1851
|
RAF inhibitors that evade paradoxical MAPK pathway activation. Nature 2015; 526:583-6. [DOI: 10.1038/nature14982] [Citation(s) in RCA: 283] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 07/28/2015] [Indexed: 12/12/2022]
|
1852
|
Vergoulidou M. More than a Decade of Tyrosine Kinase Inhibitors in the Treatment of Solid Tumors: What We Have Learned and What the Future Holds. Biomark Insights 2015; 10:33-40. [PMID: 26483606 PMCID: PMC4599592 DOI: 10.4137/bmi.s22436] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 08/02/2015] [Accepted: 08/05/2015] [Indexed: 12/11/2022] Open
Abstract
The use of tyrosine kinase inhibitors (TKIs) in the treatment of solid tumors is the expected standard of care for many types of tumors. Since the description of signal transduction pathways, followed by the development of small molecules designed to inhibit those pathways, there has been significant improvement not only in progression-free survival and overall survival but also in aiming toward chemotherapy-free treatment of solid tumors to maximize quality of life. This article reviews available TKIs and discusses toxicity, dosing, and resistance.
Collapse
Affiliation(s)
- Maria Vergoulidou
- Department of Haematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charitè Medical University, Berlin, Germany
| |
Collapse
|
1853
|
Grob JJ, Long GV, Schadendorf D, Flaherty K. Disease kinetics for decision-making in advanced melanoma: a call for scenario-driven strategy trials. Lancet Oncol 2015; 16:e522-6. [DOI: 10.1016/s1470-2045(15)00003-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 11/26/2022]
|
1854
|
Rogiers A, van den Oord JJ, Garmyn M, Stas M, Kenis C, Wildiers H, Marine JC, Wolter P. Novel Therapies for Metastatic Melanoma: An Update on Their Use in Older Patients. Drugs Aging 2015; 32:821-34. [PMID: 26442859 DOI: 10.1007/s40266-015-0304-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cutaneous melanoma is the most aggressive form of skin cancer. With age as a risk factor, melanoma is projected to become a substantial healthcare burden. The clinical course of melanoma in older patients is different from that in middle-aged and younger patients: melanomas are thicker, have higher mitotic rates and are more likely to be ulcerated. Older patients also have a higher mortality rate, yet, paradoxically, have a lower rate of lymph node metastases. After decades of no significant progress in the treatment of this devastating disease, novel insights into the mechanisms underlying the pathophysiology of metastatic melanoma have led to new and remarkably efficient therapeutic opportunities. The discovery that about half of all melanomas carry BRAF mutations led to the introduction of targeted therapy with significant improvements in clinical outcomes. Although these drugs appear to be equally effective in older patients, specific considerations regarding adverse events are required. Besides targeted therapy, immunotherapy has emerged as an alternative therapeutic option. Antibodies that block cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) can induce responses with high durability. Despite an aging immune system, older patients seem to benefit to the same degree from these treatments, apparently without increased toxicity. In this review, we focus on the epidemiology, clinicopathological features, and recent developments of systemic treatment in cutaneous melanoma with regard to older patients.
Collapse
Affiliation(s)
- Aljosja Rogiers
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | | | - Marjan Garmyn
- Department of Dermatology, University Hospitals Leuven, Leuven, Belgium
| | - Marguerite Stas
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Cindy Kenis
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Hans Wildiers
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Pascal Wolter
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
1855
|
Caunt CJ, Sale MJ, Smith PD, Cook SJ. MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road. Nat Rev Cancer 2015; 15:577-92. [PMID: 26399658 DOI: 10.1038/nrc4000] [Citation(s) in RCA: 453] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The role of the ERK signalling pathway in cancer is thought to be most prominent in tumours in which mutations in the receptor tyrosine kinases RAS, BRAF, CRAF, MEK1 or MEK2 drive growth factor-independent ERK1 and ERK2 activation and thence inappropriate cell proliferation and survival. New drugs that inhibit RAF or MEK1 and MEK2 have recently been approved or are currently undergoing late-stage clinical evaluation. In this Review, we consider the ERK pathway, focusing particularly on the role of MEK1 and MEK2, the 'gatekeepers' of ERK1/2 activity. We discuss their validation as drug targets, the merits of targeting MEK1 and MEK2 versus BRAF and the mechanisms of action of different inhibitors of MEK1 and MEK2. We also consider how some of the systems-level properties (intrapathway regulatory loops and wider signalling network connections) of the ERK pathway present a challenge for the success of MEK1 and MEK2 inhibitors, discuss mechanisms of resistance to these inhibitors, and review their clinical progress.
Collapse
Affiliation(s)
- Christopher J Caunt
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Matthew J Sale
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Paul D Smith
- AstraZeneca, Oncology iMed, Cancer Biosciences, Cancer Research UK, Li Ka Shing Centre, Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| | - Simon J Cook
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| |
Collapse
|
1856
|
Vizoso M, Esteller M. Targeting melanoma: unusual epigenetics reveals the dynamic rewiring of metastatic cells. Epigenomics 2015; 7:1079-81. [DOI: 10.2217/epi.15.67] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Miguel Vizoso
- Epigenetics & Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - Manel Esteller
- Epigenetics & Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
- Department of Physiological Sciences II, School of Medicine, University of Barcelona, 08007 Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Catalonia, Spain
| |
Collapse
|
1857
|
Abstract
PURPOSE OF REVIEW To outline the most urgent challenges in the management of advanced melanoma. RECENT FINDINGS Considerable progress in targeted and immunotherapy of advanced melanoma has opened a perspective for a cure if all molecular and medical information is integrated in a rational precision treatment algorithm. SUMMARY Bioinformatics and system biology approaches will be needed to deal with omics databases. The support of patient advocacy groups may help to increase the acceptance of large scale, routine biobanking.
Collapse
|
1858
|
Mai R, Zhou S, Zhong W, Rong S, Cong Z, Li Y, Xie Q, Chen H, Li X, Liu S, Cheng Y, Huang Y, Zhou Y, Zhang G. Therapeutic efficacy of combined BRAF and MEK inhibition in metastatic melanoma: a comprehensive network meta-analysis of randomized controlled trials. Oncotarget 2015; 6:28502-12. [PMID: 26143635 PMCID: PMC4695075 DOI: 10.18632/oncotarget.4375] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 06/03/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Several recent randomized clinical trials have preliminarily demonstrated that initial targeted therapy with combined BRAF and MEK inhibition is more effective in metastatic melanoma (MM) than single agent. To guide therapeutic decisions, we did a comprehensive network meta-analysis to identify evidence to robustly support whether combined BRAF and MEK inhibition is the best initial targeted therapeutic strategy for patients with MM. METHODS The databases of PubMed and trial registries were researched for randomized clinical trials of targeted therapy. Data of outcome were extracted on progression-free survival (PFS), objective response rate (ORR), and overall survival (OS). Network meta-analysis using a Bayesian statistical model was performed to evaluate relative hazard ratio (HR) for PFS and OS, odds ratio (OR) for ORR. RESULTS Finally, 16 eligible trials comprising 5976 participants were included in this meta-analysis. PFS were significantly prolonged in patients who received combined BRAF-MEK inhibition compared with those who received BRAF inhibition (HR: 0.58, 95%CI: 0.51-0.67, P < 0.0001) or MEK inhibition alone (HR: 0.29, 95%CI: 0.22-0.37, P < 0.0001). Combined BRAF-MEK inhibition also improved the OS over BRAF inhibition (HR: 0.67, 95%CI: 0.56-0.81, P < 0.0001) or MEK inhibition alone (HR: 0.48, 95%CI: 0.36-0.65, P < 0.0001). The ORR was superior in combined BRAF and MEK inhibition comparing with BRAF inhibition (OR: 2.00, 95%CI: 1.66-2.44, P < 0.0001) or MEK inhibition alone (OR: 20.66, 95%CI: 12.22-35.47, P < 0.0001). CONCLUSIONS This study indicates that concurrent inhibition of BRAF and MEK improved the most effective therapeutic modality as compared as single BRAF or MEK inhibition for patients with MM.
Collapse
Affiliation(s)
- Ruiqin Mai
- Department of Laboratory Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Songxia Zhou
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Weixiang Zhong
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Siming Rong
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Zhichao Cong
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yunxian Li
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Qizhi Xie
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Huanming Chen
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaoyun Li
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Shuhui Liu
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yabin Cheng
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yuanshen Huang
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Youwen Zhou
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Guohong Zhang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
1859
|
The outcomes of Polish patients with advanced BRAF-positive melanoma treated with vemurafenib in a safety clinical trial. Contemp Oncol (Pozn) 2015; 19:280-3. [PMID: 26557775 PMCID: PMC4631301 DOI: 10.5114/wo.2015.54082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 04/05/2015] [Accepted: 07/20/2015] [Indexed: 11/17/2022] Open
Abstract
AIM OF THE STUDY The BRAF inhibitor vemurafenib has improved progression-free survival and overall survival in patients with BRAFV600-mutation-positive metastatic melanoma. Here we present the results of an open-label safety study with vemurafenib in patients with metastatic melanoma enrolled in Polish oncological centres. MATERIAL AND METHODS Patients with untreated or previously treated Stage IIIC/IV BRAFV600 mutation-positive melanoma were treated with oral vemurafenib in an initial dose of 960 mg twice daily. Assessments for safety and efficacy were made every 28 days. For the survival analysis the Kaplan-Meier estimator was used with the log-rank tests for bivariate comparisons. RESULTS In total, 75 Polish patients were enrolled in the safety study across four centres. At data cut-off, 28 patients died (37%), mainly (26) due to disease progression; 33 (44%) patients continued vemurafenib after disease progression. The objective response rate was 46%, including two patients with a complete response and 29 with a partial response. Median progression-free survival was 7.4 months. The one-year overall survival rate was 61.9% (median overall survival was not reached). Seventy-three (97.3%) patients reported adverse events (AEs), and grade 3-5 toxicity was reported in 49.4% (37) patients. The most common AEs were: skin lesions (including rash and photosensitivity), arthralgia, and fatigue. CONCLUSIONS The overall safety profile and response rate of vemurafenib were comparable to those reported in previous studies of this drug. Our study confirmed the value of well-established prognostic features for overall survival, such as initial LDH (lactate dehydrogenase) level and AJCC staging.
