1
|
Mu X, Zhou Y, Yu Y, Zhang M, Liu J. The roles of cancer stem cells and therapeutic implications in melanoma. Front Immunol 2024; 15:1486680. [PMID: 39611156 PMCID: PMC11602477 DOI: 10.3389/fimmu.2024.1486680] [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: 08/26/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
Melanoma is a highly malignant skin tumor characterized by high metastasis and poor prognosis. Recent studies have highlighted the pivotal role of melanoma stem cells (MSCs)-a subpopulation of cancer stem cells (CSCs)-in driving tumor growth, metastasis, therapeutic resistance, and recurrence. Similar to CSCs in other cancers, MSCs possess unique characteristics, including specific surface markers, dysregulated signaling pathways, and the ability to thrive within complex tumor microenvironment (TME). This review explored the current landscape of MSC research, discussing the identification of MSC-specific surface markers, the role of key signaling pathways such as Wnt/β-catenin, Notch, and Hedgehog (Hh), and how interactions within the TME, including hypoxia and immune cells, contribute to MSC-mediated drug resistance and metastatic behavior. Furthermore, we also investigated the latest therapeutic strategies targeting MSCs, such as small-molecule inhibitors, immune-based approaches, and novel vaccine developments, with an emphasis on their potential to overcome melanoma progression and improve clinical outcomes. This review aims to provide valuable insights into the complex roles of MSCs in melanoma biology and offers perspectives for future research and therapeutic advances against this challenging disease.
Collapse
Affiliation(s)
- Xiaoli Mu
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yixin Zhou
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongxin Yu
- The Department of Plastic and Reconstructive Surgery, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingyi Zhang
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiyan Liu
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
The BRAF V600E Mutation Detection by quasa Sensitive Real-Time PCR Assay in Northeast Romania Melanoma Patients. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11209511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background: The prevalence of melanoma in Romanian patients is underestimated. There is a need to identify the BRAF V600E mutation to accurately treat patients with the newest approved BRAF inhibitor therapy. This is a pilot study in which we first aimed to choose the optimal DNA purification method from formalin fixation and paraffin embedding (FFPE) malignant melanoma skin samples to assess the BRAF mutation prevalence and correlate it with clinical pathological parameters. Methods: 30 FFPE samples were purified in parallel with two DNA extraction kits, a manual and a semi-automated kit. The extracted DNA in pure and optimum quantity was tested for the BRAF V600E mutation using the quantitative allele-specific amplification (quasa) method. quasa is a method for the sensitive detection of mutations that may be present in clinical samples at low levels. Results: The BRAF V600E mutation was detected in 60% (18/30) samples in patients with primary cutaneous melanoma of the skin. BRAFV600E mutation was equally distributed by gender and was associated with age >60, nodular melanoma, and trunk localization. Conclusions: The high prevalence of BRAF V600E mutations in our study group raises awareness for improvements to the national reporting system and initiation of the target therapy for patients with malignant melanoma of the skin.
Collapse
|
3
|
Tian M, Yang J, Han J, He J, Liao W. A novel immune checkpoint-related seven-gene signature for predicting prognosis and immunotherapy response in melanoma. Int Immunopharmacol 2020; 87:106821. [PMID: 32731180 DOI: 10.1016/j.intimp.2020.106821] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/09/2020] [Accepted: 07/17/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND New emergence of immunotherapy has significantly improved clinical outcome of melanoma patients with advanced and metastatic diseases. We aimed to develop a gene signature based on the expression of PD-1/PD-L1 signaling pathway genes to predict prognosis and immunotherapy response in melanoma patients. METHODS Melanoma samples from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) were used as the training set and external validation sets respectively. Prognostic genes for overall survival (OS) were identified by univariate Cox regression analysis. Then a multi-gene risk signature was established with the Least Absolute Shrinkage and Selector Operation (LASSO) regression and multivariate Cox regression. The predictive and prognostic value of gene signature was evaluated by Kaplan Meier curve, Time-dependent receiver operating characteristic (ROC) curve, and area under curve (AUC). Gene set enrichment analysis (GSEA) was performed to investigate the discrepantly enriched biological processes between low-risk and high-risk group of melanoma patients. RESULTS A seven-gene risk signature (BATF2, CTLA4, EGFR, HLA-DQB1, IKBKG, PIK3R2, PPP3CA) was constructed. The signature was an independent risk factor for OS (hazard ratio = 1.544, p < 0.001) and it could robustly predict OS in both training and validation sets. Besides, high risk scores indicated advanced clinical stage and no response to immunotherapy for melanoma patients. GSEA demonstrated that high risk score was intimately associated with immune response and immune regulation. In conclusion, the novel seven-gene signature could serve as a robust biomarker for prognosis and a potential indicator of immunotherapy response in melanoma.
