1
|
Zhao S, Chen Y, Bhojnagarwala PS, Livingston C, Jose J, Gao Y, Park DH, Herlyn M, Weiner DB. Targeting IL13Rα2 in melanoma with a bispecific T-cell engager: expression profiling and preclinical evaluation. J Immunother Cancer 2025; 13:e011073. [PMID: 40484645 PMCID: PMC12161313 DOI: 10.1136/jitc-2024-011073] [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: 11/15/2024] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Melanoma is a highly aggressive skin cancer, especially in advanced stages. While current treatments such as targeted therapies and immunotherapies have made significant progress, challenges like drug resistance and limited effectiveness in some patients persist. Therefore, ongoing development of novel therapies, particularly for late-stage melanoma, is crucial. METHODS In this study, we explored the expression of interleukin-13 receptor subunit alpha-2 (IL13Rα2) in melanoma patient-derived xenograft models. We investigated IL13Rα2 as a potential target for melanoma treatment by employing an IL13Rα2-CD3 bispecific T-cell engager (BTE). We tested the effect of IL13Rα2-CD3 BTE on T cell activity by flow cytometry. We studied the potency of IL13Rα2-CD3 BTE in tumor killing assay in vitro. For in vivo studies, we administered DNA expression cassettes encoding IL13Rα2-CD3 BTE (IL13Rα2-CD3 DNA encoding BTE (dBTE)) into immunodeficient mice for direct in vivo expression. The mice were challenged with A375 cells and then treated with IL-13Rα2-CD3 dBTE versus control and reconstituted with human peripheral blood mononuclear cells (PBMCs) or T cells. Tumor development was monitored, and T cell infiltration in the tumor was analyzed throughflow cytometry. RESULTS Our findings revealed heterogeneous expression of IL-13Rα2, particularly in samples from advanced stages of melanoma. The IL13Rα2-CD3 BTE facilitated T-cell activation and proliferation by bridging melanoma cells and T cells. We also observed the ability of IL13Rα2-CD3 BTE to direct T cells to kill multiple melanoma patient-derived cell lines through xCELLigence assay in vitro, including those with various mutations associated with late-stage metastatic melanoma. IL13Rα2-CD3 dBTE expressed in vivo led to notable tumor regression through inducing increased T-cell infiltration and activation within the tumor microenvironment. CONCLUSIONS These promising findings underscore the potential of targeting IL13Rα2 as a relevant target for the development of biologics including dBTE aimed at treating specific subsets of melanoma.
Collapse
Affiliation(s)
- Shushu Zhao
- Wistar Institute of Anatomy and Biology, Philadelphia, Pennsylvania, USA
| | - Yeqing Chen
- Wistar Institute of Anatomy and Biology, Philadelphia, Pennsylvania, USA
| | | | - Cory Livingston
- Wistar Institute of Anatomy and Biology, Philadelphia, Pennsylvania, USA
| | - Joshua Jose
- Wistar Institute of Anatomy and Biology, Philadelphia, Pennsylvania, USA
| | - Yangcheng Gao
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel H Park
- Wistar Institute of Anatomy and Biology, Philadelphia, Pennsylvania, USA
| | - Meenhard Herlyn
- Wistar Institute of Anatomy and Biology, Philadelphia, Pennsylvania, USA
| | - David B Weiner
- Wistar Institute of Anatomy and Biology, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Tovar-Parra D, Gutiérrez-Castañeda LD. Polygenic Risk Score Analysis of 37 SNPs Associated with Melanoma Risk in Colombian Population. Int J Mol Sci 2025; 26:4674. [PMID: 40429816 PMCID: PMC12112468 DOI: 10.3390/ijms26104674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/13/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Melanoma incidence is increasing, with distinct genetic and clinical patterns observed in the Latin American population. This study aimed to evaluate melanoma risk in a Colombian cohort through polygenic risk analysis using 37 variants across nine genes previously associated with melanoma. We performed polygenic risk score (PRS) analysis on 85 melanoma patients and 165 controls. Genotyping was performed for 37 melanoma-associated SNPs, and on the basis of previous GWAS reports, individual PRSs were calculated for each participant. The participants were then stratified into quartiles to examine risk gradients. In addition, phenotypic features such as eye and hair color were evaluated, and genetic models and haplotype analyses were performed, adjusting for sex and family history of cancer. PRS quartile stratification revealed a clear risk gradient. Notably, 31.8% of the melanoma cases were clustered in the highest-risk quartile (Q4), with a maximum PRS of 1.04. Variants in TYR, TYRP1, CDKN2A, and HERC2 significantly contributed to risk, and light brown eye and hair colors were strongly associated with increased melanoma risk. Moreover, a protective haplotype in the OCA2-HERC2 region was identified among males. The integration of the PRS with clinical and phenotypic factors has potential for improving melanoma risk stratification in the Colombian population, warranting further investigation in larger, diverse cohorts.
Collapse
Affiliation(s)
- David Tovar-Parra
- General Dermatology Group, Hospital Universitario Centro Dermatologico Federico Lleras Acosta E.S.E, Bogotá 111511, Colombia;
- Institut National de la Recherche Scientifique INRS, Centre Armand-Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada
| | - Luz Dary Gutiérrez-Castañeda
- General Dermatology Group, Hospital Universitario Centro Dermatologico Federico Lleras Acosta E.S.E, Bogotá 111511, Colombia;
- Research Institute, Basic Health Sciences Group, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá 111221, Colombia
| |
Collapse
|
3
|
Dycha N, Michalak-Tomczyk M, Jachuła J, Okoń E, Jarząb A, Tokarczyk J, Koch W, Gaweł-Bęben K, Kukula-Koch W, Wawruszak A. Chemopreventive and Anticancer Activity of Selected Triterpenoids in Melanoma. Cancers (Basel) 2025; 17:1625. [PMID: 40427124 PMCID: PMC12109728 DOI: 10.3390/cancers17101625] [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: 04/02/2025] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Melanoma is one of the most aggressive forms of skin cancer, characterized by high metastatic potential and resistance to conventional therapies. Natural compounds, particularly terpenoids, have gained attention for their chemopreventive potential and anticancer properties. These plant-derived compounds exhibit diverse biological activities, e.g., cell viability and proliferation inhibition, apoptosis induction, cell cycle regulation, and immune system modulation. The review evaluates the current state of the art on the chemopreventive and anticancer activity of lupane- (betulinic acid), oleanane- (oleanolic acid, β-amyrin, escin, hederagenin, glycyrrhetinic acid), and ursane-type (ursolic acid, asiatic acid, madecassic acid, α-amyrin) triterpenoids in melanoma, highlighting their mechanisms of action, therapeutic potential, and challenges in clinical application.
Collapse
Affiliation(s)
- Natalia Dycha
- Department of Pharmacognosy with Medical Plants Garden, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland; (N.D.); (W.K.-K.)
| | - Magdalena Michalak-Tomczyk
- Department of Physiology and Toxicology, The John Paul II Catholic University of Lublin, 1I Konstantynów Str., 20-708 Lublin, Poland;
| | - Jacek Jachuła
- Department of Botany, Mycology and Ecology, Institute of Biological Sciences, Maria Curie-Skłodowska University, 19 Akademicka Str., 20-033 Lublin, Poland;
| | - Estera Okoń
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland; (E.O.); (A.J.)
| | - Agata Jarząb
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland; (E.O.); (A.J.)
| | - Joanna Tokarczyk
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodźki Str., 20-093 Lublin, Poland; (J.T.); (W.K.)
| | - Wojciech Koch
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodźki Str., 20-093 Lublin, Poland; (J.T.); (W.K.)
| | - Katarzyna Gaweł-Bęben
- Department of Cosmetology, University of Information Technology and Management in Rzeszów, 2 Sucharskiego, 35-225 Rzeszów, Poland;
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy with Medical Plants Garden, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland; (N.D.); (W.K.-K.)
| | - Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland; (E.O.); (A.J.)
| |
Collapse
|
4
|
Unnisa Z, Tariq A, Sarwar N, Din I, Serhani MA, Trabelsi Z. Impact of fine-tuning parameters of convolutional neural network for skin cancer detection. Sci Rep 2025; 15:14779. [PMID: 40295678 PMCID: PMC12037876 DOI: 10.1038/s41598-025-99529-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
Melanoma skin cancer is a deadly disease with a high mortality rate. A prompt diagnosis can aid in the treatment of the disease and potentially save the patient's life. Artificial intelligence methods can help diagnose cancer at a rapid speed. The literature has employed numerous Machine Learning (ML) and Deep Learning (DL) algorithms to detect skin cancer. ML algorithms perform well for small datasets but cannot comprehend larger ones. Conversely, DL algorithms exhibit strong performance on large datasets but misclassify when applied to smaller ones. We conduct extensive experiments using a convolutional neural network (CNN), varying its parameter values to determine which set of values yields the best performance measure. We discovered that adding layers, making each Conv2D layer have multiple filters, and getting rid of dropout layers greatly improves the accuracy of the classifiers, going from 62.5% to 85%. We have also discussed the parameters that have the potential to significantly impact the model's performance. This shows how powerful it is to fine-tune the parameters of a CNN-based model. These findings can assist researchers in fine-tuning their CNN-based models for use with skin cancer image datasets.
Collapse
Affiliation(s)
- Zaib Unnisa
- Department of Computer Science and Information Technology, Superior University, Lahore, 54670, Pakistan
| | - Asadullah Tariq
- College of IT, United Arab Emirates University, 15551, Al Ain, United Arab Emirates
| | - Nadeem Sarwar
- Department of Computer Science, Bahria University, Lahore, Pakistan
| | - Irfanud Din
- Department of Computer Science, New Uzbekistan University, Tashkent, Uzbekistan.
| | | | - Zouheir Trabelsi
- College of IT, United Arab Emirates University, 15551, Al Ain, United Arab Emirates.
| |
Collapse
|
5
|
Hussain Y, You BG, Huang L, Liu X, Dormocara A, Shah KA, Ali T, Cao QR, Lee BJ, Elbehairi SEI, Iqbal H, Cui JH. Dissolving microneedles for melanoma: Most recent updates, challenges, and future perspectives. Int J Pharm 2025; 673:125382. [PMID: 39988214 DOI: 10.1016/j.ijpharm.2025.125382] [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: 09/15/2024] [Revised: 02/08/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Skin cancer is one among the common types of cancers, affecting millions of individual globally. The conventional anticancer therapy such as chemotherapy results in worst systemic and local side effects as well as inhibit the growth of healthy cells around the tumor cells. Dissolving microneedles (DMNs) is a groundbreaking technology with less invasive and more targeted features. Physically, these tiny dissolving needles deliver the anticancer payloads drug to the tumor site after its direct application on the skin surface. Specifically, the DMNs release the anticancer drug cargoes into the cancerous cell sparing the healthy cells around the tumor, thus has provided a significant contribution in the landscape of traditional skin cancer therapy. This targeted therapeutic approach of dissolving microneedles shows a significant therapeutic outcome in decreasing the growth of cancer cells in pre-clinical studies. Dissolving microneedles (DMNs) have demonstrated effectiveness in the targeted delivery of drugs, genes, and vaccines specifically at the site of skin tumors. This method mimics the localized release of adjuvants and immunomodulators, leading to significant humoral and cellular immune responses that are beneficial for skin cancer therapy. In this review, the current trends and potential roles of dissolving microneedles in delivering therapeutic agents focused on treating skin melanoma have been highlighted, drawing insights from recent literature. This emphasizes the promising applications of DMNs in enhancing treatment outcomes for skin cancer patients. Lastly, future perspectives were identified for improving the therapeutic potential and translation of DMNs into clinic.
