1
|
Zhu J, Li W, Jing J. Design, Synthesis and Anti-Melanoma Activity of Novel Annexin V Derivative with β 3-Integrin Affinity. Int J Mol Sci 2023; 24:11107. [PMID: 37446286 DOI: 10.3390/ijms241311107] [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: 06/09/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Tumor tissues often exhibit unique integrin receptor presentation during development, such as high exposures of αvβ3 and αIIbβ3 integrins. These features are not present in normal tissues. The induction of selective thrombosis and infarction in the tumor-feeding vessels, as well as specific antagonism of αvβ3 integrin on the surface of tumor endothelial cells, is a potential novel antitumor strategy. The Echistatin-Annexin V (EAV) fusion protein is a novel Annexin V (ANV) derivative that possesses a high degree of αvβ3 and αIIbβ3 integrin receptor recognition and binding characteristics while retaining the specific binding ability of the natural ANV molecule for phosphatidylserine (PS). We systematically investigated the biological effects of this novel molecule with superimposed functions on mouse melanoma. We found that EAV inhibited the viability and migration of B16F10 murine melanoma cells in a dose-dependent manner, exhibited good tumor suppressive effects in a xenograft mouse melanoma model, strongly induced tumor tissue necrosis in mice, and targeted the inhibition of angiogenesis in mouse melanoma tumor tissue. EAV exhibited stronger biological effects than natural ANV molecules in inhibiting melanoma in mice. The unique biological effects of EAV are based on its high β3-type integrin receptor-specific recognition and binding ability, as well as its highly selective binding to PS molecules. Based on these findings, we propose that EAV-mediated tumor suppression is a novel and promising antitumor strategy that targets both PS- and integrin β3-positive tumor neovascularization and the tumor cells themselves, thus providing a possible mechanism for the treatment of melanoma.
Collapse
Affiliation(s)
- Jingyi Zhu
- Beijing Key Lab of Biotechnology and Genetic Engineering, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Wenjuan Li
- Beijing Key Lab of Biotechnology and Genetic Engineering, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Jian Jing
- Beijing Key Lab of Biotechnology and Genetic Engineering, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
2
|
Vega Escobar K, Armijos PO, Milman T, Shields CL, Eagle RC. Intratumoral bacteria in uveal melanoma: A case report. Am J Ophthalmol Case Rep 2023; 30:101833. [PMID: 37139176 PMCID: PMC10149332 DOI: 10.1016/j.ajoc.2023.101833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 02/07/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023] Open
Abstract
Purpose Intratumoral bacteria and their potential application to cancer immunotherapy have been a topic of interest in recent studies. To our knowledge, bacteria in uveal melanoma have not been previously reported. Observations We describe a patient with a large choroidal melanoma, measuring 18 × 16 mm in basal dimension and 15 mm in ultrasonographic thickness, managed by plaque brachytherapy. At the time of plaque removal, a prophylactic scleral patch graft was placed to protect from anticipated scleral necrosis. Progressive ocular ischemia led to a blind and painful eye. The enucleated eye demonstrated an extensively necrotic and heavily pigmented mushroom-shaped regressed cilichoroidal mass deep to the scleral patch graft. Numerous Gram-positive cocci were noted within the regressed uveal melanoma and the adjacent sclera. Conclusions and Importance This case highlights the fact that regressed uveal melanomas can contain intra-tumoral bacteria.
Collapse
|
3
|
Arast Y, Heidary M, Tanbakousazan F, Behnamipour S, Vazirizadeh A, Pourahmad J. Selective Toxicity of Cistanche tubulosa Root Extract on Cancerous Skin Mitochondria isolated from animal Model of Melanoma. Cutan Ocul Toxicol 2022; 41:243-249. [PMID: 35796072 DOI: 10.1080/15569527.2022.2096628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
INTRODUCTION As a major public health issue, the skin cancer is a leading reason of death and has resulted in significant financial and human losses globally. Numerous environmental and internal variables may both drive and exacerbate the pathophysiology of the skin cancer. Marine herbs and animals, including marine sponges, cucumbers, and squirts, have been shown to have cytotoxic consequences on cancerous cells in prior research. PURPOSE melanoma mitochondria obtained from skin of melanoma animal model are studied in this research to see whether extracts from Cistanche tubulosa, a plant endemic to the northern coasts of the Persian Gulf, has a cytotoxic impact on them. MATERIAL AND METHOD In this study, the mitochondria isolated from melanoma cells via differential centrifugation and treated with various concentrations (1250, 2500 and 5000 µg/ml) of methanolic extract of C. tubulosa. Then MTT, ROS, MMP decline, mitochondrial swelling, cytochrome c release and flow cytometry assays were performed on them. RESULTS The results of MTT assay showed that the IC50 of C. tubulosa extract is 2500 μg/ml and C. tubulosa extract induced a selectively significant (P < 0.05) concentration-dependent decrease in the SDH activity in cancerous skin mitochondria. The ROS results also showed that all concentrations of C. tubulosa extracts significantly increased ROS production, MMP decline and the release of cytochrome c in cancer groups mitochondria. The swelling of mitochondria isolated from the cancer group was significantly increased compared to the control group. In addition, the results of apoptosis assay showed that addition of root extract of Cistanche tubulosa on melanoma cells increased apoptosis, while it had no effect on control non tumor cells. DISCUSSION AND CONCLUSION Based on these results, the presence of potentially bioactive compounds in C. tubulosa make this Persian Gulf coastal herb a strong candidate for further molecular studies and clinical research in the field of melanoma cancer therapy.
Collapse
Affiliation(s)
- Yalda Arast
- Research center of Environmental Pollutants, Qom University of Medical Sciences, Qom, Iran
| | - Mobina Heidary
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences Tehran, Iran
| | - Farahnaz Tanbakousazan
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences Tehran, Iran
| | - Somaye Behnamipour
- Research center of Environmental Pollutants, Qom University of Medical Sciences, Qom, Iran
| | - Amir Vazirizadeh
- Persian Gulf Research Institute, Marine Biology and Fishery Sciences Department, Persian Gulf University, Bandar Abbas. Iran
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences Tehran, Iran
| |
Collapse
|
4
|
Karami Fath M, Azargoonjahromi A, Jafari N, Mehdi M, Alavi F, Daraei M, Mohammadkhani N, Mueller AL, Brockmueller A, Shakibaei M, Payandeh Z. Exosome application in tumorigenesis: diagnosis and treatment of melanoma. Med Oncol 2022; 39:19. [PMID: 34982284 DOI: 10.1007/s12032-021-01621-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/28/2021] [Indexed: 12/12/2022]
Abstract
Melanoma is the most aggressive of skin cancer derived from genetic mutations in the melanocytes. Current therapeutic approaches include surgical resection, chemotherapy, photodynamic therapy, immunotherapy, biochemotherapy, and targeted therapy. However, the efficiency of these strategies may be decreased due to the development of diverse resistance mechanisms. Here, it has been proven that therapeutic monoclonal antibodies (mAbs) can improve the efficiency of melanoma therapies and also, cancer vaccines are another approach for the treatment of melanoma that has already improved clinical outcomes in these patients. The use of antibodies and gene vaccines provides a new perspective in melanoma treatment. Since the tumor microenvironment is another important factor for cancer progression and metastasis, in recent times, a mechanism has been identified to provide an opportunity for melanoma cells to communicate with remote cells. This mechanism is involved by a novel molecular structure, named extracellular vesicles (EVs). Depending on the functional status of origin cells, exosomes contain various cargos and different compositions. In this review, we presented recent progress of exosome applications in the treatment of melanoma. Different aspects of exosome therapy and ongoing efforts in this field will be discussed too.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Ali Azargoonjahromi
- Department of Nursing, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nafiseh Jafari
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Mehdi
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Fatemeh Alavi
- Department of Pathobiology, Faculty of Specialized Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mona Daraei
- Pharmacy School, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Niloufar Mohammadkhani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, 1985717443, Tehran, Iran
| | - Anna-Lena Mueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilian-University Munich, 80336, Munich, Germany
| | - Aranka Brockmueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilian-University Munich, 80336, Munich, Germany
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilian-University Munich, 80336, Munich, Germany.
