1
|
Sun JW, Zou J, Zheng Y, Yuan H, Xie YZY, Wang XN, Ou TM. Design, synthesis, and evaluation of novel quindoline derivatives with fork-shaped side chains as RNA G-quadruplex stabilizers for repressing oncogene NRAS translation. Eur J Med Chem 2024; 271:116406. [PMID: 38688064 DOI: 10.1016/j.ejmech.2024.116406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 05/02/2024]
Abstract
NRAS mutation is the second most common oncogenic factor in cutaneous melanoma. Inhibiting NRAS translation by stabilizing the G-quadruplex (G4) structure with small molecules seems to be a potential strategy for cancer therapy due to the NRAS protein's lack of a druggable pocket. To enhance the effects of previously reported G4 stabilizers quindoline derivatives, we designed and synthesized a novel series of quindoline derivatives with fork-shaped side chains by introducing (alkylamino)alkoxy side chains. Panels of experimental results showed that introducing a fork-shaped (alkylamino)alkoxy side chain could enhance the stabilizing abilities of the ligands against NRAS RNA G-quadruplexes and their anti-melanoma activities. One of them, 10b, exhibited good antitumor activity in the NRAS-mutant melanoma xenograft mouse model, showing the therapeutic potential of this kind of compounds.
Collapse
Affiliation(s)
- Jia-Wei Sun
- School of Pharmaceutical Sciences, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jing Zou
- School of Pharmaceutical Sciences, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ying Zheng
- School of Pharmaceutical Sciences, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, 510006, China
| | - Hao Yuan
- School of Pharmaceutical Sciences, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuan-Ze-Yu Xie
- School of Pharmaceutical Sciences, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xiao-Na Wang
- School of Pharmaceutical Sciences, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, 510006, China
| | - Tian-Miao Ou
- School of Pharmaceutical Sciences, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
2
|
Herlyn M, Villanueva J. Twenty years of research in melanoma therapy-From "nothing works" to cures: A personal account. Pigment Cell Melanoma Res 2023; 36:583-587. [PMID: 37726985 PMCID: PMC11187729 DOI: 10.1111/pcmr.13133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/21/2023]
Affiliation(s)
- Meenhard Herlyn
- Molecular & Cellular Oncogenesis Program, Ellen and Ronald Caplan Cancer Center, The Wistar Institute, Philadelphia, PA
| | - Jessie Villanueva
- Molecular & Cellular Oncogenesis Program, Ellen and Ronald Caplan Cancer Center, The Wistar Institute, Philadelphia, PA
| |
Collapse
|
3
|
Zaremba A, Mohr P, Gutzmer R, Meier F, Pföhler C, Weichenthal M, Terheyden P, Forschner A, Leiter U, Ulrich J, Utikal J, Welzel J, Kaatz M, Gebhardt C, Herbst R, Sindrilaru A, Dippel E, Sachse M, Meiss F, Heinzerling L, Haferkamp S, Weishaupt C, Löffler H, Kreft S, Griewank K, Livingstone E, Schadendorf D, Ugurel S, Zimmer L. Immune checkpoint inhibition in patients with NRAS mutated and NRAS wild type melanoma: a multicenter Dermatologic Cooperative Oncology Group study on 637 patients from the prospective skin cancer registry ADOREG. Eur J Cancer 2023; 188:140-151. [PMID: 37245442 DOI: 10.1016/j.ejca.2023.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/13/2023] [Accepted: 04/15/2023] [Indexed: 05/30/2023]
Abstract
BACKGROUND Melanomas frequently harbour somatic mutations in BRAF (40%) or NRAS (20%). Impact of NRAS mutations on the therapeutic outcome of immune checkpoint inhibitors (ICI) remains controversial. Potential correlation of the NRAS mutational status and programmed cell death ligand-1 (PD-L1) expression in melanoma is unknown. PATIENTS AND METHODS Advanced, non-resectable melanoma patients with known NRAS mutation status treated with first-line ICI between 06/2014 and 05/2020 in the prospective multicenter skin cancer registry ADOREG were included. Overall response rate (ORR), progression-free survival (PFS), and overall survival (OS) according to NRAS status were analysed. A multivariate Cox model was used to analyse factors associated with PFS and OS; survival was analysed using the Kaplan-Meier approach. RESULTS Among 637 BRAF wild-type patients, 310 (49%) had an NRAS mutation with Q61R (41%) and Q61K (32%). NRAS-mutated (NRASmut) melanomas were significantly more often located on the lower extremities and trunk (p = 0.001); nodular melanoma was the most common subtype (p < 0.0001). No significant differences were found for PFS and OS for anti-PD1 monotherapy (2-year PFS 39%, [95% confidence interval (CI), 33-47] in NRASmut patients and 41% [95% CI, 35-48] in NRAS-wild type (NRASwt) patients; 2-year OS was 54% [95% CI, 48-61] in NRASmut patients and 57% [95% CI, 50-64] in NRASwt patients) and anti-PD1 plus anti-CTLA4 therapy between both cohorts (2-year PFS was 54% [95% CI, 44-66] in NRASmut patients and 53% [95% CI, 41-67] in NRASwt patients; 2-year OS was 58% [95% CI, 49-70] in NRASmut patients and 62% [95% CI, 51-75] in NRASwt patients). The ORR to anti-PD1 was 35% for NRASwt patients and 26% for NRASmut patients and 34% compared to 32% for combinational therapy. Data on PD-L1 expression was available in 82 patients (13%). PD-L1 expression (>5%) was not correlated to NRAS mutational status. In multivariate analysis, elevated lactate dehydrogenase, Eastern Cooperative Oncology Group performance status ≥ 1, and brain metastases were significantly associated with a higher risk of death in all patients. CONCLUSIONS The PFS and OS were not affected by NRAS mutational status in patients treated with anti-PD1-based ICI. Similar ORR was seen in NRASwt and NRASmut patients. Tumour PD-L1 expression did not correlate with NRAS mutational status.
Collapse
Affiliation(s)
- Anne Zaremba
- Department of Dermatology, University Hospital Essen, Essen, Germany.
| | - Peter Mohr
- Department of Dermatology, Elbe Clinic Buxtehude, Buxtehude, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Hannover Medical School, Skin Cancer Centre Hannover, Hannover, Germany
| | - Friedegund Meier
- Skin Cancer Center at the University Cancer Centre Dresden and National Center for Tumor Diseases, Dresden, Germany; Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; National Center for Tumor Diseases Dresden (NCT/UCC), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Claudia Pföhler
- Saarland University Medical Center, Department of Dermatology, Homburg, Saarland, Germany
| | - Michael Weichenthal
- Department of Dermatology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | | | - Andrea Forschner
- Division of Dermatooncology, Department of Dermatology, University Medical Center, Tuebingen, Germany
| | - Ulrike Leiter
- Division of Dermatooncology, Department of Dermatology, University Medical Center, Tuebingen, Germany
| | - Jens Ulrich
- Department of Dermatology, Harz Clinic Quedlinburg, Quedlinburg, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Julia Welzel
- Department of Dermatology and Allergology, University Hospital Augsburg, Augsburg, Germany
| | - Martin Kaatz
- Department of Dermatology, Wald-Klinikum Gera, Gera, Germany
| | - Christoffer Gebhardt
- Department of Dermatology, University Hospital Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Rudolf Herbst
- Department of Dermatology, Helios Klinikum Erfurt GmbH, Erfurt, Germany
| | - Anca Sindrilaru
- Department of Dermatology, University Hospital Ulm, Ulm, Germany
| | - Edgar Dippel
- Department of Dermatology, Clinic of the City of Ludwigshafen on the Rhine gGmbH, Ludwigshafen am Rhein, Germany
| | - Michael Sachse
- Department of Dermatology, Bremerhaven Reinkenheide Hospital gGmbH, Bremerhaven, Germany
| | - Frank Meiss
- Department of Dermatology and Venereology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lucie Heinzerling
- Department of Dermatology, University Hospital Munich, Munich, Germany; Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Carsten Weishaupt
- Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Harald Löffler
- Department of Dermatology, SLK Hospital Heilbronn, Heilbronn, Germany
| | - Sophia Kreft
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | | | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Dresden, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Dresden, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Essen, Germany
| |
Collapse
|
4
|
Phadke MS, Smalley KS. Targeting NRAS Mutations in Advanced Melanoma. J Clin Oncol 2023; 41:2661-2664. [PMID: 36947724 PMCID: PMC10414701 DOI: 10.1200/jco.23.00205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 03/24/2023] Open
Affiliation(s)
- Manali S. Phadke
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Keiran S.M. Smalley
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Cutaneous Oncology, Moffitt Cancer Center & Research Institute, Tampa, FL
| |
Collapse
|
5
|
Wang J, Jiang H, Huang F, Li D, Wen X, Ding Q, Ding Y, Zhang X, Li J. Clinical features and response to systemic therapy in NRAS-mutant Chinese melanoma patients. J Cancer Res Clin Oncol 2023; 149:701-708. [PMID: 36454283 DOI: 10.1007/s00432-022-04377-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/21/2022] [Indexed: 12/05/2022]
Abstract
PURPOSE The prognosis of patients with NRAS-mutant melanoma is rather poor. Immunotherapy and targeted therapy have revolutionized anti-tumor therapy, especially for melanoma. In this study, we retrospectively summarized the real-world experience of systematic treatment for NRAS-mutant melanoma patients in this new era. PATIENTS AND METHODS The respective cohort included NRAS-mutant melanoma patients with metastatic or unresectable disease of Sun Yat-sen University Cancer Center (SYSUCC) from January 2018 to July 2022. The data about the clinical features and impact for systemic therapy of NRAS-mutant patients were collected and analyzed. RESULTS At data cutoff, 44 patients (19, 11, and 14 for acral, cutaneous, and mucosal ones, respectively) with NRAS-mutant were assessed. In addition, the median time of follow-up was 22.0 months. The immunotherapy-based combined treatment not only significantly improved the progression-free survival (PFS) (P = 0.006, HR 0.322), but was also accompanied by a higher objective response rate (ORR) (18.2%), disease control rate (DCR) (72.7%) than those of cytotoxic therapy or immunotherapy alone for advanced patients as first-line treatment. Nab-paclitaxel combined with anti-PD-1 inhibitor tended to produce better clinical benefit for the first-line treatment, especially for patients with acral melanoma. In addition, the tyrosine kinase inhibitor (TKI) combined with anti-PD-1 inhibitor also seemed to provide longer duration of response (DOR) for some patients. But combined therapy did not prolong the overall survival (OS) of NRAS-mutant patients. The combined therapy was well tolerated. Most adverse events were moderate and controllable. CONCLUSION In conclusion, PD-1 inhibitor-based combined therapy increased clinical benefit for advanced patients with NRAS-mutant melanoma.