Collapse
|
1860
|
Gallagher SJ, Tiffen JC, Hersey P. Histone Modifications, Modifiers and Readers in Melanoma Resistance to Targeted and Immune Therapy. Cancers (Basel) 2015; 7:1959-82. [PMID: 26426052 PMCID: PMC4695870 DOI: 10.3390/cancers7040870] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/17/2015] [Accepted: 09/18/2015] [Indexed: 02/06/2023] Open
Abstract
The treatment of melanoma has been revolutionized by new therapies targeting MAPK signaling or the immune system. Unfortunately these therapies are hindered by either primary resistance or the development of acquired resistance. Resistance mechanisms involving somatic mutations in genes associated with resistance have been identified in some cases of melanoma, however, the cause of resistance remains largely unexplained in other cases. The importance of epigenetic factors targeting histones and histone modifiers in driving the behavior of melanoma is only starting to be unraveled and provides significant opportunity to combat the problems of therapy resistance. There is also an increasing ability to target these epigenetic changes with new drugs that inhibit these modifications to either prevent or overcome resistance to both MAPK inhibitors and immunotherapy. This review focuses on changes in histones, histone reader proteins and histone positioning, which can mediate resistance to new therapeutics and that can be targeted for future therapies.
Collapse
Affiliation(s)
- Stuart J Gallagher
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| | - Jessamy C Tiffen
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| | - Peter Hersey
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| |
Collapse
|
1861
|
|
1862
|
The Rapid Emergence of Novel Therapeutics in Advanced Malignant Melanoma. Dermatol Ther (Heidelb) 2015; 5:151-69. [PMID: 26387031 PMCID: PMC4580658 DOI: 10.1007/s13555-015-0080-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Indexed: 02/07/2023] Open
Abstract
For decades, no cancer therapy had been shown to improve average survival in metastatic melanoma. Two critical events have occurred, the discovery of melanoma driver mutation subsets and the discovery of immune checkpoint inhibitors, which have allowed for the development of modern, effective therapies. These findings have facilitated a rapid emergence of novel therapeutics for the disease with multiple FDA approvals in the last several years. The drugs vemurafenib, trametinib, and dabrafenib, which inhibit the commonly mutated BRAF pathway, have been approved based on improvements in survival outcomes. Agents that block immune checkpoints on lymphocytes allowing for immune cell activity against melanoma have also been approved based on improved survival outcomes such as ipilimumab and nivolumab. Pembrolizumab, another immune checkpoint inhibitor, has also been approved based on the response rate and duration of response in a phase 1 trial. Further agents and combinations of approved agents are positioned to possibly further increase this tally of approved drugs. This review will discuss recently approved novel agents and select drugs in development in advanced melanoma.
Collapse
|
1863
|
Wistuba-Hamprecht K, Di Benedetto S, Schilling B, Sucker A, Schadendorf D, Garbe C, Weide B, Pawelec G. Phenotypic characterization and prognostic impact of circulating γδ and αβ T-cells in metastatic malignant melanoma. Int J Cancer 2015; 138:698-704. [PMID: 26383054 DOI: 10.1002/ijc.29818] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/04/2015] [Accepted: 08/14/2015] [Indexed: 11/07/2022]
Abstract
Human T cells carrying γδ T-cell receptors (TCRs) represent a minor population relative to those with αβ TCRs. There has been much interest recently in the possibility of using these γδ T-cells in cancer therapy because they can kill tumor cells in vitro in an MHC-unrestricted manner, and possess potential regulatory capability and antigen-presenting capacity. The presence of γδ T-cells in late-stage melanoma patients and their relationship with survival has not been extensively explored, although relatively lower percentages of total γδ T-cells and Vδ2+ cells have been reported. Here, we present a detailed analysis of associations of γδ T-cell subsets and differentiation stages with survival in Stage IV patients, compared with CD4+ and CD8+ αβ T-cells. We found an increased Vδ1:Vδ2-ratio and a decreased CD4:CD8-ratio in patients compared to healthy controls, on the basis both of relative frequencies and absolute cell counts per μL blood. Nonetheless, Kaplan-Meier analyses showed that a higher than median frequency of Vδ1+ cells was negatively associated with survival, whereas there were no positive or negative associations with frequencies of Vδ2+ cells. Correlations of cell differentiation status with survival revealed a negative association of early-differentiated Vδ1+ T cells with survival, both on the basis of relative frequencies and absolute counts. There was also a positive correlation between the frequencies of early-differentiated CD8+ αβ T-cells and survival. Our findings suggest peripheral blood frequencies of Vδ1+ T-cells as a potential prognostic marker in melanoma. The mechanisms by which higher abundance of Vδ1+ cells are associated with poorer survival require determination.
Collapse
Affiliation(s)
- Kilian Wistuba-Hamprecht
- Department of Internal Medicine II, University Medical Center, Tübingen, Germany.,Department of Dermatology, University Medical Center, Tübingen, Germany
| | | | - Bastian Schilling
- Department of Dermatology, University Hospital, West German Cancer Center, University Duisburg-Essen, and the German Cancer Consortium (DKTK), Essen, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital, West German Cancer Center, University Duisburg-Essen, and the German Cancer Consortium (DKTK), Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital, West German Cancer Center, University Duisburg-Essen, and the German Cancer Consortium (DKTK), Essen, Germany
| | - Claus Garbe
- Department of Dermatology, University Medical Center, Tübingen, Germany
| | - Benjamin Weide
- Department of Dermatology, University Medical Center, Tübingen, Germany.,Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Graham Pawelec
- Department of Internal Medicine II, University Medical Center, Tübingen, Germany
| |
Collapse
|
1864
|
Prasad CP, Mohapatra P, Andersson T. Therapy for BRAFi-Resistant Melanomas: Is WNT5A the Answer? Cancers (Basel) 2015; 7:1900-24. [PMID: 26393652 PMCID: PMC4586801 DOI: 10.3390/cancers7030868] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/09/2015] [Accepted: 09/14/2015] [Indexed: 12/18/2022] Open
Abstract
In recent years, scientists have advocated the use of targeted therapies in the form of drugs that modulate genes and proteins that are directly associated with cancer progression and metastasis. Malignant melanoma is a dreadful cancer type that has been associated with the rapid dissemination of primary tumors to multiple sites, including bone, brain, liver and lungs. The discovery that approximately 40%–50% of malignant melanomas contain a mutation in BRAF at codon 600 gave scientists a new approach to tackle this disease. However, clinical studies on patients have shown that although BRAFi (BRAF inhibitors) trigger early anti-tumor responses, the majority of patients later develop resistance to the therapy. Recent studies have shown that WNT5A plays a key role in enhancing the resistance of melanoma cells to BRAFi. The focus of the current review will be on melanoma development, signaling pathways important to acquired resistance to BRAFi, and why WNT5A inhibitors are attractive candidates to be included in combinatorial therapies for melanoma.
Collapse
Affiliation(s)
- Chandra Prakash Prasad
- Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Centre, Skåne University Hospital, Malmö SE-20502, Sweden.
| | - Purusottam Mohapatra
- Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Centre, Skåne University Hospital, Malmö SE-20502, Sweden.
| | - Tommy Andersson
- Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Centre, Skåne University Hospital, Malmö SE-20502, Sweden.
| |
Collapse
|
1865
|
Xie J, Zhang X. The Impact of Genomic Profiling for Novel Cancer Therapy--Recent Progress in Non-Small Cell Lung Cancer. J Genet Genomics 2015; 43:3-10. [PMID: 26842989 DOI: 10.1016/j.jgg.2015.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 09/02/2015] [Accepted: 09/07/2015] [Indexed: 12/31/2022]
Abstract
There is high expectation for significant improvements in cancer patient care after completion of the human genome project in 2003. Through pains-taking analyses of genomic profiles in cancer patients, a number of targetable gene alterations have been discovered, with some leading to novel therapies, such as activating mutations of EGFR, BRAF and ALK gene fusions. As a result, clinical management of cancer through targeted therapy has finally become a reality for a subset of cancers, such as lung adenocarcinomas and melanomas. In this review, we summarize how gene mutation discovery leads to new treatment strategies using non-small cell lung cancer (NSCLC) as an example. We also discuss possible future implications of cancer genome analyses.