| | - Zahra Payandeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Sawada Y, Nakamura M. Daily Lifestyle and Cutaneous Malignancies. Int J Mol Sci 2021; 22:5227. [PMID: 34069297 PMCID: PMC8156459 DOI: 10.3390/ijms22105227] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/09/2021] [Accepted: 05/13/2021] [Indexed: 12/11/2022] Open
Abstract
Daily lifestyle is a fundamental part of human life and its influence accumulates daily in the human body. We observe that a good daily lifestyle has a beneficial impact on our health; however, the actual effects of individual daily lifestyle factors on human skin diseases, especially skin cancers, have not been summarized. In this review, we focused on the influence of daily lifestyle on the development of skin cancer and described the detailed molecular mechanisms of the development or regulation of cutaneous malignancies. Several daily lifestyle factors, such as circadian rhythm disruption, smoking, alcohol, fatty acids, dietary fiber, obesity, and ultraviolet light, are known to be associated with the risk of cutaneous malignancies, malignant melanoma, squamous cell carcinoma, basal cell carcinoma, and Merkel cell carcinoma. Although the influence of some daily lifestyles on the risk of skin cancers is controversial, this review provides us a better understanding of the relationship between daily lifestyle factors and skin cancers.
Collapse
Affiliation(s)
- Yu Sawada
- Department of Dermatology, University of Occupational and Environmental Health 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555, Japan;
| | | |
Collapse
|
6
|
Ogata D, Haydu LE, Glitza IC, Patel SP, Tawbi HA, McQuade JL, Diab A, Ekmekcioglu S, Wong MK, Davies MA, Amaria RN. The efficacy of anti-programmed cell death protein 1 therapy among patients with metastatic acral and metastatic mucosal melanoma. Cancer Med 2021; 10:2293-2299. [PMID: 33686688 PMCID: PMC7982611 DOI: 10.1002/cam4.3781] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/26/2020] [Accepted: 01/28/2021] [Indexed: 12/18/2022] Open
Abstract
Background Anti‐programmed cell death protein 1 (PD‐1) antibodies are a standard treatment for metastatic melanoma patients. However, the understanding of the efficacy of anti‐PD‐1 for acral melanoma (AM) and mucosal melanoma (MM) is limited as these subtypes are relatively rare compared to cutaneous melanoma (CM). Methods This single institution, retrospective cohort study included patients with advanced AM and MM who underwent anti‐PD‐1 therapy for metastatic melanoma between 2012 and 2018. Objective responses were determined using the investigator‐assessed Response Evaluation Criteria in Solid Tumors version 1.1. Progression‐free survival (PFS) and overall survival (OS) were assessed using the Kaplan–Meier method. A Cox regression analysis was performed to identify the factors associated with survival outcomes. Results Ninety‐seven patients were identified, 38 (39%) with AM and 59 (61%) with MM. The objective response rates (ORRs) were 21.0% and 15.2% in patients with AM and MM, respectively. The median PFS and OS were 3.6 and 25.7 months for AM patients, and 3.0 and 20.1 months for MM patients, respectively. Elevated serum lactate dehydrogenase (LDH) (AM: hazard ratio [HR], 0.22; 95% confidence interval [CI], 0.06–0.87; p = 0.03, MM: HR, 0.20; 95% CI, 0.08–0.53; p = 0.001) was significantly associated with shorter OS for both subtypes. Conclusions The ORR, PFS, and OS with anti‐PD‐1 therapy were poor in patients with AM and MM compared to those previously reported clinical trials for nonacral CM. High serum LDH was associated with significantly shorter OS.
Collapse
Affiliation(s)
- Dai Ogata
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA.,Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Lauren E Haydu
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Isabella C Glitza
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer L McQuade
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Adi Diab
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Suhendan Ekmekcioglu
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Michael K Wong
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Rodabe N Amaria
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
7
|
Bognar Z, Cseh AM, Fekete K, Antus C, Bognar R, Tapodi A, Ramadan FHJ, Sumegi B, Gallyas F. Amiodarone's major metabolite, desethylamiodarone inhibits proliferation of B16-F10 melanoma cells and limits lung metastasis formation in an in vivo experimental model. PLoS One 2020; 15:e0239088. [PMID: 32977329 PMCID: PMC7518930 DOI: 10.1371/journal.pone.0239088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 08/30/2020] [Indexed: 12/27/2022] Open
Abstract
Previously, we demonstrated the in vitro anti-tumor effects of desethylamiodarone (DEA) in bladder and cervix cancer cell lines. In the present study, we intended to establish its potentiality in B16-F10 metastatic melanoma cells in vitro and in vivo. We assessed cell proliferation, apoptosis and cell cycle by using sulforhodamine B assay, Muse™ Annexin V & Dead Cell and Muse® Cell Cycle assays, respectively. We determined colony formation after crystal violet staining. For studying mechanistic aspects, immunoblotting analysis was performed. We used a C57BL/6 experimental lung metastasis model for demonstrating in vivo anti-metastatic potential of DEA. DEA inhibited in vitro proliferation and colony formation, and in vivo lung metastasizing properties of B16-F10 cells. It arrested the cells in G0/G1 phase of their cycle likely via p21 in a p53-dependent fashion, and induced caspase mediated apoptosis likely via inversely regulating Bcl-2 and Bax levels, and reducing Akt and ERK1/2 activation. In this study, we provided in vitro and in vivo experimental evidences for DEA’s potentiality in the therapy of metastatic melanomas. Since DEA is the major metabolite of amiodarone, a worldwide used antiarrhythmic drug, safety concerns could be resolved more easily for it than for a novel pharmacological agent.
Collapse
Affiliation(s)
- Zita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
- * E-mail:
| | - Anna Maria Cseh
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Csenge Antus
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Rita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Antal Tapodi
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Fadi H. J. Ramadan
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Balazs Sumegi
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
- MTA-PTE Nuclear-Mitochondrial Interactions Research Group, Pecs, Hungary
- Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
- MTA-PTE Nuclear-Mitochondrial Interactions Research Group, Pecs, Hungary
- Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary
| |
Collapse
|
8
|
Loureiro JB, Abrantes M, Oliveira PA, Saraiva L. P53 in skin cancer: From a master player to a privileged target for prevention and therapy. Biochim Biophys Acta Rev Cancer 2020; 1874:188438. [PMID: 32980466 DOI: 10.1016/j.bbcan.2020.188438] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022]
Abstract
The increasing incidence of skin cancer (SC) is a global health concern. The commonly reported side effects and resistance mechanisms have imposed the pursuit for new therapeutic alternatives. Moreover, additional preventive strategies should be adopted to strengthen prevention and reduce the rising number of newly SC cases. This review provides relevant insights on the role of p53 tumour suppressor protein in melanoma and non-melanoma skin carcinogenesis, also highlighting the therapeutic potential of p53-targeting drugs against SC. In fact, several evidences are provided demonstrating the encouraging outcomes achieved with p53-activating drugs, alone and in combination with currently available therapies in SC. Another pertinent perspective falls on targeting p53 mutations, as molecular signatures in premature phases of photocarcinogenesis, in future SC preventive approaches. Overall, this review affords a critical and timely discussion of relevant issues related to SC prevention and therapy. Importantly, it paves the way to future studies that may boost the clinical translation of p53-activating agents, making them new effective alternatives in precision medicine of SC therapy and prevention.
