2001
|
Saraceni MM, Khushalani NI, Jarkowski A. Immunotherapy in Melanoma: Recent Advances and Promising New Therapies. J Pharm Pract 2014; 28:193-203. [PMID: 24674910 DOI: 10.1177/0897190014527317] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The incidence and mortality of melanoma are on the rise. Historically, patients diagnosed with metastatic melanoma were faced with a grim prognosis, with survival rates of 15% at 5 years. Prior to 2011, no drug or therapeutic regimen had been shown to improve overall survival (OS) in metastatic melanoma. Chemotherapeutic agents, such as dacarbazine or temozolomide, are often given to patients for palliative purposes; high-dose interleukin 2 and biochemotherapy are immunotherapeutic options that could be offered to patients with a good performance status at specialized centers. Neither has been shown to impact OS, but durable complete responses are seen in a minority of patients. Since 2011, 4 new drugs have been approved by the US Food and Drug Administration for the treatment of metastatic melanoma, all of which improve survival. Three of these agents (vemurafenib, dabrafenib, and trametinib) are targeted therapies, with ipilimumab being the only new immunotherapy. With a focus on immunotherapeutic agents, this review seeks to summarize the treatment options currently available for metastatic melanoma and to examine those on the near horizon.
Collapse
Affiliation(s)
- Megan M Saraceni
- Department of Pharmacy, University of Rochester Medical Center, Rochester, NY, USA
| | | | | |
Collapse
|
2002
|
Tosti G, Cocorocchio E, Pennacchioli E, Ferrucci PF, Testori A, Martinoli C. Heat-shock proteins-based immunotherapy for advanced melanoma in the era of target therapies and immunomodulating agents. Expert Opin Biol Ther 2014; 14:955-67. [PMID: 24670226 DOI: 10.1517/14712598.2014.902928] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Heat-shock proteins (HSPs) are highly conserved, stress-induced proteins that function as chaperones, stabilizing proteins and delivering peptides. Tumor-derived HSP peptide complexes (HSPPCs) induced immunity against several malignancies in preclinical models, exhibiting activity across tumor types. AREAS COVERED HSPPC-based vaccination showed clinical activity in subsets of patients with different malignancies (e.g., gastric, colorectal, pancreatic, ovarian cancer, and glioblastoma). In Phase III clinical trials for advanced melanoma and renal cell carcinoma patients, HSPPC-based vaccine demonstrated an excellent safety profile, thus emerging as a flexible tumor- and patient-specific therapeutic approach. EXPERT OPINION Melanoma, renal clear cell carcinoma, and glioblastoma are among suitable targets for HSP-based treatment as demonstrated by immune responses and clinical activity observed in subsets of patients, mainly those with early stage of disease and limited tumor burden. In order to further improve clinical activity, combinations of HSPPC-based vaccines with mutation-driven therapies, antiangiogenic agents, or immunomodulating monoclonal antibodies should be tested in controlled clinical trials.
Collapse
Affiliation(s)
- Giulio Tosti
- Istituto Europeo di Oncologia, Melanoma and Sarcoma Division , Via Ripamonti 435, 2014i Milano , Italy +39 02 57489459 ; +39 02 94379230 ;
| | | | | | | | | | | |
Collapse
|
2003
|
Johnson GL, Stuhlmiller TJ, Angus SP, Zawistowski JS, Graves LM. Molecular pathways: adaptive kinome reprogramming in response to targeted inhibition of the BRAF-MEK-ERK pathway in cancer. Clin Cancer Res 2014; 20:2516-22. [PMID: 24664307 DOI: 10.1158/1078-0432.ccr-13-1081] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The central role of the BRAF-MEK-ERK pathway in controlling cell fate has made this pathway a primary target for deregulated activation in cancer. BRaf is activated by Ras proteins allowing Ras oncogenes to constitutively activate the pathway. Activating BRaf mutations are also frequent in several cancers, being the most common oncogenic mutation in thyroid carcinoma and melanoma. There are currently two inhibitors, vemurafenib and dabrafenib, approved for treatment of malignant melanoma having activating BRaf mutations. Concurrent administration of BRAF and MAP-ERK kinase (MEK) inhibitor (trametinib) is significantly more active in patients with BRAF-mutant melanoma than either single agent alone, but progression to resistance ultimately occurs by different mechanisms that increase the activation of extracellular signal-regulated kinase (ERK). Such adaptive changes in tumor cell signaling networks allow bypass of targeted oncoprotein inhibition. This is true with targeted inhibitors for BRaf and MEK as well as specific inhibitors for AKT, mTOR, and many receptor tyrosine kinases such as EGF receptor (EGFR) and HER2. It is this adaptive response to targeted kinase inhibitors that contributes to the failure of single-agent kinase inhibitors to have durable responses. This failure is seen in virtually all cancers treated with single-agent kinase inhibitors, most of which are not as dependent on a single signaling pathway such as BRaf-MEK-ERK in melanoma. Thus, understanding the breadth of adaptive reprogramming responses to specific targeted kinase inhibition will be critical to develop appropriate combination therapies for durable clinical responses.
Collapse
Affiliation(s)
- Gary L Johnson
- Authors' Affiliation: Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Timothy J Stuhlmiller
- Authors' Affiliation: Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Steven P Angus
- Authors' Affiliation: Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Jon S Zawistowski
- Authors' Affiliation: Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Lee M Graves
- Authors' Affiliation: Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
2004
|
Lindsay JN, Barras M. Facing the challenges of new melanoma-targeted therapies: Treatment of severe fevers associated with dabrafenib/trametinib combination therapy. J Oncol Pharm Pract 2014; 21:293-5. [DOI: 10.1177/1078155214527859] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
With the emergence of new oral therapies for metastatic melanoma to the market, as well as ongoing pre-marketing trials and special access schemes, it is important to keep up to date with the side effect profiles of these medications. A common side effect associated with the BRAF inhibitor dabrafenib is severe fever symptoms such as pyrexia and rigors/chills; however, treatment options are limited. We report a patient who was debilitated by severe pyrexia and rigors caused by dabrafenib used in combination with trametinib to treat metastatic melanoma, who was treated with low-dose steroids. To our knowledge, the use of prednisolone for the treatment and prevention of further dabrafenib-associated pyrexia is not published; however, it is a low risk and low cost option that was very effective in this case.
Collapse
Affiliation(s)
| | - Michael Barras
- Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
2005
|
Vanneste L, Wolter P, Van den Oord JJ, Stas M, Garmyn M. Cutaneous adverse effects of BRAF inhibitors in metastatic malignant melanoma, a prospective study in 20 patients. J Eur Acad Dermatol Venereol 2014; 29:61-8. [PMID: 24661317 DOI: 10.1111/jdv.12449] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/12/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND BRAF inhibitors frequently cause significant cutaneous adverse reactions. OBJECTIVE To study the timing, prevalence and response to treatment of skin lesions in patients receiving V-raf murine sarcoma viral oncogene homolog B1 (BRAF) inhibitors. METHODS We prospectively studied the cutaneous side-effects of patients with a BRAF mutant (V600E, V600K, V600R) metastatic malignant melanoma treated with a BRAF inhibitor. We systematically registered prevalence, timing of onset and response to treatment. RESULTS Twenty patients were treated for 2-52 weeks with a BRAF inhibitor. Eleven patients on vemurafenib (58%) developed cutaneous side-effects and 10 patients (42%) had more than one cutaneous adverse event. Verrucous papillomas were observed in eight patients (42%), after 1-12 weeks. We diagnosed four keratoacanthomas in two patients (11%) after 6-10 weeks and two squamous cell carcinomas in two patients (11%) after 10-16 weeks. Seven patients (37%) developed a hyperkeratotic, folliculocentric eruption after 2-8 weeks, resolving quickly under topical steroids. Four patients (21%) presented a facial erythema, two patients (11%) a seborrhoeic dermatitis-like eczema on the scalp. Three patients (16%) developed cystic lesions after 2-11 weeks. Three patients (16%) presented a hand-foot skin reaction after 4-6 weeks, which was successfully treated with topical steroids and keratolytics. Hyperkeratosis of the nipples was seen in one patient (5%). We observed phototoxic reactions after UV exposure in five patients (26%) and alopecia in two patients (11%) after 8-10 weeks. One patient on dabrafenib developed curly hairs (24 weeks), keratotic papules (1 and 36 weeks), a keratoacanthoma (4 weeks) and a hand-foot skin reaction (31 weeks). CONCLUSION Multiple cutaneous toxicities were observed in patients under BRAF inhibitors, mostly well controlled with adequate treatment. We recommend a multidisciplinary approach with regular assessments of the skin by a dermatologist. This allows early identification and adequate treatment to avoid premature discontinuation of a life-prolonging therapy.