Collapse
Affiliation(s)
- Jingwu Xie
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; The Simon Cancer Center and The Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Xiaoli Zhang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; The Simon Cancer Center and The Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
1866
|
Carlino MS, Long GV. Is chemotherapy still an option in the treatment of melanoma? Ann Oncol 2015; 26:2203-4. [PMID: 26374287 DOI: 10.1093/annonc/mdv361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- M S Carlino
- Melanoma Institute Australia, Sydney The Sydney Medical School, The University of Sydney, Sydney Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney
| | - G V Long
- Melanoma Institute Australia, Sydney The Sydney Medical School, The University of Sydney, Sydney The Mater Hospital, North Sydney, Australia
| |
Collapse
|
1867
|
Improta G, Leone I, Donia M, Gieri S, Pelosi G, Fraggetta F. New developments in the management of advanced melanoma - role of pembrolizumab. Onco Targets Ther 2015; 8:2535-43. [PMID: 26396529 PMCID: PMC4576895 DOI: 10.2147/ott.s72823] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cancer immunotherapy is now recognized to be fundamental in modern oncology, because immune system recruitment may represent a powerful and innovative strategy in cancer therapy. Pembrolizumab, a highly selective humanized monoclonal antibody directly blocking the interaction between programmed cell death-1 expressed by tumor-associated T-cells and its ligand programmed cell death-L1 present on tumor and stromal cells, was recently approved by US Food and Drug Administration for the treatment of patients with unresectable or metastatic melanoma and disease progression upon ipilimumab and BRAF inhibitor. This review will focus on the clinical development and use of pembrolizumab in the clinical practice and in the management of advanced melanoma.
Collapse
Affiliation(s)
- Giuseppina Improta
- Laboratory of Clinical Research and Advanced Diagnostics, IRCCS-CROB, Rionero in Vulture, Potenza, Italy
| | - Isabella Leone
- Laboratory of Clinical Research and Advanced Diagnostics, IRCCS-CROB, Rionero in Vulture, Potenza, Italy
| | - Marco Donia
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital, Herlev, Denmark
| | - Stefania Gieri
- Laboratory of Oncologic Technologies, IBFM-CNR, Cefalù, Potenza, Italy
| | - Giuseppe Pelosi
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy ; Department of Biomedical and Clinical Sciences "Luigi Sacco", Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
1868
|
Barras D. BRAF Mutation in Colorectal Cancer: An Update. BIOMARKERS IN CANCER 2015; 7:9-12. [PMID: 26396549 PMCID: PMC4562608 DOI: 10.4137/bic.s25248] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/17/2015] [Accepted: 08/19/2015] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is still one of the deadliest cancer-related diseases. About 10% of CRC patients are characterized by a mutation in the B-Raf proto-oncogene serine/threonine kinase (BRAF) gene resulting in a valine-to-glutamate change at the residue 600 (V600E). This mutation is also present in more than 60% of melanoma patients. BRAF inhibitors were developed and found to improve patient survival; however, most patients at the end of the track ultimately develop resistance to these inhibitors. Melanoma patients benefit from the combination of BRAF inhibitors with mitogen/extracellular signal-regulated kinase (MEK) inhibitors, among others. Unfortunately, colorectal patients do not respond much efficiently, which suggests different resistance mechanisms between the two cancer types. This review aims at shedding light on recent discoveries that improve our understanding of the BRAF mutation biology in CRC.
Collapse
Affiliation(s)
- David Barras
- SIB Swiss Institute of Bioinformatics, Bioinformatics Core Facility, Lausanne, Switzerland
| |
Collapse
|
1869
|
Cohen R, Cervera P, Svrcek M, Dumont C, Garcia ML, Chibaudel B, de Gramont A, Pocard M, Duval A, Fléjou JF, André T. [DNA mismatch repair and BRAF status in colorectal cancer: Interest for the therapeutic management?]. Bull Cancer 2015; 102:S72-81. [PMID: 26118880 DOI: 10.1016/s0007-4551(15)31220-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/09/2015] [Indexed: 12/19/2022]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related mortality in France. Recently, colorectal cancer subtyping consortium (CRCSC) identified 4 consensus molecular subtypes (CMS). CMS1 is enriched for CRC with deficient DNA mismatch repair system (dMMR) and tumors with mutated BRAF. Intriguingly, CMS1 is characterized by better relapse-free survival but worse survival after relapse, compared with the other subtypes. In this review, we provide a comprehensive overview of prognostic and predictive impacts of MMR and BRAF status. We highlight immune checkpoints inhibitors as potentially future therapeutics for CRC with deficient MMR. We also focus on the management of BRAF mutant metastatic CRC, with a particular interest on targeted therapies.
Collapse
Affiliation(s)
- Romain Cohen
- Service d'oncologie médicale, Hôpitaux universitaires de l'Est Parisien (AP-HP), Site Saint-Antoine 184, rue du Faubourg Saint-Antoine, 75571 Paris cedex 12, France; Groupe coopérateur multidisciplinaire en oncologie (GERCOR), Paris, France
| | - Pascale Cervera
- Service d'oncologie médicale, Hôpitaux universitaires de l'Est Parisien (AP-HP), Site Saint-Antoine 184, rue du Faubourg Saint-Antoine, 75571 Paris cedex 12, France
| | - Magali Svrcek
- Service d'oncologie médicale, Hôpitaux universitaires de l'Est Parisien (AP-HP), Site Saint-Antoine 184, rue du Faubourg Saint-Antoine, 75571 Paris cedex 12, France; Inserm, Unité mixte de recherche scientifique 938, Centre de recherche Saint-Antoine, Équipe Instabilité des microsatellites et cancers, Équipe labellisée par la Ligue nationale contre le cancer, Paris, France
| | - Clément Dumont
- Service d'oncologie médicale, Hôpitaux universitaires de l'Est Parisien (AP-HP), Site Saint-Antoine 184, rue du Faubourg Saint-Antoine, 75571 Paris cedex 12, France
| | - Marie-Line Garcia
- Service d'oncologie médicale, Hôpitaux universitaires de l'Est Parisien (AP-HP), Site Saint-Antoine 184, rue du Faubourg Saint-Antoine, 75571 Paris cedex 12, France
| | - Benoist Chibaudel
- Groupe coopérateur multidisciplinaire en oncologie (GERCOR), Paris, France; Institut hospitalier franco-britannique, 4 rue Kléber, 92300 Levallois-Perret, France
| | - Aimery de Gramont
- Groupe coopérateur multidisciplinaire en oncologie (GERCOR), Paris, France; Institut hospitalier franco-britannique, 4 rue Kléber, 92300 Levallois-Perret, France
| | - Marc Pocard
- Hôpital Lariboisière, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, CART, Inserm U965, Paris, France
| | - Alex Duval
- Inserm, Unité mixte de recherche scientifique 938, Centre de recherche Saint-Antoine, Équipe Instabilité des microsatellites et cancers, Équipe labellisée par la Ligue nationale contre le cancer, Paris, France
| | - Jean-François Fléjou
- Service d'oncologie médicale, Hôpitaux universitaires de l'Est Parisien (AP-HP), Site Saint-Antoine 184, rue du Faubourg Saint-Antoine, 75571 Paris cedex 12, France; Université Pierre et Marie Curie (UPMC), Paris VI, Paris, France
| | - Thierry André
- Service d'oncologie médicale, Hôpitaux universitaires de l'Est Parisien (AP-HP), Site Saint-Antoine 184, rue du Faubourg Saint-Antoine, 75571 Paris cedex 12, France; Université Pierre et Marie Curie (UPMC), Paris VI, Paris, France.
| |
Collapse
|
1870
|
Massi D, Brusa D, Merelli B, Falcone C, Xue G, Carobbio A, Nassini R, Baroni G, Tamborini E, Cattaneo L, Audrito V, Deaglio S, Mandalà M. The status of PD-L1 and tumor-infiltrating immune cells predict resistance and poor prognosis in BRAFi-treated melanoma patients harboring mutant BRAFV600. Ann Oncol 2015; 26:1980-1987. [DOI: 10.1093/annonc/mdv255] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/19/2015] [Indexed: 01/07/2023] Open
|
1871
|
Buchbinder E, Hodi FS. Cytotoxic T lymphocyte antigen-4 and immune checkpoint blockade. J Clin Invest 2015; 125:3377-83. [PMID: 26325034 DOI: 10.1172/jci80012] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The relationship between cancer and the immune system is complex and provides unique therapeutic opportunities. Cytotoxic T lymphocyte antigen-4 (CTLA-4) is a regulatory molecule that suppresses T cell effector function following initial activation by costimulatory signals. Fully human monoclonal antibodies targeting CTLA-4 have been shown to increase T cell function and antitumor responses in patients with advanced metastatic melanoma. Responses observed with such immune checkpoint therapy can follow a different pattern from that seen with cytotoxic chemotherapy or targeted therapy and may continue after therapy is discontinued. In addition, the toxicities that are associated with anti-CTLA-4 therapy may differ from those of conventional therapies and consist of inflammatory events in parts of the body that do not contain cancerous cells. Early recognition of these inflammatory events and intervention is important, and the identification of predictive biomarkers continues to be an unfulfilled need in the field of immunotherapy. Combinatorial approaches with targeted therapies, radiation therapy, chemotherapy, or other immune checkpoint agonists/antagonists have the potential to increase the efficacy of CTLA-4 blockade.
