41598-025-02334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 05/13/2025] [Indexed: 05/24/2025] Open
Abstract
Melanoma remains one of the leading causes of cancer-related mortality worldwide, necessitating advanced imaging techniques for early and accurate detection. This study assesses the dosimetry, safety, and imaging performance of a novel 68Ga-labeled α-melanocyte-stimulating hormone ([68Ga]Ga-αMSH) derivative for targeting melanocortin 1 receptors (MC1Rs) in metastatic melanoma. In this first-in-human, prospective, open-label clinical trial, 11 patients with histologically confirmed metastatic melanoma underwent whole-body PET/CT imaging following intravenous administration of the radiolabeled compound (150 ± 10 MBq). Tumor uptake, biodistribution, pharmacokinetics, and radiation dosimetry were evaluated at 60 and 120 min post-injection. Organ and tumor uptake values were measured as standardized uptake values. Radiation dose estimates were calculated using the MIRD methodology and S-values obtained from OLINDA/EXM software. Safety evaluations included monitoring adverse events, biochemical parameters, and vital signs. The radiopharmaceutical demonstrated rapid and selective uptake in metastatic melanoma lesions, achieving high tumor-to-background contrast within 60 min. Quantitative analysis showed substantial tumor uptake, with sustained activity at 120 min. High tumor-to-blood and tumor-to-muscle ratios ensured excellent lesion detectability. The kidneys exhibited the highest absorbed dose (0.0948 ± 0.0425 mSv/MBq), attributed to renal excretion, whereas the brain received the lowest dose (0.0012 ± 0.0007 mSv/MBq). Comparisons with [18F]FDG and other tracers demonstrated superior dosimetry profiles, minimizing radiation exposure and enabling repeat imaging. Also, safety monitoring revealed no serious adverse events. [68Ga]Ga-αMSH analogue exhibits excellent imaging properties, favorable pharmacokinetics, and a strong safety profile, supporting its clinical utility for PET imaging of metastatic melanoma. Its high tumor specificity and minimal off-target accumulation address limitations associated with [18F]FDG.
Collapse
Affiliation(s)
- Samaneh Zolghadri
- Radiation Application Research School, Nuclear Science and Technology Research Institute (NSTRI), Tehran, 14155-1339, Iran
| | - Mohsen Bakhshi Kashi
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasim Vahidfar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Farzanefar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Arezou Karimian
- Radiation Application Research School, Nuclear Science and Technology Research Institute (NSTRI), Tehran, 14155-1339, Iran
| | - Mohammad Hossein Mohammadi Ashnani
- Department of Environmental Science and Engineering, Faculty of Agriculture and Environment, Arak University, Arak, Iran
- Environmental Science Research Institute, Arak University, Arak, Iran
| | - Hassan Yousefnia
- Radiation Application Research School, Nuclear Science and Technology Research Institute (NSTRI), Tehran, 14155-1339, Iran.
| |
Collapse
|
8
|
Li X, Ma L, Guo J, Wei Y, Ma S, Mai Y, Gou G, Zuo W, Yang J. Synergistic anti-tumor effects of mRNA vaccine and PERK inhibitor combination in melanoma treatment. Colloids Surf B Biointerfaces 2025; 254:114808. [PMID: 40383023 DOI: 10.1016/j.colsurfb.2025.114808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/29/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
Melanoma is a highly aggressive form of skin cancer. mRNA vaccines deliver genetic material encoding specific antigens into cells, thereby triggering the host immune system to produce the antigen. Gp-100, an antigenic protein expressed on the surface of melanoma cells, serves as a target mRNA to stimulate the cytotoxic T lymphocyte (CTL) response. However, the absence of natural killer (NK) cells can lead to significant tumor cell proliferation. Gardiquimod, a TLR7 agonist, enhances NK cell cytotoxicity, promoting tumor clearance. In advanced melanoma, the unfolded protein response (UPR) often becomes dysregulated. By inhibiting protein kinase R-like ER kinase (PERK), the UPR can be disrupted, inducing apoptosis in cancer cells and shifting the tumor microenvironment (TME) towards an increased M1/M2 macrophage ratio. This study developed a cationic liposome-based mRNA vaccine (GD-LPR) using DOTMA to co-deliver gp-100 mRNA and the TLR7 agonist Gardiquimod, combined with the PERK inhibitor GSK2656157 (GSK), for synergistic melanoma immunotherapy. GD-LPR achieved 95 % mRNA encapsulation efficiency and demonstrated enhanced dendritic cell maturation and NK cell activation both in vitro and in vivo. In subcutaneous melanoma models, GD-LPR+GSK reduced tumor volume and prolonged survival by modulating the tumor microenvironment (TME): increasing CD8+ T cells (Fig. 3 f), repolarizing M2 to M1 macrophages (Fig. 4 f), and suppressing IL-10 while elevating pro-inflammatory cytokines (IL-2, IFN-γ, TNF-α). Mechanistically, GSK inhibited PERK/ATF-4 signaling, synergizing with GD-LPR to suppress lung metastasis. The combination of the GD-LPR vaccine and GSK provides new potential strategies for treating melanoma, particularly in subcutaneous tumors and lung metastases.
Collapse
Affiliation(s)
- Xiaolong Li
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Lanlan Ma
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jueshuo Guo
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yaya Wei
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shijie Ma
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yaping Mai
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Guojing Gou
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Wenbao Zuo
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Jianhong Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
9
|
Prasai A, Baral N, Elajami MK, Vallabhaneni E, Mitchell JD, Iqbal U, Bahmad HF, Welty FK, Elajami TK. Trends of all-cause, melanoma-specific, and cardiovascular mortality in melanoma patients from 2005 to 2020. Cancer Epidemiol 2025; 97:102848. [PMID: 40381435 DOI: 10.1016/j.canep.2025.102848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Recent advances in melanoma treatment, including immunotherapy and targeted therapy, have significantly improved survival among melanoma patients after 2010. However, these changes may have influenced mortality trends, including those related to cardiovascular (CV) events. OBJECTIVE In this study, we assess mortality trends including CV mortality in melanoma patients. METHODS Using data from the Surveillance, Epidemiology, and End Results (SEER) 17 registry, we examined melanoma incidence and CV, melanoma-specific, and all-cause mortalities from 2005 to 2020. We utilized Joinpoint software to estimate the annual percentage change (APC). RESULTS Our study included 299,993 melanoma patients (173,889 males; 126,104 females). The incidence of melanoma increased by 0.6 % annually (95 % CI: 0.3-1.5, p < 0.05) from 2005 to 2019, followed by a 6.3 % decrease (95 % CI: -9.9 to -0.7, p < 0.05) in 2020. All-cause mortality increased annually by 8.3 % (95 % CI: 6.7-11.3, p < 0.05) from 2005 to 2010, then by 3.3 % (95 % CI: 2.8-3.7, p < 0.05) after 2010. Melanoma-specific mortality increased by 2.0 % annually (95 % CI: 0.8-3.5, p < 0.05) from 2005 until 2013, after which it declined by 5.1 % (95 % CI: -7.3 to -3.8, p < 0.05). CV mortality increased by 7.1 % annually (95 % CI: 4.6-9.4, p < 0.05) from 2005 to 2020. Trends were similar across sexes, with a non-significant higher APC in CV mortality noted among females from 2017 to 2020. CONCLUSIONS Our study shows despite the significant decrease in all-cause and melanoma-specific mortalities after 2010, likely reflecting the benefits of modern therapies, CV mortality continued to rise. These findings underscore the need for long-term surveillance and CV risk management in melanoma patients.
Collapse
Affiliation(s)
- Astha Prasai
- Department of Internal Medicine, Rochester Regional Health, Rochester, NY, USA
| | - Nischit Baral
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, USA
| | - Mohamad K Elajami
- Department of Internal Medicine, Hartford Hospital, Hartford, CT, USA
| | - Esha Vallabhaneni
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, USA
| | - Joshua D Mitchell
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Uzma Iqbal
- Department of Internal Medicine, Rochester Regional Health, Rochester, NY, USA; Department of Cardiology, Sands, Constellation Heart Institute, Rochester Regional Health, Rochester, NY, USA
| | - Hisham F Bahmad
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Francine K Welty
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tarec K Elajami
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, USA.
| |
Collapse
|
10
|
Nazari M, Alikhani M, Nekooei S, Abnous K, Taghdisi SM, Saljooghi AS, Ramezani M, Alibolandi M. Synthesis of theranostic covalent organic framework for Tumor-targeted Chemo-photodynamic therapy. Int J Pharm 2025; 676:125621. [PMID: 40254192 DOI: 10.1016/j.ijpharm.2025.125621] [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: 02/12/2025] [Revised: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Covalent organic frameworks (COFs) are a novel class of organic porous materials that, in recent years, have gained much attention for their applications as nanocarriers toward nanomedicine development. Inspired by this, we introduce for the first time a novel theranostic nanoplatform that combines iodine ligand 5-amino-2,4,6-triiodoisophthalic acid (ATIPA)-decorated porphyrin-based covalent organic frameworks (pCOF-I) designed for effective photodynamic therapy (PDT), doxorubicin (DOX) encapsulation, and computed tomography (CT) imaging toward melanoma treatment. In the design of this COF, we have successfully integrated the iodine ligand with porphyrin. The synthesized mesoporous nanoplatform was loaded with DOX and further modified by COOH-PEG-NH2, which was conjugated with the AS1411 aptamer to be targeted to B16F0 melanoma cells. Comprehensive characterizations verified the successful synthesis and controlled release of DOX from the synthesized COF. In vitro evaluation against B16F0 showed combined chemo-PDT therapy. In addition, higher cellular uptake and toxicity were observed for the targeted platform compared to the non-targeted one towards B16F0. The porphyrin molecules imparted to the pCOF-I nanoparticles (NPs) a significant capacity for light-induced reactive oxygen species (ROS) generation, demonstrating remarkable PDT efficacy in both in vivo and in vitro environments. An in vivo investigation on B16F0 ectopic tumor model of melanoma in mice confirmed the potential for showed combined chemo-PDT therapy chemo-PDT in preclinical stage while approving guided delivery and tumor accumulation of AS1411 aptamer-tagged systems. On the other hand, the prepared platform demonstrated desirable CT-scan imaging of B16F0 tumorized mice 6 and 24 h post-injection. Notably, this is the first report of an AS1411 aptamer-targeted pCOF-I system for CT imaging-guided combined chemo-PDT, marking a significant step forward in multimodal cancer treatment strategies.
Collapse
Affiliation(s)
- Mahsa Nazari
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mina Alikhani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sh Saljooghi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Yan T, Cheng J, Liu H, Wang Y, Zhang C, Huang D, Liu J, Wang Z. Multifunctional Janus Hydrogels: Surface Design Strategies for Next-Generation Clinical Solutions. Gels 2025; 11:343. [PMID: 40422363 DOI: 10.3390/gels11050343] [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: 04/01/2025] [Revised: 05/03/2025] [Accepted: 05/04/2025] [Indexed: 05/28/2025] Open
Abstract
Janus hydrogels, distinguished by their dual-sided structure with distinct physical and chemical properties, have garnered significant attention in the medical field, particularly for applications in drug delivery, tissue engineering, and wound healing. Their ability to simultaneously perform multiple functions, such as targeted drug release and biomimetic tissue interaction, positions them as a promising platform for advanced therapeutic strategies. The growing interest in these hydrogels is primarily driven by their multifunctionality and capacity to address complex biological needs. This review delves into the design, fabrication methods, and applications of Janus hydrogels in medicine, focusing on their potential to overcome the limitations of conventional therapies and providing a comprehensive overview of their role in contemporary biomedical applications.
Collapse
Affiliation(s)
- Taoxu Yan
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Junyao Cheng
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Haoming Liu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Yifan Wang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Chuyue Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Da Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Jianheng Liu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Zheng Wang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
12
|
Bartha Á, Weltz B, Betancourt LH, Gil J, Pinto de Almeida N, Bianchini G, Szeitz B, Szadai L, Pla I, Kemény LV, Jánosi ÁJ, Hong R, Rajeh A, Nogueira F, Doma V, Woldmar N, Guedes J, Újfaludi Z, Kim Y, Szarvas T, Pahi Z, Pankotai T, Szasz AM, Sanchez A, Baldetorp B, Tímár J, Németh IB, Kárpáti S, Appelqvist R, Domont GB, Pawlowski K, Wieslander E, Malm J, Fenyo D, Horvatovich P, Marko-Varga G, Győrffy B. Melanoma Proteomics Unveiled: Harmonizing Diverse Data Sets for Biomarker Discovery and Clinical Insights via MEL-PLOT. J Proteome Res 2025. [PMID: 40322912 DOI: 10.1021/acs.jproteome.4c00749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Using several melanoma proteomics data sets we created a single analysis platform that enables the discovery, knowledge build, and validation of diagnostic, predictive, and prognostic biomarkers at the protein level. Quantitative mass-spectrometry-based proteomic data was obtained from five independent cohorts, including 489 tissue samples from 394 patients with accompanying clinical metadata. We established an interactive R-based web platform that enables the comparison of protein levels across diverse cohorts, and supports correlation analysis between proteins and clinical metadata including survival outcomes. By comparing differential protein levels between metastatic, primary tumor, and nonmalignant samples in two of the cohorts, we identified 274 proteins showing significant differences among the sample types. Further analysis of these 274 proteins in lymph node metastatic samples from a third cohort revealed that 45 proteins exhibited a significant effect on patient survival. The three most significant proteins were HP (HR = 4.67, p = 2.8e-06), LGALS7 (HR = 3.83, p = 2.9e-05), and UBQLN1 (HR = 3.2, p = 4.8e-05). The user-friendly interactive web platform, accessible at https://www.tnmplot.com/melanoma, provides an interactive interface for the analysis of proteomic and clinical data. The MEL-PLOT platform, through its interactive capabilities, streamlines the creation of a comprehensive knowledge base, empowering hypothesis formulation and diligent monitoring of the most recent advancements in the domains of biomedical research and drug development.