Collapse
Affiliation(s)
- J B Loureiro
- LAQV/REQUIMTE, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - M Abrantes
- Biophysics Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Clinical Academic Center of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; CNC.IBILI Consortium/Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - P A Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, Universidade de Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - L Saraiva
- LAQV/REQUIMTE, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
| |
Collapse
|
9
|
Takahashi A, Namikawa K, Ogata D, Nakano E, Jinnai S, Nakama K, Tsutsui K, Muto Y, Mizuta H, Yamazaki N. Real-world efficacy and safety data of nivolumab and ipilimumab combination therapy in Japanese patients with advanced melanoma. J Dermatol 2020; 47:1267-1275. [PMID: 32725685 DOI: 10.1111/1346-8138.15521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022]
Abstract
The efficacy and safety of nivolumab + ipilimumab combination therapy were retrospectively examined in Japanese patients with unresectable advanced melanoma in clinical practice. Fifty-seven patients with advanced melanoma received the nivolumab + ipilimumab combination therapy. The primary site was cutaneous, mucosal, uveal and unknown in 35, 16, two and four patients, respectively. The overall response rate was 26.3%, with complete response observed in two (3.5%) patients, partial response in 13 (22.8%), stable disease in 12 (21.1%) and progressive disease in 30 (52.6%). The response rate in the treatment-naive and prior systemic therapy group was 40.7% and 13.3%, respectively. For those treated with a single immune checkpoint inhibitor followed by the nivolumab + ipilimumab combination therapy as second-line therapy after disease progression, the response rate was 18.8%. Median progression-free survival (PFS) and overall survival (OS) in all patients was 3.3 and 14 months, respectively. Median PFS in the treatment-naive and prior systemic therapy groups was 13 and 2 months, respectively. Median OS was unreached in the treatment-naive group and was 6.3 months in prior systemic therapy groups. There was no significant difference in PFS and OS for non-acral, acral and mucosal melanoma. Adverse events occurred in 86% of patients; 56.1% were grade 3 or worse. The response rate in an actual clinical setting, including the prior systemic therapy group, was lower than that in the global study and the Japanese phase II study. However, in the treatment-naive group, the rate was equivalent to that in the Japanese phase II study. PFS and OS in the treatment-naive group were comparable with those in the global study and Japanese phase II study, suggesting that the treatment was effective. The proportion of grade 3 and 4 immune-related adverse events was as high as that in the global study and Japanese phase II study.
Collapse
Affiliation(s)
- Akira Takahashi
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Dai Ogata
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Eiji Nakano
- Department of Dermatology, The Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Shunichi Jinnai
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kenta Nakama
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Keita Tsutsui
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yusuke Muto
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Haruki Mizuta
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
10
|
Fufa TD, Baxter LL, Wedel JC, Gildea DE, Loftus SK, Pavan WJ. MEK inhibition remodels the active chromatin landscape and induces SOX10 genomic recruitment in BRAF(V600E) mutant melanoma cells. Epigenetics Chromatin 2019; 12:50. [PMID: 31399133 PMCID: PMC6688322 DOI: 10.1186/s13072-019-0297-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/28/2019] [Indexed: 01/03/2023] Open
Abstract
Background The MAPK/ERK signaling pathway is an essential regulator of numerous cell processes that are crucial for normal development as well as cancer progression. While much is known regarding MAPK/ERK signal conveyance from the cell membrane to the nucleus, the transcriptional and epigenetic mechanisms that govern gene expression downstream of MAPK signaling are not fully elucidated. Results This study employed an integrated epigenome analysis approach to interrogate the effects of MAPK/ERK pathway inhibition on the global transcriptome, the active chromatin landscape, and protein–DNA interactions in 501mel melanoma cells. Treatment of these cells with the small-molecule MEK inhibitor AZD6244 induces hyperpigmentation, widespread gene expression changes including alteration of genes linked to pigmentation, and extensive epigenomic reprogramming of transcriptionally distinct regulatory regions associated with the active chromatin mark H3K27ac. Regulatory regions with differentially acetylated H3K27ac regions following AZD6244 treatment are enriched in transcription factor binding motifs of ETV/ETS and ATF family members as well as the lineage-determining factors MITF and SOX10. H3K27ac-dense enhancer clusters known as super-enhancers show similar transcription factor motif enrichment, and furthermore, these super-enhancers are associated with genes encoding MITF, SOX10, and ETV/ETS proteins. Along with genome-wide resetting of the active enhancer landscape, MEK inhibition also results in widespread SOX10 recruitment throughout the genome, including increased SOX10 binding density at H3K27ac-marked enhancers. Importantly, these MEK inhibitor-responsive enhancers marked by H3K27ac and occupied by SOX10 are located near melanocyte lineage-specific and pigmentation genes and overlap numerous human SNPs associated with pigmentation and melanoma phenotypes, highlighting the variants located within these regions for prioritization in future studies. Conclusions These results reveal the epigenetic reprogramming underlying the re-activation of melanocyte pigmentation and developmental transcriptional programs in 501mel cells in response to MEK inhibition and suggest extensive involvement of a MEK-SOX10 axis in the regulation of these processes. The dynamic chromatin changes identified here provide a rich genomic resource for further analyses of the molecular mechanisms governing the MAPK pathway in pigmentation- and melanocyte-associated diseases. Electronic supplementary material The online version of this article (10.1186/s13072-019-0297-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Temesgen D Fufa
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA.,Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Laura L Baxter
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Julia C Wedel
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Derek E Gildea
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Stacie K Loftus
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
11
|
Antunes LCM, Cartell A, de Farias CB, Bakos RM, Roesler R, Schwartsmann G. Tropomyosin-Related Kinase Receptor and Neurotrophin Expression in Cutaneous Melanoma Is Associated with a Poor Prognosis and Decreased Survival. Oncology 2019; 97:26-37. [PMID: 31071716 DOI: 10.1159/000499384] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 03/05/2019] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Normally, activation of tropomyosin-related kinase (TRK) receptors by neurotrophins (NTs) stimulates intracellular pathways involved in cell survival and proliferation. Dysregulation of NT/TRK signaling may affect neoplasm prognosis. Data on NT and TRK expression in melanomas are limited, and it is unclear whether NT/TRK signaling pathways are involved in the origin and progression of this neoplasm. METHODS We examined whether NT/TRK expression differs across different cutaneous melanoma grades and subtypes, and whether it is associated with melanoma prognosis and survival. A cross-sectional study was performed in which the expression of TrkA, TrkB, nerve growth factor (NGF), and brain-derived neurotrophic factor (BDNF) was analyzed by immunohistochemistry of 154 melanoma samples. We investigated NT/TRK expression associations with prognostic factors for melanoma, relapse-free survival (RFS), and overall survival (OS). RESULTS Of the 154 melanoma samples, 77 (55.4%) were TrkA immunopositive, 81 (58.3%) were TrkB immunopositive, 113 (81.3%) were BDNF immunopositive, and 104 (75.4%) were NGF immunopositive. We found NT/TRK expression associated strongly with several clinical prognostic factors, including the tumor-node-metastasis stage (p < 0.001), histological subtype (p < 0.001), and Clark level (p < 0.05), as well as with a worse OS (p < 0.05 for all, except TrkB) and RFS (p < 0.05 for all). CONCLUSIONS Our results show strong associations of NT/TRK expression with melanoma stage progression and a poor prognosis.