Collapse
Affiliation(s)
- L Vanneste
- Department of Dermatology, University Hospitals Leuven, Leuven, Belgium
| | | | | | | | | |
Collapse
|
2006
|
Cheng H, Chang Y, Zhang L, Luo J, Tu Z, Lu X, Zhang Q, Lu J, Ren X, Ding K. Identification and Optimization of New Dual Inhibitors of B-Raf and Epidermal Growth Factor Receptor Kinases for Overcoming Resistance against Vemurafenib. J Med Chem 2014; 57:2692-703. [DOI: 10.1021/jm500007h] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Huimin Cheng
- Institute of Chemical
Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Yu Chang
- University of Macau, Avenue Padre
Tomás Pereira Taipa, Macau, China
| | - Lianwen Zhang
- Institute of Chemical
Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Jinfeng Luo
- Institute of Chemical
Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Zhengchao Tu
- Institute of Chemical
Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Xiaoyun Lu
- Institute of Chemical
Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Qingwen Zhang
- University of Macau, Avenue Padre
Tomás Pereira Taipa, Macau, China
| | - Jibu Lu
- Shenyang Pharmaceutical University, #103 Wenhua Road, Shenyang 110016, China
| | - Xiaomei Ren
- Institute of Chemical
Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Ke Ding
- Institute of Chemical
Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Avenue, Guangzhou 510530, China
| |
Collapse
|
2007
|
Holderfield M, Nagel TE, Stuart DD. Mechanism and consequences of RAF kinase activation by small-molecule inhibitors. Br J Cancer 2014; 111:640-5. [PMID: 24642617 PMCID: PMC4134487 DOI: 10.1038/bjc.2014.139] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/18/2014] [Accepted: 02/24/2014] [Indexed: 02/06/2023] Open
Abstract
Despite the clinical success of RAF inhibitors in BRAF-mutated melanomas, attempts to target RAF kinases in the context of RAS-driven or otherwise RAF wild-type tumours have not only been ineffective, but RAF inhibitors appear to aggravate tumorigenesis in these settings. Subsequent preclinical investigation has revealed several regulatory mechanisms, feedback pathways and unexpected enzymatic quirks in the MAPK pathway, which may explain this paradox. In this review, we cover the various proposed molecular mechanisms for the RAF paradox, the clinical consequences and strategies to overcome it.
Collapse
Affiliation(s)
- M Holderfield
- UCSF Helen Diller Family Comprehensive Cancer Research, University of California San Francisco, San Francisco, CA 94143-0128, USA
| | - T E Nagel
- Novartis Institutes for Biomedical Research, Emeryville, CA 94523, USA
| | - D D Stuart
- Novartis Institutes for Biomedical Research, Emeryville, CA 94523, USA
| |
Collapse
|
2008
|
Abstract
The RAS-RAF-MEK-ERK pathway is considered to be the most important signal transduction pathway in melanoma, and alterations in this pathway via various genetic mutations, such as BRAF and NRAS mutations, are known to be important drivers of melanomagenesis. As MEK is an essential intermediary kinase protein within this pathway, inhibition of MEK has been of a great interest as a molecular target therapy in melanoma. In fact, trametinib, a selective MEK inhibitor, has been shown to have a survival benefit over cytotoxic chemotherapy in patients with V600 BRAF-mutant metastatic melanoma, leading to the FDA approval for this patient population. MEK inhibitors may also be useful in treatment of advanced melanoma harboring other genetic mutations, such as NRAS and GNAQ/GNA11 mutations. Here, we review and discuss the preclinical and clinical data regarding MEK inhibitors and their role in the treatment of advanced melanoma.
Collapse
Affiliation(s)
- April K S Salama
- Division of Medical Oncology, Duke University Medical Center, DUMC 3476, Durham, NC, 27710, USA,
| | | |
Collapse
|
2009
|
Abstract
Hairy cell leukemia (HCL), a B cell malignancy comprising 2 % of all leukemias, has become quite exciting recently with regard to the development of new targets for therapy. This review will focus on advancements made within the past 1-2 years in targeted therapy for this disease. These advances may be grouped into two very difference categories, namely targeting of CD22 with the recombinant immunotoxin moxetumomab pasudotox, and targeting of the mutated BRAF component of the MAP kinase pathway. Moxetumomab pasudotox in phase I testing was recently reported to be associated with an overall response rate of 86 % and a complete remission (CR) rate of 46 % in 28 patients with relapsed and refractory HCL. Many of the CRs are without minimal residual disease (MRD). Severe or dose limiting toxicity was not observed on this trial, but a completely reversible and largely asymptomatic form of grade 2 hemolytic uremic syndrome occurred in two patients during retreatment. This agent has commenced phase III multicenter testing to validate its phase I results. An extensive number of studies have documented the V600E mutation in nearly all HCL patients, but not in similar hematologic malignancies. The thymidine kinase inhibitor vemurafenib, which inhibits the V600E mutant of BRAF, was reported to induce a CR in multiply relapsed and refractory HCL, with nearly complete clearing of MRD. One additional partial and one additional complete remission were subsequently reported.
Collapse
Affiliation(s)
- Robert J Kreitman
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
2010
|
Agarwala SS, Eggermont AMM, O'Day S, Zager JS. Metastatic melanoma to the liver: a contemporary and comprehensive review of surgical, systemic, and regional therapeutic options. Cancer 2014; 120:781-9. [PMID: 24301420 DOI: 10.1002/cncr.28480] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 10/15/2013] [Indexed: 12/17/2022]
Abstract
Effective management of hepatic metastases from ocular and cutaneous melanoma remains a major therapeutic challenge. Treatment options include hepatic resection, hepatic intra-arterial (HIA) chemotherapy, chemoembolization, and hepatic perfusions. Evaluating the efficacy of these interventions is limited by the retrospective nature of most of the data, although controlled phase 3 studies are starting to emerge. Studies of hepatic resection are strongly suggestive of a survival benefit following surgery in selected patients. Effective systemic agents for metastatic cutaneous melanoma are available and supported by randomized controlled phase 3 trials. In contrast, no active systemic treatment has yet been identified for metastatic ocular melanoma. HIA and intravenous delivery of fotemustine have been compared in a randomized phase 3 trial in patients with unresectable metastases from melanoma, but no differences between the 2 approaches were observed. Hepatic arterial chemoembolization appears only to be moderately effective according to uncontrolled studies; targeting patients with less liver involvement may improve outcomes. A recent phase 3 study showed a significant improvement in hepatic progression-free survival with percutaneous hepatic perfusion compared with best alternative care in patients with metastatic melanoma; however, the overall survival analysis was confounded by crossover of control patients to active treatment. In conclusion, hepatic resection offers the possibility of long-term survival in carefully selected patients with liver-limited metastases from melanoma. In patients with unresectable cutaneous melanoma, effective systemic therapy is the best treatment option. For patients with unresectable ocular melanoma, regional treatments are likely to assume a greater role until effective systemic treatments are identified.
Collapse
Affiliation(s)
- Sanjiv S Agarwala
- Department of Hematology/Oncology, St. Luke's University Health Network, Bethlehem, Pennsylvania
| | | | | | | |
Collapse
|
2011
|
Abstract
OBJECTIVES To review the diagnosis, staging, and treatments (both standard and novel) for advanced melanoma and discuss the nursing role in the care of patients with melanoma. DATA SOURCES Published research and education articles, on-line journals, recent texts, and references from pertinent articles. CONCLUSION Most melanomas are diagnosed at early localized stages when surgery alone can be curative. For patients diagnosed with metastatic disease, treatment options have been limited and generally considered ineffective. Recent developments in tumor genetics and a greater understanding of the role of the immune system in cancer have translated to better treatments. IMPLICATIONS FOR NURSING PRACTICE Nurses play a key role in ensuring that patients with melanoma understand their diagnosis, treatment recommendations (including supportive care, palliative chemotherapy, immunotherapies), and participation in clinical trials.
Collapse
Affiliation(s)
- Krista M Rubin
- Center for Melanoma, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
2012
|
Tomei S, Wang E, Delogu LG, Marincola FM, Bedognetti D. Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma. Expert Opin Biol Ther 2014; 14:663-86. [DOI: 10.1517/14712598.2014.890586] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
2013
|
Zimmer L, Haydu LE, Menzies AM, Scolyer RA, Kefford RF, Thompson JF, Schadendorf D, Long GV. Incidence of New Primary Melanomas After Diagnosis of Stage III and IV Melanoma. J Clin Oncol 2014; 32:816-23. [DOI: 10.1200/jco.2013.49.5572] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Purpose New primary melanomas (NPMs) have developed in some patients with metastatic melanoma treated with BRAF inhibitors. We sought to determine the background incidence of spontaneous NPMs after a diagnosis of American Joint Committee on Cancer/International Union Against Cancer stage III or IV melanoma in patients not treated with a BRAF inhibitor. Patients and Methods Patients diagnosed with stage III or IV melanoma at Melanoma Institute Australia between 1983 and 2008 were analyzed, and those who received a BRAF inhibitor were excluded. Results Two hundred twenty-nine (5%) of 4,215 patients with stage III melanoma and 43 (1%) of 3,563 patients with stage IV melanoma had at least one NPM after diagnosis of stage III or IV disease. The 6-month, 1-year, and 10-year cumulative incidence rates of developing an NPM after stage III melanoma were 1.2% (95% CI, 0.86% to 1.51%), 1.8% (95% CI, 1.44% to 2.26%), and 5.9% (95% CI, 5.08% to 6.74%), respectively. The 3-month, 6-month, and 1-year cumulative incidence rates of NPM after diagnosis of stage IV melanoma were 0.2% (95% CI, 0.07% to 0.36%), 0.3% (95% CI, 0.15% to 0.51%), and 0.4% (95% CI, 0.25% to 0.7%), respectively. In both patients with stage III and stage IV melanoma, male patients and patients with a prior history of multiple primaries had a higher incidence of NPM. Conclusion Patients with stage III and stage IV melanoma remain at risk for development of further primary melanomas, particularly if they have a history of multiple primary melanomas before stage III or IV disease. The incidence rates are lower than those reported in patients receiving BRAF inhibitors. However, the results must be compared with caution because dermatologic assessment is more frequent in BRAF inhibitor trials.