Collapse
Affiliation(s)
- Maolang Tian
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiangping Yang
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiaqi Han
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jinlan He
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenjun Liao
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
4
|
Fratangelo F, Camerlingo R, Carriero MV, Pirozzi G, Palmieri G, Gentilcore G, Ragone C, Minopoli M, Ascierto PA, Motti ML. Effect of ABT-888 on the apoptosis, motility and invasiveness of BRAFi-resistant melanoma cells. Int J Oncol 2018; 53:1149-1159. [PMID: 29956724 PMCID: PMC6065454 DOI: 10.3892/ijo.2018.4457] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/12/2018] [Indexed: 12/26/2022] Open
Abstract
Melanoma is a molecularly heterogeneous disease with many genetic mutations and altered signaling pathways. Activating mutations in the BRAF oncogene are observed in approximately 50% of cutaneous melanomas and the use of BRAF inhibitor (BRAFi) compounds has been reported to improve the outcome of patients with BRAF-mutated metastatic melanoma. However, the majority of these patients develop resistance within 6-8 months following the initiation of BRAFi treatment. In this study, we examined the possible use of the poly(ADP-ribose) polymerase 1 (PARP1) inhibitor, ABT-888 (veliparib), as a novel molecule that may be successfully employed in the treatment of BRAFi-resistant melanoma cells. Sensitive and resistant to BRAFi dabrafenib A375 cells were exposed to increasing concentrations of ABT-888. Cell viability and apoptosis were assessed by MTT assay and Annexin V-FITC analysis, respectively. The cell migratory and invasive ability was investigated using the xCELLigence technology and Boyden chamber assays, respectively. ABT-888 was found to reduce cell viability and exhibited pro-apoptotic activity in melanoma cell lines, independently from the BRAF/NRAS mutation status, in a dose-dependent manner, with the maximal effect being reached in the 25-50 µM concentration range. Moreover, ABT-888 promoted apoptosis in both the sensitive and resistant A375 cells, suggesting that ABT-888 may be useful in the treatment of BRAFi-resistant subsets of melanoma cells. Finally, in accordance with the involvement of PARP1 in actin cytoskeletal machinery, we found that the cytoskeletal organization, motility and invasive capability of both the A375 and A375R cells decreased upon exposure to 5 µM ABT-888 for 24 h. On the whole, the findings of this study highlight the pivotal role of PARP1 in the migration and invasion of melanoma cells, suggesting that ABT-888 may indeed be effective, not only as a pro-apoptotic drug for use in the treatment of BRAFi-resistant melanoma cells, but also in suppressing their migratory and invasive activities.
Collapse
Affiliation(s)
| | - Rosa Camerlingo
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
| | | | - Giuseppe Pirozzi
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
| | - Giuseppe Palmieri
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry, National Research Council, 07100 Sassari, Italy
| | - Giusy Gentilcore
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
- Division of Translational Medicine, Sidra Medical and Research Centre, 26999 Doha, Qatar
| | - Concetta Ragone
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
- Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', 81100 Caserta
| | - Michele Minopoli
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
| | | | - Maria Letizia Motti
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry, National Research Council, 07100 Sassari, Italy
- Division of Translational Medicine, Sidra Medical and Research Centre, 26999 Doha, Qatar
- Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', 81100 Caserta
- Department of Sport Science and Wellness, University 'Parthenope', 80133 Naples, Italy
| |
Collapse
|
5
|
Figueiredo CR, Azevedo RA, Mousdell S, Resende-Lara PT, Ireland L, Santos A, Girola N, Cunha RLOR, Schmid MC, Polonelli L, Travassos LR, Mielgo A. Blockade of MIF-CD74 Signalling on Macrophages and Dendritic Cells Restores the Antitumour Immune Response Against Metastatic Melanoma. Front Immunol 2018; 9:1132. [PMID: 29875777 PMCID: PMC5974174 DOI: 10.3389/fimmu.2018.01132] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/04/2018] [Indexed: 12/15/2022] Open
Abstract
Mounting an effective immune response against cancer requires the activation of innate and adaptive immune cells. Metastatic melanoma is the most aggressive form of skin cancer. While immunotherapies have shown a remarkable success in melanoma treatment, patients develop resistance by mechanisms that include the establishment of an immune suppressive tumor microenvironment. Thus, understanding how metastatic melanoma cells suppress the immune system is vital to develop effective immunotherapies against this disease. In this study, we find that macrophages (MOs) and dendritic cells (DCs) are suppressed in metastatic melanoma and that the Ig-CDR-based peptide C36L1 is able to restore MOs and DCs' antitumorigenic and immunogenic functions and to inhibit metastatic growth in lungs. Specifically, C36L1 treatment is able to repolarize M2-like immunosuppressive MOs into M1-like antitumorigenic MOs, and increase the number of immunogenic DCs, and activated cytotoxic T cells, while reducing the number of regulatory T cells and monocytic myeloid-derived suppressor cells in metastatic lungs. Mechanistically, we find that C36L1 directly binds to the MIF receptor CD74 which is expressed on MOs and DCs, disturbing CD74 structural dynamics and inhibiting MIF signaling on these cells. Interfering with MIF-CD74 signaling on MOs and DCs leads to a decrease in the expression of immunosuppressive factors from MOs and an increase in the capacity of DCs to activate cytotoxic T cells. Our findings suggest that interfering with MIF-CD74 immunosuppressive signaling in MOs and DCs, using peptide-based immunotherapy can restore the antitumor immune response in metastatic melanoma. Our study provides the rationale for further development of peptide-based therapies to restore the antitumor immune response in metastatic melanoma.