Collapse
|
1872
|
He J, Ahuja N. Personalized Approaches to Gastrointestinal Cancers: Importance of Integrating Genomic Information to Guide Therapy. Surg Clin North Am 2015; 95:1081-94. [PMID: 26315525 DOI: 10.1016/j.suc.2015.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancers are characterized by complex tumor heterogeneity driven by subclones with differential genotypes and phenotypes, which then drives cancer behavior. As genomic strategies become feasible on smaller samples such as biopsies, coupled with decreasing costs of these approaches, clinicians will increasingly use genomic information to drive therapeutic decision making. Early applications of such personalized approaches are discussed. Genetic testing of high-risk family members may identify patients with germline mutations who can have prophylactic surgeries as a cancer prevention strategy. This article discusses examples of successful targeted therapy. Clinical trials need to incorporate genetic testing to stratify patients into different groups.
Collapse
Affiliation(s)
- Jin He
- Department of Surgery, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Blalock 1202, Baltimore, MD 21287, USA
| | - Nita Ahuja
- Department of Surgery, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Blalock 685, Baltimore, MD 21287, USA.
| |
Collapse
|
1873
|
Hrustanovic G, Olivas V, Pazarentzos E, Tulpule A, Asthana S, Blakely CM, Okimoto RA, Lin L, Neel DS, Sabnis A, Flanagan J, Chan E, Varella-Garcia M, Aisner DL, Vaishnavi A, Ou SHI, Collisson EA, Ichihara E, Mack PC, Lovly CM, Karachaliou N, Rosell R, Riess JW, Doebele RC, Bivona TG. RAS-MAPK dependence underlies a rational polytherapy strategy in EML4-ALK-positive lung cancer. Nat Med 2015; 21:1038-47. [PMID: 26301689 PMCID: PMC4734742 DOI: 10.1038/nm.3930] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 07/23/2015] [Indexed: 12/20/2022]
Abstract
One strategy for combating cancer-drug resistance is to deploy rational polytherapy up front that suppresses the survival and emergence of resistant tumor cells. Here we demonstrate in models of lung adenocarcinoma harboring the oncogenic fusion of ALK and EML4 that the GTPase RAS-mitogen-activated protein kinase (MAPK) pathway, but not other known ALK effectors, is required for tumor-cell survival. EML4-ALK activated RAS-MAPK signaling by engaging all three major RAS isoforms through the HELP domain of EML4. Reactivation of the MAPK pathway via either a gain in the number of copies of the gene encoding wild-type K-RAS (KRAS(WT)) or decreased expression of the MAPK phosphatase DUSP6 promoted resistance to ALK inhibitors in vitro, and each was associated with resistance to ALK inhibitors in individuals with EML4-ALK-positive lung adenocarcinoma. Upfront inhibition of both ALK and the kinase MEK enhanced both the magnitude and duration of the initial response in preclinical models of EML4-ALK lung adenocarcinoma. Our findings identify RAS-MAPK dependence as a hallmark of EML4-ALK lung adenocarcinoma and provide a rationale for the upfront inhibition of both ALK and MEK to forestall resistance and improve patient outcomes.
Collapse
Affiliation(s)
- Gorjan Hrustanovic
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Victor Olivas
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Evangelos Pazarentzos
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Asmin Tulpule
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Saurabh Asthana
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Collin M Blakely
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Ross A Okimoto
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Luping Lin
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Dana S Neel
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Amit Sabnis
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Jennifer Flanagan
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Elton Chan
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Marileila Varella-Garcia
- Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
- Department of Pathology, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
| | - Dara L Aisner
- Department of Pathology, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
| | - Aria Vaishnavi
- Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
| | - Sai-Hong I Ou
- Division of Hematology-Oncology, University of California Irvine School of Medicine, Orange, California, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, California, USA
| | - Eric A Collisson
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| | - Eiki Ichihara
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Philip C Mack
- University of California Davis School of Medicine
- Comprehensive Cancer Center, Sacramento, California, USA
| | - Christine M Lovly
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Niki Karachaliou
- Cancer Biology and Precision Medicine Program Catalan Institute of Oncology Hospital Germans Trias i Pujol Badalona, Barcelona, Spain
| | - Rafael Rosell
- Cancer Biology and Precision Medicine Program Catalan Institute of Oncology Hospital Germans Trias i Pujol Badalona, Barcelona, Spain
| | - Jonathan W Riess
- University of California Davis School of Medicine
- Comprehensive Cancer Center, Sacramento, California, USA
| | - Robert C Doebele
- Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
| | - Trever G Bivona
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California, USA
| |
Collapse
|
1874
|
Dummer R, Hauschild A, Lindenblatt N, Pentheroudakis G, Keilholz U. Cutaneous melanoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2015; 26 Suppl 5:v126-32. [PMID: 26314774 DOI: 10.1093/annonc/mdv297] [Citation(s) in RCA: 276] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- R Dummer
- Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - A Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - N Lindenblatt
- Division of Plastic and Reconstructive Surgery, University Hospital Zürich, Zürich, Switzerland
| | | | - U Keilholz
- Charité Comprehensive Cancer Center, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
1875
|
Zappasodi R, Merghoub T. Alphavirus-based vaccines in melanoma: rationale and potential improvements in immunotherapeutic combinations. Immunotherapy 2015; 7:981-97. [DOI: 10.2217/imt.15.64] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Immune checkpoint blockade has formally demonstrated the clinical benefit of immunotherapy against melanoma. New immunotherapeutic modalities are currently explored to improve the management of relapsing/refractory patients. Potent antitumor vaccines would have the advantage to promote long-lasting tumor control while limiting autoimmunity. Alphavirus vectors and nonreplicating particles offer versatile platforms to deliver antigen expression and immunize against cancer. They have shown promising preclinical results and initial proof of clinical activity in melanoma. The growing number of clinically available immunomodulatory agents provides a tremendous opportunity to exploit and revisit anticancer vaccines in the setting of powerful immunotherapeutic combinations. Accelerating the evaluation of alphavirus-based vaccines in patients with immune sensitive, but still very deadly malignancies, such as melanoma, is thus extremely important.
Collapse
Affiliation(s)
- Roberta Zappasodi
- Ludwig Collaborative & Swim Across America Laboratory, New York, NY, USA
| | - Taha Merghoub
- Ludwig Collaborative & Swim Across America Laboratory, New York, NY, USA
- Melanoma & Immunotherapeutics Service MSKCC, New York, NY, USA
| |
Collapse
|
1876
|
Michielin O, Hoeller C. Gaining momentum: New options and opportunities for the treatment of advanced melanoma. Cancer Treat Rev 2015; 41:660-70. [DOI: 10.1016/j.ctrv.2015.05.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/26/2015] [Accepted: 05/27/2015] [Indexed: 12/31/2022]
|
1877
|
Zia Y, Chen L, Daud A. Future of combination therapy with dabrafenib and trametinib in metastatic melanoma. Expert Opin Pharmacother 2015; 16:2257-63. [DOI: 10.1517/14656566.2015.1085509] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
1878
|
Dienstmann R, Lassen U, Cebon J, Desai J, Brown MP, Evers S, Su F, Zhang W, Boisserie F, Lestini B, Schostack K, Meresse V, Tabernero J. First-in-Man Dose-Escalation Study of the Selective BRAF Inhibitor RG7256 in Patients with BRAF V600-Mutated Advanced Solid Tumors. Target Oncol 2015; 11:149-56. [PMID: 26310975 DOI: 10.1007/s11523-015-0381-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND BRAF mutations are a validated target for cancer therapy. A second-generation BRAF inhibitor with an improved preclinical safety profile (RG7256) was evaluated in a first-in-man study in order to determine the safety, efficacy, pharmacokinetics and pharmacodynamics in patients with BRAF V600-mutated advanced solid tumors. PATIENTS AND METHODS Patients received RG7256 orally over 8 dose levels from 200 mg once a day (QD) to 2400 mg twice a day (BID) (50-, 100- and 150-mg tablets) using a classic 3 + 3 dose escalation design. RESULTS In total, 45 patients were enrolled; most (87 %) had advanced melanoma (94 % BRAF V600E). RG7256 was rapidly absorbed, with limited accumulation and dose-proportional increase in exposure up to 1950 mg BID. The maximal tolerated dose (MTD) was not reached. The most common drug-related adverse events (AEs) were dyspepsia (20 %), dry skin (18 %), rash (18 %), fatigue (16 %) and nausea (13 %), mainly grade 1. Three patients (7 %) developed cutaneous squamous cell carcinoma. Photosensitivity, arthralgia and increased liver enzyme levels were each observed in only one patient each. Of 44 evaluable patients, 14 (32 %) had a partial response (melanoma and thyroid cancer). At high dose levels (>1200 mg BID), 10 of 16 (63 %) patients had a partial response. A decrease in maximum standardized uptake value (SUVmax) on FDG-PET of ≥25 % was observed in 19 of 37 patients. On-treatment reductions in pERK were documented in eight of ten paired tumor samples. CONCLUSIONS RG7256 has a favorable safety profile compared to other BRAF inhibitors while maintaining clinical activity, and MTD was not reached. The excessive pill burden needed to provide the desired exposure, and thus concerns about patient compliance, limited further development of this agent. Study Identifier: ClinicalTrials.gov (NCT01143753).