Collapse
Affiliation(s)
- Jiuhong Wang
- Biotherapy Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Radiotherapy, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Hang Jiang
- Biotherapy Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Fuxue Huang
- Biotherapy Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Radiation Oncology and Therapy, Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
| | - Dandan Li
- Biotherapy Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xizhi Wen
- Biotherapy Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Qiuyue Ding
- Biotherapy Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Ya Ding
- Biotherapy Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xiaoshi Zhang
- Biotherapy Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jingjing Li
- Biotherapy Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
6
|
Zob DL, Augustin I, Caba L, Panzaru MC, Popa S, Popa AD, Florea L, Gorduza EV. Genomics and Epigenomics in the Molecular Biology of Melanoma-A Prerequisite for Biomarkers Studies. Int J Mol Sci 2022; 24:ijms24010716. [PMID: 36614156 PMCID: PMC9821083 DOI: 10.3390/ijms24010716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Melanoma is a common and aggressive tumor originating from melanocytes. The increasing incidence of cutaneous melanoma in recent last decades highlights the need for predictive biomarkers studies. Melanoma development is a complex process, involving the interplay of genetic, epigenetic, and environmental factors. Genetic aberrations include BRAF, NRAS, NF1, MAP2K1/MAP2K2, KIT, GNAQ, GNA11, CDKN2A, TERT mutations, and translocations of kinases. Epigenetic alterations involve microRNAs, non-coding RNAs, histones modifications, and abnormal DNA methylations. Genetic aberrations and epigenetic marks are important as biomarkers for the diagnosis, prognosis, and prediction of disease recurrence, and for therapeutic targets. This review summarizes our current knowledge of the genomic and epigenetic changes in melanoma and discusses the latest scientific information.
Collapse
Affiliation(s)
- Daniela Luminita Zob
- Department of Medical Oncology, AI. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Iolanda Augustin
- Department of Medical Oncology, AI. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
- Correspondence: (I.A.); (L.C.)
| | - Lavinia Caba
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Correspondence: (I.A.); (L.C.)
| | - Monica-Cristina Panzaru
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Setalia Popa
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Alina Delia Popa
- Nursing Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Laura Florea
- Department of Nephrology-Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Eusebiu Vlad Gorduza
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| |
Collapse
|
7
|
Wang H, Tran TT, Duong KT, Nguyen T, Le UM. Options of Therapeutics and Novel Delivery Systems of Drugs for the Treatment of Melanoma. Mol Pharm 2022; 19:4487-4505. [PMID: 36305753 DOI: 10.1021/acs.molpharmaceut.2c00775] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Melanoma is one of the most severe cancerous diseases. The cells employ multiple signaling pathways, such as ERK, HGF/c-MET, WNT, and COX-2 to cause the cell proliferation, survival, and metastasis. Treatment of melanoma, including surgery, chemotherapy, immunotherapy, radiation, and targeted therapy, is based on 4 major or 11 substages of the disease. Fourteen drugs, including dacarbazine, interferon α-2b, interleukin-12, ipilimumab, peginterferon α-2b, vemurafenib, trametinib, talimogene laherparepvec, cobimetinib, pembrolizumab, dabrafenib, binimetinib, encorafenib, and nivolumab, have been approved by the FDA for the treatment of melanoma. All of them are in conventional dosage forms of injection solutions, suspensions, oral tablets, or capsules. Major drawbacks of the treatment are side effects of the drugs and patients' incompliance to them. These are consequences of high doses and long-term treatments for the diseases. Currently more than 350 NCI-registered clinical trials are being carried out to treat advanced and/or metastatic melanoma using novel treatment methods, such as immune cell therapy, cancer vaccines, and new therapeutic targets. In addition, novel delivery systems using biomaterials of the approved drugs have been developed attempting to increase the drug delivery, targeting, stability, bioavailability, thus potentially reducing the toxicity and increasing the treatment effectiveness. Nanoparticles and liposomes have been emerging as advanced delivery systems which can improve drug stability and systemic circulation time. In this review, the most recent findings in the options for treatment and development of novel drug delivery systems for the treatment of melanoma are comprehensively discussed.
Collapse
Affiliation(s)
- Hongbin Wang
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California 95757, United States.,Master of Pharmaceutical Sciences College of Graduate Study, California Northstate University, 9700 West Taron Drive, Elk Grove, California 95757, United States
| | - Tuan T Tran
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California 95757, United States
| | - Katherine T Duong
- CVS Pharmacy, 18872 Beach Boulevard, Huntington Beach, California 92648, United States
| | - Trieu Nguyen
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California 95757, United States
| | - Uyen M Le
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California 95757, United States
| |
Collapse
|
8
|
McGrail K, Granado-Martínez P, Esteve-Puig R, García-Ortega S, Ding Y, Sánchez-Redondo S, Ferrer B, Hernandez-Losa J, Canals F, Manzano A, Navarro-Sabaté A, Bartrons R, Yanes O, Pérez-Alea M, Muñoz-Couselo E, Garcia-Patos V, Recio JA. BRAF activation by metabolic stress promotes glycolysis sensitizing NRAS Q61-mutated melanomas to targeted therapy. Nat Commun 2022; 13:7113. [PMID: 36402789 PMCID: PMC9675737 DOI: 10.1038/s41467-022-34907-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/08/2022] [Indexed: 11/21/2022] Open
Abstract
NRAS-mutated melanoma lacks a specific line of treatment. Metabolic reprogramming is considered a novel target to control cancer; however, NRAS-oncogene contribution to this cancer hallmark is mostly unknown. Here, we show that NRASQ61-mutated melanomas specific metabolic settings mediate cell sensitivity to sorafenib upon metabolic stress. Mechanistically, these cells are dependent on glucose metabolism, in which glucose deprivation promotes a switch from CRAF to BRAF signaling. This scenario contributes to cell survival and sustains glucose metabolism through BRAF-mediated phosphorylation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-2/3 (PFKFB2/PFKFB3). In turn, this favors the allosteric activation of phosphofructokinase-1 (PFK1), generating a feedback loop that couples glycolytic flux and the RAS signaling pathway. An in vivo treatment of NRASQ61 mutant melanomas, including patient-derived xenografts, with 2-deoxy-D-glucose (2-DG) and sorafenib effectively inhibits tumor growth. Thus, we provide evidence for NRAS-oncogene contributions to metabolic rewiring and a proof-of-principle for the treatment of NRASQ61-mutated melanoma combining metabolic stress (glycolysis inhibitors) and previously approved drugs, such as sorafenib.
Collapse
Affiliation(s)
- Kimberley McGrail
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Paula Granado-Martínez
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Rosaura Esteve-Puig
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,Present Address: MAJ3 Capital S.L, Barcelona, 08018 Spain
| | - Sara García-Ortega
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Yuxin Ding
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Sara Sánchez-Redondo
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.7719.80000 0000 8700 1153Present Address: Microenvironment & Metastasis Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Berta Ferrer
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.411083.f0000 0001 0675 8654Anatomy Pathology Department, Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Javier Hernandez-Losa
- grid.411083.f0000 0001 0675 8654Anatomy Pathology Department, Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Francesc Canals
- grid.411083.f0000 0001 0675 8654Proteomics Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, 08035 Spain
| | - Anna Manzano
- grid.418284.30000 0004 0427 2257Department of Physiological Sciences, University of Barcelona, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Aura Navarro-Sabaté
- grid.418284.30000 0004 0427 2257Department of Physiological Sciences, University of Barcelona, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Ramón Bartrons
- grid.418284.30000 0004 0427 2257Department of Physiological Sciences, University of Barcelona, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Oscar Yanes
- grid.410367.70000 0001 2284 9230Universitat Rovira i Virgili, Department of Electronic Engineering, IISPV, Tarragona, Spain ,grid.413448.e0000 0000 9314 1427CIBER on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Mileidys Pérez-Alea
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,Present Address: Advance Biodesign, 69800 Saint-Priest, France
| | - Eva Muñoz-Couselo
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.411083.f0000 0001 0675 8654Clinical Oncology Program, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Vicenç Garcia-Patos
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.411083.f0000 0001 0675 8654Dermatology Department, Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Juan A. Recio
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| |
Collapse
|
9
|
Monti N, Verna R, Piombarolo A, Querqui A, Bizzarri M, Fedeli V. Paradoxical Behavior of Oncogenes Undermines the Somatic Mutation Theory. Biomolecules 2022; 12:662. [PMID: 35625590 PMCID: PMC9138429 DOI: 10.3390/biom12050662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 12/04/2022] Open
Abstract
The currently accepted theory on the influence of DNA mutations on carcinogenesis (the Somatic Mutation Theory, SMT) is facing an increasing number of controversial results that undermine the explanatory power of mutated genes considered as "causative" factors. Intriguing results have demonstrated that several critical genes may act differently, as oncogenes or tumor suppressors, while phenotypic reversion of cancerous cells/tissues can be achieved by modifying the microenvironment, the mutations they are carrying notwithstanding. Furthermore, a high burden of mutations has been identified in many non-cancerous tissues without any apparent pathological consequence. All things considered, a relevant body of unexplained inconsistencies calls for an in depth rewiring of our theoretical models. Ignoring these paradoxes is no longer sustainable. By avoiding these conundrums, the scientific community will deprive itself of the opportunity to achieve real progress in this important biomedical field. To remedy this situation, we need to embrace new theoretical perspectives, taking the cell-microenvironment interplay as the privileged pathogenetic level of observation, and by assuming new explanatory models based on truly different premises. New theoretical frameworks dawned in the last two decades principally focus on the complex interaction between cells and their microenvironment, which is thought to be the critical level from which carcinogenesis arises. Indeed, both molecular and biophysical components of the stroma can dramatically drive cell fate commitment and cell outcome in opposite directions, even in the presence of the same stimulus. Therefore, such a novel approach can help in solving apparently inextricable paradoxes that are increasingly observed in cancer biology.