Collapse
Affiliation(s)
- Yaseen Hussain
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ben-Gang You
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Linyu Huang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Xiaoyin Liu
- School of Radiation Medicine and Protection of Soochow University, Suzhou 215123, China
| | - Amos Dormocara
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Kiramat Ali Shah
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Tariq Ali
- Department of Civil and Environmental Engineering, Shantou University, Shantou, Guangdong 515063, China
| | - Qing-Ri Cao
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Beom-Jin Lee
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | | | - Haroon Iqbal
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China.
| | - Jing-Hao Cui
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
6
|
Li L, Xu Q, Zhang X, Jiang Y, Zhang L, Guo J, Liu H, Jiang B, Li S, Peng Q, Jiang N, Wang J. AIEgen-self-assembled nanoparticles with anti-PD-L1 antibody functionalization realize enhanced synergistic photodynamic therapy and immunotherapy against malignant melanoma. Mater Today Bio 2025; 30:101387. [PMID: 39742147 PMCID: PMC11683329 DOI: 10.1016/j.mtbio.2024.101387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/17/2024] [Accepted: 12/05/2024] [Indexed: 01/03/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) become integral in clinical practice, yet their application in cancer therapy is constrained by low overall response rates and the primary resistance of cancers to ICIs. Herein, this study proposes aggregation-induced emission (AIE)-based nanoparticles (NPs) for a more effective and synergistic approach combining immunotherapy and photodynamic therapy (PDT) to achieve higher responses than anti-PD-L1 monotherapy. The TBP@aPD-L1 NPs are constructed by functionalizing azide group-modified TBP-2 (TBP-N3) with anti-PD-L1 antibodies via the DBCO-S-S-PEG2000-COOH linker. The anti-PD-L1 target the tumor cells and promote the TBP-N3 accumulation in tumors for enhanced PDT. Notably, the TBP-N3, featuring aggregation-induced emission, boosts reactive oxygen species (ROS) generation through both type I and type II processes for enhanced PDT. The TBP@aPD-L1-mediated PDT induces more powerful effects of direct tumor cell-killing and further elicits effective immunogenic cell death (ICD), which exerts anti-tumor immunity by activating T cells for ICI treatment and reshapes the tumor immune microenvironment (TIME), thereby enhancing the efficacy of PD-L1 blockade of anti-PD-L1. Consequently, TBP@aPD-L1 NPs demonstrated significantly enhanced inhibition of tumor growth in the mouse model of malignant melanoma (MM). Our NPs act as a facile and effective drug delivery platform for enhanced immunotherapy combined with enhanced PDT in treating MM.
Collapse
Affiliation(s)
- Lu Li
- Department of Immunology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Qing Xu
- Department of Immunology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xiuzhen Zhang
- Hunan University of Medicine General Hospital, Hunan, 418000, PR China
| | - Yuan Jiang
- Department of Immunology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - La Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Jiao Guo
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Haichuan Liu
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Bin Jiang
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Shenglong Li
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Qiling Peng
- Bijie Municipal Health Bureau, Guizhou, 551700, PR China
| | - Ning Jiang
- Department of Pathology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, 400016, PR China
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Jianwei Wang
- Department of Immunology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| |
Collapse
|
7
|
Lee SH, Park GS, Lee R, Hong S, Han S, Lee YM, Nah SY, Han SG, Oh JW. Gintonin-Enriched Panax ginseng Extract Induces Apoptosis in Human Melanoma Cells by Causing Cell Cycle Arrest and Activating Caspases. Foods 2025; 14:381. [PMID: 39941974 PMCID: PMC11816885 DOI: 10.3390/foods14030381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Gintonin, a non-saponin glycolipoprotein from Panax ginseng, acts as a lysophosphatidic acid ligand. However, its anticancer effects, especially in melanoma, remain unclear. This study investigated the anti-proliferative effects and intracellular signaling mechanisms of a gintonin-enriched fraction (GEF) from Panax ginseng in human melanoma cell lines. In vitro, GEF treatment significantly inhibited cell proliferation, reduced clonogenic potential, and delayed wound healing in melanoma cells. Flow cytometry and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining showed that GEF induced apoptosis, as evidenced by increased apoptotic cell populations and nuclear changes. GEF also caused cell cycle arrest in the G1 phase for A375 cells and the G2/M phase for A2058 cells. It triggered apoptotic signaling via activation of caspase-3, -9, poly (ADP-ribose) polymerase cleavage, and downregulation of B cell lymphoma-2 (Bcl-2). GEF treatment also raised intracellular reactive oxygen species (ROS) levels and mitochondrial stress, which were mitigated by N-acetyl cysteine (NAC), an ROS inhibitor. In vivo, GEF suppressed tumor growth in A375- and A2058-xenografted mice without toxicity. These findings suggest that GEF from Panax ginseng has potential antitumor effects in melanoma by inducing apoptosis and cell cycle arrest, presenting a promising therapeutic avenue.
Collapse
Affiliation(s)
- Su-Hyun Lee
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (S.-H.L.); (S.H.); (S.H.); (Y.-M.L.)
| | - Gyun-Seok Park
- Department of Bio-Resources and Food Science, Konkuk University, 120 Neungdong-ro, Gwangjn-gu, Seoul 05029, Republic of Korea;
| | - Rami Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (R.L.); (S.-Y.N.)
| | - Seongwoo Hong
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (S.-H.L.); (S.H.); (S.H.); (Y.-M.L.)
| | - Sumin Han
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (S.-H.L.); (S.H.); (S.H.); (Y.-M.L.)
| | - Yoon-Mi Lee
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (S.-H.L.); (S.H.); (S.H.); (Y.-M.L.)
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (R.L.); (S.-Y.N.)
| | - Sung-Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Jae-Wook Oh
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (S.-H.L.); (S.H.); (S.H.); (Y.-M.L.)
| |
Collapse
|
8
|
Lyu Z, Mahenderan A, Radhakrishnan AKGK, Chin YS, Yin C. Swimming upregulates APOL3 through regulating macrophage polarization to inhibit glycolysis and the development of melanoma. 3 Biotech 2024; 14:307. [PMID: 39583207 PMCID: PMC11582285 DOI: 10.1007/s13205-024-04150-z] [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: 08/29/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
This study investigated the role of swimming exercise in regulating melanoma tumour growth and glycolysis in cancer cells, the specific mechanism involved was also studied. In our study, a murine melanoma tumour model was established to assess the impact of swimming on tumour growth. The mRNA and protein expressions were assessed using qRT-PCR, western blot, and IHC. The metabolic behavior of melanoma cells was examined through lactic acid level measurements and glucose consumption assessments. CCK-8 and colony formation assays were used to detect cell viability and proliferation. ELISA was employed to determine the levels of cytokines secreted by macrophages. The interaction between APOL3 and STAT3 was analyzed by dual luciferase reporter gene and ChIP assays. Our results demonstrated that swimming exercise suppressed melanoma growth in mice by suppressing glycolysis, which might be related to APOL3 upregulation. In addition, downregulation of APOL3 in melanoma was associated with poor prognosis, and APOL3 overexpression markedly suppressed melanoma cell proliferation by reducing glucose uptake and lactate production in vitro. Mechanistically, STAT3 directly down-regulated APOL3 transcription. Swimming upregulated APOL3 by inactivating the IL-6R-STAT3 signaling axis in melanoma cells by inhibiting the secretion of IL-6 by M2 macrophages. As expected, IL-6 secreted by M2 macrophages promoted glycolysis in melanoma cells by reducing APOL3 expression. In summary, swimming inactivated the IL-6R/STAT3 signaling axis in melanoma cells by inhibiting the secretion of IL-6 by M2 macrophages, which could suppress the growth of melanoma in the body by upregulating APOL3 to inhibit glycolysis.
Collapse
Affiliation(s)
- Zhenlei Lyu
- Hainan Vocational University of Science and Technology, College of Medicine, No.18 Qiongshan Avenue, Meilan District, Haikou, 571100 Hainan People’s Republic of China
- Sports Science Programme, Faculty of Sports Science and Recreation, Universiti Teknologi MARA, 40450 Selangor, Shah Alam Malaysia
| | - Appukutty Mahenderan
- Sports Science Programme, Faculty of Sports Science and Recreation, Universiti Teknologi MARA, 40450 Selangor, Shah Alam Malaysia
| | | | - Yit Siew Chin
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang Selangor Malaysia, 43400 Seri Kembangan, Malaysia
| | - Chao Yin
- Hainan Vocational University of Science and Technology, College of Medicine, No.18 Qiongshan Avenue, Meilan District, Haikou, 571100 Hainan People’s Republic of China
| |
Collapse
|
9
|
Huang J, Gao Z, Xuan J, Gao N, Wei C, Gu J. Metabolic insights into tumor lymph node metastasis in melanoma. Cell Oncol (Dordr) 2024; 47:2099-2112. [PMID: 39704926 DOI: 10.1007/s13402-024-01027-4] [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] [Accepted: 12/08/2024] [Indexed: 12/21/2024] Open
Abstract
Although accounting for only a small amount of skin cancers, melanoma contributes prominently to skin cancer-related deaths, which are mostly caused by metastatic diseases, and lymphatic metastasis constitutes the main route. In this review, we concentrate on the metabolic mechanisms of tumor lymph node (LN) metastasis in melanoma. Two hypotheses of melanoma LN metastasis are introduced, which are the premetastatic niche (PMN) and parallel progression model. Dysregulation of oxidative stress, lactic acid concentration, fatty acid synthesis, amino acid metabolism, autophagy, and ferroptosis construct the metabolic mechanisms in LN metastasis of melanoma. Moreover, melanoma cells also promote LN metastasis by interacting with non-tumor cells through metabolic reprogramming in TIME. This review will deepen our understanding of the mechanism of lymph node metastasis in melanoma.
Collapse
Affiliation(s)
- Jiayi Huang
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Zixu Gao
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Jiangying Xuan
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Ningyuan Gao
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Chuanyuan Wei
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.
| | - Jianying Gu
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.
| |
Collapse
|
10
|
Mathlouthi S, Kuryk L, Prygiel M, Lupo MG, Zasada AA, Pesce C, Ferri N, Rinner B, Salmaso S, Garofalo M. Extracellular vesicles powered cancer immunotherapy: Targeted delivery of adenovirus-based cancer vaccine in humanized melanoma model. J Control Release 2024; 376:777-793. [PMID: 39481685 DOI: 10.1016/j.jconrel.2024.10.057] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Malignant melanoma, a rapidly spreading form of skin cancer, is becoming more prevalent worldwide. While surgery is successful in treating early-stage melanoma, patients with advanced disease have only a 20 % chance of surviving beyond five years. Melanomas with mutations in the NRAS gene are characterized for a more aggressive tumor biology, poorer prognosis and shorter survival. Hence, new therapeutic strategies are needed, especially for this specific group of patients. Novel approaches, such as cancer vaccines, offer promising solutions by stimulating the anti-tumor immune response. Nevertheless, their clinical efficacy is still modest and more effective approaches are required. Herein, we propose the systemic administration of the adenovirus-based cancer vaccine complexed in extracellular vesicles (EVs) with the aim of achieving a targeted therapeutic effect. The vaccine was based on previously tested oncolytic adenovirus Ad5/3-D24-ICOSL-CD40L in combination with melanoma-specific antigens targeting NRAS mutations to enhance the anticancer effect. The antineoplastic properties of the oncolytic vaccine were evaluated in xenograft MUG Mel-2 melanoma BALB/c nude mice. Moreover, to mimic the tumor microenvironment, while investigating at the same time immune cell infiltration and drug penetration, we established a 3D co-culture model based on human NRAS mutated MUG Mel-2 spheroids and PBMCs (HLA matched), which displayed a synergistic effect when treated with the cancer vaccine compared to relative controls. Subsequently, we investigated the systemic delivery of the vaccine in EV formulations in a humanized NSG MUG Mel-2 melanoma mouse model. Our study provides a promising strategy for a tumor-targeted vaccine delivery by EVs, resulting in improved anticancer efficacy and increased infiltration of tumor-infiltrating lymphocytes. This study explores the potential of EVs for the selective delivery of cancer vaccines against malignancies, such as NRAS melanoma. Overall, this research could pave the way for applying autologous EVs as a safe and efficacious tool for targeted cancer therapy.
