1951
|
Abstract
The treatment and outcomes for people with metastatic melanoma have changed considerably in the past few years with the introduction of targeted anticancer drugs. About half of the patients with metastatic melanoma will have activating mutations in the BRAF gene. These people may benefit from a BRAF inhibitor (vemurafenib or dabrafenib) or a MEK inhibitor (trametinib). Addition of a MEK inhibitor to a BRAF inhibitor improves progression-free survival and alters the adverse effect profile. Ipilimumab is another drug indicated for metastatic melanoma. It works by altering the patient's own immune response to the tumour. Toxicities are common with these drugs and include arthralgias, fatigue, photosensitivity, squamous cell carcinomas, fever, diarrhoea, pruritus and immune-related adverse effects.
Collapse
|
1952
|
Abstract
PURPOSE OF REVIEW Treatment options for metastatic melanoma depend on the clinical course of the disease and the molecular profile such as mutations of the BRAF gene. In this article, we review the current state of targeted therapy with kinase inhibitors. RECENT FINDINGS Despite major advancements in targeted therapy of metastatic melanoma, most patients relapse and show progressive disease after 5-7 months with single inhibition of BRAF or MEK. Acquired resistance is virtually universal and mediated by diverse mitogen-activated protein kinase-dependent or independent mechanisms. Recent evidence favours concurrent targeting of BRAF and MEK in patients with BRAFV600-mutated melanoma instead of BRAF inhibitor monotherapy. The combination delays the onset of acquired resistance, resulting in increased progression-free and overall survival. A growing number of early trials evaluate the efficacy of inhibitors targeting additional pathways such as phospho-inositide 3-kinase/AKT in conjunction with BRAF or MEK. Even though consistent and mature phase III study results are not yet available for these combinations, the repertoire of targeted therapy in metastatic melanoma is wide and promising. SUMMARY The short era of single BRAF inhibition in BRAF-mutated melanoma is soon taken over by dual concurrent inhibition of MEK and BRAF.
Collapse
Affiliation(s)
- Markus V Heppt
- Department of Dermatology and Allergy, Ludwig-Maximilian University, Munich, Germany
| | | | | | | |
Collapse
|
1953
|
Ugurel S, Loquai C, Kähler K, Hassel J, Berking C, Zimmer L, Haubitz I, Satzger I, Müller-Brenne T, Mikhaimer N, Becker J, Kilian K, Schadendorf D, Heinzerling L, Kaatz M, Utikal J, Göppner D, Pföhler C, Pflugfelder A, Mössner R, Gutzmer R. A multicenter DeCOG study on predictors of vemurafenib therapy outcome in melanoma: pretreatment impacts survival. Ann Oncol 2015; 26:573-82. [DOI: 10.1093/annonc/mdu573] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
1954
|
Abstract
ABSTRACT Cancers exhibit differences in metastatic behavior and drug sensitivity that correlate with certain tumor-specific variables such as differentiation grade, growth rate/extent and molecular regulatory aberrations. In practice, patient management is based on the past results of clinical trials adjusted for these biomarkers. Here, it is proposed that treatment strategies could be fine-tuned upfront simply by quantifying tumorigenic spatial (cell growth) and temporal (genetic stability) control losses, as predicted by genetic defects of cell-cycle-regulatory gatekeeper and genome-stabilizing caretaker tumor suppressor genes, respectively. These differential quantifications of tumor dysfunction may in turn be used to create a tumor-specific ‘periodic table’ that guides rational formulation of survival-enhancing anticancer treatment strategies.