Collapse
Affiliation(s)
- Luís Carlos Moreira Antunes
- Graduate Program in Medical Sciences, Faculty of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil,
- Hematology and Oncology Service, Santa Maria University Hospital, Federal University of Santa Maria, Santa Maria, Brazil,
| | - André Cartell
- Department of Pathology, Porto Alegre Clinical Hospital, Porto Alegre, Brazil
| | - Caroline Brunetto de Farias
- Cancer and Neurobiology Laboratory, Experimental Research Center, Porto Alegre Clinical Hospital, Porto Alegre, Brazil
| | - Renato Marchiori Bakos
- Graduate Program in Medical Sciences, Faculty of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Department of Dermatology, Porto Alegre Clinical Hospital, Porto Alegre, Brazil
| | - Rafael Roesler
- Cancer and Neurobiology Laboratory, Experimental Research Center, Porto Alegre Clinical Hospital, Porto Alegre, Brazil
- Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Gilberto Schwartsmann
- Graduate Program in Medical Sciences, Faculty of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Cancer and Neurobiology Laboratory, Experimental Research Center, Porto Alegre Clinical Hospital, Porto Alegre, Brazil
- Department of Internal Medicine, Faculty of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
12
|
Abstract
OPINION STATEMENT Melanoma has several clinically and pathologically distinguishable subtypes, which also differ genetically. Mutation patterns vary among different melanoma subtypes, and efficacy of immune-checkpoint inhibitors differs depending on the subtype of melanoma. In spite of the recent revolution of systemic therapies for advanced melanoma, access to innovative agents is still restricted in many countries. This review article aimed to describe the epidemiology and current status of systemic therapies for melanoma in Japan, where melanoma is rare, but access to innovative agents is available. Acral and mucosal melanomas, which are common in Asian populations, predominantly occur in sun-protected areas and share several biological features. Both the melanomas harbor KIT mutation in approximately 15% of the cases; BRAF or NRAS mutation is found in approximately 10-15% of acral melanoma, but these mutations are less frequent in mucosal melanoma. Combined use of BRAF and MEK inhibitors is one of the standards of care for patients with advanced BRAF-mutant melanoma. In patients with melanoma harboring KIT mutation in exon 11 or 13, KIT inhibitors can be a treatment option; however, none of them have been approved in Japan. Immune-checkpoint inhibitors are expected to be less effective against acral and mucosal melanomas because their somatic mutation burden is lower than those in non-acral cutaneous melanomas. A recently completed phase II trial of nivolumab and ipilimumab combination therapy in 30 Japanese patients with melanoma, including seven with acral and 12 with mucosal melanoma, demonstrated an objective response rate of 43%. Regarding oncolytic viruses, canerpaturev (C-REV, also known as HF10) and talimogene laherparepvec (T-VEC) are currently under review in early phase trials. In the adjuvant setting, dabrafenib plus trametinb combination, nivolumab monotherapy, and pembrolizumab monotherapy were approved in July, August, and December 2018 in Japan, respectively. However, most of the adjuvant phase III trials excluded patients with mucosal melanoma. A phase III trial of adjuvant therapy with locoregional interferon (IFN)-β versus surgery alone is ongoing in Japan (JCOG1309, J-FERON), in which IFN-β is injected directly into the site of the primary tumor postoperatively, so that it would be drained through the untreated lymphatic route to the regional node basin. After the recent approval of these new agents, the JCOG1309 trial will be revised to focus on patients with stage II disease. In conclusion, acral and mucosal melanomas have been treated based on the available medical evidence for the treatment of non-acral cutaneous melanomas. Considering the differences in genetic backgrounds and therapeutic efficacy of immunotherapy, specialized therapeutic strategies for these subtypes of melanoma should be established in the future.
Collapse
Affiliation(s)
- Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| |
Collapse
|
13
|
Namikawa K, Kiyohara Y, Takenouchi T, Uhara H, Uchi H, Yoshikawa S, Takatsuka S, Koga H, Wada N, Minami H, Hatsumichi M, Asada S, Namba Y, Yamazaki N. Efficacy and safety of nivolumab in combination with ipilimumab in Japanese patients with advanced melanoma: An open-label, single-arm, multicentre phase II study. Eur J Cancer 2018; 105:114-126. [PMID: 30447539 DOI: 10.1016/j.ejca.2018.09.025] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 09/23/2018] [Indexed: 12/19/2022]
Abstract
AIM The aim of the study was to evaluate the efficacy and safety of nivolumab combined with ipilimumab in treatment-naïve Japanese patients with advanced melanoma. METHODS In this multicentre, single-arm study, treatment-naïve Japanese patients with unresectable stage III/IV or recurrent melanoma received nivolumab (1 mg/kg) plus ipilimumab (3 mg/kg) every 3 weeks for four doses, followed by biweekly doses of nivolumab (3 mg/kg). The primary end-point was centrally assessed objective response rate (ORR). Secondary end-points included overall survival (OS), progression-free survival (PFS), disease control rate and safety. RESULTS The subtypes of the thirty patients enrolled were: 12, mucosal; eight, non-acral cutaneous; seven, acral; two, uveal and one, unknown primary melanoma. The ORR was 43.3% (95% confidence interval [CI]: 25.5, 62.6) with central and local assessment. The centrally and locally assessed disease control rate (95% CI) were 73.3% (54.1, 87.7) and 86.7% (69.3, 96.2), respectively. At the median follow-up period of 14.1 months (range 5.2-27.7), median OS and centrally assessed PFS were not reached. OS (95% CI) at 6, 12, 18 and 24 months was 93.3% (75.9, 98.3), 83.3% (64.5, 92.7), 72.9% (50.0, 86.5) and 65.6% (40.4, 82.2), respectively. Treatment-related adverse events (AEs) occurred in all patients. Grade III-IV and serious AEs occurred, mostly during the combination phase, in 23 (76.7%) and 20 (66.7%) patients, respectively. No treatment-related deaths occurred. CONCLUSIONS This study confirmed the efficacy and safety of nivolumab plus ipilimumab in treatment-naïve Japanese patients with advanced melanoma including rare subtypes. Incidence rates for grade III-IV AEs were high but manageable with appropriate medical attention and treatment. TRIAL REGISTRATION JapicCTI-152869.
Collapse
Affiliation(s)
- Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Yoshio Kiyohara
- Dermatology Division, Shizuoka Cancer Center Hospital, 1007 Shimonagakubo, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan.
| | - Tatsuya Takenouchi
- Department of Dermatology, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Niigata 961-8566, Japan.
| | - Hisashi Uhara
- Department of Dermatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| | - Hiroshi Uchi
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Shusuke Yoshikawa
- Dermatology Division, Shizuoka Cancer Center Hospital, 1007 Shimonagakubo, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan.
| | - Sumiko Takatsuka
- Department of Dermatology, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Niigata 961-8566, Japan.
| | - Hiroshi Koga
- Department of Dermatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| | - Naoko Wada
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Hironobu Minami
- Department Medical Oncology/Hematology, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan.
| | - Masahiro Hatsumichi
- Ono Pharmaceutical Co. Ltd., 1-8-2 Kyutaromachi, Chuo-ku, Osaka 541-8564, Japan.
| | - Suguru Asada
- Ono Pharmaceutical Co. Ltd., 1-8-2 Kyutaromachi, Chuo-ku, Osaka 541-8564, Japan.
| | - Yoshinobu Namba
- Ono Pharmaceutical Co. Ltd., 1-8-2 Kyutaromachi, Chuo-ku, Osaka 541-8564, Japan.