Collapse
Affiliation(s)
- Lisa Zimmer
- Lisa Zimmer and Dirk Schadendorf, University Hospital, University Duisburg-Essen, Essen, Germany; Lauren E. Haydu, Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, John F. Thompson, and Georgina V. Long, Melanoma Institute Australia; Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, and Georgina V. Long, Sydney Medical School, The University of Sydney; Richard A. Scolyer and John F. Thompson, Royal Prince Alfred Hospital; Lauren E. Haydu and John F. Thompson, The University of
| | - Lauren E. Haydu
- Lisa Zimmer and Dirk Schadendorf, University Hospital, University Duisburg-Essen, Essen, Germany; Lauren E. Haydu, Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, John F. Thompson, and Georgina V. Long, Melanoma Institute Australia; Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, and Georgina V. Long, Sydney Medical School, The University of Sydney; Richard A. Scolyer and John F. Thompson, Royal Prince Alfred Hospital; Lauren E. Haydu and John F. Thompson, The University of
| | - Alexander M. Menzies
- Lisa Zimmer and Dirk Schadendorf, University Hospital, University Duisburg-Essen, Essen, Germany; Lauren E. Haydu, Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, John F. Thompson, and Georgina V. Long, Melanoma Institute Australia; Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, and Georgina V. Long, Sydney Medical School, The University of Sydney; Richard A. Scolyer and John F. Thompson, Royal Prince Alfred Hospital; Lauren E. Haydu and John F. Thompson, The University of
| | - Richard A. Scolyer
- Lisa Zimmer and Dirk Schadendorf, University Hospital, University Duisburg-Essen, Essen, Germany; Lauren E. Haydu, Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, John F. Thompson, and Georgina V. Long, Melanoma Institute Australia; Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, and Georgina V. Long, Sydney Medical School, The University of Sydney; Richard A. Scolyer and John F. Thompson, Royal Prince Alfred Hospital; Lauren E. Haydu and John F. Thompson, The University of
| | - Richard F. Kefford
- Lisa Zimmer and Dirk Schadendorf, University Hospital, University Duisburg-Essen, Essen, Germany; Lauren E. Haydu, Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, John F. Thompson, and Georgina V. Long, Melanoma Institute Australia; Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, and Georgina V. Long, Sydney Medical School, The University of Sydney; Richard A. Scolyer and John F. Thompson, Royal Prince Alfred Hospital; Lauren E. Haydu and John F. Thompson, The University of
| | - John F. Thompson
- Lisa Zimmer and Dirk Schadendorf, University Hospital, University Duisburg-Essen, Essen, Germany; Lauren E. Haydu, Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, John F. Thompson, and Georgina V. Long, Melanoma Institute Australia; Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, and Georgina V. Long, Sydney Medical School, The University of Sydney; Richard A. Scolyer and John F. Thompson, Royal Prince Alfred Hospital; Lauren E. Haydu and John F. Thompson, The University of
| | - Dirk Schadendorf
- Lisa Zimmer and Dirk Schadendorf, University Hospital, University Duisburg-Essen, Essen, Germany; Lauren E. Haydu, Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, John F. Thompson, and Georgina V. Long, Melanoma Institute Australia; Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, and Georgina V. Long, Sydney Medical School, The University of Sydney; Richard A. Scolyer and John F. Thompson, Royal Prince Alfred Hospital; Lauren E. Haydu and John F. Thompson, The University of
| | - Georgina V. Long
- Lisa Zimmer and Dirk Schadendorf, University Hospital, University Duisburg-Essen, Essen, Germany; Lauren E. Haydu, Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, John F. Thompson, and Georgina V. Long, Melanoma Institute Australia; Alexander M. Menzies, Richard A. Scolyer, Richard F. Kefford, and Georgina V. Long, Sydney Medical School, The University of Sydney; Richard A. Scolyer and John F. Thompson, Royal Prince Alfred Hospital; Lauren E. Haydu and John F. Thompson, The University of
| |
Collapse
|
2014
|
Haq R, Fisher DE, Widlund HR. Molecular pathways: BRAF induces bioenergetic adaptation by attenuating oxidative phosphorylation. Clin Cancer Res 2014; 20:2257-63. [PMID: 24610826 DOI: 10.1158/1078-0432.ccr-13-0898] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancers acquire mutations in cooperating pathways that sustain their growth and survival. To support continued proliferation, tumor cells adapt their metabolism to balance energy production with their augmented biosynthetic needs. Although most normal differentiated cells use mitochondrial oxidative phosphorylation (OXPHOS) as the bioenergetic source, cancer cells have been proposed to rely principally on cytoplasmic glycolysis. The molecular basis for this shift, termed the Warburg effect, is the subject of intense investigation, because mechanistic understanding may lead to novel approaches to target the altered metabolism of cancer cells. Recently, mutations BRAF(V600E) have emerged as a major regulator of metabolic homeostasis. Melanoma cells may use a metabolic shift to circumvent BRAF(V600E)-induced senescence though limiting their reliance on OXPHOS and promote proliferation. Furthermore, BRAF(V600E) acts to suppress expression of the melanocyte master regulator microphthalmia-associated transcription factor (MITF) and the mitochondrial biogenesis coactivator PGC1α. Accordingly, therapeutic inhibition of BRAF(V600E) reverses metabolic reprogramming in melanoma cells and elevates OXPHOS through increased MITF-PGC1α levels. BRAF-targeted drugs modulate the metabolic state of malignant melanoma cells, and counteracting these adaptive responses using pharmacologic agents may prove useful in combinatorial therapeutic strategies. Clin Cancer Res; 20(9); 2257-63. ©2014 AACR.
Collapse
Affiliation(s)
- Rizwan Haq
- Authors' Affiliations: Massachusetts General Hospital, Department of Dermatology, Cutaneous Biology Research Center, Harvard Medical School, Charlestown; Center for Melanoma, Massachusetts General Hospital Cancer Center; and Brigham and Women's Hospital, Department of Dermatology, Harvard Skin Diseases Research Center, Harvard Medical School, Boston, Massachusetts
| | | | | |
Collapse
|
2015
|
Meador CB, Micheel CM, Levy MA, Lovly CM, Horn L, Warner JL, Johnson DB, Zhao Z, Anderson IA, Sosman JA, Vnencak-Jones CL, Dahlman KB, Pao W. Beyond histology: translating tumor genotypes into clinically effective targeted therapies. Clin Cancer Res 2014; 20:2264-75. [PMID: 24599935 DOI: 10.1158/1078-0432.ccr-13-1591] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Increased understanding of intertumoral heterogeneity at the genomic level has led to significant advancements in the treatment of solid tumors. Functional genomic alterations conferring sensitivity to targeted therapies can take many forms, and appropriate methods and tools are needed to detect these alterations. This review provides an update on genetic variability among solid tumors of similar histologic classification, using non-small cell lung cancer and melanoma as examples. We also discuss relevant technological platforms for discovery and diagnosis of clinically actionable variants and highlight the implications of specific genomic alterations for response to targeted therapy.
Collapse
Affiliation(s)
- Catherine B Meador
- Authors' Affiliations: Departments of Cancer Biology, Medicine, Biomedical Informatics, and Pathology, Microbiology, and Immunology; Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2016
|
Affiliation(s)
- Peter Hersey
- Kolling Institute, and Melanoma Institute of Australia, University of Sydney, NSW 2065, Australia.
| |
Collapse
|
2017
|
Thumar J, Shahbazian D, Aziz SA, Jilaveanu LB, Kluger HM. MEK targeting in N-RAS mutated metastatic melanoma. Mol Cancer 2014; 13:45. [PMID: 24588908 PMCID: PMC3945937 DOI: 10.1186/1476-4598-13-45] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 02/25/2014] [Indexed: 12/22/2022] Open
Abstract
Background Gain of function mutations in B-RAF and N-RAS occur frequently in melanoma, leading to mitogen activating protein kinase (MAPK) pathway activation, and this pathway is the target of drugs in development. Our purpose was to study clinical characteristics of patients with mutations in this pathway and to determine activity of inhibitors of B-RAF and MEK in short term cultures grown from tumors of some of these patients. Methods Clinical and pathologic data were collected retrospectively on melanoma patients tested for B-RAF and N-RAS mutations at the Yale Cancer Center and associations with survival were determined. We studied in vitro activity of the pan-RAF inhibitor, RAF265, and the MEK inhibitor, MEK162, in 22 melanoma short term cultures. We further characterized the effect of MEK inhibition on apoptosis and growth of melanoma cultures. Results In a cohort of 144 metastatic melanoma patients we found that patients with N-RAS mutant melanoma had a worse prognosis. These patients were more likely to have brain metastases at the time of presentation with metastatic disease than their N-RAS-wild-type counterparts. All N-RAS mutant melanoma cultures tested in our study (n = 7) were sensitive to MEK inhibition162. Exposure to MEK162 reduced ERK1/2 phosphorylation, and induced apoptosis. Clonogenic survival was significantly reduced in sensitive melanoma cell cultures. Conclusions The prognosis of patients with melanoma expressing constitutively active N-RAS is poor, consistent with studies performed at other institutions. N-RAS mutant melanoma cultures appear to be particularly sensitive to MEK162, supporting ongoing clinical trials with MEK162 in N-RAS mutated melanoma.