Collapse
Affiliation(s)
- Richard J Epstein
- *Clinical Informatics & Research Centre, The Kinghorn Cancer Centre, 370 Victoria St, Darlinghurst 2010, Sydney, Australia
- Laboratory of Genome Evolution, Garvan Institute for Medical Research, 384 Victoria St, Darlinghurst 2010, Sydney, Australia
- Department of Oncology, & UNSW Clinical School, St Vincent's Hospital, 390 Victoria St, Darlinghurst 2010 Sydney, Australia
| |
Collapse
|
1955
|
Abstract
INTRODUCTION Selective inhibition of the MAPK pathway with either BRAF or MEK inhibition has emerged as a key component for the treatment of BRAF-mutant metastatic melanoma. New evidence suggests that the combination of BRAF and MEK inhibitors improves tumor response rate and progression-free survival, while potentially attenuating some of the serious adverse events observed with monotherapy. AREAS COVERED This review covers the current data on the efficacy and safety of the selective BRAF (vemurafenib and dabrafenib) and MEK (trametinib) inhibitors as well as the available data on BRAF inhibitor + MEK inhibitor combination therapy (dabrafenib + trametinib and vemurafenib + cobimetinib). The efficacy, safety and toxicity data are discussed from Phase I, Phase II and Phase III trials of these drugs. EXPERT OPINION Combination therapy with the BRAF and MEK inhibitors improves response rates and progression-free survival in patients with BRAF-mutant metastatic melanoma. Some of the serious adverse events, in particular, the incidence of cutaneous squamous cell carcinoma, are attenuated with combination therapy, whereas milder side effects such as pyrexia can be more common with combination therapy. Although dose reductions and dose interruptions are slightly more common with combination therapy, overall data supports the notion that combination therapy is safe and improves the outcomes for patients compared to single agent BRAF inhibitors.
Collapse
Affiliation(s)
- Lesly A Dossett
- Moffitt Cancer Center, Complex General Surgical Oncology , 12902 Magnolia Drive, Tampa, FL 33612 , USA
| | | | | |
Collapse
|
1956
|
Johnson AS, Crandall H, Dahlman K, Kelley MC. Preliminary results from a prospective trial of preoperative combined BRAF and MEK-targeted therapy in advanced BRAF mutation-positive melanoma. J Am Coll Surg 2015; 220:581-93.e1. [PMID: 25797743 DOI: 10.1016/j.jamcollsurg.2014.12.057] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 12/22/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND We conducted a prospective trial of BRAF and mitogen-activated protein kinase kinase (MEK) targeted therapy in advanced, operable BRAF mutation-positive melanoma to determine feasibility, tumor response rates, and biomarkers of response and resistance. STUDY DESIGN Thirteen patients with locally or regionally advanced BRAF mutation-positive melanoma received dabrafenib 150 mg po bid for 14 days, followed by dabrafenib plus trametinib 2 mg po daily for 14 days before operation. Biopsies and tumor measurements were obtained at baseline and days 14 and 28. Formalin-fixed paraffin embedded specimens were analyzed with hematoxylin and eosin, Ki-67, cleaved caspase-3, CD8, phosphorylated extracellular signal-regulated kinase (ERK), and phosphorylated MEK immunostains. RESULTS Therapy was tolerated well, with toxicity ≥ grade 3 in 2 of 13 (15%) patients. All 12 patients receiving >14 days of therapy had substantial reduction in tumor volume (65% at day 14 and 78% at day 28) and underwent resection. After 14 days of dabrafenib therapy, there was a marked reduction in viable melanoma cells and a CD8 T-cell--rich infiltrate. Proliferation of the residual melanoma cells was reduced and apoptosis was increased. The cells continued to express phosphorylated ERK and phosphorylated MEK consistent with incomplete mitogen-activated protein kinase pathway inhibition. CONCLUSIONS Preoperative targeted therapy of advanced BRAF-mutant melanoma is feasible, well tolerated, induces brisk tumor responses, and facilitates correlative science. A CD8 T-cell-rich infiltrate indicates a potential immune-mediated mechanism of action. Both proliferation and apoptosis were inhibited, but the mitogen-activated protein kinase pathway remained activated, suggesting intrinsic resistance in a subset of tumor cells. Additional investigation of the anti-tumor immune response during targeted therapy and the mechanisms of intrinsic resistance can yield novel therapeutic strategies.
Collapse
Affiliation(s)
- Adam S Johnson
- Department of Pathology, Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Holly Crandall
- Department of Clinical Trials Shared Resource, Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Kimberly Dahlman
- Department of Cancer Biology, Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Mark C Kelley
- Department of Surgery, Division of Surgical Oncology and Endocrine Surgery, Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, TN.
| |
Collapse
|
1957
|
Affiliation(s)
- Mario Sznol
- From the Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (M.S.)