Collapse
Affiliation(s)
- Sara Mathlouthi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Lukasz Kuryk
- Department of Virology, National Institute of Public Health NIH - National Research Institute, Chocimska 24, 00-791 Warsaw, Poland
| | - Marta Prygiel
- Department of Virology, National Institute of Public Health NIH - National Research Institute, Chocimska 24, 00-791 Warsaw, Poland
| | - Maria Giovanna Lupo
- Department of Medicine, University of Padova, Via Giustiniani 2, Padua 35131, Italy
| | - Aleksandra Anna Zasada
- Department of Virology, National Institute of Public Health NIH - National Research Institute, Chocimska 24, 00-791 Warsaw, Poland
| | - Cristiano Pesce
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Nicola Ferri
- Department of Medicine, University of Padova, Via Giustiniani 2, Padua 35131, Italy; Veneto Institute of Molecular Medicine, Via Orus 2, 35131 Padua, Italy
| | - Beate Rinner
- Division of Biomedical Research, Medical University of Graz, Roseggerweg 48, 8036 Graz, Austria
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy.
| |
Collapse
|
11
|
Zhang L, Khoo CS, Xiahou Z, Reddy N, Li Y, Lv J, Sun M, Fan H, Zhang X. Antioxidant and anti-melanogenesis activities of extracts from Leonurus japonicus Houtt. Biotechnol Genet Eng Rev 2024; 40:2888-2909. [PMID: 37066895 DOI: 10.1080/02648725.2023.2202544] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/10/2023] [Indexed: 04/18/2023]
Abstract
Leonurus japonicus Houtt is an important anti-skin pigmentation herb used in traditional Chinese medicine. However, the molecular basis for this activity is complex and not fully understood. In this study, water and ethanol extracts and polysaccharide extract from L. japonicus (LJPs) were analyzed by LC-MS/MS and HPLC-DAD separately. Cytotoxicity was analyzed by using CCK-8, antioxidant activity using flow cytometer, anti-MMPs, anti-tyrosinase and signalling pathway analysis using Western blotting to investigate their anti-melanogenesis function. The results showed that the water and ethanol extracts contained alkaloids, flavonoids, and phenolic acids. The LJPs mainly contain glucose, fucose, glucuronic acid, mannose, threonine and arginine, and structure characterization by FITR analyses indicated that LJPs have β- or α-D-glycosidic bonds and contain pyranose rings. The L. japonicus extracts displayed high cell viability at their maximum concentration. The water extract and polysaccharides significantly reduced lipopolysaccharide (LPS)-induced intracellular reactive oxygen species (ROS) content and exhibited a cytoprotective role. Also, these extracts displayed higher matrix metalloproteinase-2 (anti-MMP-2), anti-MMP-9 and anti-tyrosinase activities. Furthermore, the polysaccharides displayed significantly greater inhibitory effect on intracellular ROS and tyrosinase protein expression than α-arbutin and ursolic acid used for the clinical treatment of skin pigmentation. This study also investigated the polysaccharide inhibition of melanin synthesis by repressing the expression of melanocytic lineage-specific transcription factor (MITF) and melanogenic enzymes via modulation of the phosphoinositide 3-kinase (PI3K-Akt-mTOR) and β-catenin pathways. The overall results indicate that L. japonicus is a promising candidate for anti-pigmentation treatment.
Collapse
Affiliation(s)
- Lin Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Institute of Chinese Medicine, Beijing, China
| | | | - ZhiKai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Narsimha Reddy
- School of Science, Parramatta Campus, Western Sydney University, Penrith, Australia
| | - Yue Li
- Department of Instrument Management & Analysis, Beijing Institute for Drug Control, Beijing, China
| | - Jingjing Lv
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Meihe Sun
- Yuzhou Lianyun Technology Co. Ltd, Henan, China
| | - Heming Fan
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Institute of Chinese Medicine, Beijing, China
| | - Xian Zhang
- Department of Instrument Management & Analysis, Beijing Port Drug Inspection Institute of The People's Republic of China, Beijing, China
- Beijing Institute for Drug Control, Beijing, China
- Beijing Center for Vaccine Control, Beijing, China
| |
Collapse
|
12
|
Teixeira MC, Lameirinhas NS, Carvalho JPF, Luís J, Oliveira H, Oliveira JM, Silvestre AJD, Vilela C, Freire CSR. Biobased hydrogel bioinks of pectin, nanocellulose and lysozyme nanofibrils for the bioprinting of A375 melanoma cell-laden 3D in vitro platforms. Int J Biol Macromol 2024; 282:136958. [PMID: 39490881 DOI: 10.1016/j.ijbiomac.2024.136958] [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: 06/30/2024] [Revised: 09/24/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Melanoma is one of the most aggressive types of skin cancer, and the need for advanced platforms to study this disease and to develop new treatments is rising. 3D bioprinted tumor models are emerging as advanced tools to tackle these needs, with the design of adequate bioinks being a fundamental step to address this challenging process. Thus, this work explores the synergy between two biobased nanofibers, nanofibrillated cellulose (NFC) and lysozyme amyloid nanofibrils (LNFs), to create pectin nanocomposite hydrogel bioinks for the 3D bioprinting of A375 melanoma cell-laden living constructs. The incorporation of LNFs (5, 10 or 15 wt%) on a pectin-NFC suspension originates inks with enhanced rheological properties (shear viscosity and yield point) and proper shear-thinning behavior. The crosslinked hydrogels mimic the stiffness of melanoma tissues, being stable under physiological and cell-culture conditions, and non-cytotoxic towards A375 melanoma cells. P-NFC-LNFs (10 %) reveals good printability (Pr = 0.89) and printing accuracy (51 ± 2 %), and when loaded with A375 cells (3 × 106 cells mL-1) the bioink originates 3D-constructs with high cell viability (92 ± 1 %) after 14 days. The potential of the constructs as 3D in vitro platforms is corroborated by a drug-screening test with doxorubicin, where cells within the model displayed high sensitivity to the drug.
Collapse
Affiliation(s)
- Maria C Teixeira
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Nicole S Lameirinhas
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - João P F Carvalho
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Jorge Luís
- CICECO - Aveiro Institute of Materials, EMaRT Group - Emerging: Materials, Research, Technology, School of Design, Management and Production Technologies Northern Aveiro, University of Aveiro, 3720-509 Oliveira de Azeméis, Portugal
| | - Helena Oliveira
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | - José Martinho Oliveira
- CICECO - Aveiro Institute of Materials, EMaRT Group - Emerging: Materials, Research, Technology, School of Design, Management and Production Technologies Northern Aveiro, University of Aveiro, 3720-509 Oliveira de Azeméis, Portugal
| | - Armando J D Silvestre
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Carla Vilela
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Carmen S R Freire
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
13
|
Aguilera-Durán G, Hernández-Castro S, Loera-García BV, Rivera-Vargas A, Alvarez-Baltazar JM, Cuevas-Flores MDR, Romo-Mancillas A. Ursolic acid interaction with transcription factors BRAF, V600E, and V600K: a computational approach towards new potential melanoma treatments. J Mol Model 2024; 30:373. [PMID: 39387972 DOI: 10.1007/s00894-024-06165-y] [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: 05/21/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
CONTEXT Melanoma is one of the cancers with the highest mortality rate for its ability to metastasize. Several targets have undergone investigation for the development of drugs against this pathology. One of the main targets is the kinase BRAF (RAF, rapidly accelerated fibrosarcoma). The most common mutation in melanoma is BRAFV600E and has been reported in 50-90% of patients with melanoma. Due to the relevance of the BRAFV600E mutation, inhibitors to this kinase have been developed, vemurafenib-OMe and dabrafenib. Ursolic acid (UA) is a pentacyclic triterpene with a privileged structure, the pentacycle scaffold, which allows to have a broad variety of biological activity; the most studied is its anticancer capacity. In this work, we reported the interaction profile of vemurafenib-OMe, dabrafenib, and UA, to define whether UA has binding capacity to BRAFWT, BRAFV600E, and BRAFV600K. Homology modeling of BRAFWT, V600E, and V600K; molecular docking; and molecular dynamics simulations were carried out and interactions and residues relevant to the binding of the inhibitors were obtained. We found that UA, like the inhibitors, presents hydrogen bond interactions, and hydrophobic interactions of van der Waals, and π-stacking with I463, Q530, C532, and F583. The ΔG of ursolic acid in complex with BRAFV600K (- 63.31 kcal/mol) is comparable to the ΔG of the selective inhibitor dabrafenib (- 63.32 kcal/mol) in complex to BRAFV600K and presents a ΔG like vemurafenib-OMe with BRAFWT and V600E. With this information, ursolic acid could be considered as a lead compound for design cycles and to optimize the binding profile and the selectivity towards mutations for the development of new selective inhibitors for BRAFV600E and V600K to new potential melanoma treatments. METHODS The homology modeling calculations were executed on the public servers I-TASSER and ROBETTA, followed by molecular docking calculations using AutoGrid 4.2.6, AutoDockGPU 1.5.3, and AutoDockTools 1.5.6. Molecular dynamics and metadynamics simulations were performed in the Desmond module of the academic version of the Schrödinger-Maestro 2020-4 program, utilizing the OPLS-2005 force field. Ligand-protein interactions were evaluated using Schrödinger-Maestro program, LigPlot + , and PLIP (protein-ligand interaction profiler). Finally, all of the protein figures presented in this article were made in the PyMOL program.
Collapse
Affiliation(s)
- Giovanny Aguilera-Durán
- Laboratorio de Química Cuántica y Modelado Molecular, Unidad Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, 98160, Zacatecas, Mexico.
- Grupo de Diseño Asistido Por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, 76010, Querétaro, Mexico.
| | - Stephanie Hernández-Castro
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de Las Campanas S/N, 76010, Querétaro, Mexico
- Grupo de Diseño Asistido Por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, 76010, Querétaro, Mexico
| | - Brenda V Loera-García
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Zona Universitaria, 78210, San Luis Potosí, Mexico
| | - Alex Rivera-Vargas
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de Las Campanas S/N, 76010, Querétaro, Mexico
- Grupo de Diseño Asistido Por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, 76010, Querétaro, Mexico
| | - J M Alvarez-Baltazar
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de Las Campanas S/N, 76010, Querétaro, Mexico
- Grupo de Diseño Asistido Por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, 76010, Querétaro, Mexico
| | - Ma Del Refugio Cuevas-Flores
- Laboratorio de Química Cuántica y Modelado Molecular, Unidad Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, 98160, Zacatecas, Mexico
| | - Antonio Romo-Mancillas
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de Las Campanas S/N, 76010, Querétaro, Mexico.
- Grupo de Diseño Asistido Por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, 76010, Querétaro, Mexico.
| |
Collapse
|
14
|
Rajabally FZ, Flynn J, Abbasakoor F, Ghanty S. Dysphagia in an 80-Year-Old Woman: A Rare Case of Metastatic Melanoma Presenting in the Small Bowel. Cureus 2024; 16:e71373. [PMID: 39534845 PMCID: PMC11556734 DOI: 10.7759/cureus.71373] [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] [Accepted: 10/13/2024] [Indexed: 11/16/2024] Open
Abstract
This report discusses the case of an 80-year-old female presenting with non-specific gastrointestinal symptoms and who was ultimately diagnosed as having metastatic melanoma of the jejunum. Notably, on admission, the patient had failed to report a past medical history of melanoma on her back a few years prior, and this significantly impacted the diagnostic process. This case highlights the challenges in diagnosing metastatic melanoma. It adds to the literature by underscoring the rarity and diagnostic complexity of this condition, which in this particular case presented as a jejunal mass.