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| |
Collapse
|
14
|
Dimitriou F, Krattinger R, Ramelyte E, Barysch MJ, Micaletto S, Dummer R, Goldinger SM. The World of Melanoma: Epidemiologic, Genetic, and Anatomic Differences of Melanoma Across the Globe. Curr Oncol Rep 2018; 20:87. [PMID: 30250984 DOI: 10.1007/s11912-018-0732-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW As cancer remains an increasing problem in industrial countries, the incidence of melanoma has risen rapidly in many populations during the last decades and still continues to rise. Current strategies aiming to control the disease have largely focused on improving the understanding of the interplay of causal factors for this cancer. RECENT FINDINGS Cutaneous melanoma shows clear differences in incidence, mortality, genomic profile, and anatomic presentation, depending on the country of residence, ethnicity, and socioeconomic status. Known risk factors are multiple atypical nevi, positive family and/or personal history, immune suppressive diseases or treatments, and fair skin phenotype. Besides new adjuvant therapeutic options, changed attitude toward leisure and sun exposure, primary prevention, and early detection are major contributors to disease control. Melanoma is a disease of multifactorial causality and heterogeneous presentation. Its subtypes differ in origin, anatomical site, role of UV radiation, and mutational profile. Better understanding of these differences may improve prevention strategies and therapeutic developments.
Collapse
Affiliation(s)
- Florentia Dimitriou
- Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091, Zurich, Switzerland
| | - Regina Krattinger
- Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091, Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091, Zurich, Switzerland
| | - Marjam J Barysch
- Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091, Zurich, Switzerland
| | - Sara Micaletto
- Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091, Zurich, Switzerland
| | - Simone M Goldinger
- Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091, Zurich, Switzerland.
| |
Collapse
|
15
|
Zuo Q, Liu J, Huang L, Qin Y, Hawley T, Seo C, Merlino G, Yu Y. AXL/AKT axis mediated-resistance to BRAF inhibitor depends on PTEN status in melanoma. Oncogene 2018; 37:3275-3289. [PMID: 29551771 DOI: 10.1038/s41388-018-0205-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 12/25/2017] [Accepted: 02/13/2018] [Indexed: 12/21/2022]
Abstract
Multiple genetic mutations within melanoma not only cause lesion-specific responses to targeted therapy but also alter the molecular route of resistance to that therapy. Inactivation of PTEN occurs in up to 30% of melanomas, frequently with a concurrent activating BRAF mutation. PTEN loss regulates both acquired and intrinsic drug resistance. Here we show that AXL/AKT axis mediated-resistance to BRAF inhibitor (BRAFi) depends upon PTEN status in melanoma. Hyperactivation of both ERK and AKT pathways was associated with BRAFi resistance in melanoma with wildtype PTEN. The PTEN-impaired melanoma cells required only the ERK resistance mechanism. Moreover, we identified AXL as a key upstream effector of AKT pathway-associated resistance to BRAFi in melanoma with wildtype PTEN, but not in melanoma with impaired PTEN. Notably, we confirmed that blocking AXL by shRNA and a small molecular inhibitor could rescue the sensitivity of resistant melanoma cells with wildtype PTEN to BRAFi and inhibit their growth in vitro and in vivo. Our study has uncovered a mechanism by which PTEN status contributes to acquired resistance to BRAFi and offers a rational strategy to guide clinical testing in pre-identified subsets of patients who relapse during treatment with BRAFi. The identified protein AXL represents a promising therapeutic target for BRAF mutant melanoma patients with wildtype PTEN.
Collapse
Affiliation(s)
- Qiang Zuo
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institutes, National Institutes of Health, Bethesda, MD, 20892-4264, USA.,Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Jing Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Liping Huang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institutes, National Institutes of Health, Bethesda, MD, 20892-4264, USA.,Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Yifei Qin
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institutes, National Institutes of Health, Bethesda, MD, 20892-4264, USA
| | - Teresa Hawley
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institutes, National Institutes of Health, Bethesda, MD, 20892-4264, USA
| | - Claire Seo
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institutes, National Institutes of Health, Bethesda, MD, 20892-4264, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institutes, National Institutes of Health, Bethesda, MD, 20892-4264, USA
| | - Yanlin Yu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institutes, National Institutes of Health, Bethesda, MD, 20892-4264, USA.
| |
Collapse
|
16
|
Han X, Han Y, Zheng Y, Sun Q, Ma T, Dai L, Zhang J, Xu L. Use of phosphodiesterase type 5 inhibitors and risk of melanoma: a meta-analysis of observational studies. Onco Targets Ther 2018; 11:711-720. [PMID: 29440918 PMCID: PMC5804137 DOI: 10.2147/ott.s142637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Phosphodiesterase type 5 inhibitor (PE5i) administration may stimulate the proliferation and survival of melanocytes. However, discrepancies remain regarding the association between PDE5i use and melanoma risk in observational studies in humans. Aim To evaluate the association between PDE5i use and melanoma in a meta-analysis. Materials and methods Studies were identified by searching the PubMed and Embase databases. A random-effects model was applied to synthesize the data. A stratified study was performed to evaluate the influence of study characteristics on outcomes. Results Four prospective cohort studies and three case–control studies with 1,534,615 male participants and 16,053 melanoma cases were incorporated. Patients who received a PDE5i had a significantly increased risk for melanoma (adjusted risk ratio [RR] =1.12, 95% CI =1.03–1.33, P=0.008) with moderate heterogeneity (I2=54%). Cohort studies (adjusted RR =1.22, 95% CI =1.02–1.46, P=0.03) largely contributed to this result rather than case–control studies. Subsequent stratified analyses revealed that sildenafil was associated with an increased risk of melanoma (adjusted RR =1.26, 95% CI =1.07–1.50, P=0.007), but tadalafil and vardenafil were not. Also, PDE5i use was associated with a significantly increased risk of in situ melanoma (adjusted RR =1.31, 95% CI =1.01–1.69, P=0.04), but not of localized or nonlocalized melanoma. Conclusion PDE5i use may be associated with a significantly increased risk for melanoma in men. However, further research is needed to determine whether the association is causative.
Collapse
Affiliation(s)
- Xinming Han
- Plastic Surgery Department, Beijing Tongren Hospital, Capital Medical University, Beijing
| | - Yan Han
- Plastic and Reconstructive Surgery Department, Chinese PLA General Hospital, Beijing, China
| | - Yongsheng Zheng
- Plastic Surgery Department, Beijing Tongren Hospital, Capital Medical University, Beijing
| | - Qiang Sun
- Plastic Surgery Department, Beijing Tongren Hospital, Capital Medical University, Beijing
| | - Tao Ma
- Plastic Surgery Department, Beijing Tongren Hospital, Capital Medical University, Beijing
| | - Li Dai
- Plastic Surgery Department, Beijing Tongren Hospital, Capital Medical University, Beijing
| | - Junyi Zhang
- Plastic Surgery Department, Beijing Tongren Hospital, Capital Medical University, Beijing
| | - Lianji Xu
- Plastic Surgery Department, Beijing Tongren Hospital, Capital Medical University, Beijing
| |
Collapse
|
17
|
Bruno W, Martinuzzi C, Andreotti V, Pastorino L, Spagnolo F, Dalmasso B, Cabiddu F, Gualco M, Ballestrero A, Bianchi-Scarrà G, Queirolo P, Grillo F, Mastracci L, Ghiorzo P. Heterogeneity and frequency of BRAF mutations in primary melanoma: Comparison between molecular methods and immunohistochemistry. Oncotarget 2017; 8:8069-8082. [PMID: 28039443 PMCID: PMC5352383 DOI: 10.18632/oncotarget.14094] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/24/2016] [Indexed: 11/25/2022] Open
Abstract
Finding the best technique to identify BRAF mutations with a high sensitivity and specificity is mandatory for accurate patient selection for target therapy. BRAF mutation frequency ranges from 40 to 60% depending on melanoma clinical characteristics and detection technique used.Intertumoral heterogeneity could lead to misinterpretation of BRAF mutational status; this is especially important if testing is performed on primary specimens, when metastatic lesions are unavailable.Aim of this study was to identify the best combination of methods for detecting BRAF mutations (among peptide nucleic acid - PNA-clamping real-time PCR, immunohistochemistry and capillary sequencing) and investigate BRAF mutation heterogeneity in a series of 100 primary melanomas and a subset of 25 matched metastatic samples.Overall, we obtained a BRAF mutation frequency of 62%, based on the combination of at least two techniques. Concordance between mutation status in primary and metastatic tumor was good but not complete (67%), when agreement of at least two techniques were considered. Next generation sequencing was used to quantify the threshold of detected mutant alleles in discordant samples. Combining different methods excludes that the observed heterogeneity is technique-based. We propose an algorithm for BRAF mutation testing based on agreement between immunohistochemistry and PNA; a third molecular method could be added in case of discordance of the results. Testing the primary tumor when the metastatic sample is unavailable is a good option if at least two methods of detection are used, however the presence of intertumoral heterogeneity or the occurrence of additional primaries should be carefully considered.