Collapse
Affiliation(s)
- Carlos R. Figueiredo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Ricardo A. Azevedo
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Sasha Mousdell
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Pedro T. Resende-Lara
- Laboratory of Computational Biology and Bioinformatics, Federal University of ABC, Santo André, Brazil
- Laboratoire de Biologie et Pharmacologie Appliquées (LBPA), UMR 8113, Ecole Normale Supérieure, Cachan, France
| | - Lucy Ireland
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Almudena Santos
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Natalia Girola
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Rodrigo L. O. R. Cunha
- Chemical Biology Laboratory, Natural and Human Sciences Center, Federal University of ABC, Santo André, Brazil
| | - Michael C. Schmid
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Luciano Polonelli
- Unit of Biomedical, Biotechnological and Translational Sciences, Department of Medicine and Surgery, Universitá degli Studi di Parma, Parma, Italy
| | - Luiz R. Travassos
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Ainhoa Mielgo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
6
|
Zhang Y, Cai P, Liang T, Wang L, Hu L. TIM-3 is a potential prognostic marker for patients with solid tumors: A systematic review and meta-analysis. Oncotarget 2018; 8:31705-31713. [PMID: 28423646 PMCID: PMC5458241 DOI: 10.18632/oncotarget.15954] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 02/21/2017] [Indexed: 01/01/2023] Open
Abstract
Accumulated studies have demonstrated the important role of T cell immunoglobulin- and mucin-domain-containing molecule-3 (TIM-3) in various solid tumors and indicated its correlation with patients’ survival. To further verify the prognostic significance of TIM-3 in cancer patients and its correlation with tumor, we performed this meta-analysis including seven studies searched from PubMed, Web of Science, and Embase till July 2016. A total of 869 patients were used to analyze the association between TIM-3 expression and patients’ overall survival (OS). The pooled results showed that higher expression of TIM-3 was significantly correlated to shorter OS (7 studies, HR=1.89; 95% CI: 1.38-2.57; P< 0.001). In addition, higher TIM-3 expression was associated with advanced tumor stage (3 studies, III/IV vs. I/II, RR=2.02; 95% CI: 1.45–2.81; P< 0.001). In conclusion, our study highlights the role of TIM-3 as a potential prognostic marker and a promising therapeutic target in solid tumors.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pengcheng Cai
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Liang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lin Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lihua Hu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
7
|
Treatment of melanoma with selected inhibitors of signaling kinases effectively reduces proliferation and induces expression of cell cycle inhibitors. Med Oncol 2017; 35:7. [PMID: 29214525 PMCID: PMC5719123 DOI: 10.1007/s12032-017-1069-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 11/30/2017] [Indexed: 01/01/2023]
Abstract
Cancer treatment often tends to involve direct targeting enzymes essential for the growth and proliferation of cancer cells. The aim of this study was the recognition of the possible role of selected protein kinases: PI3K, ERK1/2, and mTOR in cell proliferation and cell cycle in malignant melanoma. We investigated the role of protein kinase inhibitors: U0126 (ERK1/2), LY294002 (PI3K), rapamycin (mTOR), everolimus (mTOR), GDC-0879 (B-RAF), and CHIR-99021 (GSK3beta) in cell proliferation and expression of crucial regulatory cell cycle proteins in human melanoma cells: WM793 (VGP) and Lu1205 (metastatic). They were used either individually or in various combinations. The study on the effect of signaling kinases inhibitors on proliferation—BrdU ELISA test after 48–72 h. Their effect on the expression of cell cycle regulatory proteins: cyclin D1 and D3, cyclin-dependent kinase CDK4 and CDK6, and cell cycle inhibitors: p16, p21, and p27, was studied at the protein level (western blot). Treatment of melanoma cells with protein kinase inhibitors led to significantly decreased cell proliferation except the use of a GSK-3β kinase inhibitors—CHIR-99021. The significant decrease in the expression of selected cyclins and cyclin-dependent kinases (CDKs) with parallel increase in the expression of some of cyclin-dependent kinases inhibitors and in consequence meaningful reduction in melanoma cell proliferation by the combinations of inhibitors of signaling kinases clearly showed the crucial role of AKT, ERK 1/2, and mTOR signal transduction in melanoma progression. The results unanimously indicate those pathways as an important target for treatment of melanoma.