Collapse
Affiliation(s)
| | - Jonathan Flynn
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, IRL
| | | | - Shabir Ghanty
- Radiology, Clinic Darné Floréal C-Care, Floréal, MUS
| |
Collapse
|
15
|
Russano F, Rastrelli M, Dall'Olmo L, Del Fiore P, Gianesini C, Vecchiato A, Mazza M, Tropea S, Mocellin S. Therapeutic Treatment Options for In-Transit Metastases from Melanoma. Cancers (Basel) 2024; 16:3065. [PMID: 39272923 PMCID: PMC11394241 DOI: 10.3390/cancers16173065] [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: 08/08/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
In-transit metastases (ITM) in melanoma present a significant therapeutic challenge due to their advanced stage and complex clinical nature. From traditional management with surgical resection, ITM treatment has evolved with the advent of systemic therapies such as immune checkpoint inhibitors and targeted therapies, which have markedly improved survival outcomes. This study aims to review and highlight the efficacy of both systemic and locoregional treatment approaches for ITM. Methods include a comprehensive review of clinical studies examining the impact of treatments like immune checkpoint inhibitors, targeted therapies, Isolated Limb Perfusion, and electrochemotherapy. The results indicate that combining systemic therapies with locoregional treatments enhances both local disease control and overall survival rates. The introduction of modern immunotherapies has not diminished the effectiveness of locoregional therapies but rather improved patient outcomes when used in conjunction. The conclusions emphasize that a multidisciplinary approach integrating systemic and locoregional therapies offers a promising strategy for optimizing the management of ITM in melanoma patients. This integrated treatment model not only improves survival rates but also enhances the quality of life for patients, suggesting a shift in standard care practices toward more comprehensive therapeutic regimens.
Collapse
Affiliation(s)
- Francesco Russano
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Marco Rastrelli
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
- Department of Surgical, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, 35128 Padova, Italy
| | - Luigi Dall'Olmo
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
- Department of Surgical, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, 35128 Padova, Italy
| | - Paolo Del Fiore
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Carlomaria Gianesini
- Department of Surgical, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, 35128 Padova, Italy
| | - Antonella Vecchiato
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Marcodomenico Mazza
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Saveria Tropea
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Simone Mocellin
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
- Department of Surgical, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, 35128 Padova, Italy
| |
Collapse
|
16
|
Kolathur KK, Nag R, Shenoy PV, Malik Y, Varanasi SM, Angom RS, Mukhopadhyay D. Molecular Susceptibility and Treatment Challenges in Melanoma. Cells 2024; 13:1383. [PMID: 39195270 PMCID: PMC11352263 DOI: 10.3390/cells13161383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 08/29/2024] Open
Abstract
Melanoma is the most aggressive subtype of cancer, with a higher propensity to spread compared to most solid tumors. The application of OMICS approaches has revolutionized the field of melanoma research by providing comprehensive insights into the molecular alterations and biological processes underlying melanoma development and progression. This review aims to offer an overview of melanoma biology, covering its transition from primary to malignant melanoma, as well as the key genes and pathways involved in the initiation and progression of this disease. Utilizing online databases, we extensively explored the general expression profile of genes, identified the most frequently altered genes and gene mutations, and examined genetic alterations responsible for drug resistance. Additionally, we studied the mechanisms responsible for immune checkpoint inhibitor resistance in melanoma.
Collapse
Affiliation(s)
- Kiran Kumar Kolathur
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences (MCOPS), Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India;
| | - Radhakanta Nag
- Department of Microbiology, College of Basic Science & Humanities, Odisha University of Agriculture & Technology (OUAT), Bhubaneswar 751003, Odisha, India;
| | - Prathvi V Shenoy
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences (MCOPS), Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India; (P.V.S.); (Y.M.)
| | - Yagya Malik
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences (MCOPS), Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India; (P.V.S.); (Y.M.)
| | - Sai Manasa Varanasi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (S.M.V.); (R.S.A.)
| | - Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (S.M.V.); (R.S.A.)
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (S.M.V.); (R.S.A.)
| |
Collapse
|
17
|
Pierce ES, Jindal C, Choi YM, Cassidy K, Efird JT. Pathogenic mechanisms and etiologic aspects of Mycobacterium avium subspecies paratuberculosis as an infectious cause of cutaneous melanoma. MEDCOMM - ONCOLOGY 2024; 3:e72. [PMID: 38831791 PMCID: PMC11145504 DOI: 10.1002/mog2.72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/17/2024] [Indexed: 06/05/2024]
Abstract
Infectious etiologies have previously been proposed as causes of both melanoma and non-melanoma skin cancer. This exploratory overview explains and presents the evidence for the hypothesis that a microorganism excreted in infected ruminant animal feces, Mycobacterium avium subspecies paratuberculosis (MAP), is the cause of some cases of cutaneous melanoma (CM). Occupational, residential, and recreational contact with MAP-contaminated feces, soil, sand, and natural bodies of water may confer a higher rate of CM. Included in our hypothesis are possible reasons for the differing rates and locations of CM in persons with white versus nonwhite skin, why CM develops underneath nails and in vulvar skin, why canine melanoma is an excellent model for human melanoma, and why the Bacille Calmette-Guérin (BCG) vaccine has demonstrated efficacy in the prevention and treatment of CM. The pathogenic mechanisms and etiologic aspects of MAP, as a transmittable agent underlying CM risk, are carefully deliberated in this paper. Imbalances in gut and skin bacteria, genetic risk factors, and vaccine prevention/therapy are also discussed, while acknowledging that the evidence for a causal association between MAP exposure and CM remains circumstantial.
Collapse
Affiliation(s)
- Ellen S. Pierce
- Independent Physician Researcher, Spokane Valley, Washington, USA
| | - Charulata Jindal
- School of Medicine and Public Health, University of Sydney, Sydney, New South Wales, Australia
| | - Yuk Ming Choi
- Provider Services, Signify Health, Dallas, Texas, USA
| | - Kaitlin Cassidy
- VA Boston Healthcare System, Cooperative Studies Program Coordinating Center, Boston, Massachusetts, USA
| | - Jimmy T. Efird
- VA Boston Healthcare System, Cooperative Studies Program Coordinating Center, Boston, Massachusetts, USA
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
18
|
Ren Z, Xu Z, Chang X, Liu J, Xiao W. STC1 competitively binding βPIX enhances melanoma progression via YAP nuclear translocation and M2 macrophage recruitment through the YAP/CCL2/VEGFA/AKT feedback loop. Pharmacol Res 2024; 204:107218. [PMID: 38768671 DOI: 10.1016/j.phrs.2024.107218] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 05/22/2024]
Abstract
This study investigates the role of Stanniocalcin-1 (STC1) in melanoma progression, with a focus on its impact on metastasis, angiogenesis, and immune evasion. Systematic bioinformatics analysis revealed the potential influence of STC1 dysregulation on prognosis, immune cell infiltration, response to immune therapy, and cellular functions. In vitro assays were conducted to assess the proliferation, invasion, migration, and angiogenesis capabilities of A375 cells. In vivo experiments utilizing C57BL/6 J mice established a lung metastasis model using B16-F10 cells to evaluate macrophage infiltration and M2 polarization. A Transwell co-culture system was employed to explore the crosstalk between melanoma and macrophages. Molecular interactions among STC1, YAP, βPIX, and CCL2 are investigated using mass spectrometry, Co-Immunoprecipitation, Dual-Luciferase Reporter Assay, and Chromatin Immunoprecipitation experiments. STC1 was found to enhance lung metastasis by promoting the recruitment and polarization of M2 macrophages, thereby fostering an immunosuppressive microenvironment. Mechanistically, STC1 competes with YAP for binding to βPIX within the KER domain in melanoma cells, leading to YAP activation and subsequent CCL2 upregulation. CCL2-induced M2 macrophages secrete VEGFA, which enhances tumor vascularization and increases STC1 expression via the AKT signaling pathway in melanoma cells, establishing a pro-metastatic feedback loop. Notably, STC1-induced YAP activation increases PD-L1 expression, promoting immune evasion. Silencing STC1 enhances the efficacy of PD-1 immune checkpoint therapy in mice. This research elucidates STC1's role in melanoma metastasis and its complex interactions with tumor-associated macrophages, proposing STC1 as a potential therapeutic target for countering melanoma metastasis and augmenting the efficacy of PD-1 immunotherapy.
Collapse
Affiliation(s)
- Zhaozhou Ren
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Zhijie Xu
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Xiyue Chang
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Jie Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Wan'an Xiao
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China.
| |
Collapse
|
19
|
Lins FV, Bispo ECI, Rodrigues NS, Silva MVS, Carvalho JL, Gelfuso GM, Saldanha-Araujo F. Ibrutinib Modulates Proliferation, Migration, Mitochondrial Homeostasis, and Apoptosis in Melanoma Cells. Biomedicines 2024; 12:1012. [PMID: 38790974 PMCID: PMC11117653 DOI: 10.3390/biomedicines12051012] [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: 04/07/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Ibrutinib, a tyrosine kinase inhibitor with a broad spectrum of action, has been successfully explored to treat hematological and solid cancers. Herein, we investigated the anti-cancer effect of Ibrutinib on melanoma cell lines. Cytotoxicity was evaluated using the MTT assay. Apoptosis, mitochondrial membrane potential, reactive oxygen species (ROS) production, cell proliferation, and cell cycle stages were determined by flow cytometry. LDH release and Caspase 3/7 activity were determined by colorimetric and luminescent assays, respectively. Cell migration was evaluated by wound scratch assay. Gene expression was determined by real-time PCR. Gene Ontology (GO) enrichment analysis of melanoma clinical samples was performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID). MTT assays showed that Ibrutinib is toxic for MeWo, SK-MEL-28, and WM164 cells. The annexin V/PI staining, Caspase 3/7 activity, and LDH release in MeWo cells revealed that apoptosis is the primary mechanism of death caused by Ibrutinib. Corroborating such observation, we identified that Ibrutinib treatment impairs the mitochondrial membrane potential of such cells and significantly increases the transcriptional levels of the pro-apoptotic factors ATM, HRK, BAX, BAK, CASP3, and CASP8. Furthermore, Ibrutinib showed antimetastatic potential by inhibiting the migration of MeWo cells. Finally, we performed a functional enrichment analysis and identified that the differential expression of Ibrutinib-target molecules is associated with enrichment of apoptosis and necrosis pathways in melanoma samples. Taken together, our results clearly suggest that Ibrutinib can be successfully explored as an effective therapeutic approach for melanomas.
Collapse
Affiliation(s)
- Fernanda Vitelli Lins
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elizabete Cristina Iseke Bispo
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
| | - Naomí Souza Rodrigues
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
| | - Maria Victória Souto Silva
- Laboratório Interdisciplinar de Biociências, Faculdade de Medicina, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (M.V.S.S.); (J.L.C.)
| | - Juliana Lott Carvalho
- Laboratório Interdisciplinar de Biociências, Faculdade de Medicina, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (M.V.S.S.); (J.L.C.)
| | - Guilherme Martins Gelfuso
- Laboratório de Medicamentos, Alimentos e Cosméticos, Universidade de Brasília, Brasília 70910-900, DF, Brazil;
| | - Felipe Saldanha-Araujo
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
| |
Collapse
|
20
|
Viegas J, Sarmento B. Bridging the gap between testing and clinics exploring alternative pre-clinical models in melanoma research. Adv Drug Deliv Rev 2024; 208:115295. [PMID: 38527625 DOI: 10.1016/j.addr.2024.115295] [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: 01/10/2024] [Revised: 02/20/2024] [Accepted: 03/21/2024] [Indexed: 03/27/2024]
Abstract
Melanoma, the deadliest form of skin cancer, poses a significant clinical challenge for the development of effective treatments. Conventional in vivo animal studies have shown limited translational relevance to humans, raising strength to pre-clinical models for melanoma research. This review provides an in-depth analysis of alternative pre-clinical models including in vitro and ex vivo platforms such as reconstructed skin, spheroids, organoids, organotypic models, skin-on-a-chip, and bioprinting. Through a comprehensive analysis, the specific attributes, advantages, and limitations of each model are elucidated. It discusses the points related to the uniqueness advantages, from capturing complex interactions between melanoma cells and their microenvironment to enabling high-throughput drug screening and personalized medicine approaches. This review is structured covering firstly the roadmap to identify the co-occurrence of discovering new melanoma treatments and the development of its models, secondly it covers a comparative between the most used models followed by a section discussing each of them: the in vitro and ex vivo models. It intends to serve as an asset for researchers of melanoma field and clinicians involved in melanoma therapy, offering insights into the diverse preclinical models available for optimizing their integration into the translational pipeline.