Collapse
Affiliation(s)
- Rodrigo Dienstmann
- Vall d'Hebron University Hospital, Medical Oncology, Barcelona, Spain.
- , P. Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| | - Ulrik Lassen
- Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | | | | | - Michael P Brown
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
| | - Stefan Evers
- Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Fei Su
- Pharma Research & Early Development, Roche Innovation Center New York, New York, NY, USA
- Oncology Correlative Science Lead, Novartis Pharmaceutical Corporation, East Hanover, NJ, USA
| | - Weijiang Zhang
- Pharma Research & Early Development, Roche Innovation Center New York, New York, NY, USA
| | - Frederic Boisserie
- Pharma Research & Early Development, Roche Innovation Center New York, New York, NY, USA
| | - Brian Lestini
- Pharma Research & Early Development, Roche Innovation Center New York, New York, NY, USA
- Oncology Global Clinical Research, Bristol-Myers Squibb, New York, NY, USA
| | - Kathleen Schostack
- Pharma Research & Early Development, Roche Innovation Center New York, New York, NY, USA
- Global Development, Oncology, Bayer HealthCare Pharmaceuticals, Inc, Whippany, NJ, USA
| | - Valerie Meresse
- Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Josep Tabernero
- Vall d'Hebron University Hospital, Medical Oncology, Barcelona, Spain.
- , P. Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| |
Collapse
|
1879
|
Abstract
Colorectal cancer has been characterized as a genetically heterogeneous disease, with a large diversity in molecular pathogenesis resulting in differential responses to therapy. However, the currently available validated biomarkers KRAS, BRAF, and microsatellite instability do not sufficiently cover this extensive heterogeneity and are therefore not suitable to successfully guide personalized treatment. Recent studies have focused on novel targets and rationally designed combination strategies. Furthermore, a more comprehensive analysis of the underlying biology of the disease revealed distinct phenotypic differences within subgroups of patients harboring the same genetic driver mutation with both prognostic and predictive relevance. Accordingly, patient stratification based on molecular intrinsic subtypes rather than on single gene aberrations holds promise to improve the clinical outcome of patients with colorectal cancer.
Collapse
|
1880
|
Lugowska I, Koseła-Paterczyk H, Kozak K, Rutkowski P. Trametinib: a MEK inhibitor for management of metastatic melanoma. Onco Targets Ther 2015; 8:2251-9. [PMID: 26347206 PMCID: PMC4556032 DOI: 10.2147/ott.s72951] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
This review presents the current data on the efficacy and safety of the selective mitogen-activated extracellular signal-regulated kinase (MEK) inhibitor trametinib in patients with metastatic BRAF V600-positive melanoma. The pharmacological, safety, and efficacy data come from the Phase I, II, and III studies of trametinib monotherapy, as well as those in combination with the BRAF inhibitor dabrafenib. The most common adverse effects of trametinib therapy are rash, dermatitis, diarrhea, and fatigue. The Phase III METRIC study showed significant improvement in overall survival and progression-free survival in favor of trametinib over standard dacarbazine or paclitaxel chemotherapy. Therefore, trametinib was approved by the US Food and Drug Administration and European Medicines Agency as a single agent for the treatment of patients with V600E-mutated metastatic melanoma. Progression-free survival and response rates for trametinib monotherapy were lower than those noted with BRAF inhibitors. The second step in developing trametinib was to use the combination of trametinib with the BRAF inhibitor, eg, dabrafenib, to postpone the progression on MEK or BRAF inhibitors. The recently published data showed significant improvement in overall survival and progression-free survival in favor of the combination of trametinib and dabrafenib over vemurafenib therapy or dabrafenib alone, with good tolerance. The US Food and Drug Administration has approved the combination of dabrafenib (150 mg orally twice daily) and trametinib (2 mg orally once daily) for the treatment of patients with BRAF V600E/K-mutant metastatic melanoma, and their use seems to be currently the best approach. While BRAF-MEK inhibition is a standard, molecular targeted therapy in BRAF-mutated melanomas, its future utility has to be established in the rapidly changing landscape of immunotherapeutics.
Collapse
Affiliation(s)
- Iwona Lugowska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland ; Department of Epidemiology, Institute of Mother and Child, Warsaw, Poland
| | - Hanna Koseła-Paterczyk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| | - Katarzyna Kozak
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| |
Collapse
|
1881
|
Weiss SA, Pavlick AC. Dabrafenib for the treatment of melanoma. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1067136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
1882
|
Comparative Aspects of BRAF Mutations in Canine Cancers. Vet Sci 2015; 2:231-245. [PMID: 29061943 PMCID: PMC5644641 DOI: 10.3390/vetsci2030231] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/14/2015] [Accepted: 08/19/2015] [Indexed: 01/07/2023] Open
Abstract
Activating mutations of the BRAF gene lead to constitutive activation of the MAPK pathway. The characterization and discovery of BRAF mutations in a variety of human cancers has led to the development of specific inhibitors targeting the BRAF/MAPK pathway and dramatically changed clinical outcomes in BRAF-mutant melanoma patients. Recent discovery of BRAF mutation in canine cancers underscores the importance of MAPK pathway activation as an oncogenic molecular alteration evolutionarily conserved between species. A comparative approach using the domestic dog as a spontaneous cancer model will provide new insights into the dysregulation of BRAF/MAPK pathway in carcinogenesis and facilitate in vivo studies to evaluate therapeutic strategies targeting this pathway's molecules for cancer therapy. The BRAF mutation in canine cancers may also represent a molecular marker and therapeutic target in veterinary oncology. This review article summarizes the current knowledge on BRAF mutations in human and canine cancers and discusses the potential applications of this abnormality in veterinary oncology.
Collapse
|
1883
|
Ramsdale R, Jorissen RN, Li FZ, Al-Obaidi S, Ward T, Sheppard KE, Bukczynska PE, Young RJ, Boyle SE, Shackleton M, Bollag G, Long GV, Tulchinsky E, Rizos H, Pearson RB, McArthur GA, Dhillon AS, Ferrao PT. The transcription cofactor c-JUN mediates phenotype switching and BRAF inhibitor resistance in melanoma. Sci Signal 2015; 8:ra82. [PMID: 26286024 DOI: 10.1126/scisignal.aab1111] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Most patients with BRAF-mutant metastatic melanoma display remarkable but incomplete and short-lived responses to inhibitors of the BRAF kinase or the mitogen-activated protein kinase kinase (MEK), collectively BRAF/MEK inhibitors. We found that inherent resistance to these agents in BRAF(V600)-mutant melanoma cell lines was associated with high abundance of c-JUN and characteristics of a mesenchymal-like phenotype. Early drug adaptation in drug-sensitive cell lines grown in culture or as xenografts, and in patient samples during therapy, was consistently characterized by down-regulation of SPROUTY4 (a negative feedback regulator of receptor tyrosine kinases and the BRAF-MEK signaling pathway), increased expression of JUN and reduced expression of LEF1. This coincided with a switch in phenotype that resembled an epithelial-mesenchymal transition (EMT). In cultured cells, these BRAF inhibitor-induced changes were reversed upon removal of the drug. Knockdown of SPROUTY4 was sufficient to increase the abundance of c-JUN in the absence of drug treatment. Overexpressing c-JUN in drug-naïve melanoma cells induced similar EMT-like phenotypic changes to BRAF inhibitor treatment, whereas knocking down JUN abrogated the BRAF inhibitor-induced early adaptive changes associated with resistance and enhanced cell death. Combining the BRAF inhibitor with an inhibitor of c-JUN amino-terminal kinase (JNK) reduced c-JUN phosphorylation, decreased cell migration, and increased cell death in melanoma cells. Gene expression data from a panel of melanoma cell lines and a patient cohort showed that JUN expression correlated with a mesenchymal gene signature, implicating c-JUN as a key mediator of the mesenchymal-like phenotype associated with drug resistance.