Collapse
Affiliation(s)
| | | | | | | | - Harriet M Kluger
- Section of Medical Oncology, Yale Cancer Center, Yale School of Medicine, 333 Cedar Street, WWW213, New Haven, CT 06520, USA.
| |
Collapse
|
2018
|
Escuin-Ordinas H, Atefi M, Fu Y, Cass A, Ng C, Huang RR, Yashar S, Comin-Anduix B, Avramis E, Cochran AJ, Marais R, Lo RS, Graeber TG, Herschman HR, Ribas A. COX-2 inhibition prevents the appearance of cutaneous squamous cell carcinomas accelerated by BRAF inhibitors. Mol Oncol 2014; 8:250-60. [PMID: 24345644 PMCID: PMC3943738 DOI: 10.1016/j.molonc.2013.11.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 10/31/2013] [Accepted: 11/12/2013] [Indexed: 10/25/2022] Open
Abstract
Keratoacanthomas (KAs) and cutaneous squamous cell carcinomas (cuSCCs) develop in 15-30% of patients with BRAF(V600E) metastatic melanoma treated with BRAF inhibitors (BRAFi). These lesions resemble mouse skin tumors induced by the two-stage DMBA/TPA skin carcinogenesis protocol; in this protocol BRAFi accelerates tumor induction. Since prior studies demonstrated cyclooxygenase 2 (COX-2) is necessary for DMBA/TPA tumor induction, we hypothesized that COX-2 inhibition might prevent BRAFi-accelerated skin tumors. Celecoxib, a COX-2 inhibitor, significantly delayed tumor acceleration by the BRAFi inhibitor PLX7420 and decreased tumor number by 90%. Tumor gene expression profiling demonstrated that celecoxib partially reversed the PLX4720-induced gene signature. In PDV cuSCC cells, vemurafenib (a clinically approved BRAFi) increased ERK phosphorylation and soft agar colony formation; both responses were greatly decreased by celecoxib. In clinical trials trametinib, a MEK inhibitor (MEKi) increases BRAFi therapy efficacy in BRAF(V600E) melanomas and reduces BRAFi-induced KA and cuSCC frequency. Trametinib also reduced vemurafenib-induced PDV soft agar colonies, but less efficiently than celecoxib. The trametinb/celecoxib combination was more effective than either inhibitor alone. In conclusion, celecoxib suppressed both BRAFi-accelerated skin tumors and soft-agar colonies, warranting its testing as a chemopreventive agent for non-melanoma skin lesions in patients treated with BRAFi alone or in combination with MEKi.
Collapse
Affiliation(s)
- Helena Escuin-Ordinas
- Department of Medicine (Division of Hematology-Oncology), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Mohammad Atefi
- Department of Medicine (Division of Hematology-Oncology), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Yong Fu
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Ashley Cass
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Charles Ng
- Department of Medicine (Division of Hematology-Oncology), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Rong Rong Huang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Sharona Yashar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Begonya Comin-Anduix
- Department of Surgery (Division of Surgical-Oncology), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Earl Avramis
- Department of Surgery (Division of Surgical-Oncology), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Alistair J Cochran
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | - Roger S Lo
- Department of Medicine (Division of Dermatology), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Harvey R Herschman
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
| | - Antoni Ribas
- Department of Medicine (Division of Hematology-Oncology), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Department of Surgery (Division of Surgical-Oncology), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
| |
Collapse
|
2019
|
Abstract
In the past decade, major advances have been made in the understanding of melanoma. New predisposition genes have been reported and key somatic events, such as BRAF mutation, directly translated into therapeutic management. Surgery for localised melanoma and regional lymph node metastases is the standard of care. Sentinel-node biopsy provides precise staging, but has not been reported to affect survival. The effect of lymph-node dissection on survival is a topic of investigation. Two distinct approaches have emerged to try to extend survival in patients with metastatic melanoma: immunomodulation with anti-CTLA4 monoclonal antibodies, and targeted therapy with BRAF inhibitors or MEK inhibitors for BRAF-mutated melanoma. The combination of BRAF inhibitors and MEK inhibitors might improve progression-free survival further and, possibly, increase overall survival. Response patterns differ substantially-anti-CTLA4 immunotherapy can induce long-term responses, but only in a few patients, whereas targeted drugs induce responses in most patients, but nearly all of them relapse because of pre-existing or acquired resistance. Thus, the long-term prognosis of metastatic melanoma remains poor. Anti-PD1 and anti-PDL1 antibodies have emerged as breakthrough drugs for melanoma that have high response rates and long durability. Biomarkers that have predictive value remain elusive in melanoma, although emerging data for adjuvant therapy indicate that interferon sensitivity is associated with ulceration of the primary melanoma. Intense investigation continues for clinical and biological markers that predict clinical benefit of immunotherapeutic drugs, such as interferon alfa or anti-CTLA4 antibodies, and the mechanisms that lead to resistance of targeted drugs.
Collapse
Affiliation(s)
- Alexander M M Eggermont
- Melanoma Unit and INSERM U981, Gustave Roussy Cancer Institute, Grand Paris, Villejuif, France; Erasmus University Medical Centre, Rotterdam, Netherlands.
| | - Alan Spatz
- Department of Pathology, McGill University & Lady Davis Institute for Medical Research, Montreal, QC, Canada
| | - Caroline Robert
- Melanoma Unit and INSERM U981, Gustave Roussy Cancer Institute, Grand Paris, Villejuif, France
| |
Collapse
|
2020
|
Menzies AM, Long GV. Dabrafenib and Trametinib, Alone and in Combination for BRAF-Mutant Metastatic Melanoma. Clin Cancer Res 2014; 20:2035-43. [DOI: 10.1158/1078-0432.ccr-13-2054] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
2021
|
Ackerman A, Klein O, McDermott DF, Wang W, Ibrahim N, Lawrence DP, Gunturi A, Flaherty KT, Hodi FS, Kefford R, Menzies AM, Atkins MB, Long GV, Sullivan RJ. Outcomes of patients with metastatic melanoma treated with immunotherapy prior to or after BRAF inhibitors. Cancer 2014; 120:1695-701. [DOI: 10.1002/cncr.28620] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/17/2013] [Accepted: 10/29/2013] [Indexed: 12/27/2022]
Affiliation(s)
- Allison Ackerman
- Department of Medicine, Division of Oncology; Beth Israel Deaconess Medical Center; Boston Massachusetts
| | | | - David F. McDermott
- Department of Medicine, Division of Oncology; Beth Israel Deaconess Medical Center; Boston Massachusetts
| | - Wei Wang
- Department of Medicine, Division of Sleep Medicine; Brigham and Women's Hospital; Boston Massachusetts
| | - Nageatte Ibrahim
- Department of Medical Oncology, Melanoma Disease Center; Dana-Farber Cancer Institute; Boston Massachusetts
| | - Donald P. Lawrence
- Center for Melanoma; Massachusetts General Hospital Cancer Center; Boston Massachusetts
| | - Anasuya Gunturi
- Department of Medicine, Division of Oncology; Beth Israel Deaconess Medical Center; Boston Massachusetts
| | - Keith T. Flaherty
- Center for Melanoma; Massachusetts General Hospital Cancer Center; Boston Massachusetts
| | - F. Stephen Hodi
- Department of Medical Oncology, Melanoma Disease Center; Dana-Farber Cancer Institute; Boston Massachusetts
| | - Richard Kefford
- Westmead Hospital; Sydney Australia
- Westmead Institute for Cancer Research, Westmead Millennium Institute; Westmead Hospital; Sydney Australia
- Discipline of Medicine; The University of Sydney; Sydney Australia
- Melanoma Institute Australia; North Sydney Australia
| | - Alexander M. Menzies
- Discipline of Medicine; The University of Sydney; Sydney Australia
- Melanoma Institute Australia; North Sydney Australia
| | | | - Georgina V. Long
- Westmead Institute for Cancer Research, Westmead Millennium Institute; Westmead Hospital; Sydney Australia
- Discipline of Medicine; The University of Sydney; Sydney Australia
- Melanoma Institute Australia; North Sydney Australia
| | - Ryan J. Sullivan
- Center for Melanoma; Massachusetts General Hospital Cancer Center; Boston Massachusetts
| |
Collapse
|
2022
|
Ma XH, Piao SF, Dey S, McAfee Q, Karakousis G, Villanueva J, Hart LS, Levi S, Hu J, Zhang G, Lazova R, Klump V, Pawelek JM, Xu X, Xu W, Schuchter LM, Davies MA, Herlyn M, Winkler J, Koumenis C, Amaravadi RK. Targeting ER stress-induced autophagy overcomes BRAF inhibitor resistance in melanoma. J Clin Invest 2014; 124:1406-17. [PMID: 24569374 DOI: 10.1172/jci70454] [Citation(s) in RCA: 368] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 11/22/2013] [Indexed: 01/30/2023] Open
Abstract
Melanomas that result from mutations in the gene encoding BRAF often become resistant to BRAF inhibition (BRAFi), with multiple mechanisms contributing to resistance. While therapy-induced autophagy promotes resistance to a number of therapies, especially those that target PI3K/mTOR signaling, its role as an adaptive resistance mechanism to BRAFi is not well characterized. Using tumor biopsies from BRAF(V600E) melanoma patients treated either with BRAFi or with combined BRAF and MEK inhibition, we found that BRAFi-resistant tumors had increased levels of autophagy compared with baseline. Patients with higher levels of therapy-induced autophagy had drastically lower response rates to BRAFi and a shorter duration of progression-free survival. In BRAF(V600E) melanoma cell lines, BRAFi or BRAF/MEK inhibition induced cytoprotective autophagy, and autophagy inhibition enhanced BRAFi-induced cell death. Shortly after BRAF inhibitor treatment in melanoma cell lines, mutant BRAF bound the ER stress gatekeeper GRP78, which rapidly expanded the ER. Disassociation of GRP78 from the PKR-like ER-kinase (PERK) promoted a PERK-dependent ER stress response that subsequently activated cytoprotective autophagy. Combined BRAF and autophagy inhibition promoted tumor regression in BRAFi-resistant xenografts. These data identify a molecular pathway for drug resistance connecting BRAFi, the ER stress response, and autophagy and provide a rationale for combination approaches targeting this resistance pathway.