Collapse
Affiliation(s)
- Áron Bartha
- Department of Bioinformatics, Semmelweis University, Budapest 1085, Hungary
- Department of Pediatrics, Semmelweis University, Budapest 1085, Hungary
| | - Boglárka Weltz
- Department of Bioinformatics, Semmelweis University, Budapest 1085, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, H-1117, Budapest, Hungary
| | - Lazaro Hiram Betancourt
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
| | - Jeovanis Gil
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
| | - Natália Pinto de Almeida
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
| | | | - Beáta Szeitz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, 1085, Hungary
| | - Leticia Szadai
- Department of Dermatology and Allergology, University of Szeged, Szeged, 6720, Hungary
| | - Indira Pla
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Department of Biomedical Engineering, Faculty of Engineering, LTH, Lund University, Lund, 22363, Sweden
| | - Lajos V Kemény
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, Budapest, 1085, Hungary
- Department of Physiology, Semmelweis University, Budapest, 1094, Hungary
- Department of Dermatology, Venerology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, 1085, Hungary
| | - Ágnes Judit Jánosi
- Department of Dermatology and Allergology, University of Szeged, Szeged, 6720, Hungary
| | - Runyu Hong
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, New York 10016, United States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | - Ahmad Rajeh
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Fábio Nogueira
- Proteomics Unit, Institute of Chemistry and Research Center for Precision Medicine, Institute of Biophysics Carlos Chagas Filho, Federal Univesity of Rio de Janeiro, Rio de Janeiro, 21941-170, Brazil
| | - Viktória Doma
- Department of Dermatology, Venerology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, 1085, Hungary
| | - Nicole Woldmar
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Chemistry Institute Federal, University of Rio de Janeiro, Rio de Janiero, 21941-909, Brazil
| | - Jéssica Guedes
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
- Chemistry Institute Federal, University of Rio de Janeiro, Rio de Janiero, 21941-909, Brazil
| | - Zsuzsanna Újfaludi
- University of Szeged, Albert Szent-Györgyi Clinical Centre, Department of Pathology, 6720, Szeged, Hungary
| | - Yonghyo Kim
- Drug Discovery Platform Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Tibor Szarvas
- Department of Urology, Semmelweis University, Budapest, 1082, Hungary
- Department of Urology, University of Duisburg-Essen, Essen, 45147, Germany
| | - Zoltan Pahi
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged H-6725, Hungary
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Genome Integrity and DNA Repair Core Group, University of Szeged, Budapesti út 9, Szeged H-6728, Hungary
| | - Tibor Pankotai
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged H-6725, Hungary
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Genome Integrity and DNA Repair Core Group, University of Szeged, Budapesti út 9, Szeged H-6728, Hungary
- Competence Centre of the Life Sciences Cluster of the Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Dugonics tér 13, Szeged H-6720, Hungary
| | - A Marcell Szasz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1085 Budapest, Hungary
| | - Aniel Sanchez
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, Malmö,205 02, Sweden
| | - Bo Baldetorp
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund,221 84, Sweden
| | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Faculty of Medicine, Semmelweis University, Budapest,1085, Hungary
| | - István Balázs Németh
- Department of Dermatology and Allergology, University of Szeged, Szeged, 6720, Hungary
| | - Sarolta Kárpáti
- Department of Dermatology, Venerology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, 1085, Hungary
| | - Roger Appelqvist
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
| | - Gilberto Barbosa Domont
- Proteomics Unit, Institute of Chemistry and Research Center for Precision Medicine, Institute of Biophysics Carlos Chagas Filho, Federal Univesity of Rio de Janeiro, Rio de Janeiro, 21941-170, Brazil
| | - Krzysztof Pawlowski
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, Malmö,205 02, Sweden
- Department of Biochemistry and Microbiology, Warsaw University of Life Sciences, Warszawa,02-787, Poland
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9148, United States
| | - Elisabet Wieslander
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, Malmö,205 02, Sweden
| | - Johan Malm
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Lund 21428, Sweden
| | - David Fenyo
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, New York 10016, United States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | - Peter Horvatovich
- University of Groningen, Groningen Research Institute of Pharmacy, Analytical Biochemistry, Groningen, 9711, The Netherlands
| | - György Marko-Varga
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Budapest 1085, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, H-1117, Budapest, Hungary
- Dept. of Biophysics, Medical School, University of Pecs, H-7624, Pecs, Hungary
| |
Collapse
|
13
|
Reinhard S, Utikal JS, Zaremba A, Lodde G, von Wasielewski I, Klespe KC, Meier F, Haferkamp S, Kähler KC, Herbst R, Gebhardt C, Sindrilaru A, Dippel E, Angela Y, Mohr P, Pfoehler C, Forschner A, Kaatz M, Schell B, Gesierich A, Loquai C, Hassel JC, Ulrich J, Meiss F, Schley G, Heinzerling LM, Sachse M, Welzel J, Weishaupt C, Sunderkötter C, Michl C, Lindhof HH, Kreuter A, Heppt MV, Wenk S, Mauch C, Berking C, Nedwed AS, Gutzmer R, Leiter U, Schadendorf D, Ugurel S, Weichenthal M, Haist M, Fleischer MI, Lang B, Grabbe S, Stege H. First-line checkpoint inhibitor therapy in metastatic acral lentiginous melanoma compared to other types of cutaneous melanoma: A multicenter study from the prospective skin cancer registry ADOREG. Eur J Cancer 2025; 220:115356. [PMID: 40121837 DOI: 10.1016/j.ejca.2025.115356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Melanoma is the main cause of skin cancer-related death. Treatment with immune checkpoint inhibitors (CPI) has improved the prognosis in recent years. However, subtypes of melanoma differ in their response. Acral lentiginous melanoma (ALM) has a worse prognosis compared to cutaneous melanoma other than ALM (CM) and is therefore of particular relevance. AIMS To evaluate the efficacy of CPI in first-line treatment of patients with advanced ALM compared CM. METHODS Retrospective analysis of patients with metastatic ALM (n = 45) or CM (n = 328) who received first-line CPI therapy from the multicenter prospective skin cancer registry ADOREG. Study endpoints were best overall response (BOR), progression-free survival (PFS) and overall survival (OS). RESULTS ALM patients had significantly higher rates of ulcerated tumors, loco regional metastases and fewer BRAF-mutated tumors compared to CM patients. Combined CPI was administered in 48.9 % ALM patients and 39.3 % of CM patients, while the remaining patients received PD-1 monotherapy. OS trended to be shorter in patients with ALM (18.1 vs. 43.8 months, p = 0.10) with no significant differences in PFS (7.0 vs. 11.5 months, p = 0.21). In patients with CM, median OS with combined CPI was not reached, whereas the median OS after PD-1 monotherapy was 37.8 months (p = 0.22). Conversely, in patients with ALM, OS with combined CPI was 17.8 months, compared to 26 months with PD-1 monotherapy (p = 0.15). There were no significant differences in BOR between patients with ALM or CM. CONCLUSION Analysis of this real-world cohort of patients with metastatic melanoma showed a trend towards poorer survival outcomes upon first-line treatment with CPI in ALM compared to cutaneous melanoma of other subtypes.
Collapse
Affiliation(s)
- Sören Reinhard
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | - Jochen Sven Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Anne Zaremba
- Department of Dermatology, Venereology and Allergology, University Hospital Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Georg Lodde
- Department of Dermatology, Venereology and Allergology, University Hospital Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Imke von Wasielewski
- Skin Cancer Center Hannover, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Kai Christian Klespe
- Skin Cancer Center Hannover, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Katharina C Kähler
- Department of Dermatology, Skin Cancer Center, University Hospital Schleswig-Holstein - Campus Kiel, Kiel, Germany
| | - Rudolf Herbst
- Department of Dermatology, HELIOS Hospital Erfurt, Erfurt, Germany
| | - Christoffer Gebhardt
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anca Sindrilaru
- Department of Dermatology, University Hospital of Ulm, Ulm, Germany
| | - Edgar Dippel
- Department of Dermatology, Ludwigshafen City Hospital, Ludwigshafen, Germany
| | - Yenny Angela
- Department of Dermatology, Muehlenkreiskliniken Minden and Ruhr University Bochum, Minden, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe Kliniken Buxtehude, Buxtehude, Germany
| | - Claudia Pfoehler
- Department of Dermatology, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Andrea Forschner
- Center for Dermatooncology, Department of Dermatology, Eberhard-Karls University of Tübingen, Tubingen, Germany
| | - Martin Kaatz
- Department of Dermatology, DRK Hospital Chemnitz-Rabenstein, Rabenstein, Germany
| | - Beatrice Schell
- Department of Dermatology, Wald-Klinikum Gera, Gera 07546, Germany
| | - Anja Gesierich
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Carmen Loquai
- Department of Dermatology, Klinikum Bremen-Ost, Gesundheit Nord gGmbH, Bremen, Germany
| | - Jessica C Hassel
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Jens Ulrich
- Department of Dermatology and Allergy, Harzklinikum Dorothea Christiane Erxleben GmbH, Quedlinburg, Germany
| | - Frank Meiss
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gaston Schley
- Department of Dermatology, HELIOS Hospital Schwerin, Schwerin, Germany
| | - Lucie M Heinzerling
- Department of Dermatology and Allergy, University Hospital, Ludwig-Maximilian Universität Munich, München, Germany
| | - Michael Sachse
- Department of Dermatology, Hospital Bremerhaven Reinkenheide, Bremerhaven, Germany
| | - Julia Welzel
- Department of Dermatology, University Hospital Augsburg, Augsburg, Germany
| | - Carsten Weishaupt
- Department of Dermatology, University Hospital of Muenster, Muenster, Germany
| | - Cord Sunderkötter
- Department of Dermatology and Venereology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Christiane Michl
- Department of Dermatology and Venereology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | | | - Alexander Kreuter
- Department of Dermatology, Venerology and Allergology, Helios St. Elisabeth Klinik Oberhausen, University Witten-Herdecke, Oberhausen, Germany
| | - Markus V Heppt
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN) and CCC Alliance WERA, Bavarian Cancer Research Center (BZKF), Erlangen 91052, Germany
| | - Saskia Wenk
- Department of Dermatology, Medical Center Klinikum Darmstadt, Teaching Hospital Goethe-University Frankfurt, Darmstadt, Germany
| | - Cornelia Mauch
- Department of Dermatology, Ruhr-Universität Bochum, Bochum, Germany
| | - Carola Berking
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN) and CCC Alliance WERA, Bavarian Cancer Research Center (BZKF), Erlangen 91052, Germany
| | - Annekathrin Silvia Nedwed
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center Mainz, Mainz, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Muehlenkreiskliniken Minden and Ruhr University Bochum, Minden, Germany
| | - Ulrike Leiter
- Center for Dermatooncology, Department of Dermatology, Eberhard-Karls University of Tübingen, Tubingen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Michael Weichenthal
- Department of Dermatology, Skin Cancer Center, University Hospital Schleswig-Holstein - Campus Kiel, Kiel, Germany
| | - Maximilian Haist
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | | | - Berenice Lang
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | - Henner Stege
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany.
| |
Collapse
|
14
|
Jansen P, Galetzka W, Thielmann CM, Murali R, Zaremba A, Standl F, Lodde G, Möller I, Sucker A, Paschen A, Hadaschik E, Ugurel S, Zimmer L, Livingstone E, Schadendorf D, Stang A, Griewank KG. pTERT mutational status is associated with survival in stage IV melanoma patients receiving first-line immune therapy. Eur J Cancer 2025; 220:115337. [PMID: 40056560 DOI: 10.1016/j.ejca.2025.115337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND TERT promoter mutations are the most prevalent mutations in melanoma. In this study, we investigated clinical characteristics and survival after first line therapies in a cohort of melanoma patients with known TERT promoter (pTERT) mutation status. METHODS Sequencing data from 2013 to 2021 covering 29 genes and the pTERT status was assessed and 774 melanomas patients with known pTERT status and clinical data were analyzed. Progression free survival (PFS) and overall survival (OS) of 374 melanoma patients in AJCC-stage IV who received first-line immune checkpoint inhibitors (ICI, anti-CTLA4 /anti-PD1 combination therapy or anti-PD1 monotherapy) or targeted therapy (TT) were assessed applying Cox uni-/ multivariable analyses and Kaplan-Meier curves. RESULTS The cohort included 573 cutaneous, 69 mucosal, 37 acral and 95 MUP (melanomas of unknown primary) melanoma patients with a median observational time from first diagnosis to patient death or censoring of 38.5 months. TERT promoter mutations were identified in 476 melanomas (61.5 %). Survival analysis of 374 patients with stage IV disease undergoing first-line systemic therapy (ICI or TT) suggested prolonged PFS and OS for patients with pTERT mutation positive tumors (pTERT(+)). Particularly, pTERT(+) patients receiving anti-CTLA4/anti-PD1 therapy showed mPFS of 14.8 months (95 % CI: 7.1-40.3) and mOS of 105.2 months (95 % CI: 27.6-not reached) compared to pTERT(-) patients with mPFS of 5.5 months (95 % CI: 2.7-10.0) and mOS of 14.7 months (95 % CI: 11.7-24.1). CONCLUSIONS Our findings suggest that presence of a pTERT mutation in melanomas might favor PFS and OS after first line ICI with the greatest improvement after receiving anti-CTLA4 / anti-PD1. If validated in larger prospective studies, pTERT mutation status may be a valuable prognostic marker for stage IV melanoma patients.
Collapse
Affiliation(s)
- Philipp Jansen
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Wolfgang Galetzka
- Institute for medical informatics, biometry and epidemiology, University Hospital Essen, Essen, Germany
| | - Carl M Thielmann
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Rajmohan Murali
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Anne Zaremba
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Fabian Standl
- Graduate Center of Medicine and Health, Technical University Munich, Munich, Germany
| | - Georg Lodde
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Inga Möller
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Eva Hadaschik
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Andreas Stang
- Institute for medical informatics, biometry and epidemiology, University Hospital Essen, Essen, Germany
| | - Klaus G Griewank
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany.
| |
Collapse
|
15
|
Liu Y, Wang Q, Li Q, Ren P. Role of ELP6 in tumour progression and impact on ERK1/2 signalling pathway inhibitors in skin cutaneous melanoma. Oncol Lett 2025; 29:250. [PMID: 40177137 PMCID: PMC11962575 DOI: 10.3892/ol.2025.14996] [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: 07/31/2024] [Accepted: 03/05/2025] [Indexed: 04/05/2025] Open
Abstract
Elongator acetyltransferase complex subunit 6 (ELP6), a subunit of the elongator complex, can increase the migratory potential of melanoma cells in vitro. However, the clinical relevance of ELP6 in patients with melanoma remains unclear. The present study aimed to investigate the role of ELP6 expression in melanoma progression and association with patient survival rates. Transcriptomic data from patients with melanoma available in The Cancer Genome Atlas, Gene Expression Profiling Interactive Analysis and cBioPortal databases were analysed to evaluate the associations between ELP6 expression levels and patient survival. In vitro experiments were conducted using short hairpin RNAs to downregulate ELP6, with a focus on cell viability, cell cycle regulation and the ERK1/2 signalling pathway. ELP6 expression levels were significantly elevated in patients with melanoma and were associated with poor survival outcomes. Knockdown of ELP6 resulted in decreased expression levels of p42 MAPK, reduced cell viability, G1 phase cell cycle arrest and led to reduced responsiveness to the MEK1/2 inhibitor U0126. ELP6 promotes melanoma progression via the ERK1/2 signalling pathway. Therefore, assessing ELP6 expression may offer potential therapeutic strategies for patients with melanoma.