Collapse
Affiliation(s)
- Juliana Viegas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
21
|
Provance OK, Oria VO, Tran TT, Caulfield JI, Zito CR, Aguirre-Ducler A, Schalper KA, Kluger HM, Jilaveanu LB. Vascular mimicry as a facilitator of melanoma brain metastasis. Cell Mol Life Sci 2024; 81:188. [PMID: 38635031 PMCID: PMC11026261 DOI: 10.1007/s00018-024-05217-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/19/2024]
Abstract
Melanoma has the highest propensity among solid tumors to metastasize to the brain. Melanoma brain metastases (MBM) are a leading cause of death in melanoma and affect 40-60% of patients with late-stage disease. Therefore, uncovering the molecular mechanisms behind MBM is necessary to enhance therapeutic interventions. Vascular mimicry (VM) is a form of neovascularization linked to invasion, increased risk of metastasis, and poor prognosis in many tumor types, but its significance in MBM remains poorly understood. We found that VM density is elevated in MBM compared to paired extracranial specimens and is associated with tumor volume and CNS edema. In addition, our studies indicate a relevant role of YAP and TAZ, two transcriptional co-factors scarcely studied in melanoma, in tumor cell-vasculogenesis and in brain metastasis. We recently demonstrated activation of the Hippo tumor suppressor pathway and increased degradation of its downstream targets YAP and TAZ in a metastasis impaired cell line model. In the current study we establish the utility of anti-YAP/TAZ therapy in mouse models of metastatic melanoma whereby treatment effectively inhibits VM and prolongs survival of mice with MBM. The data presented herein suggest that VM may be an important and targetable mechanism in melanoma and that VM inhibition might be useful for treating MBM, an area of high unmet clinical need, thus having important implications for future treatment regimens for these patients.
Collapse
Affiliation(s)
- Olivia K Provance
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
| | - Victor O Oria
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Thuy T Tran
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
| | - Jasmine I Caulfield
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
| | - Christopher R Zito
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
- Department of Biology, School of Arts, Sciences, Business, and Education, University of Saint Joseph, West Hartford, CT, USA
| | - Adam Aguirre-Ducler
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Harriet M Kluger
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
| | - Lucia B Jilaveanu
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA.
| |
Collapse
|
22
|
Berns HM, Watkins-Chow DE, Lu S, Louphrasitthiphol P, Zhang T, Brown KM, Moura-Alves P, Goding CR, Pavan WJ. Single-cell profiling of MC1R-inhibited melanocytes. Pigment Cell Melanoma Res 2024; 37:291-308. [PMID: 37972124 DOI: 10.1111/pcmr.13141] [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: 07/12/2023] [Revised: 09/15/2023] [Accepted: 10/05/2023] [Indexed: 11/19/2023]
Abstract
The human red hair color (RHC) trait is caused by increased pheomelanin (red-yellow) and reduced eumelanin (black-brown) pigment in skin and hair due to diminished melanocortin 1 receptor (MC1R) function. In addition, individuals harboring the RHC trait are predisposed to melanoma development. While MC1R variants have been established as causative of RHC and are a well-defined risk factor for melanoma, it remains unclear mechanistically why decreased MC1R signaling alters pigmentation and increases melanoma susceptibility. Here, we use single-cell RNA sequencing (scRNA-seq) of melanocytes isolated from RHC mouse models to define a MC1R-inhibited Gene Signature (MiGS) comprising a large set of previously unidentified genes which may be implicated in melanogenesis and oncogenic transformation. We show that one of the candidate MiGS genes, TBX3, a well-known anti-senescence transcription factor implicated in melanoma progression, binds both E-box and T-box elements to regulate genes associated with melanogenesis and senescence bypass. Our results provide key insights into further mechanisms by which melanocytes with reduced MC1R signaling may regulate pigmentation and offer new candidates of study toward understanding how individuals with the RHC phenotype are predisposed to melanoma.
Collapse
Affiliation(s)
- H Matthew Berns
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Dawn E Watkins-Chow
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sizhu Lu
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Pakavarin Louphrasitthiphol
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Kevin M Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Pedro Moura-Alves
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, PT, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, PT, Portugal
| | - Colin R Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
23
|
Sun S, He Y, Xu J, Leng S, Liu Y, Wan H, Yan L, Xu Y. Enhancing cell pyroptosis with biomimetic nanoparticles for melanoma chemo-immunotherapy. J Control Release 2024; 367:470-485. [PMID: 38290565 DOI: 10.1016/j.jconrel.2024.01.057] [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: 11/27/2023] [Revised: 01/11/2024] [Accepted: 01/26/2024] [Indexed: 02/01/2024]
Abstract
Despite the fact that immunotherapy has significantly improved the prognosis of melanoma patients, the non-response rate of monoimmunotherapy is considerably high due to insufficient tumor immunogenicity. Therefore, it is necessary to develop alternative methods of combination therapy with enhanced antitumor efficiency and less systemic toxicity. In this study, we reported a cancer cell membrane-coated zeolitic imidazole framework-8 (ZIF-8) encapsulating pyroptosis-inducer oxaliplatin (OXA) and immunomodulator imiquimod (R837) for chemoimmunotherapy. With the assistance of DNA methyltransferase inhibitor decitabine (DCT), upregulated Gasdermin E (GSDME) was cleaved by OXA-activated caspase-3, further inducing tumor cell pyroptosis, then localized antitumor immunity was enhanced by immune adjuvant R837, followed by triggering systemic antitumor immune responses. These results provided a proof-of-concept for the use of cell membrane-coated biomimetic nanoparticles as a promising drug carrier of combination therapy and a potential insight for pyroptosis-based melanoma chemo-immunotherapy.
Collapse
Affiliation(s)
- Shiquan Sun
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China; Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Yong He
- R&D Department of 3D printing, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China
| | - Jiaqi Xu
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China
| | - Shaolong Leng
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yu Liu
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China
| | - Huanhuan Wan
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China
| | - Leping Yan
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China.
| | - Yunsheng Xu
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China.
| |
Collapse
|
24
|
Ichiki N, Saigo C, Hanamatsu Y, Iwata H, Takeuchi T. Inducing Melanoma Cell Apoptosis by ERp57/PDIA3 Antibody in the Presence of CPI-613 and Hydroxychloroquine. J Cancer 2024; 15:1779-1785. [PMID: 38434963 PMCID: PMC10905412 DOI: 10.7150/jca.92252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/04/2024] [Indexed: 03/05/2024] Open
Abstract
The combination of the cancer mitochondrial metabolic inhibitor CPI-613 and hydroxychloroquine has tumor-suppressive effects on clear cell sarcoma, which shares pathobiological properties with melanoma. Therefore, we intended to examine the effects of a combination of CPI-613 and hydroxychloroquine on the growth of melanoma cells in the present study. However, cell death was not induced in melanoma cells. Therefore, a monoclonal antibody, ICT, that induced apoptosis in melanoma cells in combination with CPI-613 and hydroxychloroquine was developed. Immunoprecipitation, mass spectrometry, and small interfering RNA (siRNA)-mediated gene silencing demonstrated that ICT targeted Endoplasmic Reticulum Resident Protein 57/ Protein Disulfide Isomerase Family A Member 3 (ERp57/PDIA3), which was first identified as being upregulated by metabolic depletion stress and is localized on the cell surface during immunogenic cell death. The combination of CPI-613 and hydroxychloroquine enhanced the localization of ERp57/PDIA3 to the surface of melanoma cells. siRNA-mediated downregulation of ERp57/PDIA3 did not significantly induce ICT-mediated apoptosis in melanoma cells in the presence of CPI-613 and hydroxychloroquine. Therefore, the ICT antibody acts as a tumor suppressor in melanoma cells by targeting the cell membrane ERp57/PDIA3, expression of which was enhanced by the combination of CPI-613 and hydroxychloroquine.
Collapse
Affiliation(s)
- Naohisa Ichiki
- Department of Dermatology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Chiemi Saigo
- Department of Pathology and Translational Research, Gifu University Graduate School of Medicine, Gifu, Japan
- The United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
- Center for One Medicine Innovative Translational Research; COMIT, Gifu University, Gifu, Japan
| | - Yuki Hanamatsu
- Department of Pathology and Translational Research, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroaki Iwata
- Department of Dermatology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tamotsu Takeuchi
- Department of Pathology and Translational Research, Gifu University Graduate School of Medicine, Gifu, Japan
- Center for One Medicine Innovative Translational Research; COMIT, Gifu University, Gifu, Japan
| |
Collapse
|
25
|
Farnoush N, Khosravi-Mashizi M, Rahmani A, Barahman M, Soleymani S, Asadian F, Shirinzadeh-Dastgiri A, Vakili-Ojarood M, HaghighiKian SM, Naseri A, Aghasipour M, Shiri A, Aghili K, Neamatzadeh H. Updated Meta-Analysis of VDR FokI and TaqI Variants and Their Association with Melanoma Risk. ACTA MEDICA (HRADEC KRALOVE) 2024; 67:113-124. [PMID: 40179841 DOI: 10.14712/18059694.2025.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
BACKGROUND Research suggests that melanoma patients with low vitamin D levels exhibit a higher risk of tumor ulceration and increased tumor mitotic rates. This has led to investigations into the vitamin D receptor (VDR) gene concerning its potential link to melanoma susceptibility. This meta-analysis aims to explore the association between VDR FokI and TaqI polymorphisms and melanoma risk, with an emphasis on the need for research in diverse populations to enhance our conclusions regarding interactions between skin phenotypes and VDR variations. METHODS A comprehensive literature search was conducted in databases, including PubMed, Scopus, and Web of Science, for studies linking VDR polymorphisms to melanoma risk, up to February 1, 2024. Keywords used included "Melanoma", "VDR", and various genetic terms. Quantitative synthesis was performed with Comprehensive Meta-Analysis (Version 4.0) and a significance threshold set at p < 0.05. RESULTS A total of twenty-one case-control studies involving 8,813 melanoma cases and 7,973 controls were included. Twelve studies on FokI had 4,642 cases and 4,534 controls, while nine TaqI studies included 4,171 cases and 3,439 controls. The results show a significant association between the VDR FokI polymorphism and increased melanoma risk across four genetic models (allele model: OR = 1.128, 95% CI 1.026-1.241; P = 0.013; homozygote model: OR = 1.166, 95% CI 1.020-1.332; P = 0.025; heterozygote model: OR = 1.255, 95% CI 1.046-1.507; P = 0.015; dominant model: OR = 1.243, 95% CI 1.052-1.470; P = 0.011). In contrast, the TaqI polymorphism showed no significant association with melanoma risk in the general population. CONCLUSIONS This meta-analysis suggests that the VDR FokI polymorphism is linked to an increased susceptibility to melanoma, while the TaqI variant does not show a significant association. Future research should explore the interactions between VDR polymorphisms, skin phenotypes, and melanoma risk in diverse populations, with larger and more varied studies needed to confirm these findings and enhance our understanding of genetic factors affecting melanoma susceptibility.