Collapse
|
8
|
Rodríguez-Cerdeira C, Carnero Gregorio M, López-Barcenas A, Sánchez-Blanco E, Sánchez-Blanco B, Fabbrocini G, Bardhi B, Sinani A, Guzman RA. Advances in Immunotherapy for Melanoma: A Comprehensive Review. Mediators Inflamm 2017; 2017:3264217. [PMID: 28848246 PMCID: PMC5564072 DOI: 10.1155/2017/3264217] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/21/2017] [Accepted: 04/03/2017] [Indexed: 12/18/2022] Open
Abstract
Melanomas are tumors originating from melanocytes and tend to show early metastasis secondary to the loss of cellular adhesion in the primary tumor, resulting in high mortality rates. Cancer-specific active immunotherapy is an experimental form of treatment that stimulates the immune system to recognize antigens on the surface of cancer cells. Current experimental approaches in immunotherapy include vaccines, biochemotherapy, and the transfer of adoptive T cells and dendritic cells. Several types of vaccines, including peptide, viral, and dendritic cell vaccines, are currently under investigation for the treatment of melanoma. These treatments have the same goal as drugs that are already used to stimulate the proliferation of T lymphocytes in order to destroy tumor cells; however, immunotherapies aim to selectively attack the tumor cells of each patient. In this comprehensive review, we describe recent advancements in the development of immunotherapies for melanoma, with a specific focus on the identification of neoantigens for the prediction of their elicited immune responses. This review is expected to provide important insights into the future of immunotherapy for melanoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ardiana Sinani
- Dermatology Service, Military Medical Unit, University Trauma Hospital, Tirana, Albania
| | | |
Collapse
|
9
|
Guo B, Hui Q, Zhang Y, Chang P, Tao K. miR-194 is a negative regulator of GEF-H1 pathway in melanoma. Oncol Rep 2016; 36:2412-20. [PMID: 27573550 DOI: 10.3892/or.2016.5020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 08/01/2016] [Indexed: 11/05/2022] Open
Abstract
The incidence and associated mortality of melanoma continues to increase worldwide. At present, there is no curative therapy for advanced stage of melanoma. It is necessary to find new indicators of prognosis and therapeutic targets. Increasing evidence shows that miRNA can provide potential candidate biomarkers for melanoma and therapeutic targets. GEF-H1, a regulator of RhoA, as oncogenic driver in melanoma, promotes the growth and invasion of melanoma. miR-194 is a tumor-suppressor gene in multiple tumors, such as bladder and non-small cell lung cancer, and clear cell renal cell carcinoma. In the present study, we demonstrated that GEF-H1 serves as target of miR-194. Overexpression of miR-194 downregulates the GEF-H1/RhoA pathway, inhibits melanoma cancer cell proliferation and metastasis. Furthermore, miR-194 expression is negatively associated with tumor-node-metastasis (TNM) stages. Briefly, our findings provided new theoretical basis for melanoma treatment.