Collapse
Affiliation(s)
- Ying Liu
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Qinrong Wang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Qian Li
- Department of Pharmacy, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Peng Ren
- Department of Urology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou 556000, P.R. China
| |
Collapse
|
16
|
Thomas JL, Heagerty AHM, Goldberg Oppenheimer P. Emerging Technologies for Timely Point-of-Care Diagnostics of Skin Cancer. GLOBAL CHALLENGES (HOBOKEN, NJ) 2025; 9:2400274. [PMID: 40352638 PMCID: PMC12065104 DOI: 10.1002/gch2.202400274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/07/2025] [Indexed: 05/14/2025]
Abstract
Skin cancer is a global health crisis and a leading cause of morbidity and mortality worldwide. A leading factor of malignancy remains the UV radiation, driving various biomolecular changes. With shifting population behaviors, deficiency in screening programs and reliance on self-presentation, climate change and the ageing world populace, global incidents have been surging alarmingly. There is an urgent need for new technologies to achieve timely intervention through rapid and accurate diagnostics of skin cancer. Raman spectroscopy has been emerging as a highly promising analytical technology for diagnostic applications, poised to outpace the current costly, invasive and slow procedures, frequently hindered by varying sensitivity, specificity and lack of portability. Herein, complex and intricate progress are overviewed and consolidated across medical and engineering disciplines with a focus on the latest advances in the traditional and emerging skin cancer diagnostics. Methods detecting structural and chemical responses are categorized along with emerging chemo-biophysical sensing techniques. Particular attention is drawn to Raman spectroscopy, as a non-invasive, rapid and accurate sensing of molecular fingerprints in dermatological matrix with an additional focus on artificial intelligence, as a decision support tool collectively, laying the platform toward development and rapid translation of point-of-care diagnostic technologies for skin cancer to real-world applications.
Collapse
Affiliation(s)
- Jarrod L. Thomas
- Advanced Nanomaterials Structures and Applications LaboratoriesSchool of Chemical EngineeringCollege of Engineering and Physical SciencesUniversity of BirminghamEdgbastonBirminghamB15 2TTUK
- Healthcare Technologies InstituteInstitute of Translational MedicineMindelsohn WayBirminghamB15 2THUK
| | - Adrian H. M. Heagerty
- Department of DermatologyUniversity Hospitals Birmingham NHS Foundation TrustMindelsohn WayBirminghamB15 2GWUK
- Institute of Inflammation and AgeingCollege of Medical and Dental SciencesMindelsohn WayBirminghamB15 2GWUK
| | - Pola Goldberg Oppenheimer
- Advanced Nanomaterials Structures and Applications LaboratoriesSchool of Chemical EngineeringCollege of Engineering and Physical SciencesUniversity of BirminghamEdgbastonBirminghamB15 2TTUK
- Healthcare Technologies InstituteInstitute of Translational MedicineMindelsohn WayBirminghamB15 2THUK
| |
Collapse
|
17
|
Drews MA, Baumgarten A, Zensen S, Opitz M, Bos D, Zimmer L, Ugurel S, Haubold J, Schadendorf D, Livingstone E, Schaarschmidt BM. Adverse effects of systemic advanced melanoma therapies-do BRAF/MEK inhibitors increase the incidence of mesenteric panniculitis? Eur Radiol 2025:10.1007/s00330-025-11642-w. [PMID: 40310541 DOI: 10.1007/s00330-025-11642-w] [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: 12/17/2024] [Revised: 02/11/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025]
Abstract
OBJECTIVES BRAF/MEK inhibitors (BRAFi/MEKi) and PD-1 and CTLA-4 immune checkpoint inhibitors (ICI) have revolutionized malignant melanoma treatment and improved patients' clinical outcome significantly. However, these therapies are associated with substance class-specific side effects. Here, selected cases indicate a correlation between the incidence of mesenteric panniculitis (MP) and BRAFi/MEKi treatment. As MP can mimic or conceal underlying malignancy, the aim of the present study was to confirm a potential correlation with BRAFi/MEKi or ICI in a retrospective, observational analysis of melanoma patients. MATERIALS AND METHODS In a monocentric retrospective study, abdominal CTs of 490 melanoma patients receiving first-line treatment with ICI (nivolumab, ipilimumab, pembrolizumab, nivolumab/ipilimumab) or BRAFi/MEKi (dabrafenib/trametinib, vemurafenib/cobimetinib, encorafenib/binimetinib) in the adjuvant or advanced situation were evaluated for MP development comparing baseline imaging prior therapy start and follow-up imaging under therapy. MP was defined as an unilocular mesenteric mass characterized by small tissue nodules with increased density of the adjacent fat and a surrounding pseudo-capsule. RESULTS 384 melanoma patients with ICI (161 women, median age at therapy start: 62 years, IQR: 21 years) and 106 patients with BRAFi/MEKi first-line therapy (46 women, median age: 58 years, IQR: 18 years) were evaluated. MP incidence was significantly higher following BRAFi/MEKi treatment compared to ICI (7.5% vs. 2.9%, p = 0.04). No significance was detected comparing time until MP development from therapy start (174 days, IQR: 518 days [BRAFi/MEKi] vs. 207 days, IQR: 298 days [ICI], p > 0.05). CONCLUSION Our study demonstrates a significant increase in MP development following BRAFi/MEKi treatment compared to ICI in patients with melanoma. As this benign condition can mimic or even conceal malignancy, awareness of its appearance is important. KEY POINTS Question BRAF/MEK and immune checkpoint inhibitors have revolutionized melanoma treatment but are associated with various side effects, yet data regarding the development of mesenteric panniculitis are scarce. Findings BRAF/MEK inhibitor treatment is associated with a significantly higher rate of mesenteric panniculitis compared to immune checkpoint inhibitor treatment in advanced melanoma. Clinical relevance BRAF/MEK inhibitor-treated patients are at risk for development of mesenteric panniculitis. As this benign finding can mimic or conceal malignancy, awareness of it is important especially in these patients.
Collapse
Affiliation(s)
- Marcel Alexander Drews
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany.
| | - Alexander Baumgarten
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Sebastian Zensen
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Marcel Opitz
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Denise Bos
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Johannes Haubold
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | | | - Benedikt M Schaarschmidt
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| |
Collapse
|
18
|
Batut L, Sanchez S, Lalun N, Saber Cherif L, Bouland N, Durlach A, Dormoy V. Evaluation of the Smoking Exposure Effects on Carcinogenesis Markers According to the Localization of Melanocytic Lesions. Exp Dermatol 2025; 34:e70111. [PMID: 40312931 DOI: 10.1111/exd.70111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
Melanoma is considered to be the most lethal skin cancer, and smoking is one of the most important public health issues, but its potential carcinogenic role in melanoma is still discussed. Our study aims to determine whether direct tobacco smoke exposure has an impact on melanocytic lesions regarding atypia and proliferation by analysing three markers of interest: DNA ploidy index, MCM6 and the α5-nicotinic acetylcholine receptor (CHRNA5). 90 patients with surgically removed melanocytic lesions were selected. Their smoking exposure data were collected. The expression of all three markers was analysed in lesions directly exposed to tobacco smoke and compared with lesions protected from exogen contact. No difference was found between lesions chronically exposed to smoke and those protected. In the smoker group, CHRNA5 expression (p = 0.25) and MCM6 expression (p = 0.24) were not statistically different depending on the location of lesions. There was also no difference in the DNA ploidy index (p = 0.3). Therefore, tobacco smoke does not seem to have an impact on CHRNA5 expression, proliferation and atypia markers in melanocytic lesions.
Collapse
Affiliation(s)
- Laurine Batut
- Service de Dermatologie, CHU Robert Debré, Reims, France
| | | | - Nathalie Lalun
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
| | - Lynda Saber Cherif
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
| | - Nicole Bouland
- Université de Reims Champagne-Ardenne, Laboratoire D'anatomie Pathologique, Reims, France
| | - Anne Durlach
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
- Service Anatomocytopathologie, CHU Robert Debré, Reims, France
| | - Valérian Dormoy
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
| |
Collapse
|
19
|
Lodde G, Albrecht LJ, Schadendorf D. [Treatment of metastatic melanoma: update 2025]. Dtsch Med Wochenschr 2025; 150:562-569. [PMID: 40262755 DOI: 10.1055/a-2500-0927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Immune checkpoint inhibition and targeted therapy with BRAF/MEK inhibition for BRAF-mutated melanoma have significantly improved progression-free and overall survival in patients with metastatic melanoma. Current research focuses on novel treatment strategies for PD-1 resistance, neoadjuvant approaches, and cellular therapies. 10-year follow-up data of randomized clinical trials show that both combined CTLA-4 and PD-1 immune checkpoint inhibition and PD-1 immune checkpoint inhibition alone can achieve long-term survival in metastatic melanoma. Potential surrogate markers of long-term response include a progression-free survival at 3 years after start of treatment and a reduction in tumour burden of at least 80%. The management of PD-1 resistance remains a challenge. Advances in molecular pathology have led to the identification of new therapeutic targets. Several cellular therapies are currently being evaluated in clinical trials as alternatives for melanoma patients refractory to immune checkpoint inhibition or targeted BRAF/MEK inhibition. In BRAF-mutant melanoma, combined BRAF/MEK inhibition is an alternative to immune checkpoint inhibition. Real-world data and clinical trial results on treatment sequencing suggest that immune checkpoint inhibition may improve survival in the first line setting, particularly in the absence of prior adjuvant systemic therapy. Adjuvant treatment leads to improved progression-free survival in melanoma patients while overall survival data are still pending. Neoadjuvant treatment seems to be a promising alternative to conventional adjuvant therapy for specific subgroups of melanoma patients. Participation in clinical trials offers patients the best opportunity to benefit from the latest treatment options.
Collapse
|
20
|
Marranci A, Maresca L, Lodovichi S, Luserna di Rorà AG, Stecca B, Poliseno L. PARP1 in melanoma: Mechanistic insights and implications for basic and clinical research. Cancer Lett 2025; 617:217599. [PMID: 40024566 DOI: 10.1016/j.canlet.2025.217599] [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: 11/25/2024] [Revised: 02/04/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Targeted therapies and immunotherapies have revolutionized the treatment of metastatic melanoma and have set a successful example for the treatment of other cancers. A similar breakthrough was achieved with the advent of PARP inhibitors (PARPi) in breast and ovarian cancer. Recent evidence highlights the critical role of PARP1 in melanoma initiation and progression. High PARP1 expression correlates with aggressive melanoma characteristics and poor patient outcomes. Preclinical and clinical data suggest that PARPi, alone or in combination, can effectively reduce melanoma cell viability and inhibit tumor growth. However, integrating PARPi with current treatment approaches and identifying patients who could benefit the most from such combinations remain underexplored areas of investigation. This review highlights the need for further basic and clinical research on PARP1 in melanoma, to better understand its role and to tackle major challenges in the field, such as resistance to targeted therapies and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Andrea Marranci
- Oncohematology Unit, Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, 56017, Pisa, Italy. http://www.fpscience.it/
| | - Luisa Maresca
- Tumor Cell Biology Unit, Core Research Laboratory (CRL), Institute for Cancer Research and Prevention (ISPRO), 50139, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, 50139, Florence, Italy
| | - Samuele Lodovichi
- Department of Biosciences, University of Milan, 20133, Milan, Italy; Institute of Clinical Physiology, National Research Council (CNR-IFC), 56124, Pisa, Italy
| | | | - Barbara Stecca
- Tumor Cell Biology Unit, Core Research Laboratory (CRL), Institute for Cancer Research and Prevention (ISPRO), 50139, Florence, Italy
| | - Laura Poliseno
- Institute of Clinical Physiology, National Research Council (CNR-IFC), 56124, Pisa, Italy; Oncogenomics Unit, Core Research Laboratory, Institute for Cancer Research and Prevention (ISPRO), 56124, Pisa, Italy.
| |
Collapse
|
21
|
Xiang M, Li Z, Su J, Dai X, Zhang Y, Tan J, Han X, Xie J, Tang Y, Zhang S, Guo S, Xing HR, Li J, Wang J. KLF12 transcriptional activation by a novel LncRNA A930015D03Rik enhances melanoma metastasis. Int J Biol Macromol 2025; 309:142686. [PMID: 40185449 DOI: 10.1016/j.ijbiomac.2025.142686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/26/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025]
Abstract
Melanoma metastasis remains a poorly understood yet fatal hallmark of cancer progression, with limited therapeutic strategies targeting its underlying mechanisms. While the transcription factor KLF12 shows paradoxical roles across malignancies, its context-dependent functions in melanoma-particularly its regulatory interplay with extracellular vesicle (EV)-driven intercellular communication-have not been systematically explored. To address this gap, we investigated how metastatic melanoma cells exploit KLF12-mediated pathways through EV cargo transfer to propagate aggressive phenotypes. Our research results indicate that highly metastatic cells transfer lncRNA A930015D03Rik through exosomes, acting as a sponge for miR-204-5p, which promotes the expression of KLF12. The transcriptional activation of KLF12 facilitates the activation of critical pro-cancer pathways such as inflammation and NF-κB, while inhibiting the tumor-suppressive mechanisms of P53 and oxidative phosphorylation. This ultimately enhances the migration and invasion capabilities of low-metastatic tumor cells, driving the malignant progression of melanoma. In this study, we identified a novel tumor-derived EVs lncRNA, A930015D03Rik, which can enhance the expression of KLF12 through a ceRNA mechanism and influence the post-translational regulation of KLF12, thereby modulating the plasticity of tumor metastasis. This exosome-genome feedforward circuit explains KLF12's microenvironment-contingent pro-metastatic function, offering significant insights for the development of future therapeutic strategies targeting tumor metastasis.