Collapse
|
2023
|
Rebecca VW, Massaro RR, Fedorenko IV, Sondak VK, Anderson ARA, Kim E, Amaravadi RK, Maria-Engler SS, Messina JL, Gibney GT, Kudchadkar RR, Smalley KSM. Inhibition of autophagy enhances the effects of the AKT inhibitor MK-2206 when combined with paclitaxel and carboplatin in BRAF wild-type melanoma. Pigment Cell Melanoma Res 2014; 27:465-78. [PMID: 24490764 DOI: 10.1111/pcmr.12227] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 01/24/2014] [Indexed: 12/13/2022]
Abstract
This study investigates the mechanism of action behind the long-term responses (12-16 months) of two BRAF WT melanoma patients to the AKT inhibitor MK-2206 in combination with paclitaxel and carboplatin. Although single agent MK-2206 inhibited phospho-AKT signaling, it did not impact in vitro melanoma growth or survival. The combination of MK-2206 with paclitaxel and carboplatin was cytotoxic in long-term colony formation and 3D spheroid assays, and induced autophagy. Autophagy was initially protective with autophagy inhibitors and deletion of ATG5 found to enhance cytotoxicity. Although prolonged autophagy induction (>6 days) led to caspase-dependent apoptosis, drug resistant clones still emerged. Autophagy inhibition enhanced the cell death response through reactive oxygen species and could be reversed by anti-oxidants. We demonstrate for the first time that AKT inhibition in combination with chemotherapy may have clinical activity in BRAF WT melanoma and show that an autophagy inhibitor may prevent resistance to these drugs.
Collapse
Affiliation(s)
- Vito W Rebecca
- Department of Molecular Oncology, The Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2024
|
Chaminade A, Conte H, Jouary T, Fauconneau A, Darrigade A, Beylot‐Barry M, Pham‐Ledard A. BRAF
inhibitors‐induced panniculitis: a cutaneous side effect mimicking subcutaneous melanoma metastasis. J Eur Acad Dermatol Venereol 2014; 29:392-393. [DOI: 10.1111/jdv.12397] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- A. Chaminade
- Service de Dermatologie Hôpital Haut‐Lévêque CHU de Bordeaux Univ. Bordeaux Bordeaux France
| | - H. Conte
- Service de Dermatologie Hôpital Saint‐André CHU de Bordeaux Univ. Bordeaux Bordeaux France
| | - T. Jouary
- Service de Dermatologie Hôpital Saint‐André CHU de Bordeaux Univ. Bordeaux Bordeaux France
| | - A. Fauconneau
- Service de Dermatologie Hôpital Haut‐Lévêque CHU de Bordeaux Univ. Bordeaux Bordeaux France
| | - A.‐S. Darrigade
- Service de Dermatologie Hôpital Haut‐Lévêque CHU de Bordeaux Univ. Bordeaux Bordeaux France
- Service de Pathologie CHU de Bordeaux Univ Bordeaux Pessac France
| | - M. Beylot‐Barry
- Service de Dermatologie Hôpital Haut‐Lévêque CHU de Bordeaux Univ. Bordeaux Bordeaux France
| | - A. Pham‐Ledard
- Service de Dermatologie Hôpital Haut‐Lévêque CHU de Bordeaux Univ. Bordeaux Bordeaux France
| |
Collapse
|
2025
|
Bello DM, Dematteo RP, Ariyan CE. The GIST of targeted therapy for malignant melanoma. Ann Surg Oncol 2014; 21:2059-67. [PMID: 24531699 DOI: 10.1245/s10434-013-3373-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Indexed: 12/19/2022]
Abstract
The high response rates to the tyrosine kinase inhibitor imatinib in KIT-mutated gastrointestinal stromal tumors (GIST) has led to a paradigm shift in cancer treatment. In a parallel fashion, the field of melanoma is shifting with the utilization of targeted therapy to treat BRAF-mutated melanoma. We reviewed published literature in PubMed on GIST and melanoma, with a focus on both past and current clinical trials. The data presented centers on imatinib, vemurafenib, and most recently dabrafenib, targeting KIT and BRAF mutations and their outcomes in GIST and melanoma. The BRAF(V600E) melanoma mutation, like the KIT exon 11 mutation in GIST, has the highest response to therapy. High response rates with inhibition of KIT in GIST have not been recapitulated in KIT-mutated melanoma. Median time to resistance to targeted agents occurs in ~7 months with BRAF inhibitors and 2 years for imatinib in GIST. In GIST, the development of secondary mutations leads to resistance; however, there have been no similar gatekeeper mutations found in melanoma. Although surgery remains an important component of the treatment of early GIST and melanoma, surgeons will need to continue to define the thresholds and timing for operation in the setting of metastatic disease with improved targeted therapies. Combination treatment strategies may result in more successful clinical outcomes in the management of melanoma in the future.
Collapse
Affiliation(s)
- Danielle M Bello
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | |
Collapse
|
2026
|
Connolly K, Brungs D, Szeto E, Epstein RJ. Anticancer activity of combination targeted therapy using cetuximab plus vemurafenib for refractory BRAF (V600E)-mutant metastatic colorectal carcinoma. ACTA ACUST UNITED AC 2014; 21:e151-4. [PMID: 24523613 DOI: 10.3747/co.21.1661] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Mismatch-repair-deficient colorectal cancers often contain kinase-activating V600E BRAF mutations, but no clinical utility has yet been demonstrated in this setting for monotherapy using oral braf kinase inhibitors such as vemurafenib or dabrafenib. Recent studies have indicated that tumour resistance to braf inhibition is mediated by upregulated epidermal growth factor receptor (egfr) signalling, disruption of which is a routine treatment strategy in KRAS wild-type colorectal cancer. In this report, we describe the clinical course of a heavily pretreated patient who elected to receive off-label dual-targeted braf- and egfr-inhibitory therapy with good tolerance and apparent clinical benefit.
Collapse
Affiliation(s)
- K Connolly
- Department of Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, and UNSW Clinical School, Sydney, Australia
| | - D Brungs
- Department of Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, and UNSW Clinical School, Sydney, Australia
| | - E Szeto
- Department of Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, and UNSW Clinical School, Sydney, Australia
| | - R J Epstein
- Department of Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, and UNSW Clinical School, Sydney, Australia
| |
Collapse
|
2027
|
Combination therapies for the treatment of advanced melanoma: a review of current evidence. Biochem Res Int 2014; 2014:307059. [PMID: 24693430 PMCID: PMC3944792 DOI: 10.1155/2014/307059] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 12/23/2013] [Indexed: 02/07/2023] Open
Abstract
The treatment of advanced melanoma has been revolutionised in recent years with the advent of a range of new therapies. BRAF inhibitors, such as vemurafenib, have demonstrated improvements in the overall survival of patients with advanced melanoma that harbour a BRAF V600 mutation. Alongside these targeted therapies, novel immune-checkpoint inhibitors, such as ipilimumab, have also been developed and have produced similarly improved outcomes for patients. For the first time in the history of melanoma, monotherapy with each of these drugs has produced improvements in the overall survival of patients with advanced disease. Building on this initial success, there has been intense interest in developing combination therapies predominantly with either dual blockade of the MAPK oncogenic pathway or dual immune-checkpoint blockade. The current evidence for the use of these combination therapies will be presented here.