Collapse
Affiliation(s)
- Rachel Ramsdale
- Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, St. Andrew's Place, East Melbourne, Victoria 3002, Australia. Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Robert N Jorissen
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Melbourne, Victoria 3052, Australia. Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia
| | - Frederic Z Li
- Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, St. Andrew's Place, East Melbourne, Victoria 3002, Australia. Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia
| | - Sheren Al-Obaidi
- Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, St. Andrew's Place, East Melbourne, Victoria 3002, Australia. Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Teresa Ward
- Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, St. Andrew's Place, East Melbourne, Victoria 3002, Australia. Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Karen E Sheppard
- Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, St. Andrew's Place, East Melbourne, Victoria 3002, Australia. Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia
| | - Patricia E Bukczynska
- Molecular Therapeutics and Biomarkers Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Richard J Young
- Molecular Therapeutics and Biomarkers Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Samantha E Boyle
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Cancer Development and Treatment Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Department of Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia
| | - Mark Shackleton
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia. Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Cancer Development and Treatment Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Department of Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia
| | - Gideon Bollag
- Plexxikon Inc., 91 Bolivar Drive, Berkeley, CA 94710, USA
| | - Georgina V Long
- Melanoma Institute Australia, Sydney, New South Wales 2060, Australia. University of Sydney, Sydney, New South Wales 2006, Australia
| | - Eugene Tulchinsky
- Department of Cancer Studies and Molecular Medicine, University of Leicester, Leicester LE2 7LX, UK
| | - Helen Rizos
- Melanoma Institute Australia, Sydney, New South Wales 2060, Australia. Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Richard B Pearson
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia. Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia. Cancer Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Grant A McArthur
- Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, St. Andrew's Place, East Melbourne, Victoria 3002, Australia. Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia. Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Amardeep S Dhillon
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia. Department of Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia
| | - Petranel T Ferrao
- Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, St. Andrew's Place, East Melbourne, Victoria 3002, Australia. Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia. Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. Department of Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria 3010, Australia.
| |
Collapse
|
1884
|
Rivers JK. The Forecast Is Bright: Recent Advances in Melanoma Treatment. J Cutan Med Surg 2015; 19:435-9. [PMID: 26271962 DOI: 10.1177/1203475415599152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
1885
|
Buchbinder EI, Sosman JA, Lawrence DP, McDermott DF, Ramaiya NH, Van den Abbeele AD, Linette GP, Giobbie-Hurder A, Hodi FS. Phase 2 study of sunitinib in patients with metastatic mucosal or acral melanoma. Cancer 2015; 121:4007-15. [PMID: 26264378 DOI: 10.1002/cncr.29622] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/06/2015] [Accepted: 06/15/2015] [Indexed: 11/12/2022]
Abstract
BACKGROUND Patients with mucosal and acral melanomas have limited treatment options and a poor prognosis. Mutations of the KIT oncogene in these melanoma subtypes provide a potential therapeutic target. METHODS A multicenter phase 2 trial of sunitinib was conducted in patients with unresectable stage III or IV melanoma of a mucosal or acral primary origin. Patients were treated in 2 cohorts: cohort A received sunitinib at a dose of 50 mg daily for 4 weeks of a 6-week cycle, and cohort B received sunitinib at a dose of 37.5 mg daily on a continuous basis. Dose reductions were permitted for treatment-related toxicities, and tumor assessments were performed every 2 months. RESULTS Fifty-two patients were enrolled: 21 in cohort A and 31 in cohort B. Four patients had confirmed partial responses, which lasted 5 to 10 months (1 with a KIT mutation). In both cohorts, the proportion of patients alive and progression-free at 2 months was 52% (95% confidence interval, 38%-66%); this was significantly larger than the hypothesized null of 5%. There was no significant difference in response or overall survival between the 25% of patients with a KIT mutation and those without one (response rate, 7.7% vs 9.7%; overall survival, 6.4 vs 8.6 months). The overall disease control rate was 44%, and a high rate of toxicity was associated with the treatment. CONCLUSIONS Sunitinib showed activity in the treatment of mucosal and acral melanoma that was not dependent on the presence of a KIT mutation. However, the medication was poorly tolerated, and there were no prolonged responses. Cancer 2015;121:4007-4015. © 2015 American Cancer Society.
Collapse
Affiliation(s)
| | - Jeffrey A Sosman
- Hematology-Oncology, Vanderbilt University, Nashville, Tennessee
| | - Donald P Lawrence
- Hematology-Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - David F McDermott
- Hematology-Oncology, Beth Israel-Deaconess Medical Center, Boston, Massachusetts
| | - Nikhil H Ramaiya
- Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Annick D Van den Abbeele
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Gerald P Linette
- Hematology-Oncology, Washington University in St. Louis, St. Louis, Missouri
| | - Anita Giobbie-Hurder
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
1886
|
Palmieri G, Ombra M, Colombino M, Casula M, Sini M, Manca A, Paliogiannis P, Ascierto PA, Cossu A. Multiple Molecular Pathways in Melanomagenesis: Characterization of Therapeutic Targets. Front Oncol 2015; 5:183. [PMID: 26322273 PMCID: PMC4530319 DOI: 10.3389/fonc.2015.00183] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/26/2015] [Indexed: 12/12/2022] Open
Abstract
Molecular mechanisms involved in pathogenesis of malignant melanoma have been widely studied and novel therapeutic treatments developed in recent past years. Molecular targets for therapy have mostly been recognized in the RAS–RAF–MEK–ERK and PI3K–AKT signaling pathways; small-molecule inhibitors were drawn to specifically target key kinases. Unfortunately, these targeted drugs may display intrinsic or acquired resistance and various evidences suggest that inhibition of a single effector of the signal transduction cascades involved in melanoma pathogenesis may be ineffective in blocking the tumor growth. In this sense, a wider comprehension of the multiple molecular alterations accounting for either response or resistance to treatments with targeted inhibitors may be helpful in assessing, which is the most effective combination of such therapies. In the present review, we summarize the known molecular mechanisms underlying either intrinsic and acquired drug resistance either alternative roads to melanoma pathogenesis, which may become targets for innovative anticancer approaches.
Collapse
Affiliation(s)
- Giuseppe Palmieri
- Unità di Genetica dei Tumori, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche , Sassari , Italy
| | - MariaNeve Ombra
- Istituto di Scienze dell'Alimentazione, Consiglio Nazionale delle Ricerche , Avellino , Italy
| | - Maria Colombino
- Unità di Genetica dei Tumori, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche , Sassari , Italy
| | - Milena Casula
- Unità di Genetica dei Tumori, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche , Sassari , Italy
| | - MariaCristina Sini
- Unità di Genetica dei Tumori, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche , Sassari , Italy
| | - Antonella Manca
- Unità di Genetica dei Tumori, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche , Sassari , Italy
| | - Panagiotis Paliogiannis
- Dipartimento di Scienze Chirurgiche, Microchirurgiche e Mediche, Università di Sassari , Sassari , Italy
| | | | - Antonio Cossu
- Dipartimento di Scienze Chirurgiche, Microchirurgiche e Mediche, Università di Sassari , Sassari , Italy
| |
Collapse
|
1887
|
Grimaldi AM, Simeone E, Ascierto PA. Vemurafenib plus cobimetinib in the treatment of mutated metastatic melanoma: the CoBRIM trial. Melanoma Manag 2015; 2:209-215. [PMID: 30190850 DOI: 10.2217/mmt.15.22] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The concomitant inhibition of both BRAF and MEK can produce a more durable and greater tumor response than BRAF monotherapy while reducing BRAF inhibitor-related toxicity. Further evidence of the benefits of combined MEK and BRAF inhibition have been provided by the CoBRIM trial in which median progression-free survival was significantly increased with vemurafenib plus cobimetinib compared with vemurafenib alone (9.9 vs 6.2 months; hazard ratio for death or progression: 0.51; 95% CI: 0.39-0.68; p < 0.001) in 495 patients with advanced BRAF-mutated melanoma. Overall survival data in the CoBRIM trial were immature at time of final progression-free survival analysis but showed an hazard ratio for death of 0.65 (95% CI: 0.42-1.00; p = 0.046; boundary p < 0.0000037). Combination therapy was well tolerated with a reduced incidence of cutaneous squamous-cell carcinoma/keratoacanthoma. This combination may be a starting point for novel combination strategies with immunotherapies and other targeted therapies.
Collapse
Affiliation(s)
- Antonio M Grimaldi
- Melanoma, Cancer Immunotherapy & Innovative Therapies Unit, Istituto Nazionale Tumori Fondazione "G Pascale", Napoli, Italy
| | - Ester Simeone
- Melanoma, Cancer Immunotherapy & Innovative Therapies Unit, Istituto Nazionale Tumori Fondazione "G Pascale", Napoli, Italy
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy & Innovative Therapies Unit, Istituto Nazionale Tumori Fondazione "G Pascale", Napoli, Italy
| |
Collapse
|
1888
|
Bowyer S, Lee R, Fusi A, Lorigan P. Dabrafenib and its use in the treatment of metastatic melanoma. Melanoma Manag 2015; 2:199-208. [PMID: 30190849 DOI: 10.2217/mmt.15.21] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Approximately 50% of melanomas have mutations in the gene encoding BRAF. In recent years, new targeted therapies have transformed the landscape of metastatic melanoma treatment. Dabrafenib, a potent kinase inhibitor of mutated BRAF, has been showed to have high response rates with a rapid onset of response, as well as improved overall and progression-free survival when compared with chemotherapy. Dabrafenib in combination with trametinib, a MEK inhibitor, has demonstrated higher responses and improved clinical efficacy compared with monotherapy. Toxicity is distinct compared with chemotherapy but manageable. This article summarizes the pharmacology, key clinical trial data as well as practical experience with dabrafenib in clinical practice, and future directions.
Collapse
Affiliation(s)
- Samantha Bowyer
- Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK.,Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Rebecca Lee
- Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK.,Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Alberto Fusi
- Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK.,Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Paul Lorigan
- Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK.,Department of Medical Oncology, University of Manchester, Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester, M20 4BX, UK.,Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK.,Department of Medical Oncology, University of Manchester, Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester, M20 4BX, UK
| |
Collapse
|
1889
|
Kim KB. Nivolumab in the treatment of advanced melanoma. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1063416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
1890
|
Berghoff AS, Preusser M. The future of targeted therapies for brain metastases. Future Oncol 2015; 11:2315-27. [DOI: 10.2217/fon.15.127] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Brain metastases (BM) are an increasing challenge in the management of patients with advanced cancer. Treatment options for BM are limited and mainly focus on the application of local therapies. Systemic therapies including targeted therapies are only poorly investigated, as patients with BM were frequently excluded from clinical trials. Several targeted therapies have shown promising activity in patients with BM. In the present review we discuss existing and emerging targeted therapies for the most frequent BM primary tumor types. We focus on challenges in the conduction of clinical trials on targeted therapies in BM patients such as patient selection, combination with radiotherapy, the obstacles of the blood–brain barrier and the definition of study end points.