Collapse
Affiliation(s)
- Nazila Farnoush
- Department of General Surgery, Babol University of Medical Sciences, Babol, Iran
| | - Mehdi Khosravi-Mashizi
- Department of General Surgery, School of Medicine Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Rahmani
- Department of Plastic Surgery, Iranshahr University of Medical Sciences, Iranshahr, Iran.
| | - Maedeh Barahman
- Department of Radiation Oncology, Firoozgar Clinical Research Development Center (FCRDC), Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Soleymani
- Department of General Surgery, School of Medicine Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Asadian
- Department of Medical Laboratory Sciences, School of Paramedical Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Shirinzadeh-Dastgiri
- Department of Surgery, School of Medicine, Shohadaye Haft-e Tir Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Vakili-Ojarood
- Department of Surgery, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Seyed Masoud HaghighiKian
- Department of General Surgery, School of Medicine Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhosein Naseri
- Department of Colorectal Surgery, Imam Reza Hospital, AJA University of Medical Sciences, Tehran, Iran
| | - Maryam Aghasipour
- Department of Cancer Biology, College of Medicine, University of Cincinnati, Ohio, USA
| | - Amirmasoud Shiri
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kazem Aghili
- Department of Radiology, Shahid Rahnamoun Hospital, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Neamatzadeh
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
26
|
Ceci C, García-Chico C, Atzori MG, Lacal PM, Lista S, Santos-Lozano A, Graziani G, Pinto-Fraga J. Impact of Physical Exercise on Melanoma Hallmarks: Current Status of Preclinical and Clinical Research. J Cancer 2024; 15:1-19. [PMID: 38164270 PMCID: PMC10751671 DOI: 10.7150/jca.88559] [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: 07/27/2023] [Accepted: 10/16/2023] [Indexed: 01/03/2024] Open
Abstract
In recent years, accumulating evidence from preclinical and clinical studies consistently indicated that physical activity/exercise plays a crucial role in reducing the incidence and recurrence of various malignancies, by exerting a beneficial modulation of cancer hallmarks. Moreover, physical activity is suggested to attenuate certain adverse effects of anticancer therapy, including the reduction of cardiovascular toxicity and symptoms related to depression and anxiety, among others, while preserving muscular strength. In the case of melanoma, the relationship with physical activity has been critically debated. Historically, several cohort studies and meta-analyses reported a positive association between physical activity/exercise and melanoma risk. This association was primarily attributed to outdoor activities that may expose the skin to UV radiation, a well-known risk factor for melanocyte transformation. However, more recent evidence does not support such association and recognizes physical activity/exercise role in both melanoma prevention and progression. Nevertheless, sun protection is recommended during outdoor training to minimize UV radiation exposure. This narrative review summarizes preclinical and clinical data about physical activity effects on melanoma hallmarks. Specifically, experimental evidence is reported concerning (i) invasion and metastasis, (ii) reprogramming of energy metabolism, (iii) angiogenesis, (iv) resistance to cell death, (v) evasion from immune destruction, and (vi) tumor-promoting inflammation.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Celia García-Chico
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | | | | | - Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - José Pinto-Fraga
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| |
Collapse
|
27
|
Gosman LM, Țăpoi DA, Costache M. Cutaneous Melanoma: A Review of Multifactorial Pathogenesis, Immunohistochemistry, and Emerging Biomarkers for Early Detection and Management. Int J Mol Sci 2023; 24:15881. [PMID: 37958863 PMCID: PMC10650804 DOI: 10.3390/ijms242115881] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Cutaneous melanoma (CM) is an increasingly significant public health concern. Due to alarming mortality rates and escalating incidence, it is crucial to understand its etiology and identify emerging biomarkers for improved diagnosis and treatment strategies. This review aims to provide a comprehensive overview of the multifactorial etiology of CM, underscore the importance of early detection, discuss the molecular mechanisms behind melanoma development and progression, and shed light on the role of the potential biomarkers in diagnosis and treatment. The pathogenesis of CM involves a complex interplay of genetic predispositions and environmental exposures, ultraviolet radiation exposure being the predominant environmental risk factor. The emergence of new biomarkers, such as novel immunohistochemical markers, gene mutation analysis, microRNA, and exosome protein expressions, holds promise for improved early detection, and prognostic and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Laura Maria Gosman
- Doctoral School, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Pathology, Saint Pantelimon Clinical Emergency Hospital, 021659 Bucharest, Romania
| | - Dana-Antonia Țăpoi
- Department of Pathology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Pathology, University Emergency Hospital, 050098 Bucharest, Romania
| | - Mariana Costache
- Department of Pathology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Pathology, University Emergency Hospital, 050098 Bucharest, Romania
| |
Collapse
|
28
|
Ta N, Jiang X, Zhang Y, Wang H. Ferroptosis as a promising therapeutic strategy for melanoma. Front Pharmacol 2023; 14:1252567. [PMID: 37795022 PMCID: PMC10546212 DOI: 10.3389/fphar.2023.1252567] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023] Open
Abstract
Malignant melanoma (MM) is the most common and deadliest type of skin cancer and is associated with high mortality rates across all races and ethnicities. Although present treatment options combined with surgery provide short-term clinical benefit in patients and early diagnosis of non-metastatic MM significantly increases the probability of survival, no efficacious treatments are available for MM. The etiology and pathogenesis of MM are complex. Acquired drug resistance is associated with a pool prognosis in patients with advanced-stage MM. Thus, these patients require new therapeutic strategies to improve their treatment response and prognosis. Multiple studies have revealed that ferroptosis, a non-apoptotic form of regulated cell death (RCD) characterized by iron dependant lipid peroxidation, can prevent the development of MM. Recent studies have indicated that targeting ferroptosis is a promising treatment strategy for MM. This review article summarizes the core mechanisms underlying the development of ferroptosis in MM cells and its potential role as a therapeutic target in MM. We emphasize the emerging types of small molecules inducing ferroptosis pathways by boosting the antitumor activity of BRAFi and immunotherapy and uncover their beneficial effects to treat MM. We also summarize the application of nanosensitizer-mediated unique dynamic therapeutic strategies and ferroptosis-based nanodrug targeting strategies as therapeutic options for MM. This review suggests that pharmacological induction of ferroptosis may be a potential therapeutic target for MM.
Collapse
Affiliation(s)
- Na Ta
- Department of Neurosurgery, The Affiliated Hospital of Chifeng University, Chifeng, China
| | - Xiaodong Jiang
- Department of Anatomy, College of Basic Medicine, Chifeng University Health Science Center, Chifeng, China
| | - Yongchun Zhang
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Chifeng University, Chifeng, China
| | - Hongquan Wang
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
29
|
Wang J, Liao H, Ban J, Li S, Xiong X, He Q, Shi X, Shen H, Yang S, Sun C, Liu L. Multifunctional Near-Infrared Dye IR-817 Encapsulated in Albumin Nanoparticles for Enhanced Imaging and Photothermal Therapy in Melanoma. Int J Nanomedicine 2023; 18:4949-4967. [PMID: 37693889 PMCID: PMC10488832 DOI: 10.2147/ijn.s425013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023] Open
Abstract
Background Near-infrared cyanine dyes have high sensitivity and spatial resolution imaging capabilities, but they also have unavoidable drawbacks such as photobleaching, low water solubility, fluorescence quenching, and toxic side effects. As an effective biologic drug carrier, albumin combines with cyanine dyes to form albumin@dye nanoparticles. These nanoparticles can alleviate the aforementioned issues and are widely used in tumor imaging and photothermal therapy. Methods Herein, a newly synthesized near-infrared dye IR-817 was combined with bovine serum albumin (BSA) to create BSA@IR-817 nanoparticles. Through the detection of fluorescence emission and absorption, the optimal concentration and ratio of BSA and IR-817 were determined. Subsequently, dynamic light scattering (DLS) measurements and scanning electron microscopy (SEM) were used for the physical characterization of the BSA@IR-817 nanoparticles. Finally, in vitro and in vivo experiments were conducted to assess the fluorescence imaging and photothermal therapeutic potential of BSA@IR-817 nanoparticles. Results IR-817 was adsorbed onto the BSA carrier by covalent conjugation and supramolecular encapsulation, resulting in the formation of dispersed, homogeneous, and stable nanoparticles with a particle size range of 120-220 nm. BSA@IR-817 not only improved the poor water solubility, fluorescence quenching, and toxic side effects of IR-817 but also enhanced the absorption and fluorescence emission peaks in the near-infrared region, as well as the fluorescence in the visible spectrum. In addition, BSA@IR-817 combined with laser 808 irradiation was able to convert light energy into heat energy with temperatures exceeding 50 °C. By creating a mouse model of subcutaneous melanoma, it was discovered that the tumor inhibition rate of BSA@IR-817 was greater than 99% after laser irradiation and that it achieved nearly complete tumor ablation without causing significant toxicity. Conclusion Our research, therefore, proposes the use of safe and effective photothermal nanoparticles for the imaging, diagnosis, and treatment of melanoma, and offers a promising strategy for future biomedical applications.
Collapse
Affiliation(s)
- Jianv Wang
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Hongye Liao
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Jieming Ban
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Sen Li
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Xia Xiong
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Qingqing He
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Xinyu Shi
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Hongping Shen
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Sijin Yang
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Changzhen Sun
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Li Liu
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| |
Collapse
|
30
|
Król W, Machelak W, Zielińska M. GDF11 as a friend or an enemy in the cancer biology? Biochim Biophys Acta Rev Cancer 2023; 1878:188944. [PMID: 37356738 DOI: 10.1016/j.bbcan.2023.188944] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 06/27/2023]
Abstract
The Growth and Differential Factor 11 (GDF11) is a recently discovered representative of Transforming Growth Factor β superfamily. The highest expression of GDF11 is detected in the nervous system, bladder, seminal vesicles and muscles whereas the lowest in the testis, liver or breast. GDF11 role in physiology is still not clear. GDF11 is a crucial factor in embryogenesis, cell cycle control and apoptosis, inasmuch it mainly targets cell retain stemness features, managing to the cell differentiation and the maturation. GDF11 is entangled in lipid metabolism, inflammatory processes and aging. GDF11 is strongly related to carcinogenesis and its expression in tumors is intruded. GDF11 can promote cancer growth in the colon or inhibit the cell proliferation in breast cancer. The aberrated expression is probably allied with the impaired maturation. In this article we summarized an impact of GDF11 on the tumor cells and review the all attitudes connecting GDF11 with carcinogenesis.
Collapse
Affiliation(s)
- Wojciech Król
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Weronika Machelak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Marta Zielińska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
31
|
Udrea AM, Smarandache A, Dinache A, Mares C, Nistorescu S, Avram S, Staicu A. Photosensitizers-Loaded Nanocarriers for Enhancement of Photodynamic Therapy in Melanoma Treatment. Pharmaceutics 2023; 15:2124. [PMID: 37631339 PMCID: PMC10460031 DOI: 10.3390/pharmaceutics15082124] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Malignant melanoma poses a significant global health burden. It is the most aggressive and lethal form of skin cancer, attributed to various risk factors such as UV radiation exposure, genetic modifications, chemical carcinogens, immunosuppression, and fair complexion. Photodynamic therapy is a promising minimally invasive treatment that uses light to activate a photosensitizer, resulting in the formation of reactive oxygen species, which ultimately promote cell death. When selecting photosensitizers for melanoma photodynamic therapy, the presence of melanin should be considered. Melanin absorbs visible radiation similar to most photosensitizers and has antioxidant properties, which undermines the reactive species generated in photodynamic therapy processes. These characteristics have led to further research for new photosensitizing platforms to ensure better treatment results. The development of photosensitizers has advanced with the use of nanotechnology, which plays a crucial role in enhancing solubility, optical absorption, and tumour targeting. This paper reviews the current approaches (that use the synergistic effect of different photosensitizers, nanocarriers, chemotherapeutic agents) in the photodynamic therapy of melanoma.
Collapse
Affiliation(s)
- Ana Maria Udrea
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (A.M.U.); (A.D.); (S.N.)
