801
|
Abstract
Unprecedented advances in the treatment of melanoma and the large number of investigational therapies entering clinical studies not only represent outstanding achievements, but also create major challenges for clinical research in melanoma. The challenges for accrual and for developing important new data in trials include the relatively low incidence of melanoma compared with other diseases, a shrinking pool of patients for trials because of the high efficacy of standard of care therapy, requirements for larger studies and longer duration of follow-up to detect signals of activity or establish efficacy, and suboptimal predictive biomarkers for the vast number of new combinations and new agents. The cost of new treatments remains a major concern, particularly because current standard of care involves doublets of targeted therapy or immune therapy, and clinically meaningful further increases in efficacy may require development of triplets or larger multidrug combinations. Toxicities of the current doublets, particularly for immune therapy, may limit development of some multidrug regimens or may require novel solutions such as sequencing or alternating schedules. The activity of first-line therapies may push development of new drugs or combinations into the second-line setting or into subgroups with suboptimal response to the first-line doublets as identified by predictive clinical variables or tissue biomarkers.
Collapse
|
802
|
Targeted Therapies in Combination With Immune Therapies for the Treatment of Metastatic Melanoma. Cancer J 2017; 23:59-62. [PMID: 28114256 DOI: 10.1097/ppo.0000000000000245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In recent years, the field of oncology has witnessed many breakthroughs in the treatment of advanced malignancies, particularly in patients with advanced melanoma. Targeted and immune checkpoint therapies have emerged as the primary treatment strategies for these patients. Molecular profiling of melanoma is incorporated into routine practice to identify potential therapeutic targets, and patients are offered either a targeted or immune checkpoint inhibitor therapy approach. Both strategies have limitations where not all patients experience durable responses. Preclinical data have demonstrated the ability of targeted therapy to enhance activity of effector T cells, reduce immunosuppressive cytokine production, and increase tumor cell antigen presentation, which can augment antitumor immunity. In vivo models have shown synergy with improved tumor control when targeted and immune checkpoint agents are combined. Therefore, combination strategies with targeted and immune checkpoint therapy may improve patient outcomes. Early clinical data with anti-programmed cell-death protein 1/programmed cell-death ligand 1 agents in combination with targeted inhibitors appear to have acceptable toxicity rates and the potential for enhanced antitumor activity. This review explores the current status of preclinical and clinical development for these combination approaches in patients with advanced melanoma.
Collapse
|
803
|
Abstract
Oncogene-targeted therapy is a major component of precision oncology, and although patients with metastatic melanoma have experienced improved outcomes with this strategy, there are a number of potential therapeutic targets currently under study that may further increase the drug armamentarium for this patient population. In this review, we discuss the landscape of targeted therapies for patients with advanced melanoma, focusing on oncogene mutation-specific targets. In patients with typical BRAF V600-mutant melanoma, combination BRAF and MEK inhibition has surpassed outcomes compared with monotherapy with BRAF or MEK inhibition alone, and current strategies seek to address inevitable resistance mechanisms. For patients with NRAS-mutant melanoma, MEK inhibitor monotherapy and combined MEK and CDK4/6 inhibition are burgeoning strategies; for patients with KIT-mutant melanoma, tyrosine kinase inhibition is being leveraged, and for NF-1-mutant melanoma, mTOR and MEK inhibition is being actively evaluated. In patients with atypical, non-V600 BRAF-mutant melanoma, MEK inhibitor monotherapy is the potential novel targeted approach on the horizon. For advanced uveal melanoma, novel targets such as IMCgp100 and glembatumumab have shown activity in early studies. We review additional strategies that remain in the preclinical and early clinical pipeline, so there is much hope for the future of targeted agents for distinct molecular cohorts of patients with advanced melanoma.
Collapse
|
804
|
Pike E, Hamidi V, Saeterdal I, Odgaard-Jensen J, Klemp M. Multiple treatment comparison of seven new drugs for patients with advanced malignant melanoma: a systematic review and health economic decision model in a Norwegian setting. BMJ Open 2017; 7:e014880. [PMID: 28827234 PMCID: PMC5724191 DOI: 10.1136/bmjopen-2016-014880] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 05/31/2017] [Accepted: 07/03/2017] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE To assess the relative effectiveness and cost-effectiveness of seven new drugs (cobimetinib, dabrafenib, ipilimumab, nivolumab, pembrolizumab, trametinib and vemurafenib) used for treatment of patients with advanced malignant melanoma in the Norwegian setting. DESIGN A multiple technology assessment. PATIENTS Patients with advanced malignant melanoma aged 18 or older. DATA SOURCES A systematic search for randomised controlled trials in relevant bibliographic databases. METHODS We performed network meta-analyses using both direct and indirect evidence with dacarbazine as a common comparator. We ranked the different treatments in terms of their likelihood of leading to the best results for each endpoint. The cost-utility analysis was based on a probabilistic discrete-time Markov cohort model. The model calculated the costs and quality-adjusted life years (QALYs) with different treatment strategies from a healthcare perspective. Sensitivity analysis was performed by means of Monte Carlo simulation. RESULTS Monotherapies with a programmed cell death 1 (PD-1) immune-checkpoint-inhibitor had a higher probability of good performance for overall survival than monotherapies with ipilimumab or BRAF/MEK inhibitors. The combination treatments had all similar levels of effectiveness to the PD-1 immune-checkpoint-inhibitors.PD-1 immune-checkpoint-inhibitors are more effective and more costly compared with ipilimumab in monotherapy. Nivolumab in combination with ipilimumab had higher costs and the same level of effectiveness as the PD-1 immune-checkpoint-inhibitors in monotherapy.BRAF/MEK inhibitor combinations (dabrafenib and trametinib or vemurafenib and cobimetinib) had both similar effectiveness and cost-effectiveness; however, the combination therapies are more likely to give higher quality adjusted life year gains than BRAF or MEK inhibitor monotherapies, but to a higher cost. CONCLUSIONS None of the drugs investigated can be considered cost-effective at what has normally been considered a reasonable willingness-to-pay (WTP) in Norway. Price reductions (from the official list prices) in the region of 63%-84% would be necessary for these drugs to be cost-effective at a WTP of €55 850 per QALY.