Collapse
Affiliation(s)
- William Bruno
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Claudia Martinuzzi
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Virginia Andreotti
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Lorenza Pastorino
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Bruna Dalmasso
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Marina Gualco
- Department of Pathology, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Alberto Ballestrero
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Giovanna Bianchi-Scarrà
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Paola Queirolo
- Department of Medical Oncology, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Federica Grillo
- Department of Surgical and Diagnostic Sciences, Pathology Unit, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Luca Mastracci
- Department of Surgical and Diagnostic Sciences, Pathology Unit, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| |
Collapse
|
18
|
Potential therapeutic targets of epithelial-mesenchymal transition in melanoma. Cancer Lett 2017; 391:125-140. [PMID: 28131904 DOI: 10.1016/j.canlet.2017.01.029] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/02/2017] [Accepted: 01/18/2017] [Indexed: 12/16/2022]
Abstract
Melanoma is a cutaneous neoplastic growth of melanocytes with great potential to invade and metastasize, especially when not treated early and effectively. Epithelial-mesenchymal transition (EMT) is the process by which melanocytes lose their epithelial characteristics and acquire mesenchymal phenotypes. Mesenchymal protein expression increases the motility, invasiveness, and metastatic potential of melanoma. Many pathways play a role in promotion of mesenchymal protein expression including RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, Wnt/β-catenin, and several others. Downstream effectors of these pathways induce expression of EMT transcription factors including Snail, Slug, Twist, and Zeb that promote repression of epithelial and induction of mesenchymal character. Emerging research has demonstrated that a variety of small molecule inhibitors as well as phytochemicals can influence the progression of EMT and may even reverse the process, inducing re-expression of epithelial markers. Phytochemicals are of particular interest as supplementary treatment options because of their relatively low toxicities and anti-EMT properties. Modulation of EMT signaling pathways using synthetic small molecules and phytochemicals is a potential therapeutic strategy for reducing the aggressive progression of metastatic melanoma. In this review, we discuss the emerging pathways and transcription factor targets that regulate EMT and evaluate potential synthetic small molecules and naturally occurring compounds that may reduce metastatic melanoma progression.
Collapse
|
19
|
Oliveira S, Coelho P, Prudêncio C, Vieira M, Soares R, Guerreiro SG, Fernandes R. Melanoma and obesity: Should antioxidant vitamins be addressed? Life Sci 2016; 165:83-90. [DOI: 10.1016/j.lfs.2016.09.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 09/20/2016] [Accepted: 09/22/2016] [Indexed: 01/14/2023]
|
20
|
Harris Z, Donovan MG, Branco GM, Limesand KH, Burd R. Quercetin as an Emerging Anti-Melanoma Agent: A Four-Focus Area Therapeutic Development Strategy. Front Nutr 2016; 3:48. [PMID: 27843913 PMCID: PMC5086580 DOI: 10.3389/fnut.2016.00048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/10/2016] [Indexed: 12/21/2022] Open
Abstract
Replacing current refractory treatments for melanoma with new prevention and therapeutic approaches is crucial in order to successfully treat this aggressive cancer form. Melanoma develops from neural crest cells, which express tyrosinase – a key enzyme in the pigmentation pathway. The tyrosinase enzyme is highly active in melanoma cells and metabolizes polyphenolic compounds; tyrosinase expression thus makes feasible a target for polyphenol-based therapies. For example, quercetin (3,3′,4′,5,7-pentahydroxyflavone) is a highly ubiquitous and well-classified dietary polyphenol found in various fruits, vegetables, and other plant products including onions, broccoli, kale, oranges, blueberries, apples, and tea. Quercetin has demonstrated antiproliferative and proapoptotic activity in various cancer cell types. Quercetin is readily metabolized by tyrosinase into various compounds that promote anticancer activity; additionally, given that tyrosinase expression increases during tumorigenesis, and its activity is associated with pigmentation changes in both early- and late-stage melanocytic lesions, it suggests that quercetin can be used to target melanoma. In this review, we explore the potential of quercetin as an anti-melanoma agent utilizing and extrapolating on evidence from previous in vitro studies in various human malignant cell lines and propose a “four-focus area strategy” to develop quercetin as a targeted anti-melanoma compound for use as either a preventative or therapeutic agent. The four areas of focus include utilizing quercetin to (i) modulate cellular bioreduction potential and associated signaling cascades, (ii) affect transcription of relevant genes, (iii) regulate epigenetic processes, and (iv) develop effective combination therapies and delivery modalities/protocols. In general, quercetin could be used to exploit tyrosinase activity to prevent, and/or treat, melanoma with minimal additional side effects.
Collapse
Affiliation(s)
- Zoey Harris
- Department of Nutritional Sciences, University of Arizona , Tucson, AZ , USA
| | - Micah G Donovan
- Department of Nutritional Sciences, University of Arizona , Tucson, AZ , USA
| | | | - Kirsten H Limesand
- Department of Nutritional Sciences, University of Arizona , Tucson, AZ , USA
| | - Randy Burd
- Department of Nutritional Sciences, University of Arizona , Tucson, AZ , USA
| |
Collapse
|
21
|
Li J, Li J, Aipire A, Gao L, Huo S, Luo J, Zhang F. Phenylethanoid Glycosides from Cistanche tubulosa Inhibits the Growth of B16-F10 Cells both in Vitro and in Vivo by Induction of Apoptosis via Mitochondria-dependent Pathway. J Cancer 2016; 7:1877-1887. [PMID: 27698928 PMCID: PMC5039372 DOI: 10.7150/jca.15512] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 07/24/2016] [Indexed: 01/06/2023] Open
Abstract
Cistanche tubulosa phenylethanoid glycosides (CTPG) have been shown various biological activities including anti-allergy, hepatoprotective activity and bone regeneration. However, the anti-tumor activity of CTPG needs to be investigated. CTPG was used to treat B16-F10 cells both in vitro and in vivo. We found that CTPG dramatically changed the morphology of B16-F10 cells, and significantly reduced the viability of B16-F10 cells in a dose-dependent and time-dependent manner, which might be mediated by CTPG-induced apoptosis and cell cycle arrest. After CTPG treatment, the expressions of BAX and BCL-2 were up-regulated and down-regulated, respectively. Moreover, mitochondrial membrane potential was reduced and ROS generation was increased. Consequently, the levels of cytochrome c and cleaved-caspase-3 and -9 were up-regulated by CTPG treatment but not for cleaved-caspase-8. We further observed that CTPG significantly inhibited the tumor growth in vivo and improved the survival rate of tumor mice. We also observed that CTPG promoted the proliferation of splenocytes and increased the proportions of CD4+ and CD8+ T cells in spleens of tumor mice. The results showed that CTPG induced the apoptosis of B16-F10 cells through mitochondria-dependent pathway, suggesting that CTPG could be a potential candidate for treatment of cancer.