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (C.M.); (S.A.)
| | - Adriana Smarandache
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (A.M.U.); (A.D.); (S.N.)
| | - Andra Dinache
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (A.M.U.); (A.D.); (S.N.)
| | - Catalina Mares
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (C.M.); (S.A.)
| | - Simona Nistorescu
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (A.M.U.); (A.D.); (S.N.)
| | - Speranta Avram
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (C.M.); (S.A.)
| | - Angela Staicu
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (A.M.U.); (A.D.); (S.N.)
| |
Collapse
|
32
|
Castellani G, Buccarelli M, Arasi MB, Rossi S, Pisanu ME, Bellenghi M, Lintas C, Tabolacci C. BRAF Mutations in Melanoma: Biological Aspects, Therapeutic Implications, and Circulating Biomarkers. Cancers (Basel) 2023; 15:4026. [PMID: 37627054 PMCID: PMC10452867 DOI: 10.3390/cancers15164026] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Melanoma is an aggressive form of skin cancer resulting from the malignant transformation of melanocytes. Recent therapeutic approaches, including targeted therapy and immunotherapy, have improved the prognosis and outcome of melanoma patients. BRAF is one of the most frequently mutated oncogenes recognised in melanoma. The most frequent oncogenic BRAF mutations consist of a single point mutation at codon 600 (mostly V600E) that leads to constitutive activation of the BRAF/MEK/ERK (MAPK) signalling pathway. Therefore, mutated BRAF has become a useful target for molecular therapy and the use of BRAF kinase inhibitors has shown promising results. However, several resistance mechanisms invariably develop leading to therapeutic failure. The aim of this manuscript is to review the role of BRAF mutational status in the pathogenesis of melanoma and its impact on differentiation and inflammation. Moreover, this review focuses on the mechanisms responsible for resistance to targeted therapies in BRAF-mutated melanoma and provides an overview of circulating biomarkers including circulating tumour cells, circulating tumour DNA, and non-coding RNAs.
Collapse
Affiliation(s)
- Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Maria Beatrice Arasi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Maria Elena Pisanu
- High Resolution NMR Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Bellenghi
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Carla Lintas
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| |
Collapse
|
33
|
Xie Y, Gong C, Xia Y, Zhou Y, Ye T, Mei T, Chen H, Chen J. α-Mangostin Suppresses Melanoma Growth, Migration, and Invasion and Potentiates the Anti-tumor Effect of Chemotherapy. Int J Med Sci 2023; 20:1220-1234. [PMID: 37575275 PMCID: PMC10416720 DOI: 10.7150/ijms.80940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 07/27/2023] [Indexed: 08/15/2023] Open
Abstract
Purpose: Melanoma is a highly malignant tumor, which metastasizes and has poor prognosis in late-stage cancer patients. α-Mangostin possesses pharmacological properties, including antioxidant, anti-infective, and anticarcinogenic activities. We investigated α-Mangostin effect on melanoma growth, migration, and invasion and its possible molecular mechanism. Methods: Melanoma cells growth inhibition was determined by the colorimetric 4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction assay. Morphological changes of α-Mangostin-treated melanoma cells were evaluated by transmission electron microscopy and JC-1 staining. Cell apoptosis and cell cycle arrest were assessed by flow cytometry. The effect of α-Mangostin on tumor cells migration and invasion was observed by migration and invasion in vitro assay. Furthermore, the nude and C57BL/6 mouse subcutaneous melanoma models were used to evaluate the in vivo anti-tumor effect of α-Mangostin. Western blot and real time-PCR were performed to analyze the influence of α-Mangostin on some of the common signaling pathways in melanoma cell lines. Signaling pathways were further verified in dissected tumor tissues. Results: α-Mangostin inhibited in vitro melanoma cells proliferation, migration, and invasion of melanoma cells, induced cell cycle arrest in G0/G1 phase, and caused mitochondrial swelling and membrane depolarization, whereas it effectively suppressed melanoma growth in xenografted mice. In addition, α-Mangostin potentiated the in vitro and in vivo anti-tumor effects of cisplatin both in vitro and in vivo. Mechanistically, α-Mangostin down-regulated expression of RAS protein and mRNA, as well as phosphorylation of PI3K in A375, B16F10, M14 and SK-MEL-2 cells. MITF protein and mRNA were inhibited only in M14 cells. Conclusion: α-Mangostin suppresses melanoma cells growth, migration and invasion, and synergistically enhances the anti-tumor effect of chemotherapy, whose mechanism may be mediated through inhibiting Ras, PI3K and MITF.
Collapse
Affiliation(s)
- Yuxiu Xie
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chongwen Gong
- Department of Oncology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Xia
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhan Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Ye
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Mei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Jing Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Țăpoi DA, Gheorghișan-Gălățeanu AA, Dumitru AV, Ciongariu AM, Furtunescu AR, Marin A, Costache M. Primary Undifferentiated/Dedifferentiated Cutaneous Melanomas-A Review on Histological, Immunohistochemical, and Molecular Features with Emphasis on Prognosis and Treatment. Int J Mol Sci 2023; 24:9985. [PMID: 37373134 DOI: 10.3390/ijms24129985] [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: 05/29/2023] [Revised: 06/04/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Diagnosing cutaneous melanoma is usually straightforward based on these malignancies' histopathological and immunohistochemical features. Nevertheless, melanomas can imitate various other neoplasms, sometimes lacking the expression of conventional melanocytic markers and expressing non-melanocytic ones. Furthermore, divergent differentiation is more often encountered in metastatic melanomas and is still poorly described in primary cutaneous melanomas, and little is known about these patients' prognosis and therapeutic approach. Therefore, we reviewed the literature on undifferentiated/dedifferentiated cutaneous melanomas, and we discuss the histological, immunohistochemical, and molecular profiles of undifferentiated/dedifferentiated cutaneous melanomas to understand these peculiar lesions better and improve their diagnostic algorithm. In addition to this, we also discuss how different genetic mutations may influence prognosis and become potential therapeutic targets.
Collapse
Affiliation(s)
- Dana Antonia Țăpoi
- Department of Pathology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Pathology, University Emergency Hospital, 050098 Bucharest, Romania
| | - Ancuța-Augustina Gheorghișan-Gălățeanu
- Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- C.I. Parhon National Institute of Endocrinology, 011863 Bucharest, Romania
| | - Adrian Vasile Dumitru
- Department of Pathology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Pathology, University Emergency Hospital, 050098 Bucharest, Romania
| | - Ana Maria Ciongariu
- Department of Pathology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Pathology, University Emergency Hospital, 050098 Bucharest, Romania
| | - Andreea Roxana Furtunescu
- Doctoral School, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Dermatology, Victor Babes Clinical Hospital, 030303 Bucharest, Romania
| | - Andrei Marin
- Department of Plastic Surgery, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mariana Costache
- Department of Pathology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Pathology, University Emergency Hospital, 050098 Bucharest, Romania
| |
Collapse
|
35
|
Yang L, Cao ZJ, Zhang Y, Zhou JK, Tian J. Disulfidptosis-related classification patterns and tumor microenvironment characterization in skin cutaneous melanoma. Melanoma Manag 2023; 10:MMT65. [PMID: 38230203 PMCID: PMC10789442 DOI: 10.2217/mmt-2023-0006] [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: 08/21/2023] [Accepted: 11/28/2023] [Indexed: 01/18/2024] Open
Abstract
Aim To identify distinct disulfidptosis-molecular subtypes and develop a novel prognostic signature. Methods/materials We integrated into this study multiple SKCM transcriptomic datasets from the Cancer Genome Atlas database and Gene Expression Omnibus dataset. The consensus clustering algorithm was applied to categorize SKCM patients into different DRG subtypes. Results Three distinct DRG subtypes were identified, which were correlated to different clinical outcomes and signaling pathways. Then, a disulfidptosis-relaed signature and nomogram were constructed, which could accurately predict the individual OS of patients with SKCM. The high-risk group was less sensitive to immunotherapy than the low-risk group. Conclusion The signature can assist healthcare professionals in making more accurate and individualized treatment choices for patients with SKCM.
Collapse
Affiliation(s)
- Li Yang
- Department of Dermatology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Zi-jian Cao
- Department of Dermatology, The 63600 Hospital of PLA, Lanzhou, 732750, China
| | - Yuan Zhang
- Department of Oncology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Jin-ke Zhou
- Department of Dermatology, The 63600 Hospital of PLA, Lanzhou, 732750, China
| | - Jun Tian
- Department of Dermatology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| |
Collapse
|
36
|
Bolovan LM, Ceausu M, Stanciu AE, Panait ME, Busca A, Hotnog CM, Bleotu C, Gales LN, Georgescu MT, Prunoiu VM, Brasoveanu LI, Voinea SC. Correlation Studies between S100 Protein Level and Soluble MIA or Tissue MelanA and gp100 (HMB45) Expression in Cutaneous Melanoma. J Pers Med 2023; 13:898. [PMID: 37373887 DOI: 10.3390/jpm13060898] [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: 04/30/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
(1) Background: Cutaneous melanoma (CM) originates from melanocytes and causes 90% of skin cancer deaths; therefore, the comparison of different soluble and tissue markers could be valuable in the detection of melanoma progression and therapy monitoring. The present study is focused on the potential correlations between soluble S100B and MIA protein levels in different melanoma stages or with tissue expression of S100, gp100 (HMB45), and MelanA biomarkers. (2) Methods: Soluble S100B and MIA levels were evaluated by means of immunoassay methods in blood samples from 176 patients with CM, while tissue expressions of S100, MelanA, and gp100 (HMB45) were detected by means of immunohistochemistry in 76 melanomas. (3) Results: Soluble S100B correlated with MIA in stages III (r = 0.677, p < 0.001) and IV (r = 0.662, p < 0.001) but not in stages I and II; however, 22.22% and 31.98% of stage I and II patients, respectively, had high values for at least one of the two soluble markers. S100 tissue expression correlated with both MelanA (r = 0.610, p < 0.001) and HMB45 (r = 0.476, p < 0.01), while HMB45 and MelanA also significantly positively correlated (r = 0.623, p < 0.001). (4) Conclusions: Blood levels of S100B and MIA corroborated with melanoma tissue markers expression could help to improve the stratification process for patients with a high risk of tumor progression.
Collapse
Affiliation(s)
- Lucica Madalina Bolovan
- Carcinogenesis and Molecular Biology Department, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| | - Mihai Ceausu
- Pathology Department, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| | - Adina Elena Stanciu
- Carcinogenesis and Molecular Biology Department, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| | - Marieta Elena Panait
- Cancer Biology Department, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| | - Antonela Busca
- Cancer Biology Department, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| | - Camelia Mia Hotnog
- Center of Immunology, "Stefan S. Nicolau" Institute of Virology, Romanian Academy, 030304 Bucharest, Romania
| | - Coralia Bleotu
- Cellular and Molecular Pathology Department, "Stefan S. Nicolau" Institute of Virology, Romanian Academy, 030304 Bucharest, Romania
| | - Laurentia Nicoleta Gales
- Oncology Department, University of Medicine and Pharmacy "Carol Davila" Bucharest, 050474 Bucharest, Romania
- Oncology Department, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", 252 Fundeni Ave, 022328 Bucharest, Romania
| | - Mihai Teodor Georgescu
- Oncology Department, University of Medicine and Pharmacy "Carol Davila" Bucharest, 050474 Bucharest, Romania
- Oncology Department, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", 252 Fundeni Ave, 022328 Bucharest, Romania
| | - Virgiliu Mihail Prunoiu
- Oncological Surgery Department, University of Medicine and Pharmacy "Carol Davila" Bucharest, 050474 Bucharest, Romania
- Oncological Surgery Department, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| | - Lorelei Irina Brasoveanu
- Center of Immunology, "Stefan S. Nicolau" Institute of Virology, Romanian Academy, 030304 Bucharest, Romania
| | - Silviu Cristian Voinea
- Oncological Surgery Department, University of Medicine and Pharmacy "Carol Davila" Bucharest, 050474 Bucharest, Romania
- Oncological Surgery Department, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", 022328 Bucharest, Romania
| |
Collapse
|
37
|
Berns HM, Watkins-Chow DE, Lu S, Louphrasitthiphol P, Zhang T, Brown KM, Moura-Alves P, Goding CR, Pavan WJ. Loss of MC1R signaling implicates TBX3 in pheomelanogenesis and melanoma predisposition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.10.532018. [PMID: 37090624 PMCID: PMC10120706 DOI: 10.1101/2023.03.10.532018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
The human Red Hair Color (RHC) trait is caused by increased pheomelanin (red-yellow) and reduced eumelanin (black-brown) pigment in skin and hair due to diminished melanocortin 1 receptor (MC1R) function. In addition, individuals harboring the RHC trait are predisposed to melanoma development. While MC1R variants have been established as causative of RHC and are a well-defined risk factor for melanoma, it remains unclear mechanistically why decreased MC1R signaling alters pigmentation and increases melanoma susceptibility. Here, we use single-cell RNA-sequencing (scRNA-seq) of melanocytes isolated from RHC mouse models to reveal a Pheomelanin Gene Signature (PGS) comprising genes implicated in melanogenesis and oncogenic transformation. We show that TBX3, a well-known anti-senescence transcription factor implicated in melanoma progression, is part of the PGS and binds both E-box and T-box elements to regulate genes associated with melanogenesis and senescence bypass. Our results provide key insights into mechanisms by which MC1R signaling regulates pigmentation and how individuals with the RHC phenotype are predisposed to melanoma.