Collapse
Affiliation(s)
- Bingyu Guo
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenhe, Shenyang, Liaoning 110016, P.R. China
| | - Qiang Hui
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenhe, Shenyang, Liaoning 110016, P.R. China
| | - Yu Zhang
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenhe, Shenyang, Liaoning 110016, P.R. China
| | - Peng Chang
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenhe, Shenyang, Liaoning 110016, P.R. China
| | - Kai Tao
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenhe, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
10
|
Casula M, Colombino M, Manca A, Caracò C, Botti G, Ascierto PA, Lissia A, Cossu A, Palmieri G. Low Levels of Genetic Heterogeneity in Matched Lymph Node Metastases from Patients with Melanoma. J Invest Dermatol 2016; 136:1917-1920. [PMID: 27265006 DOI: 10.1016/j.jid.2016.05.103] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 05/19/2016] [Accepted: 05/20/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Milena Casula
- Istituto di Chimica Biomolecolare, CNR, Sassari, Italy
| | | | | | | | | | | | - Amelia Lissia
- Dipartimento di Chirurgia, Microchirurgia e Scienze Mediche, Università di Sassari, Sassari, Italy
| | - Antonio Cossu
- Dipartimento di Chirurgia, Microchirurgia e Scienze Mediche, Università di Sassari, Sassari, Italy
| | | | | |
Collapse
|
11
|
Carletto B, Berton J, Ferreira TN, Dalmolin LF, Paludo KS, Mainardes RM, Farago PV, Favero GM. Resveratrol-loaded nanocapsules inhibit murine melanoma tumor growth. Colloids Surf B Biointerfaces 2016; 144:65-72. [PMID: 27070053 DOI: 10.1016/j.colsurfb.2016.04.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/30/2016] [Accepted: 04/01/2016] [Indexed: 10/22/2022]
Abstract
In this study, resveratrol-loaded nanocapsules were developed and its antitumor activity tested on a melanoma mice model. These nanocapsules were spherically-shaped and presented suitable size, negative charge and high encapsulation efficiency for their use as a modified-release system of resveratrol. Nanoencapsulation leads to the drug amorphization. Resveratrol-loaded nanoparticles reduced cell viability of murine melanoma cells. There was a decrease in tumor volume, an increase in the necrotic area and inflammatory infiltrate of melanoma when resveratrol-loaded nanocapsules were compared to free resveratrol in treated mice. Nanoencapsulation of resveratrol also prevented metastasis and pulmonary hemorrhage. This modified-release technology containing resveratrol can be used as a feasible approach in order to inhibit murine melanoma tumor growth.
Collapse
Affiliation(s)
- Bruna Carletto
- Postgraduate Program in Pharmaceutical Sciences, Ponta Grossa State University/Midwestern State University, Brazil
| | - Juliana Berton
- Postgraduate Program in Pharmaceutical Sciences, Ponta Grossa State University/Midwestern State University, Brazil
| | - Tamara Nascimento Ferreira
- Postgraduate Program in Pharmaceutical Sciences, Ponta Grossa State University/Midwestern State University, Brazil
| | - Luciana Facco Dalmolin
- Postgraduate Program in Pharmaceutical Sciences, Ponta Grossa State University/Midwestern State University, Brazil
| | - Katia Sabrina Paludo
- Department of Structural, Molecular and Genetic Biology, Ponta Grossa State University, Brazil
| | | | - Paulo Vitor Farago
- Department of Pharmaceutical Sciences, Ponta Grossa State University, Brazil
| | | |
Collapse
|
12
|
Abstract
A few years ago therapeutic options in advanced melanoma were very limited and the prognosis was somber. Although recent progresses are far from providing a cure for advanced melanoma, yet these have kindled new hopes and searching for a cure does not seem unreasonable. Seven new medicines have been authorized in various regions of the world in the recent past in the therapy of advanced melanoma, over half of them acting by mechanisms involving the immune system of the host. The anti-CTLA-4 (cytotoxic T lymphocyte associated protein-4) ipilimumab has been followed by anti-PD1 (programmed death1) inhibitors, more effective and safer. Very recently, the first oncolytic immunotherapy, talimogene laherparepvec (T-VEC) has been authorized for placing on the market and a variety of combinations of the new therapies are currently being evaluated or considered. Besides, a plethora of other molecules and approaches, especially monoclonal antibodies, are in the preliminary phases of clinical investigation and are likely to bring new benefits for the treatment of this potentially fatal form of cancer.
Collapse
Affiliation(s)
- Robert Ancuceanu
- University of Medicine and Pharmacy “Carol Davila”, Bucharest, Faculty of Pharmacy, Bucharest, Romania
| | - Monica Neagu
- “Victor Babes” National Institute of Pathology, Bucharest, Romania
| |
Collapse
|