Collapse
Affiliation(s)
| | | | | | | | | | - Valeria Fedeli
- Systems Biology Group Lab, Department of Experimental Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (N.M.); (R.V.); (A.P.); (A.Q.); (M.B.)
| |
Collapse
|
10
|
Prabhash K, Saha S, Joshi A, Noronha V, Patil V, Menon N, Singh A, Shetty O, Mittal N, Chandrani P, Chougule A. NRAS mutation in differentiated thyroid cancer. CANCER RESEARCH, STATISTICS, AND TREATMENT 2022. [DOI: 10.4103/crst.crst_296_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
|
11
|
Hirata AS, Rezende-Teixeira P, Machado-Neto JA, Jimenez PC, Clair JJL, Fenical W, Costa-Lotufo LV. Seriniquinones as Therapeutic Leads for Treatment of BRAF and NRAS Mutant Melanomas. Molecules 2021; 26:7362. [PMID: 34885944 PMCID: PMC8658889 DOI: 10.3390/molecules26237362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/27/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
Isolated from the marine bacteria Serinicoccus sp., seriniquinone (SQ1) has been characterized by its selective activity in melanoma cell lines marked by its modulation of human dermcidin and induction of autophagy and apoptosis. While an active lead, the lack of solubility of SQ1 in both organic and aqueous media has complicated its preclinical evaluation. In response, our team turned its effort to explore analogues with the goal of returning synthetically accessible materials with comparable selectivity and activity. The analogue SQ2 showed improved solubility and reached a 30-40-fold greater selectivity for melanoma cells. Here, we report a detailed comparison of the activity of SQ1 and SQ2 in SK-MEL-28 and SK-MEL-147 cell lines, carrying the top melanoma-associated mutations, BRAFV600E and NRASQ61R, respectively. These studies provide a definitive report on the activity, viability, clonogenicity, dermcidin expression, autophagy, and apoptosis induction following exposure to SQ1 or SQ2. Overall, these studies showed that SQ1 and SQ2 demonstrated comparable activity and modulation of dermcidin expression. These studies are further supported through the evaluation of a panel of basal expression of key-genes related to autophagy and apoptosis, providing further insight into the role of these mutations. To explore this rather as a survival or death mechanism, autophagy inhibition sensibilized BRAF mutants to SQ1 and SQ2, whereas the opposite happened to NRAS mutants. These data suggest that the seriniquinones remain active, independently of the melanoma mutation, and suggest the future combination of their application with inhibitors of autophagy to treat BRAF-mutated tumors.
Collapse
Affiliation(s)
- Amanda S. Hirata
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, SP, Brazil; (A.S.H.); (P.R.-T.); (J.A.M.-N.)
| | - Paula Rezende-Teixeira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, SP, Brazil; (A.S.H.); (P.R.-T.); (J.A.M.-N.)
| | - João Agostinho Machado-Neto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, SP, Brazil; (A.S.H.); (P.R.-T.); (J.A.M.-N.)
| | - Paula C. Jimenez
- Institute of Marine Science, Federal University of São Paulo, Santos 11070-100, SP, Brazil;
| | - James J. La Clair
- Department of Chemistry and Biochemistry, University of California, La Jolla, San Diego, CA 92093-0358, USA;
| | - William Fenical
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, La Jolla, San Diego, CA 92093-0204, USA;
| | - Leticia V. Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, SP, Brazil; (A.S.H.); (P.R.-T.); (J.A.M.-N.)
| |
Collapse
|
12
|
Ma EZ, Hoegler KM, Zhou AE. Bioinformatic and Machine Learning Applications in Melanoma Risk Assessment and Prognosis: A Literature Review. Genes (Basel) 2021; 12:1751. [PMID: 34828357 PMCID: PMC8621295 DOI: 10.3390/genes12111751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/19/2021] [Accepted: 10/28/2021] [Indexed: 12/20/2022] Open
Abstract
Over 100,000 people are diagnosed with cutaneous melanoma each year in the United States. Despite recent advancements in metastatic melanoma treatment, such as immunotherapy, there are still over 7000 melanoma-related deaths each year. Melanoma is a highly heterogenous disease, and many underlying genetic drivers have been identified since the introduction of next-generation sequencing. Despite clinical staging guidelines, the prognosis of metastatic melanoma is variable and difficult to predict. Bioinformatic and machine learning analyses relying on genetic, clinical, and histopathologic inputs have been increasingly used to risk stratify melanoma patients with high accuracy. This literature review summarizes the key genetic drivers of melanoma and recent applications of bioinformatic and machine learning models in the risk stratification of melanoma patients. A robustly validated risk stratification tool can potentially guide the physician management of melanoma patients and ultimately improve patient outcomes.
Collapse
Affiliation(s)
| | | | - Albert E. Zhou
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, MD 21230, USA; (E.Z.M.); (K.M.H.)
| |
Collapse
|
13
|
Abstract
Neoplasia occurs as a result of genetic mutations. Research evaluating the association between gene mutations and skin cancer is limited and has produced inconsistent results. There are no established guidelines for screening skin cancer at molecular level. It should also be noted that the combinations of some mutations may play a role in skin tumors’ biology and immune response. There are three major types of skin cancer, and the originality of this study comes from its approach of each of them.
Collapse
|
14
|
Garcia-Alvarez A, Ortiz C, Muñoz-Couselo E. Current Perspectives and Novel Strategies of NRAS-Mutant Melanoma. Onco Targets Ther 2021; 14:3709-3719. [PMID: 34135599 PMCID: PMC8202735 DOI: 10.2147/ott.s278095] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022] Open
Abstract
Melanoma is the deadliest cutaneous cancer. Activating mutations in NRAS are found in 20% of melanomas. NRAS-mutant melanoma is more aggressive and, therefore, has poorer outcomes, compared to non-NRAS-mutant melanoma. Despite promising preclinical data, to date immune checkpoint inhibitors remain the standard of care for locally advanced unresectable or metastatic NRAS melanoma. Data for efficacy of immunotherapy for NRAS melanoma mainly come from retrospective cohorts with divergent conclusions. MEK inhibitors have been the most developed targeted therapy approach. Although associated with an increase in progression-free survival, MEK inhibitors do not provide any benefit in terms of overall survival. Combination strategies with PI3K-AKT-mTOR pathway and CDK4/6 inhibitors seem to increase MEK inhibitors' benefit. Nevertheless, results from clinical trials are still prelaminar. A greater comprehension of the biology and intracellular interactions of NRAS-mutant melanoma will outline novel impactful strategies which could improve prognosis of these subgroup of patients.
Collapse
Affiliation(s)
- Alejandro Garcia-Alvarez
- Vall d’Hebron University Hospital, Medical Oncology Department, Melanoma and Other Skin Tumors Unit, Vall Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| | - Carolina Ortiz
- Vall d’Hebron University Hospital, Medical Oncology Department, Melanoma and Other Skin Tumors Unit, Vall Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| | - Eva Muñoz-Couselo
- Vall d’Hebron University Hospital, Medical Oncology Department, Melanoma and Other Skin Tumors Unit, Vall Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| |
Collapse
|
15
|
Salem D, Chelvanambi M, Storkus WJ, Fecek RJ. Cutaneous Melanoma: Mutational Status and Potential Links to Tertiary Lymphoid Structure Formation. Front Immunol 2021; 12:629519. [PMID: 33746966 PMCID: PMC7970117 DOI: 10.3389/fimmu.2021.629519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/11/2021] [Indexed: 12/21/2022] Open
Abstract
Recent advances in immunotherapy have enabled rapid evolution of novel interventional approaches designed to reinvigorate and expand patient immune responses against cancer. An emerging approach in cancer immunology involves the conditional induction of tertiary lymphoid structures (TLS), which are non-encapsulated ectopic lymphoid structures forming at sites of chronic, pathologic inflammation. Cutaneous melanoma (CM), a highly-immunogenic form of solid cancer, continues to rise in both incidence and mortality rate, with recent reports supporting a positive correlation between the presence of TLS in melanoma and beneficial treatment outcomes amongst advanced-stage patients. In this context, TLS in CM are postulated to serve as dynamic centers for the initiation of robust anti-tumor responses within affected regions of active disease. Given their potential importance to patient outcome, significant effort has been recently devoted to gaining a better understanding of TLS neogenesis and the influence these lymphoid organs exert within the tumor microenvironment. Here, we briefly review TLS structure, function, and response to treatment in the setting of CM. To uncover potential tumor-intrinsic mechanisms that regulate TLS formation, we have taken the novel perspective of evaluating TLS induction in melanomas impacted by common driver mutations in BRAF, PTEN, NRAS, KIT, PRDM1, and MITF. Through analysis of The Cancer Genome Atlas (TCGA), we show expression of DNA repair proteins (DRPs) including BRCA1, PAXIP, ERCC1, ERCC2, ERCC3, MSH2, and PMS2 to be negatively correlated with expression of pro-TLS genes, suggesting DRP loss may favor TLS development in support of improved patient outcome and patient response to interventional immunotherapy.