Collapse
Affiliation(s)
- Meng Xiang
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing, China; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Ziyi Li
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, China
| | - Jie Su
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, China
| | - Xue Dai
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing, China
| | - Yuhan Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Jiyu Tan
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Xue Han
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, China
| | - Jiacheng Xie
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Yao Tang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Sicheng Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Shimeng Guo
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing, China.
| | - H Rosie Xing
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Jingyu Li
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing, China.
| | - Jianyu Wang
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, China.
| |
Collapse
|
22
|
Yang A, Chen L, Tang S, Guo X, Su H, Jiang BP, Shen XC. Light/Ultrasound Dual Responsive Carbon Dots-Based Nanovaccines for Multimodal Activation Tumor Immunotherapy of Melanoma. Adv Healthc Mater 2025; 14:e2405194. [PMID: 40200897 DOI: 10.1002/adhm.202405194] [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: 12/23/2024] [Revised: 03/26/2025] [Indexed: 04/10/2025]
Abstract
Melanoma is a highly aggressive and metastatic tumor, and immunotherapy has become the current solution. However, conventional nanovaccines do not strongly activate T cell immune responses. Therefore, development of effective therapeutic nanovaccines to activate systemic antitumor immunity is urgently required. Herein, light/ultrasound (US) dual-responsive carbon dot-based nanovaccines (Cu-N-CDs@OVA) are designed using copper-nitrogen-coordinated carbon dots composited with ovalbumin. Under 650-nm laser irradiation, Cu-N-CDs@OVA exhibited superior photothermal ablation of primary tumors, induced immunogenic cell death and released antigens by phototherapy, facilitating the maturation of dendritic cells (DCs). More importantly, Cu-N-CDs@OVA stably penetrated and diffused upon US treatment, eradicating metastatic tumors and generating low-dose reactive oxygen species to activate DCs. By integrating with the model antigen OVA, the combined multimodal treatment promotes DC maturation to activate systematic antitumor immunity. This is the first example of a light/US dual-responsive therapeutic nanovaccine that provides a paradigm for the production of personalized nanovaccines against malignant tumors.
Collapse
Affiliation(s)
- Aijia Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Li Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Shunxin Tang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Xiaolu Guo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Hongqin Su
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Bang-Ping Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Xing-Can Shen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| |
Collapse
|
23
|
Alamoudi MK, Alsaleh AA, Thyagarajan A, Alkholifi FK, Raza ML, Sahu RP. Elevated HDAC4 Expression Is Associated with Reduced T-Cell Inflamed Tumor Microenvironment Gene Signatures and Immune Checkpoint Inhibitor Effectiveness in Melanoma. Cancers (Basel) 2025; 17:1518. [PMID: 40361444 PMCID: PMC12070970 DOI: 10.3390/cancers17091518] [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: 03/26/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Melanoma remains a difficult malignancy to treat because it employs tolerance mechanisms like negative immune checkpoint (IC) molecules to avoid antitumor immune responses. Thus, immune checkpoint inhibitors (ICIs) are increasingly used to treat melanoma. However, many patients do not respond, indicating resistance mechanisms like intrinsic tumor characteristics and an immunosuppressive tumor microenvironment (TME). An inflamed TME was associated with improved ICI efficacy by upregulating the T-cell inflamed TME gene signatures, an array of genes associated with dendritic cells (DCs) and cytotoxic CD8+ T-cell-mediated anti-tumor responses. As histone deacetylases (HDACs) have been shown to play crucial roles in regulating gene expression and aberrant HDAC expression has been reported in melanoma and also implicated in the regulation of IC, programmed cell death protein 1 (PD-1), and its ligand (PD-L1) and various immune evasion genes, we investigated the relationship between T-cell inflamed TME gene signatures and the HDAC family, particularly HDAC4. Methods: We used the skin cutaneous melanoma (SKCM) database, ICI-pretreated melanoma dataset, and other platforms including cBioPortal, TIMER 2.0, TISIDB, and UALCAN for the analysis. Results: We identified that high HDAC4 expression negatively modulated the TME by decreasing the abundance of DCs and cytotoxic CD8+ T-cells. The group of melanoma patients with elevated HDAC4 expression exhibited not only poor prognosis but also diminished transcription of T-cell inflamed TME gene signatures and increased DNA methylation of T-cell inflamed TME gene signatures. Importantly, elevated HDAC4 expression was associated with decreased CD8+ T-cells and a decreased ESTIMATE immune score in ICI-pretreated melanoma patients. Conclusions: Our findings suggest that HDAC4 may transform the TME into a non-inflamed phenotype, thereby reducing ICI efficacy in melanoma. Overall, this research shows that a combination of HDAC4 inhibitors and ICIs could result in better melanoma prognosis.
Collapse
Affiliation(s)
- Mariam K. Alamoudi
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Abdulmonem A. Alsaleh
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard-Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia;
| | - Anita Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA;
| | - Faisal K. Alkholifi
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Muhammad Liaquat Raza
- Department of Infection Prevention Control, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia;
| | - Ravi P. Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA;
| |
Collapse
|
24
|
Xie T, Zhang Y, Ling N, Yuan Y, Liu W, Guo J, Wei X, Peng T, Wang Z, Dai J, Chen W, Yan M, Wu X, Wu W, Ye M, Tan W. Aptamer as a Molecular Tethering Agent Induces PrP C Aggregation and Degradation to Inhibit Melanoma Proliferation. Angew Chem Int Ed Engl 2025:e202425051. [PMID: 40304120 DOI: 10.1002/anie.202425051] [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: 12/20/2024] [Revised: 04/05/2025] [Accepted: 04/29/2025] [Indexed: 05/02/2025]
Abstract
Melanoma, a malignant tumor originating from melanocytes, is the most aggressive and deadly form of skin cancer. Previous studies have revealed that the cellular prion protein (PrPC) is frequently overexpressed in melanoma, contributing to tumor progression. This study presents the first proof of concept evidence that nucleic acid aptamers can be used to construct a molecular tethering agent that regulates PrPC protein levels by inducing membrane-bound PrPC aggregation for antimelanoma therapy. Using a screening strategy combining cell-SELEX and cell-internalization SELEX, we obtained ssDNA aptamer, TT-1e, specifically binding to melanoma cells and tissues. We identified that the binding site of TT-1e is located at the octapeptide repeat region of glycosylated PrPC. Based on the binding characteristics of TT-1e, we engineered an aptamer-based molecular tethering agent TTe-TTe. We found that TTe-TTe induces aggregation of cell surface PrPC, promoting its internalization and facilitating its lysosomal degradation. This process resulted in the inhibition of AKT pathway activation. Importantly, in vivo studies confirmed the ability of TTe-TTe to target melanoma xenografts and suppress tumor growth through this unique mechanism. Our study presents a promising strategy for targeted melanoma therapy and introduces a paradigm-shifting approach for manipulating protein levels using aptamers as molecular tethering agents.
Collapse
Affiliation(s)
- Tiantian Xie
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Yibin Zhang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Neng Ling
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China, State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China, Zhejiang Key Laboratory of Key Technologies for Visual Pathway Reconstruction,Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yijun Yuan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Weibin Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China, State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China, Zhejiang Key Laboratory of Key Technologies for Visual Pathway Reconstruction,Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Junxiao Guo
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Xianhua Wei
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Tianhuan Peng
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Zhiyan Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Jing Dai
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Wenjing Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Ming Yan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Xiaoqiu Wu
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Wencan Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China, State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China, Zhejiang Key Laboratory of Key Technologies for Visual Pathway Reconstruction,Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
25
|
Zhao Z, Li ZQ, Huang YB, Liu MM, Cao F, Bu GL, Xu PF, Fang Q, Hu ZL, Wu D, Feng GK, Liu XK. An optimized integrin α6-targeted peptide capable of delivering toxins for melanoma treatment. J Transl Med 2025; 23:495. [PMID: 40307853 PMCID: PMC12044807 DOI: 10.1186/s12967-025-06511-5] [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: 12/06/2024] [Accepted: 04/18/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Peptide-based therapeutics for melanoma have received increasing attention in medical research. However, the local delivery of such therapeutics poses unique challenges. Cell-penetrating peptides (CPPs) with the ability to selectively enter cancer cells, with sufficient stability and increased endosomal escape mechanisms, can provide a new and improved delivery strategy for therapeutic agents for treating cancer. METHODS We developed a new combination strategy for the synthesis of penetrating peptides functionalized with targeting of integrin α6. The linear peptide S5 was multimerized with 4 copies in linear sequential order spaced by GSG between each copy to yield the 4S5 peptide. The multimerized 4S5 peptide coupled with an intracellular delivery peptide (N) and endosomal escape peptide (G) was separated by a GGS spacer. This optimized peptide was called 4S5NG. The 4S5NG, EGFP or PE24 peptide-protein conjugates were purified via a C-terminal His-tag. The uptake efficacy, intracellular distribution and integrin α6-targeting ability of these 4S5NG peptides were systematically characterized via IncuCyte, flow cytometry and in vivo imaging using 4S5NG-Cy5 or 4S5NG-EGFP. Moreover, 4S5NG-incorporated Pseudomonas aeruginosa (PE24) exotoxin A generated therapeutic peptides. The antitumor efficacy and underlying mechanism were studied in cell lines and a mouse model. In addition, the effect of 4S5NG-PE24 on antitumor immunity of a healthy immune system was investigated via a mouse model. RESULTS Images of living cells and mice indicated that 4S5NG accumulated at tumor sites in vitro and in vivo and was much more effective than the S5 and 4S5 peptides. 4S5NG-PE24 induced cell pyroptosis in integrin α6-expressing melanoma through the caspase 3/gasdermin E (GSDME) signaling pathway in the absence of histological alterations in other organs. 4S5NG-PE24 also promoted the response rate of programmed cell death protein-1 (PD-1) checkpoint blockade to increase antitumor efficacy. CONCLUSIONS Collectively, these results highlight the potential use of 4S5NG to deliver the toxin PE24 to selectively eliminate integrin α6+ cells in melanoma, which may represent a novel treatment approach for melanoma patients.
Collapse
Affiliation(s)
- Zheng Zhao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Zi-Qian Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Ying-Bin Huang
- Organ Transplantation Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Meng-Meng Liu
- Department of Oncology, The Second Affiliated Hospital Jiangxi Medical College Nanchang University, Nanchang, 330000, People's Republic of China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, 330000, People's Republic of China
| | - Fei Cao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Guo-Long Bu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Peng-Fei Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Qi Fang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Zhu-Long Hu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, People's Republic of China
| | - Di Wu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Guo-Kai Feng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Xue-Kui Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
26
|
Ródenas-Herranz T, Rodriguez-Barranco M, Petrova D, Pérez-Gómez B, Ruiz-Villaverde R, Sánchez MJ. Trends in cutaneous malignant melanoma incidence, mortality and survival over three decades: a population-based study in Southern Spain. Clin Exp Dermatol 2025; 50:981-993. [PMID: 39576024 DOI: 10.1093/ced/llae516] [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: 04/22/2024] [Revised: 09/18/2024] [Accepted: 11/17/2024] [Indexed: 04/25/2025]
Abstract
BACKGROUND Cutaneous malignant melanoma (CMM) represents a global public health problem. Epidemiological studies about CMM trends tend to focus on single indicators or lack information about tumour characteristics that can help gain a more thorough understanding of CMM epidemiology. OBJECTIVES To fill this gap, we studied CMM trends in incidence, mortality and survival over three decades and that considered sex, age and tumour characteristics. METHODS Data were obtained for all patients with a new diagnosis of CMM during 1985-2017 from the population-based cancer registry in Granada (Southern Spain) and for deaths owing to CMM from the Ministry of Health of the Spanish government. Incidence, mortality and survival estimates were obtained according to sex, age group, anatomical site, histological type, Breslow index and Clark level. Observed and net survival were calculated for 1, 3 and 5 years using the Kaplan-Meier and Pohar-Perme methods, respectively. Log-linear jointpoint regression was used to assess temporal trends in age-standardized rates of incidence and mortality. RESULTS Between 1985 and 2017, 2446 patients aged ≥ 15 years were newly diagnosed with CMM. There was a significant increase in incidence, both in males and females (annual percentage of change 4.4% and 3.7%, respectively), mainly in superficial spreading and nodular melanomas, and more pronounced among those with < 1 mm Breslow index. Mortality rates increased, mainly driven by increases among males and individuals ≥ 65 years old. Nodular melanoma had the worst prognosis, with a 5-year net survival of 62.7% in the period 2008-2017 compared with survival > 90% for other subtypes. Overall 5-year net survival improved from 78.3% in 1985-1997 to 88.6% in 2008-2017, with a clear trend by Breslow index. CONCLUSIONS The incidence of melanoma in early stages in Southern Spain has increased over recent decades. This has been accompanied by noticeable improvements in mortality and survival, especially among younger age groups, suggesting the potential effectiveness of prevention strategies and new treatment regimens.
Collapse
Affiliation(s)
- Teresa Ródenas-Herranz
- Dermatology Department, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Miguel Rodriguez-Barranco
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Dafina Petrova
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Beatriz Pérez-Gómez
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Epidemiology for Chronic Diseases, National Center of Epidemiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Ricardo Ruiz-Villaverde
- Dermatology Department, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - María-José Sánchez
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| |
Collapse
|
27
|
Zhu L, Qiao G, Gao H, Jiang A, Zhang L, Wang X. Enhancing melanoma therapy with hydrogel microneedles. Front Oncol 2025; 15:1590534. [PMID: 40313257 PMCID: PMC12043666 DOI: 10.3389/fonc.2025.1590534] [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: 03/09/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Melanoma is highly invasive and resistant to conventional treatments, accounting for nearly 75% of skin cancer-related deaths globally. Traditional therapies, such as chemotherapy and immunotherapy, often exhibit limited efficacy and are associated with significant side effects due to systemic drug exposure. Microneedles (MNs), as an emerging drug delivery system, offer multiple advantages, including safety, painlessness, minimal invasiveness, and controlled drug release. Among these, hydrogel microneedles (HMNs) stand out due to their extracellular matrix-like structure and swelling-induced continuous hydrogel channels, which enable the direct delivery of therapeutic agents into the tumor microenvironment (TME). This approach enhances drug bioavailability while reducing systemic toxicity, establishing HMNs as a promising platform for melanoma treatment. This review highlights recent advancements in HMNs for melanoma therapy, focusing on their applications in biomarker extraction for early diagnosis and their role in supporting multimodal treatment strategies, such as chemotherapy, immunotherapy, phototherapy, targeted therapy, and combination therapy. Furthermore, the current matrix materials and fabrication techniques for HMNs are discussed. Finally, the limitations of HMNs in melanoma treatment are critically analyzed, and recommendations for future research and development are provided.