Collapse
Affiliation(s)
- Jinyu Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Adila Aipire
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Li Gao
- Xinjiang Laboratory of Uyghur Medical Prescription, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, Xinjiang, China
| | - Shixia Huo
- Xinjiang Laboratory of Uyghur Medical Prescription, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, Xinjiang, China
| | - Jiaojiao Luo
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Fuchun Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| |
Collapse
|
22
|
Manzano JL, Layos L, Bugés C, de Los Llanos Gil M, Vila L, Martínez-Balibrea E, Martínez-Cardús A. Resistant mechanisms to BRAF inhibitors in melanoma. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:237. [PMID: 27429963 DOI: 10.21037/atm.2016.06.07] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Patients with advanced melanoma have traditionally had very poor prognosis. However, since 2011 better understanding of the biology and epidemiology of this disease has revolutionized its treatment, with newer therapies becoming available. These newer therapies can be classified into immunotherapy and targeted therapy. The immunotherapy arsenal includes inhibitors of CTLA4, PD-1 and PDL-1, while targeted therapy focuses on BRAF and MEK. BRAF inhibitors (vemurafenib, dabrafenib) have shown benefit in terms of overall survival (OS) compared to chemotherapy, and their combination with MEK inhibitors has recently been shown to improve progression-free survival (PFS), compared with monotherapy with BRAF inhibitors. However, almost 20% of patients initially do not respond, due to intrinsic resistance to therapy and, of those who do, most eventually develop mechanisms of acquired resistance, including reactivation of the MAP kinase pathway, persistent activation of receptor tyrosine kinase (RTKS) receptor, activation of phosphatidyinositol-3OH kinase, overexpression of epidermal growth factor receptor (EGFR), and interactions with the tumor microenvironment. Herein we comment in detail on mechanisms of resistance to targeted therapy and discuss the strategies to overcome them.
Collapse
Affiliation(s)
- José Luís Manzano
- Medical Oncology Service, Catalan Institute of Oncology (ICO), Germans Trias i Pujol University Hospital, Badalona, Barcelona, Catalonia, Spain;; Health Sciences Research Institute of the Germans Trias i Pujol Foundation (IGTP), Badalona, Catalonia, Spain
| | - Laura Layos
- Medical Oncology Service, Catalan Institute of Oncology (ICO), Germans Trias i Pujol University Hospital, Badalona, Barcelona, Catalonia, Spain
| | - Cristina Bugés
- Medical Oncology Service, Catalan Institute of Oncology (ICO), Germans Trias i Pujol University Hospital, Badalona, Barcelona, Catalonia, Spain
| | - María de Los Llanos Gil
- Medical Oncology Service, Catalan Institute of Oncology (ICO), Germans Trias i Pujol University Hospital, Badalona, Barcelona, Catalonia, Spain
| | - Laia Vila
- Medical Oncology Service, Catalan Institute of Oncology (ICO), Germans Trias i Pujol University Hospital, Badalona, Barcelona, Catalonia, Spain
| | - Eva Martínez-Balibrea
- Health Sciences Research Institute of the Germans Trias i Pujol Foundation (IGTP), Badalona, Catalonia, Spain
| | - Anna Martínez-Cardús
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| |
Collapse
|
23
|
Xia Y, Li Y, Westover KD, Sun J, Chen H, Zhang J, Fisher DE. Inhibition of Cell Proliferation in an NRAS Mutant Melanoma Cell Line by Combining Sorafenib and α-Mangostin. PLoS One 2016; 11:e0155217. [PMID: 27152946 PMCID: PMC4859503 DOI: 10.1371/journal.pone.0155217] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/26/2016] [Indexed: 12/19/2022] Open
Abstract
α-Mangostin is a natural product commonly used in Asia for cosmetic and medicinal applications including topical treatment of acne and skin cancer. Towards finding new pharmacological strategies that overcome NRAS mutant melanoma, we performed a cell proliferation-based combination screen using a collection of well-characterized small molecule kinase inhibitors and α-Mangostin. We found that α-Mangostin significantly enhances Sorafenib pharmacological efficacy against an NRAS mutant melanoma cell line. The synergistic effects of α-Mangostin and Sorafenib were associated with enhanced inhibition of activated AKT and ERK, induced ER stress, and reduced autophagy, eventually leading to apoptosis. The structure of α-Mangostin resembles several inhibitors of the Retinoid X receptor (RXR). MITF expression, which is regulated by RXR, was modulated by α-Mangostin. Molecular docking revealed that α-Mangostin can be accommodated by the ligand binding pocket of RXR and may thereby compete with RXR-mediated control of MITF expression. In summary, these data demonstrate an unanticipated synergy between α-Mangostin and sorafenib, with mechanistic actions that convert a known safe natural product to a candidate combinatorial therapeutic agent.
Collapse
Affiliation(s)
- Yun Xia
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cutaneous Biology Research Center, Massachusetts General Hospital, 149 Building 13th ST, Charlestown, Massachusetts, United States of America
| | - Ying Li
- Cutaneous Biology Research Center, Massachusetts General Hospital, 149 Building 13th ST, Charlestown, Massachusetts, United States of America
- Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, No.7 Front Kangfu ST, Zhengzhou 450052, China
| | - Kenneth D. Westover
- Departments of Biochemistry and Radiation Oncology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongxiang Chen
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cutaneous Biology Research Center, Massachusetts General Hospital, 149 Building 13th ST, Charlestown, Massachusetts, United States of America
| | - Jianming Zhang
- Cutaneous Biology Research Center, Massachusetts General Hospital, 149 Building 13th ST, Charlestown, Massachusetts, United States of America
| | - David E. Fisher
- Cutaneous Biology Research Center, Massachusetts General Hospital, 149 Building 13th ST, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
24
|
Castiglione R, Ihle MA, Heydt C, Schultheis AM, Merkelbach-Bruse S, Mauch C, Büttner R. The impact of sequencing on diagnosis and treatment of malignant melanoma. Expert Rev Mol Diagn 2016; 16:423-33. [PMID: 26822148 DOI: 10.1586/14737159.2016.1147958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Melanoma is one of the clinically most important cancer types considering its high mortality rate and that it is commonly diagnosed in relatively young people. With the advent of targeted therapies and, more recently, immune checkpoint inhibitors, more treatment options are available resulting in higher patient survival rates. However, the successful application of these targeted therapies critically depends on the reliable detection of molecular aberrations. Today, massively parallel sequencing techniques enable us to analyze large sets of genes in a relatively short time. It has allowed increased knowledge of acquired somatic mutations in melanoma and has helped to identify new targets for personalized therapy, and potentially may help to predict response to immune therapies. Described here are the development of sequencing techniques, how their improvement has changed diagnosis, prognosis and management of malignant melanoma and the future perspectives of melanoma diagnostics in the routine clinical setting.