Collapse
Affiliation(s)
- Deepak Salem
- Department of Microbiology, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA, United States
| | - Manoj Chelvanambi
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ronald J Fecek
- Department of Microbiology, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA, United States
| |
Collapse
|
16
|
Zhao J, Galvez C, Beckermann KE, Johnson DB, Sosman JA. Novel insights into the pathogenesis and treatment of NRAS mutant melanoma. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021; 6:281-294. [PMID: 34485698 PMCID: PMC8415440 DOI: 10.1080/23808993.2021.1938545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION NRAS was the first mutated oncogene identified in melanoma and is currently the second most common driver mutation in this malignancy. For patients with NRASmutant advanced stage melanoma refractory to immunotherapy or with contraindications to immune-based regimens, there are few therapeutic options including low-efficacy chemotherapy regimens and binimetinib monotherapy. Here, we review recent advances in preclinical studies of molecular targets for NRAS mutant melanoma as well as the failures and successes of early-phase clinical trials. While there are no targeted therapies for NRAS-driven melanoma, there is great promise in approaches combining MEK inhibition with inhibitors of the focal adhesion kinase (FAK), inhibitors of autophagy pathways, and pan-RAF inhibitors. AREAS COVERED This review surveys new developments in all aspects of disease pathogenesis and potential treatment - including those that have failed, stalled, or progressed through various phases of preclinical and clinical development. EXPERT OPINION There are no currently approved targeted therapies for BRAF wild-type melanoma patients harboring NRAS driver mutations though an array of agents are in early phase clinical trials. The diverse strategies taken exploit combined MAP kinase signaling blockade with inhibition of cell cycle mediators, inhibition of the autophagy pathway, and alteration of kinases involved in actin cytoskeleton signaling. Future advances of developmental therapeutics into late stage trials may yield new options beyond immunotherapy for patients with advanced stage disease and NRAS mutation status.
Collapse
Affiliation(s)
- Jeffrey Zhao
- Northwestern University Feinberg School of Medicine
| | - Carlos Galvez
- Northwestern Medicine, Division of Hematology and Oncology.,Robert H. Lurie Comprehensive Cancer Center
| | - Kathryn Eby Beckermann
- Vanderbilt University Medical Center, Department of Medicine, Division of Hematology and Oncology, 1301 Medical Center Drive, Nashville, 37232, USA
| | - Douglas B Johnson
- Vanderbilt University Medical Center, Department of Medicine, Division of Hematology and Oncology, 1301 Medical Center Drive, Nashville, 37232, USA
| | - Jeffrey A Sosman
- Northwestern Medicine, Division of Hematology and Oncology.,Robert H. Lurie Comprehensive Cancer Center
| |
Collapse
|
17
|
Alicea GM, Rebecca VW. Emerging strategies to treat rare and intractable subtypes of melanoma. Pigment Cell Melanoma Res 2021; 34:44-58. [PMID: 32274887 PMCID: PMC7544642 DOI: 10.1111/pcmr.12880] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 03/18/2020] [Accepted: 03/30/2020] [Indexed: 02/06/2023]
Abstract
Melanoma is the deadliest form of skin cancer, possessing a diverse landscape of subtypes with distinct molecular signatures and levels of aggressiveness. Although immense progress has been achieved therapeutically for patients with the most common forms of this disease, little is known of how to effectively treat patients with rarer subtypes of melanoma. These subtypes include acral lentiginous (the rarest form of cutaneous melanoma; AL), uveal, and mucosal melanomas, which display variations in distribution across (a) the world, (b) patient age-groups, and (c) anatomic sites. Unfortunately, patients with these relatively rare subtypes of melanoma typically respond worse to therapies approved for the more common, non-AL cutaneous melanoma and do not have effective alternatives, and thus consequently have worse overall survival rates. Achieving durable therapeutic responses in these high-risk melanoma subtypes represents one of the greatest challenges of the field. This review aims to collate and highlight effective preclinical and/or clinical strategies against these rare forms of melanoma.
Collapse
|
18
|
Neuroblastoma RAS viral oncogene homolog mRNA is differentially spliced to give five distinct isoforms: implications for melanoma therapy. Melanoma Res 2020; 29:491-500. [PMID: 31116161 DOI: 10.1097/cmr.0000000000000623] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neuroblastoma RAS viral oncogene homolog is a commonly mutated oncogene in melanoma, and therapeutic targeting of neuroblastoma RAS viral oncogene homolog has proven difficult. We characterized the expression and phenotypic functions of five recently discovered splice isoforms of neuroblastoma RAS viral oncogene homolog in melanoma. Canonical neuroblastoma RAS viral oncogene homolog (isoform-1) was expressed to the highest degree and its expression was significantly increased in melanoma metastases compared to primary lesions. Isoform-5 expression in metastases showed a significant, positive correlation with survival and tumours over-expressing isoform-5 had significantly decreased growth in a xenograft model. In contrast, over-expression of any isoform resulted in enhanced proliferation, and invasiveness was increased with over-expression of isoform-1 or isoform-2. Downstream signalling analysis indicated that the isoforms signalled differentially through the mitogen-activated protein kinase and PI3K pathways and A375 cells over-expressing isoform-2 or isoform-5 showed resistance to vemurafenib treatment in vitro. The neuroblastoma RAS viral oncogene homolog isoforms appear to play varying roles in melanoma phenotype and could potentially serve as biomarkers for therapeutic response and disease prognosis.
Collapse
|
19
|
LoRusso PM, Schalper K, Sosman J. Targeted therapy and immunotherapy: Emerging biomarkers in metastatic melanoma. Pigment Cell Melanoma Res 2020; 33:390-402. [PMID: 31705737 DOI: 10.1111/pcmr.12847] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 10/18/2019] [Accepted: 10/31/2019] [Indexed: 12/11/2022]
Abstract
Targeted therapy directed against oncogenic BRAF mutations and immune checkpoint inhibitors have transformed melanoma therapy over the past decade and prominently improved patient outcomes. However, not all patients will respond to targeted therapy or immunotherapy and many relapse after initially responding to treatment. This unmet need presents two major challenges. First, can we elucidate novel oncogenic drivers to provide new therapeutic targets? Second, can we identify patients who are most likely to respond to current therapeutic strategies in order to both more accurately select populations and avoid undue drug exposure in patients unlikely to respond? In an effort to evaluate the current state of the field with respect to these questions, we provide an overview of some common oncogenic mutations in patients with metastatic melanoma and ongoing efforts to therapeutically target these populations, as well as a discussion of biomarkers for response to immune checkpoint inhibitors-including tumor programmed death ligand 1 expression and the future use of neoantigens as a means of truly personalized therapy. This information is becoming important in treatment decision making and provides the framework for a treatment algorithm based on the current landscape in metastatic melanoma.
Collapse
Affiliation(s)
| | - Kurt Schalper
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Jeffrey Sosman
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Medical Center, Chicago, IL, USA
| |
Collapse
|
20
|
Mancera N, Smalley KSM, Margo CE. Melanoma of the eyelid and periocular skin: Histopathologic classification and molecular pathology. Surv Ophthalmol 2019; 64:272-288. [PMID: 30578807 DOI: 10.1016/j.survophthal.2018.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/29/2022]
Abstract
Cutaneous melanoma, a potentially lethal malignancy of the periocular skin, represents only a small proportion of the roughly 87,000 new cases of cutaneous melanoma diagnosed annually in the United States. Most of our understanding of melanoma of the eyelid skin is extrapolated from studies of cutaneous melanoma located elsewhere. Recent years have witnessed major breakthroughs in molecular biology and genomics of cutaneous melanoma, some of which have led to the development of targeted therapies. The molecular insights have also kindled interest in rethinking how cutaneous melanomas are classified and assessed for risk. We provide a synopsis of the epidemiology, histopathologic classification, and clinical experience of eyelid melanoma since 1990 and then review major advances in the molecular biology of cutaneous melanoma, exploring how this impacts our understanding of classification and predicting risk.
Collapse
Affiliation(s)
- Norberto Mancera
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA.
| | - Keiran S M Smalley
- Departments of Tumor Biology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA; Cutaneous Oncology The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Curtis E Margo
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA; Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
21
|
Savoia P, Fava P, Casoni F, Cremona O. Targeting the ERK Signaling Pathway in Melanoma. Int J Mol Sci 2019; 20:ijms20061483. [PMID: 30934534 PMCID: PMC6472057 DOI: 10.3390/ijms20061483] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 03/17/2019] [Accepted: 03/19/2019] [Indexed: 12/24/2022] Open
Abstract
The discovery of the role of the RAS/RAF/MEK/ERK pathway in melanomagenesis and its progression have opened a new era in the treatment of this tumor. Vemurafenib was the first specific kinase inhibitor approved for therapy of advanced melanomas harboring BRAF-activating mutations, followed by dabrafenib and encorafenib. However, despite the excellent results of first-generation kinase inhibitors in terms of response rate, the average duration of the response was short, due to the onset of genetic and epigenetic resistance mechanisms. The combination therapy with MEK inhibitors is an excellent strategy to circumvent drug resistance, with the additional advantage of reducing side effects due to the paradoxical reactivation of the MAPK pathway. The recent development of RAS and extracellular signal-related kinases (ERK) inhibitors promises to add new players for the ultimate suppression of this signaling pathway and the control of pathway-related drug resistance. In this review, we analyze the pharmacological, preclinical, and clinical trial data of the various MAPK pathway inhibitors, with a keen interest for their clinical applicability in the management of advanced melanoma.
Collapse
Affiliation(s)
- Paola Savoia
- Department of Health Science, University of Eastern Piedmont, via Solaroli 17, 28100 Novara, Italy.
| | - Paolo Fava
- Section of Dermatology, Department of Medical Science, University of Turin, 10124 Turin, Italy.
| | - Filippo Casoni
- San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 58, 20132 Milano, Italy.
- Università Vita Salute San Raffaele, via Olgettina 58, 20132 Milano, Italy.
| | - Ottavio Cremona
- San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 58, 20132 Milano, Italy.
- Università Vita Salute San Raffaele, via Olgettina 58, 20132 Milano, Italy.
| |
Collapse
|
22
|
Mayo Z, Seyedin S, Marquardt M, Mohiuddin I, Milhem M, Liu V, Pagedar N, Anderson C. Cutaneous malignant melanoma of the oral cavity following skin graft reconstruction: Case report. Head Neck 2019; 41:E26-E29. [PMID: 30537068 DOI: 10.1002/hed.25417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/06/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Malignant melanomas on skin graft recipient sites are rare, with few cases reported in the literature. METHODS We present a case report of a patient with recurrent cutaneous melanoma in the grafted anterolateral thigh flap of the tongue. RESULTS A patient underwent hemiglossectomy with free flap reconstruction for squamous cell carcinoma of the tongue. Five years later the patient was seen with a 1-cm nodule and surrounding erythroplakia at the recipient site of the graft. Analysis revealed cutaneous malignant melanoma. Patient then related a remote history of a suspected skin melanoma of the donor leg that had been treated with excision, with unknown histology. CONCLUSION This may be the first reported case of a cutaneous malignant melanoma in the oral cavity following an anterolateral thigh free flap reconstruction, emphasizing the importance of obtaining a comprehensive history of skin cancers and closely inspecting the donor site prior to graft harvesting.