Collapse
Affiliation(s)
- H. Matthew Berns
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford, OX3 7DQ, UK
| | - Dawn E. Watkins-Chow
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sizhu Lu
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford, OX3 7DQ, UK
| | - Pakavarin Louphrasitthiphol
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford, OX3 7DQ, UK
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, 13 USA
| | - Kevin M. Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, 13 USA
| | - Pedro Moura-Alves
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford, OX3 7DQ, UK
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, PT
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, PT
| | - Colin R. Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford, OX3 7DQ, UK
| | - William J. Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
38
|
Musi A, Bongiovanni L. Extracellular Vesicles in Cancer Drug Resistance: Implications on Melanoma Therapy. Cancers (Basel) 2023; 15:1074. [PMID: 36831417 PMCID: PMC9954626 DOI: 10.3390/cancers15041074] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/29/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Extracellular vesicles (EVs) are involved in the pathogenesis of neoplastic diseases. Their role in mediating drug resistance has been widely described in several types of cancers, including melanoma. EVs can mediate drug resistance through several different mechanisms, such as drug-sequestration, transfer of pro-survival proteins and RNA, induction of cancer stem cell-like features and interaction with cells of the tumor microenvironment and immune-system. Melanoma is a highly immunogenic tumor originating from the malignant transformation of melanocytes. Several therapeutic strategies currently used in the treatment of melanoma and the combination of BRAF and MEK-inhibitors, as well as immune check-point inhibitors (ICI), have consistently improved the overall survival time of melanoma patients. However, the development of resistance is one of the biggest problems leading to a poor clinical outcome, and EVs can contribute to this. EVs isolated from melanoma cells can contain "sequestered" chemotherapeutic drugs in order to eliminate them, or bioactive molecules (such as miRNA or proteins) that have been proven to play a crucial role in the transmission of resistance to sensitive neoplastic cells. This leads to the hypothesis that EVs could be considered as resistance-mediators in sensitive melanoma cells. These findings are a pivotal starting point for further investigations to better understand EVs' role in drug resistance mechanisms and how to target them. The purpose of this review is to summarize knowledge about EVs in order to develop a deeper understanding of their underlying mechanisms. This could lead to the development of new therapeutic strategies able to bypass EV-mediated drug-resistance in melanoma, such as by the use of combination therapy, including EV release inhibitors.
Collapse
Affiliation(s)
- Alice Musi
- Department of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy
| | - Laura Bongiovanni
- Department of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584CT Utrecht, The Netherlands
| |
Collapse
|
39
|
Zob DL, Augustin I, Caba L, Panzaru MC, Popa S, Popa AD, Florea L, Gorduza EV. Genomics and Epigenomics in the Molecular Biology of Melanoma-A Prerequisite for Biomarkers Studies. Int J Mol Sci 2022; 24:ijms24010716. [PMID: 36614156 PMCID: PMC9821083 DOI: 10.3390/ijms24010716] [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: 11/07/2022] [Revised: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Melanoma is a common and aggressive tumor originating from melanocytes. The increasing incidence of cutaneous melanoma in recent last decades highlights the need for predictive biomarkers studies. Melanoma development is a complex process, involving the interplay of genetic, epigenetic, and environmental factors. Genetic aberrations include BRAF, NRAS, NF1, MAP2K1/MAP2K2, KIT, GNAQ, GNA11, CDKN2A, TERT mutations, and translocations of kinases. Epigenetic alterations involve microRNAs, non-coding RNAs, histones modifications, and abnormal DNA methylations. Genetic aberrations and epigenetic marks are important as biomarkers for the diagnosis, prognosis, and prediction of disease recurrence, and for therapeutic targets. This review summarizes our current knowledge of the genomic and epigenetic changes in melanoma and discusses the latest scientific information.
Collapse
Affiliation(s)
- Daniela Luminita Zob
- Department of Medical Oncology, AI. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Iolanda Augustin
- Department of Medical Oncology, AI. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
- Correspondence: (I.A.); (L.C.)
| | - Lavinia Caba
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Correspondence: (I.A.); (L.C.)
| | - Monica-Cristina Panzaru
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Setalia Popa
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Alina Delia Popa
- Nursing Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Laura Florea
- Department of Nephrology-Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Eusebiu Vlad Gorduza
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| |
Collapse
|
40
|
de Morais FAP, De Oliveira ACV, Balbinot RB, Lazarin-Bidóia D, Ueda-Nakamura T, de Oliveira Silva S, da Silva Souza Campanholi K, da Silva Junior RC, Gonçalves RS, Caetano W, Nakamura CV. Multifunctional Nanoparticles as High-Efficient Targeted Hypericin System for Theranostic Melanoma. Polymers (Basel) 2022; 15:polym15010179. [PMID: 36616529 PMCID: PMC9824163 DOI: 10.3390/polym15010179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Biotin, spermine, and folic acid were covalently linked to the F127 copolymer to obtain a new drug delivery system designed for HY-loaded PDT treatment against B16F10 cells. Chemical structures and binders quantification were performed by spectroscopy and spectrophotometric techniques (1NMR, HABA/Avidin reagent, fluorescamine assay). Critical micelle concentration, critical micelle temperature, size, polydispersity, and zeta potential indicate the hydrophobicity of the binders can influence the physicochemical parameters. Spermine-modified micelles showed fewer changes in their physical and chemical parameters than the F127 micelles without modification. Furthermore, zeta potential measurements suggest an increase in the physical stability of these carrier systems. The phototherapeutic potential was demonstrated using hypericin-loaded formulation against B16F10 cells, which shows that the combination of the binders on F127 copolymer micelles enhances the photosensitizer uptake and potentializes the photodynamic activity.
Collapse
Affiliation(s)
- Flávia Amanda Pedroso de Morais
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
- Department of Chemistry, State University of Maringá, Maringá 87020-900, PR, Brazil
- Correspondence: (F.A.P.d.M.); (C.V.N.); Tel.: +55-(44)-3011-3680 (F.A.P.d.M. & C.V.N.)
| | | | - Rodolfo Bento Balbinot
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
| | - Danielle Lazarin-Bidóia
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
| | - Tânia Ueda-Nakamura
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
| | - Sueli de Oliveira Silva
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
| | | | | | - Renato Sonchini Gonçalves
- Laboratory of Chemistry of Natural Products, Department of Chemistry, Center for Exact Sciences and Technology, Federal University of Maranhão, São Luís 65080-805, MA, Brazil
| | - Wilker Caetano
- Department of Chemistry, State University of Maringá, Maringá 87020-900, PR, Brazil
| | - Celso Vataru Nakamura
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
- Correspondence: (F.A.P.d.M.); (C.V.N.); Tel.: +55-(44)-3011-3680 (F.A.P.d.M. & C.V.N.)
| |
Collapse
|
41
|
Fontes SS, Nogueira ML, Dias RB, Rocha CAG, Soares MBP, Vannier-Santos MA, Bezerra DP. Combination Therapy of Curcumin and Disulfiram Synergistically Inhibits the Growth of B16-F10 Melanoma Cells by Inducing Oxidative Stress. Biomolecules 2022; 12:1600. [PMID: 36358950 PMCID: PMC9687191 DOI: 10.3390/biom12111600] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/11/2022] [Accepted: 10/16/2022] [Indexed: 10/05/2023] Open
Abstract
Oxidative stress plays a central role in the pathophysiology of melanoma. Curcumin (CUR) is a polyphenolic phytochemical that stimulates reactive oxygen species (ROS) production, while disulfiram (DSS) is a US FDA-approved drug for the treatment of alcoholism that can act by inhibiting the intracellular antioxidant system. Therefore, we hypothesized that they act synergistically against melanoma cells. Herein, we aimed to study the antitumor potential of the combination of CUR with DSS in B16-F10 melanoma cells using in vitro and in vivo models. The cytotoxic effects of different combination ratios of CUR and DSS were evaluated using the Alamar Blue method, allowing the production of isobolograms. Apoptosis detection, DNA fragmentation, cell cycle distribution, and mitochondrial superoxide levels were quantified by flow cytometry. Tumor development in vivo was evaluated using C57BL/6 mice bearing B16-F10 cells. The combinations ratios of 1:2, 1:3, and 2:3 showed synergic effects. B16-F10 cells treated with these combinations showed improved apoptotic cell death and DNA fragmentation. Enhanced mitochondrial superoxide levels were observed at combination ratios of 1:2 and 1:3, indicating increased oxidative stress. In vivo tumor growth inhibition for CUR (20 mg/kg), DSS (60 mg/kg), and their combination were 17.0%, 19.8%, and 28.8%, respectively. This study provided data on the potential cytotoxic activity of the combination of CUR with DSS and may provide a useful tool for the development of a therapeutic combination against melanoma.
Collapse
Affiliation(s)
- Sheila S. Fontes
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
| | - Mateus L. Nogueira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
| | - Rosane B. Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40110-909, BA, Brazil
| | - Clarissa A. Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40110-909, BA, Brazil
| | - Milena B. P. Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
- SENAI Institute for Innovation in Advanced Health Systems, SENAI CIMATEC, Salvador 41650-010, BA, Brazil
| | | | - Daniel P. Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
| |
Collapse
|
42
|
Lopes J, Rodrigues CMP, Gaspar MM, Reis CP. Melanoma Management: From Epidemiology to Treatment and Latest Advances. Cancers (Basel) 2022; 14:4652. [PMID: 36230575 PMCID: PMC9562203 DOI: 10.3390/cancers14194652] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Melanoma is the deadliest skin cancer, whose morbidity and mortality indicators show an increasing trend worldwide. In addition to its great heterogeneity, melanoma has a high metastatic potential, resulting in very limited response to therapies currently available, which were restricted to surgery, radiotherapy and chemotherapy for many years. Advances in knowledge about the pathophysiological mechanisms of the disease have allowed the development of new therapeutic classes, such as immune checkpoint and small molecule kinase inhibitors. However, despite the incontestable progress in the quality of life and survival rates of the patients, effectiveness is still far from desired. Some adverse side effects and resistance mechanisms are the main barriers. Thus, the search for better options has resulted in many clinical trials that are now investigating new drugs and/or combinations. The low water solubility of drugs, low stability and rapid metabolism limit the clinical potential and therapeutic use of some compounds. Thus, the research of nanotechnology-based strategies is being explored as the basis for the broad application of different types of nanosystems in the treatment of melanoma. Future development focus on challenges understanding the mechanisms that make these nanosystems more effective.
Collapse
Affiliation(s)
- Joana Lopes
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Cecília M. P. Rodrigues
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Catarina Pinto Reis
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
- Instituto de Biofísica e Engenharia Biomédica, IBEB, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|