Collapse
Affiliation(s)
- Lanqi Zhu
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guanlin Qiao
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huiyang Gao
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Aowei Jiang
- Department of Plastic and Reconstructive Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Linan Zhang
- Department of Plastic and Reconstructive Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaobing Wang
- Department of Plastic and Reconstructive Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
28
|
Ahmed RS, Ali RR, Hiwa DS, Ghafour AK, Siddiq SO, Asaad SK, Abdullah AM, Ali RM, Hammood ZD, Ali RM, Kakamad FH. Acral lentiginous melanoma with subsequent spontaneous vitiligo vulgaris: a case report and literature review. Dermatol Reports 2025. [PMID: 40244039 DOI: 10.4081/dr.2025.10125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/22/2025] [Indexed: 04/18/2025] Open
Abstract
Acral lentiginous melanoma (ALM) is an infrequent and often missed subtype of melanoma. Its association with spontaneous vitiligo vulgaris is an exceedingly rare condition. The current study presents a rare case of ALM with spontaneous vitiligo vulgaris after complete resection of the ALM. An 84-year-old male patient presented with multiple white skin lesions on the scalp, neck, trunk, and upper and lower limbs for a three-month duration. One year prior, a diagnosis of ALM was confirmed on histopathological examination with a pathological stage of T4bNxMx. The patient did not receive any treatment until the time of presentation, apart from the surgical removal of the primary tumor. No treatment was advised for the patient's vitiligo vulgaris; only observation was recommended. Clinical diagnosis of vitiligo vulgaris was made with the association of ALM. In the literature review, only several case reports of vitiligo were attributed to ALM, especially without the use of immunotherapeutic agents. Melanoma represents a notably immunogenic malignancy, provoking both humoral and cellular responses from the immune system directed against cytoplasmic and membrane antigens of melanocytes. There is a suggestion that normal melanocytes might function as neutral observers and become targets of immune responses directed against melanoma cells, leading to melanoma-associated vitiligo. It occurs more commonly as a consequence of immunologic-based therapeutic interventions or after metastatic melanoma; however, the occurrence of spontaneous vitiligo vulgaris several months following complete surgical removal of ALM is possible.
Collapse
Affiliation(s)
- Ronak Saeed Ahmed
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah; Dermatology Teaching Center for Treating Skin Diseases, Sulaimani Directorate of Health, Sulaymaniyah.
| | | | - Dilan S Hiwa
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah.
| | | | - Shvan Omar Siddiq
- Dermatology Teaching Center for Treating Skin Diseases, Sulaimani Directorate of Health, Sulaymaniyah; Kurdistan Board of Medical Specialties, Erbil.
| | - Saywan K Asaad
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah; College of Medicine, University of Sulaimani, Sulaymaniyah.
| | - Ari M Abdullah
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah; Department of Pathology, Sulaymaniyah Teaching Hospital.
| | - Rawa M Ali
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah; Department of Histopathology, Hospital for Treatment of Victims of Chemical Weapons, Halabja.
| | - Zuhair D Hammood
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah; Kscien Organization, Sulaymaniyah.
| | - Rebaz M Ali
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah; Hiwa Hospital, Ministry of Health, Sulaymaniyah.
| | - Fahmi H Kakamad
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah; College of Medicine, University of Sulaimani, Sulaymaniyah.
| |
Collapse
|
29
|
Oddershede JK, Meklenborg IK, Bastholt L, Guldbrandt LM, Schmidt H, Friis RB. Cardiotoxicity in patients with metastatic melanoma treated with BRAF/MEK inhibitors: a real-world analysis of incidence, risk factors, and reversibility. Acta Oncol 2025; 64:507-515. [PMID: 40223207 PMCID: PMC12012651 DOI: 10.2340/1651-226x.2025.42567] [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: 11/24/2024] [Accepted: 03/01/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND BRAF/MEK inhibitors (BRAFi/MEKi) improve outcome in patients with BRAF-mutated metastatic melanoma but are associated with cardiotoxicity, leading to a decline in left ventricular ejection fraction (LVEF). This study aimed to evaluate the incidence, timeline, risk factors, and reversibility of BRAFi/MEKi-induced cardiotoxicity in a real-world setting. PATIENTS/MATERIALS AND METHODS Patients with metastatic melanoma (n = 170) treated with Encorafenib/Binimetinib, Vemurafenib/Cobimetinib, or Dabrafenib/Trametinib at Aarhus and Odense University Hospital, Denmark, from 2015 to 2023 were included. Cardiac function was assessed at baseline and every 3 months during treatment with either echocardiograms or multigated acquisition scans. Cardiotoxicity was defined as a reduction of LVEF by ≥10 percentage points (pp) to an LVEF < 50% (Major cardiotoxicity) or a reduction of LVEF by ≥15 pp but remaining > 50% (Minor cardiotoxicity). RESULTS Cardiotoxicity occurred in 21% of patients, with 14% experiencing major cardiotoxicity. The mean time to LVEF decline was 187 days, with 92% of major cardiotoxicity cases occurring within the first year. Cardiotoxicity was reversible in 79% of patients following dose reduction, treatment pauses, heart failure therapy, or continued treatment with monitoring. Baseline atrial fibrillation (odds ratio 13.67, p = 0.008) was identified as a risk factor for major cardiotoxicity. INTERPRETATION BRAFi/MEKi-induced cardiotoxicity is a significant but manageable complication, often reversible with timely interventions. Routine LVEF monitoring is recommended. The majority (92%) of major cardiac events were diagnosed within the first year of treatment, which might warrant a discontinuation of routine LVEF monitoring after 1 year of BRAFi/MEKi treatment.
Collapse
Affiliation(s)
| | - Ida K Meklenborg
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | | | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Rasmus B Friis
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
30
|
Sun Q, Wang N, Poelchen J, Peter M, Novak D, Özbay Kurt FG, Bitsch R, Wu H, Wang Y, Pardo S, Han R, Liu S, Gong L, Zhang Y, Wistuba-Hamprecht K, Umansky V, Utikal JS. Neural crest-associated gene FOXD1 induces an immunosuppressive microenvironment by regulating myeloid-derived suppressor cells in melanoma. J Immunother Cancer 2025; 13:e010352. [PMID: 40210238 PMCID: PMC11987097 DOI: 10.1136/jitc-2024-010352] [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: 08/14/2024] [Accepted: 03/26/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Neural crest-associated genes play pivotal roles in tumor initiation, progression, and the intricate dynamics of the tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSC) within the TME are important in dampening T cell activity and contributing to resistance against immunotherapeutic interventions. The neural crest-associated gene Forkhead Box D1 (FOXD1) has been identified as an oncogenic factor that induces melanoma dedifferentiation and progression. However, the underlying mechanisms and the impact of FOXD1 on the antitumor immune response remain unclear. METHODS To investigate the impacts of FOXD1 on the melanoma microenvironment, we analyzed publicly available datasets from multiple platforms, including TNMplot, TIMER2.0, etc. In addition, FOXD1 was overexpressed (OE) or knocked down in melanoma cells to identify its biological functions in vitro and in vivo. Flow cytometry and arginase activity assay were used to analyze the phenotype and function of MDSC. Western blot, reverse transcription-PCR, or ELISA assays were employed to analyze the expression of FOXD1 and its downstream effectors. In vivo experiments were conducted to investigate the role of FOXD1 in melanoma progression and the influence on MDSC accumulation within the TME. RESULTS We demonstrate that increased FOXD1 levels inversely correlated with melanoma responsiveness to immunotherapy. Ex-vivo analyses unveiled that monocytes, exposed to conditioned medium from FOXD1-OE melanoma cells, effectively suppressed T cell proliferation and upregulated the expression of programmed death-ligand 1 (PD-L1) and other immunosuppressive factors. FOXD1 was identified as a direct regulator of interleukin 6 (IL6) expression, which is pivotal for MDSC induction. Blocking IL6 reversed MDSC-associated immunosuppression. Additionally, miR-581, a potential negative regulator of FOXD1, attenuated the impact of FOXD1 on IL6 expression and MDSC differentiation. In vivo experiments demonstrated that tumors derived from FOXD1 OE melanoma cells contained a significantly higher frequency of PD-L1+ MDSC compared with controls, while FOXD1 knockdown resulted in reduced tumor growth and diminished MDSC accumulation. CONCLUSION Our study elucidated a novel function of FOXD1 in melanoma pathogenesis, highlighting its role in orchestrating the immunosuppressive TME by promoting the generation of MDSC via IL6 upregulation.
Collapse
Affiliation(s)
- Qian Sun
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Nina Wang
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Juliane Poelchen
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Mareike Peter
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Daniel Novak
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Feyza Gül Özbay Kurt
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Rebekka Bitsch
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Huizi Wu
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Yiman Wang
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Sandra Pardo
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Rui Han
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University, Heidelberg, Baden-Württemberg, Germany
| | - Shibo Liu
- State Key Laboratory of Molecular Oncology, Tsinghua university, School of Basic Medical Sciences, Beijing, China
| | - Lidong Gong
- Peking University Institute of Systems Biomedicine, Beijing, China
| | - Yuxin Zhang
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Kilian Wistuba-Hamprecht
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| | - Jochen Sven Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and DKFZ Hector Cancer Institute, Heidelberg, Baden-Württemberg, Germany
- Department of Dermatology Venereology and Allergology, Heidelberg University, Mannheim, Baden-Württemberg, Germany
| |
Collapse
|
31
|
Ye M, Ren S, Luo H, Wu X, Lian H, Cai X, Ji Y. Integration of graph neural networks and transcriptomics analysis identify key pathways and gene signature for immunotherapy response and prognosis of skin melanoma. BMC Cancer 2025; 25:648. [PMID: 40205338 PMCID: PMC11983817 DOI: 10.1186/s12885-025-13611-4] [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: 06/06/2024] [Accepted: 01/29/2025] [Indexed: 04/11/2025] Open
Abstract
OBJECTIVE The assessment of immunotherapy plays a pivotal role in the clinical management of skin melanoma. Graph neural networks (GNNs), alongside other deep learning algorithms and bioinformatics approaches, have demonstrated substantial promise in advancing cancer diagnosis and treatment strategies. METHODS GNNs models were developed to predict the response to immunotherapy and to pinpoint key pathways. Utilizing the genes from these key pathways, multi-omics bioinformatics methods were employed to refine the construction of a gene signature, termed responseScore, aimed at enhancing the precision of immunotherapy response predictions. Subsequently, responseScore was explored from the perspectives of prognosis, genetic variation, pathway enrichment, and the tumor microenvironment. Concurrently, the association among 13 genes contributing to responseScore and factors such as immunotherapy response, prognosis, and the tumor microenvironment was investigated. Among these genes, PSMB6 was subjected to an in-depth analysis of its biological effect through experimental approaches like transfection and co-culture. RESULTS In the finalized model utilizing GNNs, it has revealed an AUC of 0.854 within the training dataset and 0.824 within the testing set, pinpointing key pathways such as R-HSA-70,268. The indicator named as responseScore excelled in its predictive accuracy regarding immunotherapy response and patient prognosis. Investigations into genetic variation, pathway enrichment, tumor microenvironment disclosed a profound association between responseScore and the enhancement of immune cell infiltration and anti-tumor immunity. A negative correlation was observed between the expression of PSMB6 and immune genes, with elevated PSMB6 expression correlating with poor prognosis. ELISA detection after co-cultivation experiments revealed significant reductions in the levels of cytokines IL-6 and IL-1β in specimens from the PCDH-PSMB6 group. CONCLUSION The GNNs prediction model and the responseScore developed in this research effectively indicate the immunotherapy response and prognosis for patients with skin melanoma. Additionally, responseScore provides insights into the tumor microenvironment and the characteristics of tumor immunity of melanoma. Thirteen genes identified in this study show promise as potential tumor markers or therapeutic targets. Notably, PSMB6 emerges as a potential therapeutic target for skin melanoma, where its elevated expression exhibits an inhibitory effect on the tumor immunity.
Collapse
Affiliation(s)
- Maodong Ye
- Medical Cosmetic Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Shuai Ren
- Medical Cosmetic Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Huanjuan Luo
- Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Xiumin Wu
- Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Hongwei Lian
- Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Xiangna Cai
- Medical Cosmetic Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Yingchang Ji
- Medical Cosmetic Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China.
| |
Collapse
|
32
|
Kaiser I, Rappl A, Bolay LS, Pfahlberg AB, Heppt MV, Gefeller O. The Impact of Dietary Intake of Furocoumarins and Furocoumarin-Rich Foods on the Risk of Cutaneous Melanoma: A Systematic Review. Nutrients 2025; 17:1296. [PMID: 40284161 PMCID: PMC12030158 DOI: 10.3390/nu17081296] [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: 02/27/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Furocoumarins, chemical compounds found in many plant species, have a photosensitizing effect on the skin when applied topically and, by interacting with ultraviolet radiation (UVR), stimulate melanoma cells to proliferate. Whether dietary intake of furocoumarins acts as a melanoma risk factor has been investigated in several epidemiological studies, which are synthesized in our systematic review. METHODS The study protocol was registered with PROSPERO (registration number: CRD42023428596). We conducted an in-depth literature search in three databases coupled with forward and backward citation tracking and expert consultations to identify all epidemiological studies, irrespective of their design, addressing the association between a furocoumarin-containing diet and melanoma risk. We extracted information on the study details and results in a standardized manner and evaluated the risk of bias of the results using the Joanna Briggs Institute's critical appraisal tools. RESULTS We identified 20 publications based on 19 different studies providing information on the association between dietary furocoumarin intake and melanoma risk. We refrained from a meta-analytical synthesis of the results because of the large heterogeneity in exposure assessment, operationalization of furocoumarin intake in the analyses, and analytical methods of the studies. In a qualitative synthesis, we found moderate evidence supporting the notion that dietary furocoumarin intake at higher levels acts as a risk factor for cutaneous melanoma. CONCLUSIONS Our systematic review provides an overview of the current epidemiological evidence, but it could not clearly answer whether and to what extent dietary furocoumarin intake increases melanoma risk. Future epidemiological analyses focusing on this topic require more comprehensive dietary and UVR exposure data to better characterize the individual total furocoumarin intake and its interplay with UVR exposure patterns.