Collapse
Affiliation(s)
- Anna S Berghoff
- Department for Medicine I, Comprehensive Cancer Center Central Nervous System Unit (CCC-CNS), Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center – CNS Tumors Unit, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Department for Medicine I, Comprehensive Cancer Center Central Nervous System Unit (CCC-CNS), Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center – CNS Tumors Unit, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
1891
|
Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, Garbe C, Jouary T, Hauschild A, Grob JJ, Chiarion-Sileni V, Lebbe C, Mandalà M, Millward M, Arance A, Bondarenko I, Haanen JBAG, Hansson J, Utikal J, Ferraresi V, Kovalenko N, Mohr P, Probachai V, Schadendorf D, Nathan P, Robert C, Ribas A, DeMarini DJ, Irani JG, Swann S, Legos JJ, Jin F, Mookerjee B, Flaherty K. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial. Lancet 2015; 386:444-51. [PMID: 26037941 DOI: 10.1016/s0140-6736(15)60898-4] [Citation(s) in RCA: 1025] [Impact Index Per Article: 102.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Previously, a study of ours showed that the combination of dabrafenib and trametinib improves progression-free survival compared with dabrafenib and placebo in patients with BRAF Val600Lys/Glu mutation-positive metastatic melanoma. The study was continued to assess the secondary endpoint of overall survival, which we report in this Article. METHODS We did this double-blind phase 3 study at 113 sites in 14 countries. We enrolled previously untreated patients with BRAF Val600Glu or Val600Lys mutation-positive unresectable stage IIIC or stage IV melanoma. Participants were computer-randomised (1:1) to receive a combination of dabrafenib (150 mg orally twice daily) and trametinib (2 mg orally once daily), or dabrafenib and placebo. The primary endpoint was progression-free survival and overall survival was a secondary endpoint. This study is registered with ClinicalTrials.gov, number NCT01584648. FINDINGS Between May 4, 2012, and Nov 30, 2012, we screened 947 patients for eligibility, of whom 423 were randomly assigned to receive dabrafenib and trametinib (n=211) or dabrafenib only (n=212). The final data cutoff was Jan 12, 2015, at which time 222 patients had died. Median overall survival was 25·1 months (95% CI 19·2-not reached) in the dabrafenib and trametinib group versus 18·7 months (15·2-23·7) in the dabrafenib only group (hazard ratio [HR] 0·71, 95% CI 0·55-0·92; p=0·0107). Overall survival was 74% at 1 year and 51% at 2 years in the dabrafenib and trametinib group versus 68% and 42%, respectively, in the dabrafenib only group. Based on 301 events, median progression-free survival was 11·0 months (95% CI 8·0-13·9) in the dabrafenib and trametinib group and 8·8 months (5·9-9·3) in the dabrafenib only group (HR 0·67, 95% CI 0·53-0·84; p=0·0004; unadjusted for multiple testing). Treatment-related adverse events occurred in 181 (87%) of 209 patients in the dabrafenib and trametinib group and 189 (90%) of 211 patients in the dabrafenib only group; the most common was pyrexia (108 patients, 52%) in the dabrafenib and trametinib group, and hyperkeratosis (70 patients, 33%) in the dabrafenib only group. Grade 3 or 4 adverse events occurred in 67 (32%) patients in the dabrafenib and trametinib group and 66 (31%) patients in the dabrafenib only group. INTERPRETATION The improvement in overall survival establishes the combination of dabrafenib and trametinib as the standard targeted treatment for BRAF Val600 mutation-positive melanoma. Studies assessing dabrafenib and trametinib in combination with immunotherapies are ongoing. FUNDING GlaxoSmithKline.
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Mater Hospital, Sydney, NSW, Australia.
| | | | - Helen Gogas
- Department of Medicine, University of Athens, Medical School, Athens, Greece
| | | | | | | | - Claus Garbe
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Thomas Jouary
- Department of Dermatology, Hôpital Saint André, CHU Bordeaux, Bordeaux, France
| | | | - Jean-Jacques Grob
- Aix-Marseille University and APHM Hospital CHU Timone, Marseille, France
| | - Vanna Chiarion-Sileni
- Melanoma and Oesophageal Oncology Unit, Veneto Oncology Institute-IRCCS, Padua, Italy
| | - Celeste Lebbe
- APHP Dermatology CIC Hôpital Saint Louis, University Paris Diderot, INSERM U976, Paris, France
| | - Mario Mandalà
- Papa Giovanni XXIII Cancer Center Hospital, Bergamo, Italy
| | - Michael Millward
- Sir Charles Gairdner Hospital, Hospital Avenue, Perth, WA, Australia
| | - Ana Arance
- Department of Medical Oncology, Hospital Clinic and Translational Genomics and Targeted Therapeutics in Solid Tumors, Barcelona, Spain
| | - Igor Bondarenko
- Dnepropetrovsk State Medical Academy, Dnepropetrovsk, Ukraine
| | | | - Johan Hansson
- Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jochen Utikal
- University Medical Center Mannheim, Heidelberg University, Mannheim, Germany; German Cancer Research Center, Heidelberg, Germany
| | | | | | | | - Volodymr Probachai
- Dnipropetrovsk Clinical Oncology Center of Dnipropetrovsk State Council, Dnipropetrovsk, Ukraine
| | | | | | - Caroline Robert
- Gustave Roussy, Villejuif-Paris-Sud, France; Paris Sud University, Le Kremlin Bicêtre, France
| | - Antoni Ribas
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | | | | | - Suzanne Swann
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | - Fan Jin
- Merck & Co, Kenilworth, NJ, USA
| | | | - Keith Flaherty
- Massachusetts General Hospital Cancer Center, Boston MA, USA
| |
Collapse
|
1892
|
Yélamos O, Gerami P. Predicting the outcome of melanoma: can we tell the future of a patient's melanoma? Melanoma Manag 2015; 2:217-224. [PMID: 30190851 PMCID: PMC6094684 DOI: 10.2217/mmt.15.15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cutaneous melanoma is responsible for the greatest number of skin cancer related deaths. For many years there were few therapeutic options. However, in the last years a number of new therapeutic options have emerged showing improved survival rates for advanced melanoma patients. A significant question based on these findings is whether identification and treatment of patients with biologically aggressive melanomas at an earlier clinical stage offer an opportunity for even greater improvement in overall survival. In this review, we will discuss the recent advancements in molecular strategies beyond traditional staging to identify biologically aggressive melanomas, and which are their implications in terms of predicting the prognosis of patients with melanoma.
Collapse
Affiliation(s)
- Oriol Yélamos
- Department of Dermatology, Feinberg School of Medicine, The Robert H Lurie Cancer Center, Northwestern University, 676 N. St Clair Street, Suite 1765, Chicago, IL 60611, USA
| | - Pedram Gerami
- Department of Dermatology, Feinberg School of Medicine, The Robert H Lurie Cancer Center, Northwestern University, 676 N. St Clair Street, Suite 1765, Chicago, IL 60611, USA
| |
Collapse
|
1893
|
Goyal S, Silk AW, Tian S, Mehnert J, Danish S, Ranjan S, Kaufman HL. Clinical Management of Multiple Melanoma Brain Metastases: A Systematic Review. JAMA Oncol 2015; 1:668-76. [PMID: 26181286 PMCID: PMC5726801 DOI: 10.1001/jamaoncol.2015.1206] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
IMPORTANCE The treatment of multiple brain metastases (MBM) from melanoma is controversial and includes surgical resection, stereotactic radiosurgery (SRS), and whole-brain radiation therapy (WBRT). Several new classes of agents have revolutionized the treatment of metastatic melanoma, allowing some subsets of patients to have long-term survival. Given this, management of MBM from melanoma is continually evolving. OBJECTIVE To review the current evidence regarding the treatment of MBM from melanoma. EVIDENCE REVIEW The PubMed database was searched using combinations of search terms and synonyms for melanoma, brain metastases, radiation, chemotherapy, immunotherapy, and targeted therapy published between January 1, 1995, and January 1, 2015. Articles were selected for inclusion on the basis of targeted keyword searches, manual review of bibliographies, and whether the article was a clinical trial, large observational study, or retrospective study focusing on melanoma brain metastases. Of 2243 articles initially identified, 110 were selected for full review. Of these, the most pertinent 73 articles were included. FINDINGS Patients with newly diagnosed MBM can be treated with various modalities, either alone or in combination. Level 1 evidence supports the use of SRS alone, WBRT, and SRS with WBRT. Although the addition of WBRT to SRS improves the overall brain relapse rate, WBRT has no significant impact on overall survival and has detrimental neurocognitive outcomes. Cytotoxic chemotherapy has largely been ineffective; targeted therapies and immunotherapies have been reported to have high response rates and deserve further attention in larger clinical trials. Further studies are needed to fully evaluate the efficacy of these novel regimens in combination with radiation therapy. CONCLUSIONS AND RELEVANCE At this time, the standard management for patients with MBM from melanoma includes SRS, WBRT, or a combination of both. Emerging data exist to support the notion that SRS in combination with targeted therapies or immune therapy may obviate the need for WBRT; prospective studies are required to fully evaluate the efficacy of these novel regimens in combination with radiation therapy.