Collapse
Affiliation(s)
- Zachary Mayo
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Steven Seyedin
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Michael Marquardt
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Imran Mohiuddin
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Mohammed Milhem
- Department of Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Vincent Liu
- Department of Dermatology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Nitin Pagedar
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Carryn Anderson
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| |
Collapse
|
23
|
Kitano S, Nakayama T, Yamashita M. Biomarkers for Immune Checkpoint Inhibitors in Melanoma. Front Oncol 2018; 8:270. [PMID: 30073150 PMCID: PMC6058029 DOI: 10.3389/fonc.2018.00270] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/29/2018] [Indexed: 01/05/2023] Open
Abstract
Immune checkpoint inhibitors have now become a standard therapy for malignant melanoma. However, as immunotherapies are effective in only a limited number of patients, biomarker development remains one of the most important clinical challenges. Biomarkers predicting clinical benefit facilitate appropriate selection of individualized treatments for patients and maximize clinical benefits. Many biomarkers derived from tumors and peripheral blood components have recently been reported, mainly in retrospective settings. This review summarizes the recent findings of biomarker studies for predicting the clinical benefits of immunotherapies in melanoma patients. Taking into account the complex interactions between the immune system and various cancers, it would be difficult for only one biomarker to predict clinical benefits in all patients. Many efforts to discover candidate biomarkers are currently ongoing. In the future, verification, by means of a prospective study, may allow some of these candidates to be combined into a scoring system based on bioinformatics technology.
Collapse
Affiliation(s)
- Shigehisa Kitano
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.,Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Takayuki Nakayama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Makiko Yamashita
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
24
|
Powis G. Recent Advances in the Development of Anticancer Drugs that Act against Signalling Pathways. TUMORI JOURNAL 2018; 80:69-87. [PMID: 8016910 DOI: 10.1177/030089169408000201] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cancer can be considered a disease of deranged intracellular signalling. The intracellular signalling pathways that mediate the effects of oncogenes on cell growth and transformation present attractive targets for the development of new classes of drugs for the prevention and treatment of cancer. This is a new approach to developing anticancer drugs and the potential, as well as some of the problems, inherent in the approach are discussed. Anticancer drugs that produce their effects by disrupting signalling pathways are already in clinical trial. Some properties of these drugs, as well as other inhibitors of signalling pathways under development as potential anticancer drugs, are reviewed.
Collapse
Affiliation(s)
- G Powis
- Arizona Cancer Center, University of Arizona Health Sciences Center, Tucson 85724
| |
Collapse
|
25
|
Śniegocka M, Podgórska E, Płonka PM, Elas M, Romanowska-Dixon B, Szczygieł M, Żmijewski MA, Cichorek M, Markiewicz A, Brożyna AA, Słominski AT, Urbańska K. Transplantable Melanomas in Hamsters and Gerbils as Models for Human Melanoma. Sensitization in Melanoma Radiotherapy-From Animal Models to Clinical Trials. Int J Mol Sci 2018; 19:E1048. [PMID: 29614755 PMCID: PMC5979283 DOI: 10.3390/ijms19041048] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
The focus of the present review is to investigate the role of melanin in the radioprotection of melanoma and attempts to sensitize tumors to radiation by inhibiting melanogenesis. Early studies showed radical scavenging, oxygen consumption and adsorption as mechanisms of melanin radioprotection. Experimental models of melanoma in hamsters and in gerbils are described as well as their use in biochemical and radiobiological studies, including a spontaneously metastasizing ocular model. Some results from in vitro studies on the inhibition of melanogenesis are presented as well as radio-chelation therapy in experimental and clinical settings. In contrast to cutaneous melanoma, uveal melanoma is very successfully treated with radiation, both using photon and proton beams. We point out that the presence or lack of melanin pigmentation should be considered, when choosing therapeutic options, and that both the experimental and clinical data suggest that melanin could be a target for radiosensitizing melanoma cells to increase efficacy of radiotherapy against melanoma.
Collapse
Affiliation(s)
- Martyna Śniegocka
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Ewa Podgórska
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Przemysław M Płonka
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Martyna Elas
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Bożena Romanowska-Dixon
- Department of Ophthalmology and Ocular Oncology, Medical College of Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Małgorzata Szczygieł
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Michał A Żmijewski
- Department of Histology, Medical University of Gdansk, 80-210 Gdańsk, Poland.
| | - Mirosława Cichorek
- Department of Embryology, Medical University of Gdansk, 80-210 Gdańsk, Poland.
| | - Anna Markiewicz
- Department of Ophthalmology and Ocular Oncology, Medical College of Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Anna A Brożyna
- Department of Tumor Pathology and Pathomorphology, Faculty of Health Sciences, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland.
- Department of Dermatology, Comprehensive Cancer Center Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Andrzej T Słominski
- Department of Dermatology, Comprehensive Cancer Center Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
- VA Medical Center, Birmingham, AL 35294, USA.
| | - Krystyna Urbańska
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| |
Collapse
|
26
|
Liu Q, Das M, Liu Y, Huang L. Targeted drug delivery to melanoma. Adv Drug Deliv Rev 2018; 127:208-221. [PMID: 28939379 DOI: 10.1016/j.addr.2017.09.016] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/29/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022]
Abstract
Melanoma derived from melanocytes is the most aggressive genre of skin cancer. Although the considerable advancement in the study of human cancer biology and drug discovery, most advanced melanoma patients are inevitably unable to be cured. With the emergence of nanotechnology, the use of nano-carriers is widely expected to alter the landscape of melanoma treatment. In this review, we will discuss melanoma biology, current treatment options, mechanisms behind drug resistance, and nano-based solutions for effective anti-cancer therapy, followed by challenges and perspectives in both pre-clinical and clinical settings.
Collapse
Affiliation(s)
- Qi Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC & NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Manisit Das
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC & NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
27
|
Hammerlindl H, Ravindran Menon D, Hammerlindl S, Emran AA, Torrano J, Sproesser K, Thakkar D, Xiao M, Atkinson VG, Gabrielli B, Haass NK, Herlyn M, Krepler C, Schaider H. Acetylsalicylic Acid Governs the Effect of Sorafenib in RAS-Mutant Cancers. Clin Cancer Res 2017; 24:1090-1102. [PMID: 29196297 DOI: 10.1158/1078-0432.ccr-16-2118] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 06/27/2017] [Accepted: 10/26/2017] [Indexed: 01/07/2023]
Abstract
Purpose: Identify and characterize novel combinations of sorafenib with anti-inflammatory painkillers to target difficult-to-treat RAS-mutant cancer.Experimental Design: The cytotoxicity of acetylsalicylic acid (aspirin) in combination with the multikinase inhibitor sorafenib (Nexavar) was assessed in RAS-mutant cell lines in vitro The underlying mechanism for the increased cytotoxicity was investigated using selective inhibitors and shRNA-mediated gene knockdown. In vitro results were confirmed in RAS-mutant xenograft mouse models in vivoResults: The addition of aspirin but not isobutylphenylpropanoic acid (ibruprofen) or celecoxib (Celebrex) significantly increased the in vitro cytotoxicity of sorafenib. Mechanistically, combined exposure resulted in increased BRAF/CRAF dimerization and the simultaneous hyperactivation of the AMPK and ERK pathways. Combining sorafenib with other AMPK activators, such as metformin or A769662, was not sufficient to decrease cell viability due to sole activation of the AMPK pathway. The cytotoxicity of sorafenib and aspirin was blocked by inhibition of the AMPK or ERK pathways through shRNA or via pharmacologic inhibitors of RAF (LY3009120), MEK (trametinib), or AMPK (compound C). The combination was found to be specific for RAS/RAF-mutant cells and had no significant effect in RAS/RAF-wild-type keratinocytes or melanoma cells. In vivo treatment of human xenografts in NSG mice with sorafenib and aspirin significantly reduced tumor volume compared with each single-agent treatment.Conclusions: Combination sorafenib and aspirin exerts cytotoxicity against RAS/RAF-mutant cells by simultaneously affecting two independent pathways and represents a promising novel strategy for the treatment of RAS-mutant cancers. Clin Cancer Res; 24(5); 1090-102. ©2017 AACR.
Collapse
Affiliation(s)
- Heinz Hammerlindl
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Dinoop Ravindran Menon
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Sabrina Hammerlindl
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Abdullah Al Emran
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Joachim Torrano
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | | | - Divya Thakkar
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Min Xiao
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Victoria G Atkinson
- Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Australia
| | - Brian Gabrielli
- Mater Medical Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Nikolas K Haass
- The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | | | | | - Helmut Schaider
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia. .,The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| |
Collapse
|
28
|
Muñoz-Couselo E, Adelantado EZ, Ortiz C, García JS, Perez-Garcia J. NRAS-mutant melanoma: current challenges and future prospect. Onco Targets Ther 2017; 10:3941-3947. [PMID: 28860801 PMCID: PMC5558581 DOI: 10.2147/ott.s117121] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Melanoma is one of the most common cutaneous cancers worldwide. Activating mutations in RAS oncogenes are found in a third of all human cancers and NRAS mutations are found in 15%–20% of melanomas. The NRAS-mutant subset of melanoma is more aggressive and associated with poorer outcomes, compared to non-NRAS-mutant melanoma. Although immune checkpoint inhibitors and targeted therapies for BRAF-mutant melanoma are transforming the treatment of metastatic melanoma, the ideal treatment for NRAS-mutant melanoma remains unknown. Despite promising preclinical data, current therapies for NRAS-mutant melanoma remain limited, showing a modest increase in progression-free survival but without any benefit in overall survival. Combining MEK inhibitors with agents inhibiting cell cycling and the PI3K–AKT pathway appears to provide additional benefit; in particular, a strategy of MEK inhibition and CDK4/6 inhibition is likely to be a viable treatment option in the future. Patients whose tumors had NRAS mutations had better response to immunotherapy and better outcomes than patients whose tumors had other genetic subtypes, suggesting that immune therapies – especially immune checkpoint inhibitors – may be particularly effective as treatment options for NRAS-mutant melanoma. Improved understanding of NRAS-mutant melanoma will be essential to develop new treatment strategies for this subset of patients with melanoma.