Collapse
Affiliation(s)
- Isabelle Kaiser
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (I.K.); (A.R.); (L.S.B.); (A.B.P.)
| | - Anja Rappl
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (I.K.); (A.R.); (L.S.B.); (A.B.P.)
| | - Lena S. Bolay
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (I.K.); (A.R.); (L.S.B.); (A.B.P.)
| | - Annette B. Pfahlberg
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (I.K.); (A.R.); (L.S.B.); (A.B.P.)
| | - Markus V. Heppt
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
| | - Olaf Gefeller
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (I.K.); (A.R.); (L.S.B.); (A.B.P.)
| |
Collapse
|
33
|
Sun J, Wu W, Wang Y, Zhang J, Qiu S, Guan Z, Shi C, Ma J, Xu Y. MLKL-Mediated Necroptosis Predominantly Contributes to Immune-Associated Myocardial Damage. Inflammation 2025:10.1007/s10753-025-02298-1. [PMID: 40195182 DOI: 10.1007/s10753-025-02298-1] [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: 01/06/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
Activated T cells and macrophages play a critical role in immune-associated myocarditis. However, the molecular and cellular mechanisms driving cardiomyocyte damage by immune cells remain poorly understood. In this study, we co-cultured human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with activated human peripheral blood mononuclear cells (aPBMCs) to recapitulate myocardial infiltration of immune cells. Our results demonstrated that aPBMCs induced hiPSC-CMs death in a dose- and time-dependent manner. Transcriptome analysis revealed the activation of several death pathways, including pyroptosis, apoptosis and necroptosis. The time course of immunofluorescence staining of key proteins related to different death pathways demonstrated that necroptosis was the earliest activated pathway. Pharmacological blockade of necroptosis by targeting mixed lineage kinase domain-like protein (MLKL), receptor-interacting protein kinase 1 (RIPK1) and receptor-interacting protein RIPK1 kinase 3 (RIPK3) protected hiPSC-CMs against injury induced by aPBMCs, while inhibitors of pyroptosis and apoptosis showed no protective effect. Moreover, MLKL knockdown in hiPSC-CMs prevented cell death due to aPBMCs challenge. Additionally, we validated the cardioprotective effects of blocking necroptosis in a mouse model of immune checkpoint inhibitors (ICIs)-related myocarditis using a combination of long-term anti-programmed cell death 1 (PD- 1) and anti-cytotoxic T-lymphocyte antigen- 4 (CTLA- 4) antibodies. ICIs led to elevation of myocardial injury markers in serum and activated immune cells infiltration. Furthermore, in vivo administration of a MLKL inhibitor prevented ICIs-induced myocardial injury. In conclusion, our findings suggested that MLKL-mediated necroptosis predominantly contributed to cardiomyocyte death resulting from activated immune cells. Suppressing necroptosis may be an effective therapeutic approach against myocardial damage in myocarditis.
Collapse
Affiliation(s)
- Jinglei Sun
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China
| | - Wenting Wu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China
- Department of Clinical Pharmacy, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Yi Wang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China
| | - Jiali Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China
| | - Suhua Qiu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China
| | - Zhengkun Guan
- Department of Cardiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050010, China
| | - Chenxia Shi
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China
| | - Jingtao Ma
- Department of Cardiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050010, China
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, 361 East Zhongshan Road, Shijiazhuang, 050017, Hebei, China.
| |
Collapse
|
34
|
Liang Y, Maeda O, Nishida K, Chretien B, Ando Y. Genomic profiles of patients with skin melanoma in the era of immune checkpoint inhibitors. Cancer Sci 2025; 116:1107-1114. [PMID: 39888082 PMCID: PMC11967263 DOI: 10.1111/cas.16338] [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: 07/21/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 02/01/2025] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) for treating melanoma has dramatically improved patient prognosis. The genomic profiles of patients receiving ICI therapy would provide valuable information for disease management and treatment. We investigated the genomic profiles of patients with melanoma who had received ICI therapy and explored associations with clinical features and outcomes via a large-scale nationwide database in Japan (the C-CAT database). We identified 339 patients eligible for this study. The most frequent genetic mutations were found in the BRAF (27%), TERT (24%), and NRAS (19%) genes, and the most common copy number variations (CNVs) were in the CDKN2A (36%), CDKN2B (26%), and MTAP (19%) genes. Associations with high tumor mutational burden (TMB-high) status were significant for TERT (p < 0.001), NF1 (p < 0.001), ROS1 (p = 0.015), POLE (p = 0.045), and POLD1 (p = 0.008) mutations, along with older age (≥65 years, p = 0.036). Patients with multiple metastases (two or more) were more likely to have NOTCH3 mutations (p = 0.017) and be younger than 65 years (p = 0.024). In particular, as well as younger age, patients with brain metastases were more likely to harbor BRAF mutations (p < 0.001), while those with liver metastases were more likely to harbor NOTCH3 mutations (p < 0.001) but not CDKN2B CNVs (p = 0.041). Patients with NRAS mutations were less likely to respond to ICI therapy (p = 0.014) and exhibited shorter overall survival (p = 0.006). In this population, the frequency of BRAF mutations was lower than that in fair-skinned populations, but the associations between genomic profiles, clinical features, and outcomes were similar to those previously reported in fair-skinned populations.
Collapse
Affiliation(s)
- Yao Liang
- Department of Clinical Oncology and ChemotherapyNagoya University HospitalNagoyaJapan
| | - Osamu Maeda
- Department of Clinical Oncology and ChemotherapyNagoya University HospitalNagoyaJapan
| | - Kazuki Nishida
- Department of Advanced MedicineNagoya University HospitalNagoyaJapan
| | - Basile Chretien
- Department of Advanced MedicineNagoya University HospitalNagoyaJapan
| | - Yuichi Ando
- Department of Clinical Oncology and ChemotherapyNagoya University HospitalNagoyaJapan
| |
Collapse
|
35
|
Kamali MJ, Salehi M, Fath MK. Advancing personalized immunotherapy for melanoma: Integrating immunoinformatics in multi-epitope vaccine development, neoantigen identification via NGS, and immune simulation evaluation. Comput Biol Med 2025; 188:109885. [PMID: 40010174 DOI: 10.1016/j.compbiomed.2025.109885] [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: 11/25/2024] [Revised: 01/23/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
The use of cancer vaccines represents a promising avenue in cancer immunotherapy. Advances in next-generation sequencing (NGS) technology, coupled with the development of sophisticated analysis tools, have enabled the identification of somatic mutations by comparing genetic sequences between normal and tumor samples. Tumor neoantigens, derived from these mutations, have emerged as potential candidates for therapeutic cancer vaccines. In this study, raw NGS data from two melanoma patients (NCI_3903 and NCI_3998) were analyzed using publicly available SRA datasets from NCBI to identify patient-specific neoantigens. A comprehensive pipeline was employed to select candidate peptides based on their antigenicity, immunogenicity, physicochemical properties, and toxicity profiles. These validated epitopes were utilized to design multi-epitope chimeric vaccines tailored to each patient. Peptide linkers were employed to connect the epitopes, ensuring optimal vaccine structure and function. The two-dimensional (2D) and three-dimensional (3D) structures of the chimeric vaccines were predicted and refined to ensure structural stability and immunogenicity. Furthermore, molecular docking simulations were conducted to evaluate the binding interactions between the vaccine chimeras and the HLA class I receptors, confirming their potential to elicit a robust immune response. This work highlights a personalized approach to cancer vaccine development, demonstrating the feasibility of utilizing neoantigen-based immunoinformatics pipelines to design patient-specific therapeutic vaccines for melanoma.
Collapse
Affiliation(s)
- Mohammad Javad Kamali
- Department of Medical Genetics, School of Medicine, Babol University of Medical Science, Babol, Iran
| | - Mohammad Salehi
- Department of Medical Genetics, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran.
| |
Collapse
|
36
|
Yang T, Huang Q, Cai F, Li J, Jiang L, Xia Y. Vital Characteristics Cellular Neural Network (VCeNN) for Melanoma Lesion Segmentation: A Biologically Inspired Deep Learning Approach. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025; 38:1147-1164. [PMID: 39284982 PMCID: PMC11950543 DOI: 10.1007/s10278-024-01257-w] [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: 07/01/2024] [Revised: 08/27/2024] [Accepted: 08/31/2024] [Indexed: 03/29/2025]
Abstract
Cutaneous melanoma is a highly lethal form of cancer. Developing a medical image segmentation model capable of accurately delineating melanoma lesions with high robustness and generalization presents a formidable challenge. This study draws inspiration from cellular functional characteristics and natural selection, proposing a novel medical segmentation model named the vital characteristics cellular neural network. This model incorporates vital characteristics observed in multicellular organisms, including memory, adaptation, apoptosis, and division. Memory module enables the network to rapidly adapt to input data during the early stages of training, accelerating model convergence. Adaptation module allows neurons to select the appropriate activation function based on varying environmental conditions. Apoptosis module reduces the risk of overfitting by pruning neurons with low activation values. Division module enhances the network's learning capacity by duplicating neurons with high activation values. Experimental evaluations demonstrate the efficacy of this model in enhancing the performance of neural networks for medical image segmentation. The proposed method achieves outstanding results across numerous publicly available datasets, indicating its potential to contribute significantly to the field of medical image analysis and facilitating accurate and efficient segmentation of medical imagery. The proposed method achieves outstanding results across numerous publicly available datasets, with an F1 score of 0.901, Intersection over Union of 0.841, and Dice coefficient of 0.913, indicating its potential to contribute significantly to the field of medical image analysis and facilitating accurate and efficient segmentation of medical imagery.
Collapse
Affiliation(s)
- Tongxin Yang
- Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Qilin Huang
- Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Fenglin Cai
- Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Jie Li
- Chongqing University of Science and Technology, Chongqing, 401331, China.
| | - Li Jiang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yulong Xia
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
37
|
Taskaeva I, Shatruk A, Ivanov I, Bgatova N. The effects of autophagy-modifying drugs chloroquine and lithium on the skin melanoma microenvironment. Tissue Cell 2025; 93:102753. [PMID: 39864206 DOI: 10.1016/j.tice.2025.102753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND Skin melanoma is a highly metastatic cancer with an increasing global incidence. Despite advancements in immunotherapy, new treatment strategies based on tumor biology are essential for improving outcomes and developing novel therapies. Autophagy plays a critical role in melanoma cell metabolism and affects the tumor microenvironment (TME). This study aims to evaluate the impact of autophagy-modifying drugs on extracellular matrix (ECM) remodeling and changes in the TME cytokine profile. METHODS Immunohistochemical analysis was performed using paraffin-embedded tumor samples of B16-bearing C57BL/6 mice to assess the effects of autophagy-modifying drugs, lithium or chloroquine, on the matrix degradation proteins, their main substrates, lysyl oxidase and collagen fibril formation-associated proteins. The cytokine profile of the tumor was defined to estimate the effect of autophagy-modifying drugs on the TME. RESULTS Chloroquine and lithium administration caused a decrease in the expression of matrix metalloproteinases, and chloroquine contributed to the accumulation of collagen type I. Moreover, chloroquine dramatically decreased LOX levels. Decorin expression levels were reduced in tumors of mice treated with chloroquine or lithium. Significant changes in the cytokine profile were detected after chloroquine treatment, with increased expression of IL1, IL4, IL6, M-CSF, TGFβ2 and TNF-α genes observed in the tumors. CONCLUSION Autophagy-modifying drugs affect the TME, in particular, chloroquine promotes ECM remodeling, accumulation of collagen type I deposits and probably the formation of abnormal collagen fibril structures. In addition, chloroquine-treated mice showed high expression of pro-tumorigenic cytokines and growth factors, such as IL1, IL4, IL6, M-CSF, TGFβ2 and TNF-α in the TME.
Collapse
Affiliation(s)
- Iuliia Taskaeva
- Laboratory of Ultrastructural Research, Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 6 Arbuzov St., Novosibirsk 630117, Russia.
| | - Anastasia Shatruk
- Laboratory of Ultrastructural Research, Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 6 Arbuzov St., Novosibirsk 630117, Russia
| | - Igor Ivanov
- Laboratory of Drug Metabolism and Pharmacokinetics, Federal Research Center of Fundamental and Translational Medicine, Institute of Molecular Biology and Biophysics, 2 Timakov St., Novosibirsk 630060, Russia
| | - Nataliya Bgatova
- Laboratory of Ultrastructural Research, Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 6 Arbuzov St., Novosibirsk 630117, Russia
| |
Collapse
|
38
|
Wang D, Xie A, Luo J, Li L, Zhang Z, Deng W, Yang B, Chang Y, Liang Y. Thiotaurine inhibits melanoma progression by enhancing Ca 2+ overload-induced cellular apoptosis. J Dermatol Sci 2025; 118:29-37. [PMID: 40189970 DOI: 10.1016/j.jdermsci.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Melanoma is the most dangerous type of skin cancer with poor therapy outcomes. Since malignant cells are more susceptible to Ca2+ overload than normal cells, activating Ca2+ overload-mediated apoptosis may be a promising strategy to inhibit melanoma progression. Hydrogen sulfide (H2S) donors can regulate Ca2+ channels, but their effects on melanoma cells remain unclear. OBJECTIVE To explore the effects of Thiotaurine (TTAU), an H2S donor, on melanoma cells and its underlying mechanisms. METHODS We tested the effect of TTAU by culturing melanoma cells in vitro and establishing the xenograft model of mice in vivo. Cell proliferation and apoptosis were assessed using the CCK-8 test and flow cytometry. Molecules involved in apoptosis or Ca2+-related signal transduction were analyzed by western blotting. Immunofluorescence was used to measure Ca2+ levels, mitochondrial damage, and reactive oxygen species (ROS). RESULTS TTAU significantly reduced melanoma cell viability and induced apoptosis both in vitro and in vivo. Mechanistically, TTAU increased intracellular Ca2+, upregulated transient receptor potential vanilloid 1(TRPV1), and decreased activating transcription factor 3(ATF3) by nuclear factor of activated T cell cytoplasmic 1(NFATc1). TTAU also caused mitochondrial damage and ROS overproduction, which also promoted apoptosis. CONCLUSION We first elucidate that TTAU inhibits melanoma progression by activating Ca2+ influx-NFATc1-ATF3 signaling and aggravating mitochondrial oxidative stress, in which TRPV1 may act as an amplifier for Ca2+ influx. Our research is expected to provide new ideas for the treatment of tumors such as melanoma, as well as the clinical application of reactive sulfur species-based drugs.