Collapse
|
2028
|
Basile KJ, Le K, Hartsough EJ, Aplin AE. Inhibition of mutant BRAF splice variant signaling by next-generation, selective RAF inhibitors. Pigment Cell Melanoma Res 2014; 27:479-84. [PMID: 24422853 DOI: 10.1111/pcmr.12218] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 01/07/2014] [Indexed: 01/06/2023]
Abstract
Vemurafenib and dabrafenib block MEK-ERK1/2 signaling and cause tumor regression in the majority of advanced-stage BRAF(V600E) melanoma patients; however, acquired resistance and paradoxical signaling have driven efforts for more potent and selective RAF inhibitors. Next-generation RAF inhibitors, such as PLX7904 (PB04), effectively inhibit RAF signaling in BRAF(V600E) melanoma cells without paradoxical effects in wild-type cells. Furthermore, PLX7904 blocks the growth of vemurafenib-resistant BRAF(V600E) cells that express mutant NRAS. Acquired resistance to vemurafenib and dabrafenib is also frequently driven by expression of mutation BRAF splice variants; thus, we tested the effects of PLX7904 and its clinical analog, PLX8394 (PB03), in BRAF(V600E) splice variant-mediated vemurafenib-resistant cells. We show that paradox-breaker RAF inhibitors potently block MEK-ERK1/2 signaling, G1/S cell cycle events, survival and growth of vemurafenib/PLX4720-resistant cells harboring distinct BRAF(V600E) splice variants. These data support the further investigation of paradox-breaker RAF inhibitors as a second-line treatment option for patients failing on vemurafenib or dabrafenib.
Collapse
Affiliation(s)
- Kevin J Basile
- Department of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
2029
|
Shih KC, Shastry M, Williams JT, Jelsma PF, Abram SR, Ayyanar K, Burris HA, Infante JR. Successful treatment with dabrafenib (GSK2118436) in a patient with ganglioglioma. J Clin Oncol 2014; 32:e98-e100. [PMID: 24516030 DOI: 10.1200/jco.2013.48.6852] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Kent C Shih
- Sarah Cannon Research Institute; Tennessee Oncology; and St Thomas Hospital, Nashville, TN
| | | | | | | | | | | | - Howard A Burris
- Sarah Cannon Research Institute; and Tennessee Oncology, Nashville, TN
| | - Jeffrey R Infante
- Sarah Cannon Research Institute; and Tennessee Oncology, Nashville, TN
| |
Collapse
|
2030
|
Jarkowski A, Khushalani NI. BRAF and beyond: Tailoring strategies for the individual melanoma patient. J Carcinog 2014; 13:1. [PMID: 24737949 PMCID: PMC3986540 DOI: 10.4103/1477-3163.126759] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/07/2013] [Indexed: 12/20/2022] Open
Abstract
Until recently, options for therapy in metastatic melanoma were limited. The understanding of immune check-point blockade and the discovery of molecular pathways involving driver mutations like BRAF has transformed the therapeutic landscape in this disease. Ipilimumab was the first drug shown to improve survival while vemurafenib demonstrated rapid responses never seen before in melanoma. Drugs from these classes and others are now in advanced stages of development and primed to positively impact patient survival in an incremental fashion. In this review, we highlight some of the developments during this renaissance in melanoma therapy and discuss agents of promise. Clinical challenges we face include individualizing therapy for patients, overcoming resistance to molecularly targeted therapy and developing rationale combinations or sequences of drugs. A concerted bench and bedside effort in this direction will undoubtedly keep melanoma in the forefront in an era of personalized medicine.
Collapse
Affiliation(s)
- Anthony Jarkowski
- Department of Pharmacy, James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, USA
| | | |
Collapse
|
2031
|
Mapping the molecular determinants of BRAF oncogene dependence in human lung cancer. Proc Natl Acad Sci U S A 2014; 111:E748-57. [PMID: 24550319 DOI: 10.1073/pnas.1320956111] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Oncogenic mutations in the BRAF kinase occur in 6-8% of nonsmall cell lung cancers (NSCLCs), accounting for more than 90,000 deaths annually worldwide. The biological and clinical relevance of these BRAF mutations in NSCLC is incompletely understood. Here we demonstrate that human NSCLC cells with BRAF(V600E), but not other BRAF mutations, initially are sensitive to BRAF-inhibitor treatment. However, these BRAF(V600E) NSCLC cells rapidly acquire resistance to BRAF inhibition through at least one of two discrete molecular mechanisms: (i) loss of full-length BRAF(V600E) coupled with expression of an aberrant form of BRAF(V600E) that retains RAF pathway dependence or (ii) constitutive autocrine EGF receptor (EGFR) signaling driven by c-Jun-mediated EGFR ligand expression. BRAF(V600E) cells with EGFR-driven resistance are characterized by hyperphosphorylated protein kinase AKT, a biomarker we validated in BRAF inhibitor-resistant NSCLC clinical specimens. These data reveal the multifaceted molecular mechanisms by which NSCLCs establish and regulate BRAF oncogene dependence, provide insights into BRAF-EGFR signaling crosstalk, and uncover mechanism-based strategies to optimize clinical responses to BRAF oncogene inhibition.
Collapse
|
2032
|
BRAF--a new player in hematological neoplasms. Blood Cells Mol Dis 2014; 53:77-83. [PMID: 24495477 DOI: 10.1016/j.bcmd.2014.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 12/31/2013] [Indexed: 01/07/2023]
Abstract
BRAF oncogenic kinase has become a target for specific therapy in oncology. Genetic characterization of a predominant V600E mutation in melanoma, thyroid cancer, and other tumors became a focus for developing specific inhibitors, such as vemurafenib or dabrafenib. Our knowledge regarding the role of mutated BRAF in hematological malignancies has grown quickly as a result of new genetic techniques such as next-generation sequencing. This review summarizes current knowledge regarding the role of BRAF in lymphoid and myeloid neoplasms, with a focus on hairy-cell leukemia, Langerhans cell histiocytosis, and Erdheim-Chester disease.
Collapse
|
2033
|
Le devenir des thérapeutiques ciblant la voie RAS/RAF/ MEK/ERK en cancérologie : l’exemple des mélanomes. BULLETIN DE L'ACADÉMIE NATIONALE DE MÉDECINE 2014. [DOI: 10.1016/s0001-4079(19)31345-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
2034
|
Bucheit AD, Davies MA. Emerging insights into resistance to BRAF inhibitors in melanoma. Biochem Pharmacol 2014; 87:381-9. [PMID: 24291778 DOI: 10.1016/j.bcp.2013.11.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 11/16/2013] [Accepted: 11/18/2013] [Indexed: 02/03/2023]
Abstract
Melanoma is the most aggressive form of skin cancer. The treatment of patients with advanced melanoma is rapidly evolving due to an improved understanding of molecular drivers of this disease. Somatic mutations in BRAF are the most common genetic alteration found in these tumors. Recently, two different mutant-selective small molecule inhibitors of BRAF, vemurafenib and dabrafenib, have gained regulatory approval based on positive results in randomized phase III trials. While the development of these agents represents a landmark in the treatment of melanoma, the benefit of these agents is limited by the frequent and rapid onset of resistance. The identification of several molecular mechanisms of resistance to BRAF inhibitors is rapidly leading to the clinical testing of combinatorial strategies to improve the clinical benefit of these agents. These mechanisms, and the lessons learned from the initial testing of the BRAF inhibitors, provide multiple insights that may facilitate the development of targeted therapies against other oncogenic mutations in melanoma, as well as in other cancers.
Collapse
Affiliation(s)
- Amanda D Bucheit
- Department of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, United States
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, United States; Department of Systems Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, United States.
| |
Collapse
|
2035
|
Dummer R, Goldinger SM, Turtschi CP, Eggmann NB, Michielin O, Mitchell L, Veronese L, Hilfiker PR, Felderer L, Rinderknecht JD. Vemurafenib in patients with BRAFV600 mutation-positive melanoma with symptomatic brain metastases: Final results of an open-label pilot study. Eur J Cancer 2014; 50:611-21. [DOI: 10.1016/j.ejca.2013.11.002] [Citation(s) in RCA: 216] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 10/22/2013] [Accepted: 11/02/2013] [Indexed: 10/26/2022]
|
2036
|
Cupissol D, Sassolas B, Combemale P, Modiano P, Bedane C, Derrey S, Dygai-Cochet I, Lamant L, Lubrano V, Mirabel X, Mourrégot A, Rougé Bugat ME, Siegrist S, Thariat J, Tiffet O, Truc G, Verdoni L, Mazeau-Woynar V, Planchamp F, Leccia MT. Traitements systémiques de première et de deuxième lignes des patients atteints d’un mélanome cutané métastatique (hors métastase cérébrale) : Recommandations nationales françaises. ONCOLOGIE 2014. [DOI: 10.1007/s10269-013-2360-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
2037
|
Sanmamed MF, Fernández-Landázuri S, Rodríguez C, Lozano MD, Echeveste JI, Pérez Gracia JL, Alegre E, Carranza O, Zubiri L, Martín-Algarra S, González A. Relevance of MIA and S100 serum tumor markers to monitor BRAF inhibitor therapy in metastatic melanoma patients. Clin Chim Acta 2014; 429:168-74. [DOI: 10.1016/j.cca.2013.11.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 11/27/2013] [Accepted: 11/27/2013] [Indexed: 10/25/2022]
|
2038
|
Garnett MJ, McDermott U. The evolving role of cancer cell line-based screens to define the impact of cancer genomes on drug response. Curr Opin Genet Dev 2014; 24:114-9. [PMID: 24607840 PMCID: PMC4003351 DOI: 10.1016/j.gde.2013.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 12/03/2013] [Accepted: 12/03/2013] [Indexed: 02/08/2023]
Abstract
Over the last decade we have witnessed the convergence of two powerful experimental designs toward a common goal of defining the molecular subtypes that underpin the likelihood of a cancer patient responding to treatment in the clinic. The first of these 'experiments' has been the systematic sequencing of large numbers of cancer genomes through the International Cancer Genome Consortium and The Cancer Genome Atlas. This endeavour is beginning to yield a complete catalogue of the cancer genes that are critical for tumourigenesis and amongst which we will find tomorrow's biomarkers and drug targets. The second 'experiment' has been the use of large-scale biological models such as cancer cell lines to correlate mutations in cancer genes with drug sensitivity, such that one could begin to develop rationale clinical trials to begin to test these hypotheses. It is at this intersection of cancer genome sequencing and biological models that there exists the opportunity to completely transform how we stratify cancer patients in the clinic for treatment.