Collapse
Affiliation(s)
- Eva Muñoz-Couselo
- Medical Oncology Department, Vall d'Hebron Hospital, Barcelona, Spain.,Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ester Zamora Adelantado
- Medical Oncology Department, Vall d'Hebron Hospital, Barcelona, Spain.,Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Carolina Ortiz
- Medical Oncology Department, Vall d'Hebron Hospital, Barcelona, Spain.,Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | | |
Collapse
|
29
|
Flem-Karlsen K, Tekle C, Andersson Y, Flatmark K, Fodstad Ø, Nunes-Xavier CE. Immunoregulatory protein B7-H3 promotes growth and decreases sensitivity to therapy in metastatic melanoma cells. Pigment Cell Melanoma Res 2017; 30:467-476. [PMID: 28513992 DOI: 10.1111/pcmr.12599] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/09/2017] [Indexed: 12/26/2022]
Abstract
B7-H3 (CD276) belongs to the B7 family of immunoregulatory proteins and has been implicated in cancer progression and metastasis. In this study, we found that metastatic melanoma cells with knockdown expression of B7-H3 showed modest decrease in proliferation and glycolytic capacity and were more sensitive to dacarbazine (DTIC) chemotherapy and small-molecule inhibitors targeting MAP kinase (MAPK) and AKT/mTOR pathways: vemurafenib (PLX4032; BRAF inhibitor), binimetinib (MEK-162; MEK inhibitor), everolimus (RAD001; mTOR inhibitor), and triciribidine (API-2; AKT inhibitor). Similar effects were observed in melanoma cells in the presence of an inhibitory B7-H3 monoclonal antibody, while the opposite was seen in B7-H3-overexpressing cells. Further, combining B7-H3 inhibition with small-molecule inhibitors resulted in significantly increased antiproliferative effect in melanoma cells, as well as in BRAFV600E mutated cell lines derived from patient biopsies. Our findings indicate that targeting B7-H3 may be a novel alternative to improve current therapy of metastatic melanoma.
Collapse
Affiliation(s)
- Karine Flem-Karlsen
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Christina Tekle
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Yvonne Andersson
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Kjersti Flatmark
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Øystein Fodstad
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Caroline E Nunes-Xavier
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| |
Collapse
|
30
|
Ma J, Guo W, Li C. Ubiquitination in melanoma pathogenesis and treatment. Cancer Med 2017; 6:1362-1377. [PMID: 28544818 PMCID: PMC5463089 DOI: 10.1002/cam4.1069] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 12/13/2022] Open
Abstract
Melanoma is one of the most aggressive skin cancers with fiercely increasing incidence and mortality. Since the progressive understanding of the mutational landscape and immunologic pathogenic factors in melanoma, the targeted therapy and immunotherapy have been recently established and gained unprecedented improvements for melanoma treatment. However, the prognosis of melanoma patients remains unoptimistic mainly due to the resistance and nonresponse to current available drugs. Ubiquitination is a posttranslational modification which plays crucial roles in diverse cellular biological activities and participates in the pathogenesis of various cancers, including melanoma. Through the regulation of multiple tumor promoters and suppressors, ubiquitination is emerging as the key contributor and therefore a potential therapeutic target for melanoma. Herein, we summarize the current understanding of ubiquitination in melanoma, from mechanistic insights to clinical progress, and discuss the prospect of ubiquitination modification in melanoma treatment.
Collapse
Affiliation(s)
- Jinyuan Ma
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weinan Guo
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
31
|
Dorard C, Estrada C, Barbotin C, Larcher M, Garancher A, Leloup J, Beermann F, Baccarini M, Pouponnot C, Larue L, Eychène A, Druillennec S. RAF proteins exert both specific and compensatory functions during tumour progression of NRAS-driven melanoma. Nat Commun 2017; 8:15262. [PMID: 28497782 PMCID: PMC5437303 DOI: 10.1038/ncomms15262] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 03/14/2017] [Indexed: 12/19/2022] Open
Abstract
NRAS and its effector BRAF are frequently mutated in melanoma. Paradoxically, CRAF but not BRAF was shown to be critical for various RAS-driven cancers, raising the question of the role of RAF proteins in NRAS-induced melanoma. Here, using conditional ablation of Raf genes in NRAS-induced mouse melanoma models, we investigate their contribution in tumour progression, from the onset of benign tumours to malignant tumour maintenance. We show that BRAF expression is required for ERK activation and nevi development, demonstrating a critical role in the early stages of NRAS-driven melanoma. After melanoma formation, single Braf or Craf ablation is not sufficient to block tumour growth, showing redundant functions for RAF kinases. Finally, proliferation of resistant cells emerging in the absence of BRAF and CRAF remains dependent on ARAF-mediated ERK activation. These results reveal specific and compensatory functions for BRAF and CRAF and highlight an addiction to RAF signalling in NRAS-driven melanoma. The melanoma-driver mutations in NRAS and BRAF are mutually exclusive but the contribution of RAF signalling downstream of NRAS remains to be clarified. Here, using mouse models, the authors show specific roles of each member of the RAF family at different stages of melanomagenesis.
Collapse
Affiliation(s)
- Coralie Dorard
- Institut Curie, Orsay F-91405, France.,INSERM U1021, Centre Universitaire, Orsay F-91405, France.,CNRS UMR 3347, Centre Universitaire, Orsay F-91405, France.,Université Paris Sud-11, Orsay F-91405, France.,Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay F-91405, France
| | - Charlène Estrada
- Institut Curie, Orsay F-91405, France.,INSERM U1021, Centre Universitaire, Orsay F-91405, France.,CNRS UMR 3347, Centre Universitaire, Orsay F-91405, France.,Université Paris Sud-11, Orsay F-91405, France.,Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay F-91405, France
| | - Céline Barbotin
- Institut Curie, Orsay F-91405, France.,INSERM U1021, Centre Universitaire, Orsay F-91405, France.,CNRS UMR 3347, Centre Universitaire, Orsay F-91405, France.,Université Paris Sud-11, Orsay F-91405, France.,Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay F-91405, France
| | - Magalie Larcher
- Institut Curie, Orsay F-91405, France.,INSERM U1021, Centre Universitaire, Orsay F-91405, France.,CNRS UMR 3347, Centre Universitaire, Orsay F-91405, France.,Université Paris Sud-11, Orsay F-91405, France.,Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay F-91405, France
| | - Alexandra Garancher
- Institut Curie, Orsay F-91405, France.,INSERM U1021, Centre Universitaire, Orsay F-91405, France.,CNRS UMR 3347, Centre Universitaire, Orsay F-91405, France.,Université Paris Sud-11, Orsay F-91405, France
| | - Jessy Leloup
- Institut Curie, Orsay F-91405, France.,INSERM U1021, Centre Universitaire, Orsay F-91405, France.,CNRS UMR 3347, Centre Universitaire, Orsay F-91405, France.,Université Paris Sud-11, Orsay F-91405, France.,Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay F-91405, France
| | - Friedrich Beermann
- Swiss Institute for Experimental Cancer Research (ISREC), Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Manuela Baccarini
- Max F. Perutz Laboratories, Center for Molecular Biology, University of Vienna, Vienna 1030, Austria
| | - Celio Pouponnot
- Institut Curie, Orsay F-91405, France.,INSERM U1021, Centre Universitaire, Orsay F-91405, France.,CNRS UMR 3347, Centre Universitaire, Orsay F-91405, France.,Université Paris Sud-11, Orsay F-91405, France
| | - Lionel Larue
- Institut Curie, Orsay F-91405, France.,INSERM U1021, Centre Universitaire, Orsay F-91405, France.,CNRS UMR 3347, Centre Universitaire, Orsay F-91405, France.,Université Paris Sud-11, Orsay F-91405, France.,Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay F-91405, France
| | - Alain Eychène
- Institut Curie, Orsay F-91405, France.,INSERM U1021, Centre Universitaire, Orsay F-91405, France.,CNRS UMR 3347, Centre Universitaire, Orsay F-91405, France.,Université Paris Sud-11, Orsay F-91405, France.,Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay F-91405, France
| | - Sabine Druillennec
- Institut Curie, Orsay F-91405, France.,INSERM U1021, Centre Universitaire, Orsay F-91405, France.,CNRS UMR 3347, Centre Universitaire, Orsay F-91405, France.,Université Paris Sud-11, Orsay F-91405, France.,Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay F-91405, France
| |
Collapse
|
32
|
Abstract
Treatment options for patients with metastatic melanoma, and especially BRAF-mutant melanoma, have changed dramatically in the past 5 years, with the FDA approval of eight new therapeutic agents. During this period, the treatment paradigm for BRAF-mutant disease has evolved rapidly: the standard-of-care BRAF-targeted approach has shifted from single-agent BRAF inhibition to combination therapy with a BRAF and a MEK inhibitor. Concurrently, immunotherapy has transitioned from cytokine-based treatment to antibody-mediated blockade of the cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) and, now, the programmed cell-death protein 1 (PD-1) immune checkpoints. These changes in the treatment landscape have dramatically improved patient outcomes, with the median overall survival of patients with advanced-stage melanoma increasing from approximately 9 months before 2011 to at least 2 years - and probably longer for those with BRAF-V600-mutant disease. Herein, we review the clinical trial data that established the standard-of-care treatment approaches for advanced-stage melanoma. Mechanisms of resistance and biomarkers of response to BRAF-targeted treatments and immunotherapies are discussed, and the contrasting clinical benefits and limitations of these therapies are explored. We summarize the state of the field and outline a rational approach to frontline-treatment selection for each individual patient with BRAF-mutant melanoma.