Collapse
Affiliation(s)
- Di Wang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Ansheng Xie
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Jialiang Luo
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Lei Li
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhiwen Zhang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Weiwei Deng
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Chang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yunsheng Liang
- Dermatology Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
39
|
Sayedyahossein S, Huang K, Zhang C, Karimi M, Bahmani M, O’Donnell BL, Wakefield B, Li Z, Johnston D, Leighton SE, Huver MS, Dagnino L, Sacks DB, Penuela S. Pannexin 1 crosstalk with the Hippo pathway in malignant melanoma. FEBS J 2025; 292:1633-1653. [PMID: 39786847 PMCID: PMC11971028 DOI: 10.1111/febs.17396] [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: 02/22/2024] [Revised: 08/19/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
In this study, we explored the intricate relationship between Pannexin 1 (PANX1) and the Hippo signaling pathway effector, Yes-associated protein (YAP). Analysis of The Cancer Genome Atlas (TCGA) data revealed a significant positive correlation between PANX1 mRNA and core Hippo components, Yes-associated protein 1 [YAP], Transcriptional coactivator with PDZ-binding motif [TAZ], and Hippo scaffold, Ras GTPase-activating-like protein IQGAP1 [IQGAP1], in invasive cutaneous melanoma and breast carcinoma. Furthermore, we demonstrated that PANX1 expression is upregulated in invasive melanoma cell lines and is associated with increased YAP protein levels. Notably, our investigations uncovered a previously unrecognized interaction between endogenous PANX1 and the Hippo scaffold protein IQGAP1 in melanoma cells. Moreover, our findings revealed that IQGAP1 exhibits differential expression in melanoma cells and plays a regulatory role in cellular morphology. Functional studies involving PANX1 knockdown provided compelling evidence that PANX1 modulates YAP protein levels and its cotranscriptional activity in melanoma and breast carcinoma cells. Importantly, our study highlights the potential therapeutic significance of targeting PANX1. Pharmacological inhibition of PANX1 using selective FDA-approved inhibitors or PANX1 knockdown reduced YAP levels in melanoma cells. Furthermore, our Clariom™ S analysis unveiled key genes implicated in cell proliferation, such as neuroglin1 (NRG1), β-galactoside binding protein and galectin-3 (LGALS3), that are affected in PANX1-deficient cells. In summary, our investigation delves into the intricate interplay between PANX1 and YAP in the context of invasive melanoma, offering valuable insights into potential therapeutic strategies for effective treatment.
Collapse
Affiliation(s)
- Samar Sayedyahossein
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Kenneth Huang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Christopher Zhang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Mehdi Karimi
- Department of Mathematics, Illinois State University, Normal, Illinois, USA, 61790
| | | | - Brooke L. O’Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Brent Wakefield
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Zhigang Li
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
| | - Danielle Johnston
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Stephanie E. Leighton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Matthew S. Huver
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Lina Dagnino
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - David B. Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| |
Collapse
|
40
|
Zuo L, Wang Z, Wang Y. A multi-stage multi-modal learning algorithm with adaptive multimodal fusion for improving multi-label skin lesion classification. Artif Intell Med 2025; 162:103091. [PMID: 40015211 DOI: 10.1016/j.artmed.2025.103091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 09/10/2024] [Accepted: 02/14/2025] [Indexed: 03/01/2025]
Abstract
Skin cancer is frequently occurring and has become a major contributor to both cancer incidence and mortality. Accurate and timely diagnosis of skin cancer holds the potential to save lives. Deep learning-based methods have demonstrated significant advancements in the screening of skin cancers. However, most current approaches rely on a single modality input for diagnosis, thereby missing out on valuable complementary information that could enhance accuracy. Although some multimodal-based methods exist, they often lack adaptability and fail to fully leverage multimodal information. In this paper, we introduce a novel uncertainty-based hybrid fusion strategy for a multi-modal learning algorithm aimed at skin cancer diagnosis. Our approach specifically combines three different modalities: clinical images, dermoscopy images, and metadata, to make the final classification. For the fusion of two image modalities, we employ an intermediate fusion strategy that considers the similarity between clinical and dermoscopy images to extract features containing both complementary and correlated information. To capture the correlated information, we utilize cosine similarity, and we employ concatenation as the means for integrating complementary information. In the fusion of image and metadata modalities, we leverage uncertainty to obtain confident late fusion results, allowing our method to adaptively combine the information from different modalities. We conducted comprehensive experiments using a popular publicly available skin disease diagnosis dataset, and the results of these experiments demonstrate the effectiveness of our proposed method. Our proposed fusion algorithm could enhance the clinical applicability of automated skin lesion classification, offering a more robust and adaptive way to make automatic diagnoses with the help of uncertainty mechanism. Code is available at https://github.com/Zuo-Lihan/CosCatNet-Adaptive_Fusion_Algorithm.
Collapse
Affiliation(s)
- Lihan Zuo
- School of Computer and Artificial Intelligence, Southwest Jiaotong University, Chengdu 610000, PR China
| | - Zizhou Wang
- Institute of High Performance Computing, Agency for Science, Technology and Research (A*STAR), Singapore 138632, Singapore
| | - Yan Wang
- Institute of High Performance Computing, Agency for Science, Technology and Research (A*STAR), Singapore 138632, Singapore.
| |
Collapse
|
41
|
Liu C, Liu X, Cao P, Xin H, Li X, Zhu S. Circadian rhythm related genes identified through tumorigenesis and immune infiltration-guided strategies as predictors of prognosis, immunotherapy response, and candidate drugs in skin cutaneous malignant melanoma. Front Immunol 2025; 16:1513750. [PMID: 40191195 PMCID: PMC11968383 DOI: 10.3389/fimmu.2025.1513750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/20/2025] [Indexed: 04/09/2025] Open
Abstract
Background Skin cutaneous malignant melanoma (SKCM) is among the most aggressive forms of skin cancer, notorious for its rapid progression and poor prognosis under late diagnosis. This study investigates the role of circadian rhythm-related genes (CRGs) in SKCM addressing a gap in understanding how CRGs affect tumor progression and patient outcomes. Methods An analysis of CRGs expression was conducted on SKCM samples derived from The Cancer Genome Atlas datasets(TCGA). Moreover, a correlation between various subtypes and their clinical features was identified. The study employed various bioinformatics methods, including differential expression analysis, consensus clustering, and survival analysis, to investigate the role of CRGs. The functional consequences of various CRG expression patterns were further investigated using immune infiltration analysis and gene set variation analysis (GSVA). A scoring system based on CRGs was developed to predict overall survival (OS) and treatment responses in SKCM patients. The predictive accuracy of this score system was then tested, and a nomogram was used to improve its clinical usefulness. Results Key findings from this study include significant genetic alterations in circadian rhythm-related genes (CRGs) in skin cutaneous melanoma (SKCM), such as mutations and CNVs. Two molecular subtypes with distinct clinical outcomes and immune profiles were identified. A prognostic model based on six CRGs (CMTM, TNPO1, CTBS, UTRN, HK2, and LIF) was developed and validated with TCGA and GEO datasets, showing high predictive accuracy for overall survival (OS). A high CRGs score correlated with poor OS, immune checkpoint expression, and reduced sensitivity to several chemotherapeutics, including AKT inhibitor VIII and Camptothecin. Conclusions This work provides valuable insights into the circadian regulation of SKCM and underscores the potential of CRGs as biomarkers for prognosis and targets for therapeutic interventions. The novel molecular subtypes and CRGs prognostic scoring model introduced in this study offer significant contributions to the understanding and management of SKCM.
Collapse
Affiliation(s)
- Chengling Liu
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Xingchen Liu
- Department of Pathology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Pengjuan Cao
- Department of Endocrinology and Traditional Chinese Medicine, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Haiming Xin
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Xin Li
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Sailing Zhu
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| |
Collapse
|
42
|
Zhu S, Zhu R, Wang Y, Zhu J, Zong Y, Zhu L, Guo W. Comprehensive systems biology analysis reveals splicing factor contributions to cutaneous melanoma progression. Sci Rep 2025; 15:9486. [PMID: 40108329 PMCID: PMC11923367 DOI: 10.1038/s41598-025-93695-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Cutaneous melanoma (CM) is an aggressive skin cancer with high metastatic potential and poor prognosis. Splicing factors, which regulate pre-mRNA alternative splicing (AS) events, have been suggested as potential therapeutic targets in CM. The objective of this study was to identify candidate splicing factors involved in CM through a systems biology approach and to elucidate their roles in CM progression. 390 AS events associated with patient survival were identified using bivariate Cox regression and receiver operating characteristic (ROC) analyses. 121 splicing factors significantly associated with patient prognosis were screened by univariate Cox regression analysis. A bipartite association network between AS events and splicing factors was constructed using Spearman correlation analysis. Based on the network topology, five candidate splice factors were identified. Among them, U2SURP, a poorly characterized serine/arginine-rich protein family member, was selected for further analysis in CM. Results indicated that U2SURP gene expression was significantly negatively correlated with the Immune Infiltration Score, the infiltration levels of dendritic cells, gamma-delta T cells, natural killer (NK) cells, and cytotoxic cells, as well as the expression of the immune checkpoint gene PD-1, suggesting that U2SURP may serve as a potential target for CM immunotherapy. Experimental validation showed that U2SURP mRNA and protein were overexpressed in CM cells, and silencing of U2SURP using siRNA significantly reduced CM cell survival, proliferation and migration. Furthermore, single-cell functional analysis showed that U2SURP gene expression was positively correlated with CM cell proliferation and differentiation. This study systematically identified candidate splicing factors involved in CM and provided new insights into the role of U2SURP in CM progression. These findings contribute to a deeper understanding of the pathogenesis of CM and establish new approaches for identifying splicing-related cancer therapeutic targets.
Collapse
Affiliation(s)
- Shuting Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Rui Zhu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Yanna Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Junru Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Yifan Zong
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Liucun Zhu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Wenna Guo
- School of Life Sciences, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
43
|
Zhou Y, Han W, Feng Y, Wang Y, Liu X, Sun T, Xu J. Revealing gut microbiota biomarkers associated with melanoma immunotherapy response and key bacteria-fungi interaction relationships: evidence from metagenomics, machine learning, and SHAP methodology. Front Immunol 2025; 16:1539653. [PMID: 40170844 PMCID: PMC11959079 DOI: 10.3389/fimmu.2025.1539653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Introduction The gut microbiota is associated with the response to immunotherapy in cutaneous melanoma (CM). However, gut fungal biomarkers and bacterial-fungal interactions have yet to be determined. Methods Metagenomic sequencing data of stool samples collected before immunotherapy from three independent groups of European ancestry CM patients were collected. After characterizing the relative abundances of bacteria and fungi, Linear Discriminant Analysis Effect Size (LEfSe) analysis, Random Forest (RF) model construction, and SHapley Additive exPlanations (SHAP) methodology were applied to identify biomarkers and key bacterial-fungal interactions associated with immunotherapy responders in CM. Results Diversity analysis revealed significant differences in the bacterial and fungal composition between CM immunotherapy responders and non-responders. LEfSe analysis identified 45 bacterial and 4 fungal taxa as potential biomarkers. After constructing the RF model, the AUC of models built using bacterial and fungal data separately were 0.64 and 0.65, respectively. However, when bacterial and fungal data were combined, the AUC of the merged model increased to 0.71. In the merged model, the following taxa were identified as important biomarkers: Romboutsia, Endomicrobium, Aggregatilinea, Candidatus Moduliflexus, Colwellia, Akkermansia, Mucispirillum, and Rutstroemia, which were associated with responders, whereas Zancudomyces was associated with non-responders. Moreover, the positive correlation interaction between Akkermansia and Rutstroemia is considered a key bacterial-fungal interaction associated with CM immunotherapy response. Conclusion Our results provide valuable insights for the enrichment of responders to immunotherapy in CM patients. Moreover, this study highlights the critical role of bacterial-fungal interactions in CM immunotherapy.
Collapse
Affiliation(s)
- Yuhang Zhou
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Wenjie Han
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Yun Feng
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Yue Wang
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Xiaolin Liu
- Department of Bioinformatics, Kanghui Biotechnology Co., Ltd., Shenyang, China
| | - Tao Sun
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China
| | - Junnan Xu
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China
| |
Collapse
|
44
|
Lin TL, Karmakar R, Mukundan A, Chaudhari S, Hsiao YP, Hsieh SC, Wang HC. Assessing the Efficacy of the Spectrum-Aided Vision Enhancer (SAVE) to Detect Acral Lentiginous Melanoma, Melanoma In Situ, Nodular Melanoma, and Superficial Spreading Melanoma: Part II. Diagnostics (Basel) 2025; 15:714. [DOI: https:/doi.org/10.3390/diagnostics15060714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Background: Melanoma, a highly aggressive form of skin cancer, necessitates early detection to significantly improve survival rates. Traditional diagnostic techniques, such as white-light imaging (WLI), are effective but often struggle to differentiate between melanoma subtypes in their early stages. Methods: The emergence of the Spectrum-Aided Vison Enhancer (SAVE) offers a promising alternative by utilizing specific wavelength bands to enhance visual contrast in melanoma lesions. This technique facilitates greater differentiation between malignant and benign tissues, particularly in challenging cases. In this study, the efficacy of the SAVE is evaluated in detecting melanoma subtypes including acral lentiginous melanoma (ALM), melanoma in situ (MIS), nodular melanoma (NM), and superficial spreading melanoma (SSM) compared to WLI. Results: The findings demonstrated that the SAVE consistently outperforms WLI across various key metrics, including precision, recall, F1-scorw, and mAP, making it a more reliable tool for early melanoma detection using the four different machine learning methods YOLOv10, Faster RCNN, Scaled YOLOv4, and YOLOv7. Conclusions: The ability of the SAVE to capture subtle spectral differences offers clinicians a new avenue for improving diagnostic accuracy and patient outcomes.