Collapse
Affiliation(s)
- Sharad Goyal
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Ann W. Silk
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Sibo Tian
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Janice Mehnert
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Shabbar Danish
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Sinthu Ranjan
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Howard L. Kaufman
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| |
Collapse
|
1894
|
Affiliation(s)
- Keiran S M Smalley
- Department of Tumor Biology, The Moffitt Cancer Center, Tampa, FL 33612, USA; Department of Cutaneous Oncology, The Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Vernon K Sondak
- Department of Cutaneous Oncology, The Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
1895
|
McAleer MF, Kim DW, Trinh VA, Hwu WJ. Management of melanoma brain metastases. Melanoma Manag 2015; 2:225-239. [PMID: 30190852 PMCID: PMC6094653 DOI: 10.2217/mmt.15.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Relapses in the brain remain a major obstacle to cure in many patients with advanced melanoma. At present, the management of melanoma brain metastases continues to rely heavily on surgical and radiotherapeutic interventions, which have become safer and more effective with modern imaging, surgery and radiation technologies. Additionally, novel targeted and immunotherapeutic agents, shown to generate meaningful intracranial response and survival benefit in patients with melanoma brain metastases when compared with historical controls, expand systemic treatment options for this subset of patients. These systemic therapies become particularly important when intracranial disease burden precludes neuro- or radio-surgery. Considerable multidisciplinary research effort is ongoing to improve outcomes for melanoma patients with brain metastases, a key challenge in the management of advanced melanoma.
Collapse
Affiliation(s)
- Mary Frances McAleer
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Dae W Kim
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Van A Trinh
- Clinical Pharmacy Specialist, Pharmacy Clinical Programs, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
1896
|
Abstract
PURPOSE AND DESIGN In recent years, there have been dramatic improvements in the diagnosis and treatment of patients with melanoma. The development of molecular markers and associated targeted therapies have given new hope to subsets of patients with advanced disease. Here we discuss the most important advances in molecular targeted therapy and how these developments are likely to affect the practice of the clinical surgeon. RESULTS AND CONCLUSIONS Germ-line and somatic mutations are common in melanoma and provide prognostic information that can now be harnessed to provide a more personalized approach to cancer treatment. BRAF mutation at the V600 position is the most commonly identified mutation in patients with melanoma. Treatment with targeted inhibitors in patients with BRAF-mutant melanoma has afforded dramatic responses in about half of selected patients. Unfortunately, disease control is not durable and recurrences are common. We predict an increasing role for the surgeon in the multidisciplinary treatment of patients with metastatic disease, as well as a role for molecular profiling in patients with high-risk early stage disease. Further, we are only beginning to understand the prognostic significance of various gene mutations in patients with melanoma.
Collapse
Affiliation(s)
- Danielle K DePeralta
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Genevieve M Boland
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
1897
|
Clinicopathologic features associated with efficacy and long-term survival in metastatic melanoma patients treated with BRAF or combined BRAF and MEK inhibitors. Cancer 2015. [DOI: 10.1002/cncr.29586] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
1898
|
Personalised medicine: Development and external validation of a prognostic model for metastatic melanoma patients treated with ipilimumab. Eur J Cancer 2015; 51:2086-94. [PMID: 26227432 DOI: 10.1016/j.ejca.2015.06.130] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/31/2015] [Accepted: 06/27/2015] [Indexed: 02/05/2023]
Abstract
PURPOSE The purpose of this study was to set up a prognostic model for the identification of survival predictors specific for melanoma patients treated with ipilimumab. EXPERIMENTAL DESIGN The following prospectively collected data were utilised: patient and primary tumour characteristics, relapse-free-interval, site and number of metastases, previous therapies and level of serum biomarkers (lactic dehydrogenase (LDH), C-reactive protein, β2-microglobulin, vascular endothelial growth factor (VEGF), IL2, IL6, S-100, alkaline phosphatase (ALP), transaminases, leucocyte count, lymphocytes subpopulations). A multivariate prognostic model was developed using the Cox regression model fitted to the data of 113 consecutive metastatic patients treated with ipilimumab (3 mg/kg, q3w) at Veneto Institute of Oncology (IOV). External validation was obtained using the data of 69 and 34 patients treated at European Oncology Institute (IEO) and University of Torino (UT), respectively. RESULTS Median survival was 8.3, 4.9 and 7.1 months from first ipilimumab administration at IOV, IEO and UT, respectively. Both higher baseline levels of LDH (Hazard Ratio [HR] v=1.36, 95% Confidence Interval [CI] 1.16-1.58, P<.001) and neutrophils (HR=1.76, 95% CI 1.41-2.10, P<.001) were associated with worse prognosis. Model performance was satisfactory both upon internal validation (Dxy=0.42) and external validation (Dxy=0.40). Serum LDH and neutrophil count discriminated patients who lived more (low neutrophils and low LDH) or less (high LDH or neutrophils) than 24 months. CONCLUSION Serum LDH and neutrophil count were significant independent prognostic factors. This externally validated prognostic nomogram, could help clinicians to identify the patients who would benefit most from ipilimumab and consequently to improve resource allocation. These easily available biomarkers deserve further validation.
Collapse
|
1899
|
Marzuka A, Huang L, Theodosakis N, Bosenberg M. Melanoma Treatments: Advances and Mechanisms. J Cell Physiol 2015; 230:2626-33. [DOI: 10.1002/jcp.25019] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 04/14/2015] [Indexed: 02/03/2023]
Affiliation(s)
- Alexander Marzuka
- Department of Dermatology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
- Department of Internal Medicine; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| | - Laura Huang
- Department of Dermatology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
- Department of Internal Medicine; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| | - Nicholas Theodosakis
- Department of Pathology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| | - Marcus Bosenberg
- Department of Dermatology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
- Department of Pathology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| |
Collapse
|
1900
|
Amaravadi RK, Hamilton KE, Ma X, Piao S, Portillo AD, Nathanson KL, Carlino MS, Long GV, Puzanov I, Xu X, Morrissette JJD, Tsai KY, Flaherty KT, Sosman JA, Goodman GR, McArthur GA, Rustgi AK, Metz DC, Schuchter LM, Chapman PB, Sepulveda AR. Multiple Gastrointestinal Polyps in Patients Treated with BRAF Inhibitors. Clin Cancer Res 2015. [PMID: 26202952 DOI: 10.1158/1078-0432.ccr-15-0469] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE BRAF inhibitors (BRAFi) extend survival in BRAF-mutant melanoma but can promote the growth of Ras-mutant neoplasms. This study determined if gastrointestinal polyps found in BRAFi-treated patients harbored Ras mutations. EXPERIMENTAL DESIGN Colonic and gastric polyps were identified and resected from BRAFi-treated melanoma patients. Next-generation sequencing (NGS) was performed on polyps. The ability of BRAFi to promote polyp formation was functionally characterized in Apc Min(+/-) mice. MAPK and β-catenin pathway activity was assessed by immunohistochemistry in mouse and human polyps. RESULTS Fourteen patients treated with BRAFi underwent endoscopy to assess for polyps. Seven out of 7 patients >40 years of age and treated for >2 years were found to have colonic tubular adenomas with 4 out of the 7 patients having 5 or more polyps. One patient presented with bleeding from hyperplastic gastric polyps that recurred 6 months after BRAFi rechallenge. NGS performed on polyps found no mutations in MAPK pathway genes, but found APC mutations in all tubular adenomas. A significant increase in the number of polyps was observed in BRAFi-treated compared with control-treated Apc Min(+/-) mice (20.8 ± 9.2 vs 12.8 ± 0.1; P = 0.016). No polyps were observed in BRAFi-treated wild-type mice. CONCLUSIONS BRAFi may increase the risk of developing hyperplastic gastric polyps and colonic adenomatous polyps. Due to the risk of gastrointestinal bleeding and the possibility of malignant transformation, further studies are needed to determine whether or not endoscopic surveillance should be recommended for patients treated with BRAFi.
Collapse
Affiliation(s)
- Ravi K Amaravadi
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Kathryn E Hamilton
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaohong Ma
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shengfu Piao
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Armando Del Portillo
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Katherine L Nathanson
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Matteo S Carlino
- Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, New South Wales, Australia. Melanoma Institute Australia and The University of Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia and The University of Sydney, New South Wales, Australia
| | - Igor Puzanov
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer J D Morrissette
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kenneth Y Tsai
- University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | | | - Grant R Goodman
- Genentech, Inc., South San Francisco, San Francisco, California
| | - Grant A McArthur
- Peter MacCallum Cancer Centre and University of Melbourne, Australia, Melbourne, Victoria, Australia
| | - Anil K Rustgi
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David C Metz
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lynn M Schuchter
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paul B Chapman
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Antonia R Sepulveda
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| |
Collapse
|