Collapse
|
33
|
Hosseini M, Kasraian Z, Rezvani HR. Energy metabolism in skin cancers: A therapeutic perspective. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:712-722. [PMID: 28161328 DOI: 10.1016/j.bbabio.2017.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/20/2017] [Accepted: 01/23/2017] [Indexed: 12/13/2022]
Abstract
Skin cancers are the most common cancers worldwide. The incidence of common skin cancers, including basal cell carcinomas (BCCs), squamous cell carcinomas (SCCs) and melanomas, continues to rise by 5 to 7% per year, mainly due to ultraviolet (UV) exposure and partly because of aging. This suggests an urgent necessity to improve the level of prevention and protection for skin cancers as well as developing new prognostic and diagnostic markers of skin cancers. Moreover, despite innovative therapies especially in the fields of melanoma and carcinomas, new therapeutic options are needed to bypass resistance to targeted therapies or treatment's side effects. Since reprogramming of cellular metabolism is now considered as a hallmark of cancer, some of the recent findings on the role of energy metabolism in skin cancer initiation and progression as well as its effect on the response to targeted therapies are discussed in this review. This article is part of a Special Issue entitled Mitochondria in cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.
Collapse
Affiliation(s)
- Mohsen Hosseini
- Inserm U 1035, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Zeinab Kasraian
- Inserm U 1035, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Hamid Reza Rezvani
- Inserm U 1035, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; Centre de Référence pour les Maladies Rares de la Peau, CHU de Bordeaux, France.
| |
Collapse
|
34
|
Abstract
To provide appropriate therapy and follow-up to patients with malignant melanoma, proper diagnostics are of critical importance. Targeted therapy of advanced melanoma is based on the molecular genetic analyses of tumor tissue. In addition, sequencing of genes and other genetic approaches can provide insight into the origin of melanocytic tumors and can aid in distinguishing benign from malignant lesions. In this regard, spizoid neoplasms remain a challenging entity. Aside from genetic analyses of tumor tissue, immunohistochemistry remains an essential tool in melanoma diagnostics and TNM classification. With new immunotherapies being approved for advanced melanoma, immunohistochemistry to determine PD-L1 expression has gained clinical interest. While PD-L1 expression is associated with response to PD-1 blockade, a substantial number of patients without PD-L1 expression can still experience tumor remission upon treatment. In this review, current and future developments in melanoma diagnostics with regard to molecular genetics and immunohistochemistry are summarized. The utilization of such analyses in clinical decision making is also discussed.
Collapse
Affiliation(s)
- B Schilling
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Partnerstandort des Deutschen Konsortium für Translationale Krebsforschung (DKTK), Hufelandstr. 55, 45147, Essen, Deutschland.
| | - K G Griewank
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Partnerstandort des Deutschen Konsortium für Translationale Krebsforschung (DKTK), Hufelandstr. 55, 45147, Essen, Deutschland.,Dermatopathologie bei Mainz, Bahnhofstr. 2B, 55268, Nieder-Olm, Deutschland
| |
Collapse
|
35
|
Oncogene status as a diagnostic tool in ocular and cutaneous melanoma. Eur J Cancer 2016; 57:112-7. [DOI: 10.1016/j.ejca.2016.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 01/15/2016] [Indexed: 11/24/2022]
|
36
|
Akabane H, Sullivan RJ. The Future of Molecular Analysis in Melanoma: Diagnostics to Direct Molecularly Targeted Therapy. Am J Clin Dermatol 2016; 17:1-10. [PMID: 26518880 DOI: 10.1007/s40257-015-0159-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Melanoma is a malignancy of pigment-producing cells that is driven by a variety of genetic mutations and aberrations. In most cases, this leads to upregulation of the mitogen-activated protein kinase (MAPK) pathway through activating mutations of upstream mediators of the pathway including BRAF and NRAS. With the advent of effective MAPK pathway inhibitors, including the US FDA-approved BRAF inhibitors vemurafenib and dabrafenib and MEK inhibitor trametinib, molecular analysis has become an integral part of the care of patients with metastatic melanoma. In this article, the key molecular targets and strategies to inhibit these targets therapeutically are presented, and the techniques of identifying these targets, in both tissue and blood, are discussed.
Collapse
Affiliation(s)
- Hugo Akabane
- Department of Medicine, Metrowest Medical Center, Framingham, MA, USA
| | - Ryan J Sullivan
- Center for Melanoma, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
37
|
Metastatic melanoma treatment: Combining old and new therapies. Crit Rev Oncol Hematol 2015; 98:242-53. [PMID: 26616525 DOI: 10.1016/j.critrevonc.2015.11.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 10/16/2015] [Accepted: 11/12/2015] [Indexed: 01/04/2023] Open
Abstract
Metastatic melanoma is an aggressive form of cancer characterised by poor prognosis and a complex etiology. Until 2010, the treatment options for metastatic melanoma were very limited. Largely ineffective dacarbazine, temozolamide or fotemustine were the only agents in use for 35 years. In recent years, the development of molecularly targeted inhibitors in parallel with the development of checkpoint inhibition immunotherapies has rapidly improved the outcomes for metastatic melanoma patients. Despite these new therapies showing initial promise; resistance and poor duration of response have limited their effectiveness as monotherapies. Here we provide an overview of the history of melanoma treatment, as well as the current treatments in development. We also discuss the future of melanoma treatment as we go beyond monotherapies to a combinatorial approach. Combining older therapies with the new molecular and immunotherapies will be the most promising way forward for treatment of metastatic melanoma.
Collapse
|
38
|
Kuzu OF, Nguyen FD, Noory MA, Sharma A. Current State of Animal (Mouse) Modeling in Melanoma Research. CANCER GROWTH AND METASTASIS 2015; 8:81-94. [PMID: 26483610 PMCID: PMC4597587 DOI: 10.4137/cgm.s21214] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/10/2015] [Accepted: 08/17/2015] [Indexed: 11/16/2022]
Abstract
Despite the considerable progress in understanding the biology of human cancer and technological advancement in drug discovery, treatment failure remains an inevitable outcome for most cancer patients with advanced diseases, including melanoma. Despite FDA-approved BRAF-targeted therapies for advanced stage melanoma showed a great deal of promise, development of rapid resistance limits the success. Hence, the overall success rate of melanoma therapy still remains to be one of the worst compared to other malignancies. Advancement of next-generation sequencing technology allowed better identification of alterations that trigger melanoma development. As development of successful therapies strongly depends on clinically relevant preclinical models, together with the new findings, more advanced melanoma models have been generated. In this article, besides traditional mouse models of melanoma, we will discuss recent ones, such as patient-derived tumor xenografts, topically inducible BRAF mouse model and RCAS/TVA-based model, and their advantages as well as limitations. Although mouse models of melanoma are often criticized as poor predictors of whether an experimental drug would be an effective treatment, development of new and more relevant models could circumvent this problem in the near future.
Collapse
Affiliation(s)
- Omer F Kuzu
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Felix D Nguyen
- The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mohammad A Noory
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Arati Sharma
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
39
|
Karachaliou N, Pilotto S, Teixidó C, Viteri S, González-Cao M, Riso A, Morales-Espinosa D, Molina MA, Chaib I, Santarpia M, Richardet E, Bria E, Rosell R. Melanoma: oncogenic drivers and the immune system. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:265. [PMID: 26605311 PMCID: PMC4630557 DOI: 10.3978/j.issn.2305-5839.2015.08.06] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 08/04/2015] [Indexed: 12/19/2022]
Abstract
Advances and in-depth understanding of the biology of melanoma over the past 30 years have contributed to a change in the consideration of melanoma as one of the most therapy-resistant malignancies. The finding that oncogenic BRAF mutations drive tumor growth in up to 50% of melanomas led to a molecular therapy revolution for unresectable and metastatic disease. Moving beyond BRAF, inactivation of immune regulatory checkpoints that limit T cell responses to melanoma has provided targets for cancer immunotherapy. In this review, we discuss the molecular biology of melanoma and we focus on the recent advances of molecularly targeted and immunotherapeutic approaches.
Collapse
|
40
|
Sullivan RJ, Flaherty KT. New Strategies in Melanoma: Entering the Era of Combinatorial Therapy. Clin Cancer Res 2015; 21:2424-35. [DOI: 10.1158/1078-0432.ccr-14-1650] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
41
|
Abstract
Melanoma, the deadliest form of skin cancer, is an aggressive disease that is rising in incidence. Although melanoma is a historically treatment-resistant malignancy, in recent years unprecedented breakthroughs in targeted therapies and immunotherapies have revolutionized the standard of care for patients with advanced disease. Here, we provide an overview of recent developments in our understanding of melanoma risk factors, genomics, and molecular pathogenesis and how these insights have driven advances in melanoma treatment. In addition, we review benefits and limitations of current therapies and look ahead to continued progress in melanoma prevention and therapy. Remarkable achievements in the field have already produced a paradigm shift in melanoma treatment: Metastatic melanoma, once considered incurable, can now be treated with potentially curative rather than palliative intent.
Collapse
Affiliation(s)
- Jennifer A Lo
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
42
|
Rogers MJ. Tumor-associated transplantation antigens of chemically-induced tumors: new complexities. ACTA ACUST UNITED AC 2014; 5:167-8. [PMID: 25289743 DOI: 10.1016/0167-5699(84)90008-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- M J Rogers
- Laboratory of Genetics, National Cancer Institute, Bethesda, MD 20205, USA
| |
Collapse
|
43
|
Beyond BRAF: where next for melanoma therapy? Br J Cancer 2014; 112:217-26. [PMID: 25180764 PMCID: PMC4453440 DOI: 10.1038/bjc.2014.476] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 07/23/2014] [Accepted: 08/04/2014] [Indexed: 12/22/2022] Open
Abstract
In recent years, melanoma has become a poster-child for the development of oncogene-directed targeted therapies. This approach, which has been exemplified by the development of small-molecule BRAF inhibitors and the BRAF/MEK inhibitor combination for BRAF-mutant melanoma, has brought new hope to patients. Despite these successes, treatment failure seems near inevitable in the majority of cases—even in individuals treated with the BRAF/MEK inhibitor doublet. In the current review, we discuss the future of combination strategies for patients with BRAF-mutant melanoma as well as the emerging therapeutic options for patients with NRAS-mutant and BRAF/NRAS-wild-type melanoma. We also outline some of the newest developments in the in-depth personalisation of therapy that should allow melanoma treatment to continue shaping the field precision cancer medicine.