| | | |
Collapse
|
1958
|
Spagnolo F, Ghiorzo P, Orgiano L, Pastorino L, Picasso V, Tornari E, Ottaviano V, Queirolo P. BRAF-mutant melanoma: treatment approaches, resistance mechanisms, and diagnostic strategies. Onco Targets Ther 2015; 8:157-168. [PMID: 25653539 PMCID: PMC4303458 DOI: 10.2147/ott.s39096] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BRAF inhibitors vemurafenib and dabrafenib achieved improved overall survival over chemotherapy and have been approved for the treatment of BRAF-mutated metastatic melanoma. More recently, the combination of BRAF inhibitor dabrafenib with MEK inhibitor trametinib has shown improved progression-free survival, compared to dabrafenib monotherapy, in a Phase II study and has received approval by the US Food and Drug Administration. However, even when treated with the combination, most patients develop mechanisms of acquired resistance, and some of them do not achieve tumor regression at all, because of intrinsic resistance to therapy. Along with the development of BRAF inhibitors, immunotherapy made an important step forward: ipilimumab, an anti-CTLA-4 monoclonal antibody, was approved for the treatment of metastatic melanoma; anti-PD-1 agents achieved promising results in Phase I/II trials, and data from Phase III studies will be ready soon. The availability of such drugs, which are effective regardless of BRAF status, has made the therapeutic approach more complex, as first-line treatment with BRAF inhibitors may not be the best choice for all BRAF-mutated patients. The aim of this paper is to review the systemic therapeutic options available today for patients affected by BRAF V600-mutated metastatic melanoma, as well as to summarize the mechanisms of resistance to BRAF inhibitors and discuss the possible strategies to overcome them. Moreover, since the molecular analysis of tumor specimens is now a pivotal and decisional factor in the treatment strategy of metastatic melanoma patients, the advances in the molecular detection techniques for the BRAF V600 mutation will be reported.
Collapse
Affiliation(s)
- Francesco Spagnolo
- Department of Plastic and Reconstructive Surgery, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa, Genova, Italy
- Genetics of Rare Cancers, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Laura Orgiano
- Department of Medical Oncology, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Lorenza Pastorino
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa, Genova, Italy
- Genetics of Rare Cancers, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Virginia Picasso
- Department of Medical Oncology, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Elena Tornari
- Department of Medical Oncology, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Vincenzo Ottaviano
- Department of Medical Oncology, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Paola Queirolo
- Department of Medical Oncology, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| |
Collapse
|
1959
|
McQuade J, Davies MA. Converting biology into clinical benefit: lessons learned from BRAF inhibitors. Melanoma Manag 2015; 2:241-254. [PMID: 26594316 PMCID: PMC4649930 DOI: 10.2217/mmt.15.18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The identification and pharmacological targeting of activating BRAF mutations in melanoma has led to significant improvements in patient outcomes. This perspective paper illustrates the lessons learned from the study of BRAF mutations and the development of BRAF inhibitors. The relevance of these lessons to the development of future targeted therapies is highlighted.
Collapse
Affiliation(s)
- Jennifer McQuade
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Michael A Davies
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
- *Author for correspondence: Tel.: +1 713 792 3454; Fax: +1 713 563 3454;
| |
Collapse
|
1960
|
Alexander W. Society for melanoma research and american heart association scientific sessions. P & T : A PEER-REVIEWED JOURNAL FOR FORMULARY MANAGEMENT 2015; 40:68-72. [PMID: 25628510 PMCID: PMC4296595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Among the featured topics: oncolytic immunotherapy, BRAF/MEK inhibition, and a programmed death-1 inhibitor at the Society for Melanoma Research; and anticoagulation therapy, an alternative to statins, and endocarditis in the absence of dental antibiotic prophylaxis at the American Heart Association Scientific Sessions.
Collapse
|
1961
|
Carneiro BA, Kaplan JB, Altman JK, Giles FJ, Platanias LC. Targeting mTOR signaling pathways and related negative feedback loops for the treatment of acute myeloid leukemia. Cancer Biol Ther 2015; 16:648-56. [PMID: 25801978 PMCID: PMC4622839 DOI: 10.1080/15384047.2015.1026510] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/01/2015] [Indexed: 12/29/2022] Open
Abstract
An accumulating understanding of the complex pathogenesis of acute myeloid leukemia (AML) continues to lead to promising therapeutic approaches. Among the key aberrant intracellular signaling pathways involved in AML, the phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) axis is of major interest. This axis modulates a wide array of critical cellular functions, including proliferation, metabolism, and survival. Pharmacologic inhibitors of components of this pathway have been developed over the past decade, but none has an established role in the treatment of AML. This review will discuss the preclinical data and clinical results driving ongoing attempts to exploit the PI3K/AKT/mTOR pathway in patients with AML and address issues related to negative feedback loops that account for leukemic cell survival. Targeting the PI3K/AKT/mTOR pathway is of high interest for the treatment of AML, but combination therapies with other targeted agents may be needed to block negative feedback loops in leukemia cells.