Collapse
Affiliation(s)
| | - Michaela A Ihle
- a Institute of Pathology , University Hospital Cologne , Cologne , Germany
| | - Carina Heydt
- a Institute of Pathology , University Hospital Cologne , Cologne , Germany
| | - Anne M Schultheis
- a Institute of Pathology , University Hospital Cologne , Cologne , Germany
| | | | - Cornelia Mauch
- b Clinic for Dermatology , University Hospital Cologne , Cologne , Germany
| | - Reinhard Büttner
- a Institute of Pathology , University Hospital Cologne , Cologne , Germany
| |
Collapse
|
25
|
Abstract
Tenascin-C (TNC), a multifunctional matricellular glyco-protein, is highly expressed in the majority of melanoma cell lines and has been implicated in the progression of melanoma. A growing body of evidence has implicated the role of TNC in the process of invasion and metastasis for melanoma. However, the mechanism and individual signaling pathways by which TNC drives melanoma progression have not been illuminated. Herein we provide perspectives from the investigation of TNC in other settings that may hint at the mechanistic role of TNC in this disease.
Collapse
|
26
|
Lee JJ, Sholl LM, Lindeman NI, Granter SR, Laga AC, Shivdasani P, Chin G, Luke JJ, Ott PA, Hodi FS, Mihm MC, Lin JY, Werchniak AE, Haynes HA, Bailey N, Liu R, Murphy GF, Lian CG. Targeted next-generation sequencing reveals high frequency of mutations in epigenetic regulators across treatment-naïve patient melanomas. Clin Epigenetics 2015; 7:59. [PMID: 26221190 PMCID: PMC4517542 DOI: 10.1186/s13148-015-0091-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/27/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Recent developments in genomic sequencing have advanced our understanding of the mutations underlying human malignancy. Melanoma is a prototype of an aggressive, genetically heterogeneous cancer notorious for its biologic plasticity and predilection towards developing resistance to targeted therapies. Evidence is rapidly accumulating that dysregulated epigenetic mechanisms (DNA methylation/demethylation, histone modification, non-coding RNAs) may play a central role in the pathogenesis of melanoma. Therefore, we sought to characterize the frequency and nature of mutations in epigenetic regulators in clinical, treatment-naïve, patient melanoma specimens obtained from one academic institution. RESULTS Targeted next-generation sequencing for 275 known and investigative cancer genes (of which 41 genes, or 14.9 %, encoded an epigenetic regulator) of 38 treatment-naïve patient melanoma samples revealed that 22.3 % (165 of 740) of all non-silent mutations affected an epigenetic regulator. The most frequently mutated genes were BRAF, MECOM, NRAS, TP53, MLL2, and CDKN2A. Of the 40 most commonly mutated genes, 12 (30.0 %) encoded epigenetic regulators, including genes encoding enzymes involved in histone modification (MECOM, MLL2, SETD2), chromatin remodeling (ARID1B, ARID2), and DNA methylation and demethylation (TET2, IDH1). Among the 38 patient melanoma samples, 35 (92.1 %) harbored at least one mutation in an epigenetic regulator. The genes with the highest number of total UVB-signature mutations encoded epigenetic regulators, including MLL2 (100 %, 16 of 16) and MECOM (82.6 %, 19 of 23). Moreover, on average, epigenetic genes harbored a significantly greater number of UVB-signature mutations per gene than non-epigenetic genes (3.7 versus 2.4, respectively; p = 0.01). Bioinformatics analysis of The Cancer Genome Atlas (TCGA) melanoma mutation dataset also revealed a frequency of mutations in the 41 epigenetic genes comparable to that found within our cohort of patient melanoma samples. CONCLUSIONS Our study identified a high prevalence of somatic mutations in genes encoding epigenetic regulators, including those involved in DNA demethylation, histone modification, chromatin remodeling, and microRNA processing. Moreover, UVB-signature mutations were found more commonly among epigenetic genes than in non-epigenetic genes. Taken together, these findings further implicate epigenetic mechanisms, particularly those involving the chromatin-remodeling enzyme MECOM/EVI1 and histone-modifying enzyme MLL2, in the pathobiology of melanoma.
Collapse
Affiliation(s)
- Jonathan J. Lee
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Lynette M. Sholl
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Neal I. Lindeman
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Scott R. Granter
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Alvaro C. Laga
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Priyanka Shivdasani
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Gary Chin
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Jason J. Luke
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Patrick A. Ott
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - F. Stephen Hodi
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Martin C. Mihm
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Jennifer Y. Lin
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Andrew E. Werchniak
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Harley A. Haynes
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Nancy Bailey
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Robert Liu
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - George F. Murphy
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Christine G. Lian
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| |
Collapse
|
27
|
Abstract
Spitzoid melanomas (SM) and atypical Spitz tumors (AST) are rare pediatric neoplasms. We performed a retrospective, single-institution review and report our institutional experience. We identified 10 patients (median age: 12.5 years). A sentinel node biopsy (SNB) was performed in 8/10 (80%) patients, and interestingly 7/8 (87.5%) were found to be positive for malignant cells. A complete regional lymphadenectomy was performed in all SNB-positive patients, but only 2/8 (25%) were found to have additional lymph node spread. Adjuvant therapy was administered in 5/8 SLNB-positive and 2/2 (100%) regional LN-positive cases. All patients had excellent long-term outcomes (100% survival). This report highlights the excellent outcomes associated with SNB + pediatric SM and AST.
Collapse
Affiliation(s)
- Sandeep Batra
- Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children, 705 Riley Hospital Drive, Indianapolis, IN 46202, USA.,Division of Pediatric Hematology-Oncology, Indiana University School of Medicine, Riley Hospital for Children, 705 Riley Hospital Drive, Indianapolis, IN 46202, USA.,Hematopoiesis, Hematologic Malignancies & Immunology, Indiana University Melvin & Bren Simon Cancer Center, 535 Barnhill Drive, Indianapolis, IN 46202, USA.,Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children, 705 Riley Hospital Drive, Indianapolis, IN 46202, USA
| |
Collapse
|
28
|
Sullivan R, LoRusso P, Boerner S, Dummer R. Achievements and challenges of molecular targeted therapy in melanoma. Am Soc Clin Oncol Educ Book 2015:177-186. [PMID: 25993155 DOI: 10.14694/edbook_am.2015.35.177] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The treatment of melanoma has been revolutionized over the past decade with the development of effective molecular and immune targeted therapies. The great majority of patients with melanoma have mutations in oncogenes that predominantly drive signaling through the mitogen activated protein kinase (MAPK) pathway. Analytic tools have been developed that can effectively stratify patients into molecular subsets based on the identification of mutations in oncogenes and/or tumor suppressor genes that drive the MAPK pathway. At the same time, potent and selective inhibitors of mediators of the MAPK pathway such as RAF, MEK, and ERK have become available. The most dramatic example is the development of single-agent inhibitors of BRAF (vemurafenib, dabrafenib, encorafenib) and MEK (trametinib, cobimetinib, binimetinib) for patients with metastatic BRAFV600-mutant melanoma, a subset that represents 40% to 50% of patients with metastatic melanoma. More recently, the elucidation of mechanisms underlying resistance to single-agent BRAF inhibitor therapy led to a second generation of trials that demonstrated the superiority of BRAF inhibitor/MEK inhibitor combinations (dabrafenib/trametinib; vemurafenib/cobimetinib) compared to single-agent BRAF inhibitors. Moving beyond BRAFV600 targeting, a number of other molecular subsets--such as mutations in MEK, NRAS, and non-V600 BRAF and loss of function of the tumor suppressor neurofibromatosis 1 (NF1)--are predicted to respond to MAPK pathway targeting by single-agent pan-RAF, MEK, or ERK inhibitors. As these strategies are being tested in clinical trials, preclinical and early clinical trial data are now emerging about which combinatorial approaches might be best for these patients.
Collapse
Affiliation(s)
- Ryan Sullivan
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Patricia LoRusso
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Scott Boerner
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|