Collapse
|
44
|
Griewank KG, Murali R, Puig-Butille JA, Schilling B, Livingstone E, Potrony M, Carrera C, Schimming T, Möller I, Schwamborn M, Sucker A, Hillen U, Badenas C, Malvehy J, Zimmer L, Scherag A, Puig S, Schadendorf D. TERT promoter mutation status as an independent prognostic factor in cutaneous melanoma. J Natl Cancer Inst 2014; 106:dju246. [PMID: 25217772 PMCID: PMC4200061 DOI: 10.1093/jnci/dju246] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 06/25/2014] [Accepted: 07/01/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Recently, TERT promoter mutations were identified at high frequencies in cutaneous melanoma tumor samples and cell lines. The mutations were found to have a UV-signature and to lead to increased TERT gene expression. We analyzed a large cohort of melanoma patients for the presence and distribution of TERT promoter mutations and their association with clinico-pathological characteristics. METHODS 410 melanoma tumor samples were analyzed by Sanger sequencing for the presence of TERT promoter mutations. An analysis of associations between mutation status and various clinical and pathologic variables was performed. RESULTS TERT promoter mutations were identified in 154 (43%) of 362 successfully sequenced melanomas. Mutation frequencies varied between melanoma subtype, being most frequent in melanomas arising in nonacral skin (48%) and melanomas with occult primary (50%), and less frequent in mucosal (23%), and acral (19%) melanomas. Mutations carried a UV signature (C>T or CC>TT). The presence of TERT promoter mutations was associated with factors such as BRAF or NRAS mutation (P < .001), histologic type (P = .002), and Breslow thickness (P < .001). TERT promoter mutation was independently associated with poorer overall survival in patients with nonacral cutaneous melanomas (median survival 80 months vs 291 months for wild-type; hazard ratio corrected for other covariates 2.47; 95% confidence interval [CI] = 1.29 to 4.74; P = .006). CONCLUSIONS UV-induced TERT promoter mutations are one of the most frequent genetic alterations in melanoma, with frequencies varying depending on melanoma subtype. In nonacral cutaneous melanomas, presence of TERT promoter mutations is independently associated with poor prognosis.
Collapse
Affiliation(s)
- Klaus G Griewank
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS).
| | - Rajmohan Murali
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Joan Anton Puig-Butille
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Bastian Schilling
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Elisabeth Livingstone
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Miriam Potrony
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Cristina Carrera
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Tobias Schimming
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Inga Möller
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Marion Schwamborn
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Antje Sucker
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Uwe Hillen
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Celia Badenas
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Josep Malvehy
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Lisa Zimmer
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - André Scherag
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Susana Puig
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS)
| | - Dirk Schadendorf
- Department of Dermatology,University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen and the German Cancer Consortium (KGG, BS, EL, TS, IM, MS, AS, UH, LZ, DS), Germany; Department of Pathology and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY (RM); CIBER Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain (JAPB, MP, CC, CB, JM, SP); Department of Dermatology, Hospital Clinic Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain (CC, JM, SP); Biochemistry and Molecular Genetics Department, Hospital Clinic Barcelona, IDIBAPS, Barcelona, Spain (CB); Integriertes Forschungs- und Behandlungszentrum (IFB) Sepsis und Sepsisfolgen Center for Sepsis Control and Care (CSCC) University Hospital Jena, Jena, Germany (AS).
| |
Collapse
|
45
|
Griewank KG, Scolyer RA, Thompson JF, Flaherty KT, Schadendorf D, Murali R. Genetic alterations and personalized medicine in melanoma: progress and future prospects. J Natl Cancer Inst 2014; 106:djt435. [PMID: 24511108 DOI: 10.1093/jnci/djt435] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
High-throughput sequencing technologies are providing new insights into the genetic alterations involved in melanomagenesis. It appears likely that most genetic events important in the pathogenesis of melanoma will be discovered over the next few years. Genetic analysis is also increasingly being used to direct patient care. In parallel with the discovery of new genes and the elucidation of molecular pathways important in the development of melanoma, therapies targeting these pathways are becoming available. In other words, the age of personalized medicine has arrived, characterized by molecular profiling of melanoma to identify the relevant genetic alterations and the abnormal signaling mechanisms involved, followed by selection of optimal, individualized therapies. In this review, we summarize the key genetic alterations in melanoma and the development of targeted agents against melanomas bearing specific mutations. These developments in melanoma serve as a model for the implementation of personalized medicine for patients with all cancers.
Collapse
Affiliation(s)
- Klaus G Griewank
- Affiliations of authors: Department of Dermatology, University Hospital, University Duisburg-Essen, Essen, Germany (KGG, DS); Royal Prince Alfred Hospital, Camperdown, NSW, Australia (RAS); University of Sydney, Camperdown, NSW, Australia (RAS, JFT); Melanoma Institute Australia, North Sydney, NSW, Australia (RAS, JFT); Center for Melanoma, Massachusetts General Hospital Cancer Center, Boston, MA (KTF); Department of Pathology, and Center for Molecular Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (RM)
| | | | | | | | | | | |
Collapse
|
46
|
Tomei S, Wang E, Delogu LG, Marincola FM, Bedognetti D. Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma. Expert Opin Biol Ther 2014; 14:663-86. [DOI: 10.1517/14712598.2014.890586] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
47
|
Martín-Algarra S, Fernández-Figueras MT, López-Martín JA, Santos-Briz A, Arance A, Lozano MD, Berrocal A, Ríos-Martín JJ, Espinosa E, Rodríguez-Peralto JL. Guidelines for biomarker testing in metastatic melanoma: a National Consensus of the Spanish Society of Pathology and the Spanish Society of Medical Oncology. Clin Transl Oncol 2013; 16:362-73. [DOI: 10.1007/s12094-013-1090-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 07/16/2013] [Indexed: 12/19/2022]
|
48
|
Feng Y, Lau E, Scortegagna M, Ruller C, De SK, Barile E, Krajewski S, Aza-Blanc P, Williams R, Pinkerton AB, Jackson M, Chin L, Pellecchia M, Bosenberg M, Ronai ZA. Inhibition of melanoma development in the Nras((Q61K)) ::Ink4a(-/-) mouse model by the small molecule BI-69A11. Pigment Cell Melanoma Res 2012; 26:136-42. [PMID: 23035722 DOI: 10.1111/pcmr.12033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Accepted: 09/26/2012] [Indexed: 11/30/2022]
Abstract
To date, there are no effective therapies for tumors bearing NRAS mutations, which are present in 15-20% of human melanomas. Here we extend our earlier studies where we demonstrated that the small molecule BI-69A11 inhibits the growth of melanoma cell lines. Gene expression analysis revealed the induction of interferon- and cell death-related genes that were associated with responsiveness of melanoma cell lines to BI-69A11. Strikingly, the administration of BI-69A11 inhibited melanoma development in genetically modified mice bearing an inducible form of activated Nras and a deletion of the Ink4a gene (Nras((Q61K)) ::Ink4a(-/-) ). Biweekly administration of BI-69A11 starting at 10 weeks or as late as 24 weeks after the induction of mutant Nras expression inhibited melanoma development (100 and 36%, respectively). BI-69A11 treatment did not inhibit the development of histiocytic sarcomas, which constitute about 50% of the tumors in this model. BI-69A11-resistant Nras((Q61K)) ::Ink4a(-/-) tumors exhibited increased CD45 expression, reflective of immune cell infiltration and upregulation of gene networks associated with the cytoskeleton, DNA damage response, and small molecule transport. The ability to attenuate the development of NRAS mutant melanomas supports further development of BI-69A11 for clinical assessment.
Collapse
Affiliation(s)
- Yongmei Feng
- Signal Transduction Program, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
NRAS mutant melanoma: biological behavior and future strategies for therapeutic management. Oncogene 2012; 32:3009-18. [PMID: 23069660 DOI: 10.1038/onc.2012.453] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The recent years have seen a significant shift in the expectations for the therapeutic management of disseminated melanoma. The clinical success of BRAF targeted therapy suggests that long-term disease control may one day be a reality for genetically defined subgroups of melanoma patients. Despite this progress, few advances have been made in developing targeted therapeutic strategies for the 50% of patients whose melanomas are BRAF wild-type. The most well-characterized subgroup of BRAF wild-type tumors is the 15-20% of all melanomas that harbor activating NRAS (Neuroblastoma Rat Sarcoma Virus) mutations. Emerging preclinical and clinical evidence suggests that NRAS mutant melanomas have patterns of signal transduction and biological behavior that is distinct from BRAF mutant melanomas. This overview will discuss the unique clinical and prognostic behavior of NRAS mutant melanoma and will summarize the emerging data on how NRAS-driven signaling networks can be translated into novel therapeutic strategies.
Collapse
|
50
|
Superficial spreading and nodular melanoma are distinct biological entities: a challenge to the linear progression model. Melanoma Res 2012; 22:1-8. [PMID: 22108608 DOI: 10.1097/cmr.0b013e32834e6aa0] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The classification of melanoma subtypes into prognostically relevant and therapeutically insightful categories has been a challenge since the first description of melanoma in the 1800s. One limitation has been the assumption that the two most common histological subtypes of melanoma, superficial spreading and nodular, evolve according to a linear model of progression, as malignant melanocytes spread radially and then invade vertically. However, recent clinical, pathological, and molecular data indicate that these two histological subtypes might evolve as distinct entities. Here, we review the published data that support distinct molecular characterization of superficial spreading and nodular melanoma, the clinical significance of this distinction including prognostic relevance and the therapeutic implications.
Collapse
|