Collapse
Affiliation(s)
- Mathew J Garnett
- Cancer Genome Project, Wellcome Trust Sanger Institute Hinxton, Cambridge, United Kingdom
| | - Ultan McDermott
- Cancer Genome Project, Wellcome Trust Sanger Institute Hinxton, Cambridge, United Kingdom.
| |
Collapse
|
2039
|
Kurppa KJ, Catón J, Morgan PR, Ristimäki A, Ruhin B, Kellokoski J, Elenius K, Heikinheimo K. High frequency of BRAF V600E mutations in ameloblastoma. J Pathol 2014; 232:492-8. [PMID: 24374844 PMCID: PMC4255689 DOI: 10.1002/path.4317] [Citation(s) in RCA: 220] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 12/18/2013] [Accepted: 12/19/2013] [Indexed: 12/18/2022]
Abstract
Ameloblastoma is a benign but locally infiltrative odontogenic neoplasm. Although ameloblastomas rarely metastasise, recurrences together with radical surgery often result in facial deformity and significant morbidity. Development of non-invasive therapies has been precluded by a lack of understanding of the molecular background of ameloblastoma pathogenesis. When addressing the role of ERBB receptors as potential new targets for ameloblastoma, we discovered significant EGFR over-expression in clinical samples using real-time RT-PCR, but observed variable sensitivity of novel primary ameloblastoma cells to EGFR-targeted drugs in vitro. In the quest for mutations downstream of EGFR that could explain this apparent discrepancy, Sanger sequencing revealed an oncogenic BRAF V600E mutation in the cell line resistant to EGFR inhibition. Further analysis of the clinical samples by Sanger sequencing and BRAF V600E-specific immunohistochemistry demonstrated a high frequency of BRAF V600E mutations (15 of 24 samples, 63%). These data provide novel insight into the poorly understood molecular pathogenesis of ameloblastoma and offer a rationale to test drugs targeting EGFR or mutant BRAF as novel therapies for ameloblastoma.
Collapse
Affiliation(s)
- Kari J Kurppa
- Department of Medical Biochemistry and Genetics and MediCity Research Laboratories, University of TurkuFinland
- Turku Doctoral Programme of Molecular MedicineTurku, Finland
| | - Javier Catón
- Division of Clinical and Diagnostic Sciences, KCL Dental Institute, King's College LondonUK
| | - Peter R Morgan
- Division of Clinical and Diagnostic Sciences, KCL Dental Institute, King's College LondonUK
| | - Ari Ristimäki
- Division of Pathology and Genetics, HUSLAB, Helsinki University Central Hospital, and Department of Pathology, Haartman Institute and Genome-Scale Biology, Research Programs Unit, University of HelsinkiFinland
| | - Blandine Ruhin
- Assistance Publique-Hôpitaux de Paris, Maxillofacial and Stomatology Department, Pitié-Salpêtrière Hospital, and Molecular Oral Pathophysiology, INSERM UMRS 872, Cordeliers Biomedical Institute, Paris 7 UniversityFrance
| | - Jari Kellokoski
- Department of Oral and Maxillofacial Surgery, Institute of Dentistry, University of Eastern Finland, and Department of Oral and Maxillofacial Diseases, Kuopio University HospitalFinland
| | - Klaus Elenius
- Department of Medical Biochemistry and Genetics and MediCity Research Laboratories, University of TurkuFinland
- Department of Oncology, Turku University HospitalFinland
- # These authors contributed equally to this study
| | - Kristiina Heikinheimo
- Department of Oral and Maxillofacial Surgery, Institute of Dentistry, University of Turku and Turku University Hospital, Turku, and Department of Oral Diagnostic Sciences, Institute of Dentistry, University of Eastern FinlandKuopio, Finland
- *Correspondence to: K Heikinheimo, Department of Oral and Maxillofacial Surgery, Institute of Dentistry, University of Turku, Lemminkäisenkatu 2, FI-20520 Turku, Finland. E-mail:
| |
Collapse
|
2040
|
Affiliation(s)
- Kevin B Kim
- University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
2041
|
Wilmott JS, Haydu LE, Menzies AM, Lum T, Hyman J, Thompson JF, Hersey P, Kefford RF, Scolyer RA, Long GV. Dynamics of Chemokine, Cytokine, and Growth Factor Serum Levels in BRAF-Mutant Melanoma Patients during BRAF Inhibitor Treatment. THE JOURNAL OF IMMUNOLOGY 2014; 192:2505-13. [DOI: 10.4049/jimmunol.1302616] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
2042
|
Flaherty KT, Hennig M, Lee SJ, Ascierto PA, Dummer R, Eggermont AMM, Hauschild A, Kefford R, Kirkwood JM, Long GV, Lorigan P, Mackensen A, McArthur G, O'Day S, Patel PM, Robert C, Schadendorf D. Surrogate endpoints for overall survival in metastatic melanoma: a meta-analysis of randomised controlled trials. Lancet Oncol 2014; 15:297-304. [PMID: 24485879 DOI: 10.1016/s1470-2045(14)70007-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Recent phase 3 trials have shown an overall survival benefit in metastatic melanoma. We aimed to assess whether progression-free survival (PFS) could be regarded as a reliable surrogate for overall survival through a meta-analysis of randomised trials. METHODS We systematically reviewed randomised trials comparing treatment regimens in metastatic melanoma that included dacarbazine as the control arm, and which reported both PFS and overall survival with a standard hazard ratio (HR). We correlated HRs for overall survival and PFS, weighted by sample size or by precision of the HR estimate, assuming fixed and random effects. We did sensitivity analyses according to presence of crossover, trial size, and dacarbazine dose. FINDINGS After screening 1649 reports and meeting abstracts published before Sept 8, 2013, we identified 12 eligible randomised trials that enrolled 4416 patients with metastatic melanoma. Irrespective of weighting strategy, we noted a strong correlation between the treatment effects for PFS and overall survival, which seemed independent of treatment type. Pearson correlation coefficients were 0·71 (95% CI 0·29-0·90) with a random-effects assumption, 0·85 (0·59-0·95) with a fixed-effects assumption, and 0·89 (0·68-0·97) with sample-size weighting. For nine trials without crossover, the correlation coefficient was 0·96 (0·81-0·99), which decreased to 0·93 (0·74-0·98) when two additional trials with less than 50% crossover were included. Inclusion of mature follow-up data after at least 50% crossover (in vemurafenib and dabrafenib phase 3 trials) weakened the PFS to overall survival correlation (0·55, 0·03-0·84). Inclusion of trials with no or little crossover with the random-effects assumption yielded a conservative statement of the PFS to overall survival correlation of 0·85 (0·51-0·96). INTERPRETATION PFS can be regarded as a robust surrogate for overall survival in dacarbazine-controlled randomised trials of metastatic melanoma; we postulate that this association will hold as treatment standards evolve and are adopted as the control arm in future trials. FUNDING None.