Collapse
Affiliation(s)
- Teng-Li Lin
- Department of Dermatology, Dalin Tzu Chi General Hospital, No. 2 Min-Sheng Rd., Dalin Town, Chiayi 62247, Taiwan
| | - Riya Karmakar
- Department of Mechanical Engineering, National Chung Cheng University, 168 University Rd., Min Hsiung, Chiayi 62102, Taiwan
| | - Arvind Mukundan
- Department of Mechanical Engineering, National Chung Cheng University, 168 University Rd., Min Hsiung, Chiayi 62102, Taiwan
| | - Sakshi Chaudhari
- Department of Computer Science, Sanjivani College of Engineering, Station Rd, Singapur, Kopargaon 423603, Maharashtra, India
| | - Yu-Ping Hsiao
- Department of Dermatology, Chung Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Rd., South Dist., Taichung City 40201, Taiwan
- Institute of Medicine, School of Medicine, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., South Dist., Taichung City 40201, Taiwan
| | - Shang-Chin Hsieh
- Division of General Surgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, 2 Zhongzheng 1st. Rd., Lingya District, Kaohsiung City 80284, Taiwan
| | - Hsiang-Chen Wang
- Department of Mechanical Engineering, National Chung Cheng University, 168 University Rd., Min Hsiung, Chiayi 62102, Taiwan
- Hitspectra Intelligent Technology Co., Ltd., Kaohsiung 80661, Taiwan
| |
Collapse
|
45
|
Lin TL, Karmakar R, Mukundan A, Chaudhari S, Hsiao YP, Hsieh SC, Wang HC. Assessing the Efficacy of the Spectrum-Aided Vision Enhancer (SAVE) to Detect Acral Lentiginous Melanoma, Melanoma In Situ, Nodular Melanoma, and Superficial Spreading Melanoma: Part II. Diagnostics (Basel) 2025; 15:714. [PMID: 40150057 PMCID: PMC11941011 DOI: 10.3390/diagnostics15060714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Melanoma, a highly aggressive form of skin cancer, necessitates early detection to significantly improve survival rates. Traditional diagnostic techniques, such as white-light imaging (WLI), are effective but often struggle to differentiate between melanoma subtypes in their early stages. Methods: The emergence of the Spectrum-Aided Vison Enhancer (SAVE) offers a promising alternative by utilizing specific wavelength bands to enhance visual contrast in melanoma lesions. This technique facilitates greater differentiation between malignant and benign tissues, particularly in challenging cases. In this study, the efficacy of the SAVE is evaluated in detecting melanoma subtypes including acral lentiginous melanoma (ALM), melanoma in situ (MIS), nodular melanoma (NM), and superficial spreading melanoma (SSM) compared to WLI. Results: The findings demonstrated that the SAVE consistently outperforms WLI across various key metrics, including precision, recall, F1-scorw, and mAP, making it a more reliable tool for early melanoma detection using the four different machine learning methods YOLOv10, Faster RCNN, Scaled YOLOv4, and YOLOv7. Conclusions: The ability of the SAVE to capture subtle spectral differences offers clinicians a new avenue for improving diagnostic accuracy and patient outcomes.
Collapse
Affiliation(s)
- Teng-Li Lin
- Department of Dermatology, Dalin Tzu Chi General Hospital, No. 2 Min-Sheng Rd., Dalin Town, Chiayi 62247, Taiwan;
| | - Riya Karmakar
- Department of Mechanical Engineering, National Chung Cheng University, 168 University Rd., Min Hsiung, Chiayi 62102, Taiwan; (R.K.); (A.M.)
| | - Arvind Mukundan
- Department of Mechanical Engineering, National Chung Cheng University, 168 University Rd., Min Hsiung, Chiayi 62102, Taiwan; (R.K.); (A.M.)
| | - Sakshi Chaudhari
- Department of Computer Science, Sanjivani College of Engineering, Station Rd, Singapur, Kopargaon 423603, Maharashtra, India;
| | - Yu-Ping Hsiao
- Department of Dermatology, Chung Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Rd., South Dist., Taichung City 40201, Taiwan;
- Institute of Medicine, School of Medicine, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., South Dist., Taichung City 40201, Taiwan
| | - Shang-Chin Hsieh
- Division of General Surgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, 2 Zhongzheng 1st. Rd., Lingya District, Kaohsiung City 80284, Taiwan
| | - Hsiang-Chen Wang
- Department of Mechanical Engineering, National Chung Cheng University, 168 University Rd., Min Hsiung, Chiayi 62102, Taiwan; (R.K.); (A.M.)
- Hitspectra Intelligent Technology Co., Ltd., Kaohsiung 80661, Taiwan
| |
Collapse
|
46
|
Lubrano S, Cervantes-Villagrana RD, Faraji F, Ramirez S, Sato K, Adame-Garcia SR, Officer A, Arang N, Rigiracciolo DC, Anguiano Quiroz PY, Martini C, Wang Y, Ferguson FM, Bacchiocchi A, Halaban R, Coma S, Holmen SL, Pachter JA, Aplin AE, Gutkind JS. FAK inhibition combined with the RAF-MEK clamp avutometinib overcomes resistance to targeted and immune therapies in BRAF V600E melanoma. Cancer Cell 2025; 43:428-445.e6. [PMID: 40020669 PMCID: PMC11903146 DOI: 10.1016/j.ccell.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 10/17/2024] [Accepted: 02/04/2025] [Indexed: 03/03/2025]
Abstract
Widespread BRAF mutations result in persistent RAS-RAF-MEK-ERK (MAPK) signaling in melanoma. BRAF (BRAFi) and MEK (MEKi) inhibitors are approved for BRAF V600E melanomas, including those progressing on immunotherapy; however, rapid resistance to these agents highlights the need for novel strategies. Here, transcriptome analysis of BRAF V600E melanomas from patients resistant to BRAFi and MEKi shows activation of focal adhesion signaling. Consistently, BRAFi, MEKi, and the RAF-MEK clamp avutometinib activate focal adhesion kinase (FAK) in melanoma cells. Mechanistically, inhibition of an MAPK-RhoE (RND3) feedback loop results in the adaptive activation of RhoA-FAK-AKT. In turn, FAK inhibitors (FAKi) exert potent pro-apoptotic activity when combined with MAPK pathway inhibition. FAKi plus avutometinib overcomes resistance in multiple models derived from BRAFi plus MEKi-resistant melanoma patients and immunotherapy-resistant syngeneic mouse models. These findings provide a rationale for the development of avutometinib in combination with FAKi for patients with BRAF V600E melanoma progressing on BRAFi plus MEKi or immunotherapy.
Collapse
Affiliation(s)
- Simone Lubrano
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Rodolfo Daniel Cervantes-Villagrana
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Farhoud Faraji
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Otolaryngology - Head and Neck Surgery, UC San Diego Health, La Jolla, CA 92037, USA
| | - Sydney Ramirez
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kuniaki Sato
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sendi R Adame-Garcia
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adam Officer
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nadia Arang
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Paola Y Anguiano Quiroz
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; John Muir College, University of California, San Diego, La Jolla, CA 92093, USA
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - YiYu Wang
- Department of Chemistry and Biochemistry and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Fleur M Ferguson
- Department of Chemistry and Biochemistry and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | | | - Ruth Halaban
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | - Sheri L Holmen
- Department of Surgery, University of Utah Health Sciences Center, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Andrew E Aplin
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, US
| | - J Silvio Gutkind
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
47
|
Smith-Díaz CC, Kumar A, Das A, Pace P, Chitcholtan K, Magon NJ, Hossain SM, Eccles MR, Winterbourn CC, Paumann-Page M. Peroxidasin is associated with a mesenchymal-like transcriptional phenotype and promotes invasion in metastatic melanoma. Free Radic Biol Med 2025; 229:427-440. [PMID: 39793908 DOI: 10.1016/j.freeradbiomed.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/20/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Cutaneous melanoma is a highly invasive, heterogeneous and treatment resistant cancer. It's ability to dynamically shift between transcriptional states or phenotypes results in an adaptive cell plasticity that may drive cancer cell invasion or the development of therapy resistance. The expression of peroxidasin (PXDN), an extracellular matrix peroxidase, has been proposed to be associated with the invasive metastatic melanoma phenotype. We have confirmed this association by analysing the transcriptomes of 70 metastatic melanoma cell lines with variable levels of PXDN expression. This analysis highlighted a strong association between high PXDN expression and the undifferentiated invasive melanoma phenotype. To assess the functional role of PXDN in melanoma invasion, we performed a knockout of PXDN in a highly invasive cell line (NZM40). PXDN knockout decreased the invasive potential by ∼50 % and decreased the expression of epithelial-mesenchymal transition and invasive marker genes as determined by RNAseq and substantiated by proteomics analysis. Bioinformatics analysis of differentially expressed genes following PXDN knockout highlighted decreases in genes linked to extracellular matrix formation, organization and degradation as well as signalling pathways such as the WNT pathway. This study provides compelling evidence that PXDN plays a functional role in melanoma invasion by promoting an invasive, mesenchymal-like transcriptional phenotype.
Collapse
Affiliation(s)
- Carlos C Smith-Díaz
- Mātai Hāora, Centre for Redox Biology and Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Abhishek Kumar
- Centre for Protein Research, Research Infrastructure Centre, University of Otago, Dunedin, New Zealand
| | - Andrew Das
- Mātai Hāora, Centre for Redox Biology and Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Paul Pace
- Mātai Hāora, Centre for Redox Biology and Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, University of Otago Christchurch, Christchurch, New Zealand
| | - Nicholas J Magon
- Mātai Hāora, Centre for Redox Biology and Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Sultana Mehbuba Hossain
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Michael R Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Christine C Winterbourn
- Mātai Hāora, Centre for Redox Biology and Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Martina Paumann-Page
- Mātai Hāora, Centre for Redox Biology and Medicine, University of Otago Christchurch, Christchurch, New Zealand.
| |
Collapse
|
48
|
Xu W, Wang M, Liu X, Ding Y, Fu J, Zhang P. Recent advances in chemodynamic nanotherapeutics to overcome multidrug resistance in cancers. Biomed Pharmacother 2025; 184:117901. [PMID: 39933445 DOI: 10.1016/j.biopha.2025.117901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
Multidrug resistance (MDR) has become a major challenge in cancer therapy, it results in the failure of chemotherapy and anticancer drug development. Chemodynamic therapy (CDT), an emerging cancer treatment strategy, has been reported as a novel approach for cancer treatment characterized by low toxicity and minimal side effects. By generating robust cytotoxic hydroxyl radicals (·OH) via Fenton/Fenton-like reaction, CDT may cause cellular damage and oxidative stress-induced cell death. In recent years, many therapies based on CDT and/or combined with other treatment modalities are reported and exhibit exciting treatment efficacy in cancer treatment, such as photothermal therapy, photodynamic therapy, sonodynamic therapy, chemotherapy, starvation therapy and gas therapy etc. These combination therapies exhibit synergistic effects, significantly improving anticancer outcomes compared to CDT alone. Herein, we provide a comprehensive overview of CDT-based strategies in cancer treatment, highlighting developments of CDT and CDT-based combination strategies in tumor therapy, especially in overcoming MDR challenges. Finally, the opportunities and challenges of CDT and CDT-combination therapy in the clinical application are also addressed.
Collapse
Affiliation(s)
- Wenjia Xu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Min Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Xinyu Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Yucui Ding
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Jianlong Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Peng Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| |
Collapse
|
49
|
Zhang N, Qiu X, Chen X, Du C, Dong J, Li X, Chen B, Zhang L, Zhang Y. Survival expectations in melanoma patients: a molecular prognostic model associated with aging. Discov Oncol 2025; 16:253. [PMID: 40019657 PMCID: PMC11874052 DOI: 10.1007/s12672-025-01971-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Aging and long non-coding RNAs (lncRNAs) are research hotspots in melanoma. However, no study has so far explored the relationship between melanoma prognosis and aging-related lncRNAs (ARLs). METHODS The Cancer Genome Atlas database, the GTEx database, and the HAGR database were used in this study in a combined manner. Univariate and multivariate cox regression analyses were used to screen out lncRNA signatures associated with overall survival (OS) in the primary dataset. The risk scoring model was analyzed by risk stratification and tested internally. The protein expression levels of possible target genes of ARLs were verified by immunohistochemistry analysis in HPA database. Finally, gene enrichment analysis was performed. RESULTS In the primary dataset, five OS-related lncRNA signatures (AC011481.1, USP30-AS1, EBLN3P, LINC01527, HLA-DQB1-AS1) were screened out. The survival curve showed that the high-risk group had a worse prognosis than the low-risk group. The immunohistochemical analysis revealed that reduced expression of Epidermal Growth Factor Receptor (EGFR), along with increased expression of Activating Transcription Factor 2 (ATF2) and DNA Polymerase Delta 1 (POLD1), was linked to a worse prognosis. Finally, enrichment analysis revealed that OS-related DELs were significantly enriched in the regulation of reactive oxygen metabolism, etc. The ARGs were significantly activated in the SKCM tissues. The regulation of aging in melanoma cells may be realized through ferroptosis, immunity, and autophagy and so on. CONCLUSION The ARL signature obtained in this study had better prognostic ability than individual clinical features.
Collapse
Affiliation(s)
- Nenghua Zhang
- Clinical Laboratory, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, 314033, China
| | - Xinyi Qiu
- The First School of Clinical Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xingying Chen
- Clinical Laboratory, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, 314033, China
| | - Cheng Du
- Ophthalmology Department, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, 314033, China
| | - Jingyi Dong
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaohong Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Bing Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lin Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yuyan Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
50
|
Li J, Jiang L, Ma Q, Zhang Z, Zheng S, Qiu J, Pang Y, Wang J. Evodiamine inhibits programmed cell death ligand 1 expression via the PI3K/AKT signaling pathway to regulate antitumor immunity in melanoma. Sci Rep 2025; 15:6649. [PMID: 39994441 PMCID: PMC11850830 DOI: 10.1038/s41598-025-91137-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
Malignant melanoma, a rare and aggressive skin cancer, arises from the transformation of cutaneous melanocytes and is associated with a poor prognosis. Evodiamine (EVO), a bioactive compound derived from traditional Chinese herbal medicine, has demonstrated significant inhibitory effects on various tumor cells. In this study, we aimed to investigate the potential of EVO in regulating melanoma immunity and elucidate its underlying mechanisms. Experimental results revealed that the IC50 value of EVO in B16-F10 cells for 24, 48, and 72 h were 11.73, 5.083, and 4.604 µM, respectively. EVO inhibited the proliferation, invasion, and metastasis of B16-F10 cells by more than 50%, while promoting apoptosis of higher concentration of EVO. EVO also significantly suppressed tumor growth by more than 80% and reduced spleen damage in tumor-bearing mice. Treatment with EVO led to a marked increase in T-cell subsets in the spleen, bone marrow, and tumors, with immunohistochemical (IHC) staining in particular showing about 50% higher. Furthermore, EVO inhibited the expression of programmed cell death ligand 1 (PD-L1) and the PI3K/AKT signaling pathway-related proteins in both B16-F10 cells and tumors. These findings suggest that EVO exerts antitumor effects by enhancing the tumor immune microenvironment and indicates its potential as a therapeutic agent for melanoma.
Collapse
Affiliation(s)
- Jiamin Li
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Li Jiang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Qianlong Ma
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Zhenglong Zhang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Shengping Zheng
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jing Qiu
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Yunqing Pang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China.
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China.
| | - Jing Wang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China.
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|