Collapse
Affiliation(s)
- Benedito A Carneiro
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Jason B Kaplan
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Jessica K Altman
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Francis J Giles
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Leonidas C Platanias
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
- Division of Hematology-Oncology; Department of Medicine; Jesse Brown VA Medical Center; Chicago, IL, USA
| |
Collapse
|
1962
|
Tarhini A, Corman SL, Rao S, Margolin K, Ji X, Mehta S, Botteman MF. Healthcare Resource Utilization and Associated Costs in Patients with Advanced Melanoma Receiving First-Line Ipilimumab. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/jct.2015.610091] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
1963
|
Sullivan R, LoRusso P, Boerner S, Dummer R. Achievements and challenges of molecular targeted therapy in melanoma. Am Soc Clin Oncol Educ Book 2015:177-186. [PMID: 25993155 DOI: 10.14694/edbook_am.2015.35.177] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The treatment of melanoma has been revolutionized over the past decade with the development of effective molecular and immune targeted therapies. The great majority of patients with melanoma have mutations in oncogenes that predominantly drive signaling through the mitogen activated protein kinase (MAPK) pathway. Analytic tools have been developed that can effectively stratify patients into molecular subsets based on the identification of mutations in oncogenes and/or tumor suppressor genes that drive the MAPK pathway. At the same time, potent and selective inhibitors of mediators of the MAPK pathway such as RAF, MEK, and ERK have become available. The most dramatic example is the development of single-agent inhibitors of BRAF (vemurafenib, dabrafenib, encorafenib) and MEK (trametinib, cobimetinib, binimetinib) for patients with metastatic BRAFV600-mutant melanoma, a subset that represents 40% to 50% of patients with metastatic melanoma. More recently, the elucidation of mechanisms underlying resistance to single-agent BRAF inhibitor therapy led to a second generation of trials that demonstrated the superiority of BRAF inhibitor/MEK inhibitor combinations (dabrafenib/trametinib; vemurafenib/cobimetinib) compared to single-agent BRAF inhibitors. Moving beyond BRAFV600 targeting, a number of other molecular subsets--such as mutations in MEK, NRAS, and non-V600 BRAF and loss of function of the tumor suppressor neurofibromatosis 1 (NF1)--are predicted to respond to MAPK pathway targeting by single-agent pan-RAF, MEK, or ERK inhibitors. As these strategies are being tested in clinical trials, preclinical and early clinical trial data are now emerging about which combinatorial approaches might be best for these patients.
Collapse
Affiliation(s)
- Ryan Sullivan
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Patricia LoRusso
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Scott Boerner
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
1964
|
|
1965
|
Lee RJ, Ul-Ain-Tariq N, Fusi A, Bowyer S, Lorigan P. The role of chemotherapy in the modern management of melanoma. Melanoma Manag 2014; 1:173-184. [PMID: 30190822 PMCID: PMC6094611 DOI: 10.2217/mmt.14.20] [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] [Indexed: 11/21/2022] Open
Abstract
The last 4 years have seen dramatic changes in the treatment of advanced melanoma, largely based on advances in targeted therapy and immunotherapy. This article examines the role of chemotherapy in the modern management of melanoma. We examine the evidence for promising new agents and discuss their position in the sequencing of treatment options for patients with advanced disease. In addition, we discuss the combination of chemotherapy with targeted treatments and immune therapies. Finally, we discuss future areas of research for ensuring that we maximize the potential of all agents available to us and identify new, effective treatments.
Collapse
Affiliation(s)
- Rebecca Jane Lee
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
- The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Noor Ul-Ain-Tariq
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Alberto Fusi
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Samantha Bowyer
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Paul Lorigan
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
- The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| |
Collapse
|
1966
|
Perales Palacios I, García Campos F, Michaus Oquiñena L, Blanco Guzmán S, Lantero Benedito M. [Isolation of Plesiomonas shigelloides in a case of gastroenteritis]. Rev Clin Esp 1984; 15:353-365. [PMID: 6658089 DOI: 10.1038/s41571-018-0002-6] [Citation(s) in RCA: 370] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|