Collapse
Affiliation(s)
- Keith T Flaherty
- Center for Melanoma, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Michael Hennig
- Biostatistics and Epidemiology, GlaxoSmithKline, Munich, Germany
| | - Sandra J Lee
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy-Istituto Nazionale Tumori Fondazione "G Pascale", Napoli, Italy
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | | | - Axel Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, University Hospital Kiel, Kiel, Germany
| | - Richard Kefford
- Westmead Hospital and Melanoma Institute Australia, University of Sydney, NSW, Australia
| | - John M Kirkwood
- Skin Cancer Program University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Georgina V Long
- Melanoma Institute Australia and the University of Sydney, Sydney, NSW, Australia
| | - Paul Lorigan
- Institute of Cancer Sciences, Faculty of Medical & Human Sciences, University of Manchester, Manchester, UK
| | - Andreas Mackensen
- Department of Internal Medicine 5-Hematology/Oncology, University of Erlangen, Erlangen, Germany
| | - Grant McArthur
- Peter MacCallum Cancer Institute, St Andrews Place, East Melbourne, VIC, Australia
| | - Steven O'Day
- Beverly Hills Cancer Center, Beverly Hills, CA, USA
| | - Poulam M Patel
- Academic Unit of Oncology, University of Nottingham, Nottingham, UK
| | - Caroline Robert
- Dermatology and INSERM Unit 981 Gustave Roussy Cancer Campus and Paris-Sud University Grand Paris, Villejuif, France
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany.
| |
Collapse
|
2043
|
Parmenter TJ, Kleinschmidt M, Kinross KM, Bond ST, Li J, Kaadige MR, Rao A, Sheppard KE, Hugo W, Pupo GM, Pearson RB, McGee SL, Long GV, Scolyer RA, Rizos H, Lo RS, Cullinane C, Ayer DE, Ribas A, Johnstone RW, Hicks RJ, McArthur GA. Response of BRAF-mutant melanoma to BRAF inhibition is mediated by a network of transcriptional regulators of glycolysis. Cancer Discov 2014; 4:423-33. [PMID: 24469106 DOI: 10.1158/2159-8290.cd-13-0440] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
UNLABELLED Deregulated glucose metabolism fulfills the energetic and biosynthetic requirements for tumor growth driven by oncogenes. Because inhibition of oncogenic BRAF causes profound reductions in glucose uptake and a strong clinical benefit in BRAF-mutant melanoma, we examined the role of energy metabolism in responses to BRAF inhibition. We observed pronounced and consistent decreases in glycolytic activity in BRAF-mutant melanoma cells. Moreover, we identified a network of BRAF-regulated transcription factors that control glycolysis in melanoma cells. Remarkably, this network of transcription factors, including hypoxia-inducible factor-1α, MYC, and MONDOA (MLXIP), drives glycolysis downstream of BRAF(V600), is critical for responses to BRAF inhibition, and is modulated by BRAF inhibition in clinical melanoma specimens. Furthermore, we show that concurrent inhibition of BRAF and glycolysis induces cell death in BRAF inhibitor (BRAFi)-resistant melanoma cells. Thus, we provide a proof-of-principle for treatment of melanoma with combinations of BRAFis and glycolysis inhibitors. SIGNIFICANCE BRAF is suppress glycolysis and provide strong clinical benefi t in BRAF V600 melanoma. We show that BRAF inhibition suppresses glycolysis via a network of transcription factors that are critical for complete BRAFi responses. Furthermore, we provide evidence for the clinical potential of therapies that combine BRAFis with glycolysis inhibitors.
Collapse
Affiliation(s)
- Tiffany J Parmenter
- 1Molecular Oncology Laboratory, Oncogenic Signaling and Growth Control Program, 2Translational Research Laboratory, Cancer Therapeutics Program, 3Bioinformatics Core Facility, 4The Cancer Signalling Laboratory, Oncogenic Signaling and Growth Control Program, 5Gene Regulation Laboratory, Cancer Therapeutics Program, 6Molecular Imaging and Targeted Therapeutics Laboratory, Cancer Therapeutics Program, 7Department of Cancer Imaging, Peter MacCallum Cancer Centre, East Melbourne; 8Sir Peter MacCallum Department of Oncology, Departments of 9Biochemistry and Molecular Biology, and 10Pathology, University of Melbourne, Parkville; 11Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Waurn Ponds; 12Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria; 13Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead; 14Department of Tissue Pathology & Diagnostic Oncology, Royal Prince Alfred Hospital; 15Discipline of Pathology, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; 16Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah; and 17Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2044
|
Rizos H, Menzies AM, Pupo GM, Carlino MS, Fung C, Hyman J, Haydu LE, Mijatov B, Becker TM, Boyd SC, Howle J, Saw R, Thompson JF, Kefford RF, Scolyer RA, Long GV. BRAF inhibitor resistance mechanisms in metastatic melanoma: spectrum and clinical impact. Clin Cancer Res 2014; 20:1965-77. [PMID: 24463458 DOI: 10.1158/1078-0432.ccr-13-3122] [Citation(s) in RCA: 410] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Multiple BRAF inhibitor resistance mechanisms have been described, however, their relative frequency, clinical correlates, and effect on subsequent therapy have not been assessed in patients with metastatic melanoma. EXPERIMENTAL DESIGN Fifty-nine BRAF(V600)-mutant melanoma metastases from patients treated with dabrafenib or vemurafenib were analyzed. The genetic profile of resistance mechanisms and tumor signaling pathway activity was correlated with clinicopathologic features and therapeutic outcomes. RESULTS Resistance mechanisms were identified in 58% progressing tumors and BRAF alterations were common. Gene expression analysis revealed that mitogen-activated protein kinase (MAPK) activity remained inhibited in 21% of resistant tumors, and the outcomes of patients with these tumors were poor. Resistance mechanisms also occurred in pretreatment biopsies and heterogeneity of resistance mechanisms occurred within patients and within tumors. There were no responses to subsequent targeted therapy, even when a progressing tumor had a resistance mechanism predicted to be responsive. CONCLUSIONS Selecting sequential drugs based on the molecular characteristics of a single progressing biopsy is unlikely to provide improved responses, and first-line therapies targeting multiple pathways will be required.
Collapse
Affiliation(s)
- Helen Rizos
- Authors' Affiliations: Westmead Institute for Cancer Research, The University of Sydney at Westmead Millennium Institute; Departments of Medical Oncology and Surgical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead; Melanoma Institute Australia; Disciplines of Pathology, Medicine, and Surgery, Sydney Medical School, The University of Sydney, Sydney; Departments of Melanoma and Surgical Oncology and Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2045
|
Sloot S, Fedorenko IV, Smalley KSM, Gibney GT. Long-term effects of BRAF inhibitors in melanoma treatment: friend or foe? Expert Opin Pharmacother 2014; 15:589-92. [DOI: 10.1517/14656566.2014.881471] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
2046
|
McArthur GA. Adjuvant Interferon in Melanoma: Is Duration of Therapy Important? J Clin Oncol 2014; 32:171-3. [DOI: 10.1200/jco.2013.53.5179] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Grant A. McArthur
- Peter MacCallum Cancer Centre, East Melbourne; University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
2047
|
Vella LJ, Pasam A, Dimopoulos N, Andrews M, Knights A, Puaux AL, Louahed J, Chen W, Woods K, Cebon JS. MEK Inhibition, Alone or in Combination with BRAF Inhibition, Affects Multiple Functions of Isolated Normal Human Lymphocytes and Dendritic Cells. Cancer Immunol Res 2014; 2:351-60. [DOI: 10.1158/2326-6066.cir-13-0181] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
2048
|
Ouellet D, Gibiansky E, Leonowens C, O'Hagan A, Haney P, Switzky J, Goodman VL. Population pharmacokinetics of dabrafenib, a BRAF inhibitor: Effect of dose, time, covariates, and relationship with its metabolites. J Clin Pharmacol 2014; 54:696-706. [DOI: 10.1002/jcph.263] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 01/04/2014] [Indexed: 11/09/2022]
|
2049
|
Duffy MJ, Crown J. Precision treatment for cancer: Role of prognostic and predictive markers. Crit Rev Clin Lab Sci 2014; 51:30-45. [DOI: 10.3109/10408363.2013.865700] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
2050
|
Carlino MS, Todd JR, Gowrishankar K, Mijatov B, Pupo GM, Fung C, Snoyman S, Hersey P, Long GV, Kefford RF, Rizos H. Differential activity of MEK and ERK inhibitors in BRAF inhibitor resistant melanoma. Mol Oncol 2014; 8:544-54. [PMID: 24476679 DOI: 10.1016/j.molonc.2014.01.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/06/2014] [Accepted: 01/06/2014] [Indexed: 12/17/2022] Open
Abstract
Acquired resistance to BRAF inhibitors often involves MAPK re-activation, yet the MEK inhibitor trametinib showed minimal clinical activity in melanoma patients that had progressed on BRAF-inhibitor therapy. Selective ERK inhibitors have been proposed as alternative salvage therapies. We show that ERK inhibition is more potent than MEK inhibition at suppressing MAPK activity and inhibiting the proliferation of multiple BRAF inhibitor resistant melanoma cell models. Nevertheless, melanoma cells often failed to undergo apoptosis in response to ERK inhibition, because the relief of ERK-dependent negative feedback activated RAS and PI3K signalling. Consequently, the combination of ERK and PI3K/mTOR inhibition was effective at promoting cell death in all resistant melanoma cell models, and was substantially more potent than the MEK/PI3K/mTOR inhibitor combination. Our data indicate that a broader targeting strategy concurrently inhibiting ERK, rather than MEK, and PI3K/mTOR may circumvent BRAF inhibitor resistance, and should be considered during the clinical development of ERK inhibitors.
Collapse
Affiliation(s)
- Matteo S Carlino
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales 2145, Australia; Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, New South Wales, Australia
| | - Jason R Todd
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales 2145, Australia
| | - Kavitha Gowrishankar
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales 2145, Australia
| | - Branka Mijatov
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales 2145, Australia
| | - Gulietta M Pupo
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales 2145, Australia
| | - Carina Fung
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales 2145, Australia
| | - Stephanie Snoyman
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales 2145, Australia
| | - Peter Hersey
- Kolling Institute of Medical Research, University of Sydney, St. Leonards, New South Wales, Australia; Melanoma Institute Australia, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard F Kefford
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales 2145, Australia; Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, New South Wales, Australia; Melanoma Institute Australia, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Helen Rizos
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales 2145, Australia.
| |
Collapse
|