Collapse
Affiliation(s)
- Eva Pike
- Norwegian Institute of Public Health, Oslo, Norway
| | - Vida Hamidi
- Norwegian Institute of Public Health, Oslo, Norway
| | | | | | - Marianne Klemp
- Norwegian Institute of Public Health, Oslo, Norway
- Department of Pharmacology, University of Oslo, Oslo, Norway
| |
Collapse
|
805
|
Reddy BY, Miller DM, Tsao H. Somatic driver mutations in melanoma. Cancer 2017; 123:2104-2117. [PMID: 28543693 DOI: 10.1002/cncr.30593] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/21/2016] [Accepted: 12/26/2016] [Indexed: 12/13/2022]
Abstract
Melanoma has one of the highest somatic mutational burdens among solid malignancies. Although the rapid progress in genomic research has contributed immensely to our understanding of the pathogenesis of melanoma, the clinical significance of the vast array of genomic alterations discovered by next-generation sequencing is far from being fully characterized. Most mutations prevalent in melanoma are simply neutral "passengers," which accompany functionally significant "drivers" under transforming conditions. The delineation of driver mutations from passenger mutations is critical to the development of targeted therapies. Novel advances in genomic data analysis have aided in distinguishing true driver mutations involved in tumor progression. Here, the authors review the current literature on important somatic driver mutations in melanoma, along with the implications for treatment. Cancer 2017;123:2104-17. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Bobby Y Reddy
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - David M Miller
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Hensin Tsao
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
806
|
Szczepaniak Sloane RA, Gopalakrishnan V, Reddy SM, Zhang X, Reuben A, Wargo JA. Interaction of molecular alterations with immune response in melanoma. Cancer 2017; 123:2130-2142. [PMID: 28543700 DOI: 10.1002/cncr.30681] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/10/2017] [Accepted: 02/21/2017] [Indexed: 01/01/2023]
Abstract
Major advances have been made in melanoma treatment with the use of molecularly targeted therapies and immunotherapies, and numerous regimens are now approved by the US Food and Drug Administration for patients with stage IV disease. However, therapeutic resistance remains an issue to both classes of agents, and reliable biomarkers of therapeutic response and resistance are lacking. Mechanistic insights are being gained through preclinical studies and translational research, offering potential strategies to enhance responses and survival in treated patients. A comprehensive understanding of the immune effects of common mutations at play in melanoma is critical, as is an appreciation of the molecular mechanisms contributing to therapeutic resistance to immunotherapy. These mechanisms and the interplay between them are discussed herein. Cancer 2017;123:2130-42. © 2017 American Cancer Society.
Collapse
Affiliation(s)
| | | | - Sangeetha M Reddy
- Department of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xue Zhang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexandre Reuben
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
807
|
Westphal D, Glitza Oliva IC, Niessner H. Molecular insights into melanoma brain metastases. Cancer 2017; 123:2163-2175. [PMID: 28543697 DOI: 10.1002/cncr.30594] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/19/2016] [Accepted: 12/28/2016] [Indexed: 01/26/2023]
Abstract
Substantial proportions of patients with metastatic melanoma develop brain metastases during the course of their disease, often resulting in significant morbidity and death. Despite recent advances with BRAF/MEK and immune-checkpoint inhibitors in the treatment of patients who have melanoma with extracerebral metastases, patients who have melanoma brain metastases still have poor overall survival, highlighting the need for further therapy options. A deeper understanding of the molecular pathways involved in the development of melanoma brain metastases is required to develop more brain-specific therapies. Here, the authors summarize the currently known preclinical data and describe steps involved in the development of melanoma brain metastases. Only by knowing the molecular background is it possible to design new therapeutic agents that can be used to improve the outcome of patients with melanoma brain metastases. Cancer 2017;123:2163-75. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Dana Westphal
- Department of Dermatology, Carl Gustav Carus Medical Center, Technical University of Dresden, Dresden, Germany.,Center for Regenerative Therapies, Technical University of Dresden, Dresden, Germany
| | - Isabella C Glitza Oliva
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heike Niessner
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany
| |
Collapse
|
808
|
Abstract
PURPOSE OF REVIEW The therapeutic landscape for metastatic melanoma has been revolutionized in recent years. This review will discuss existing evidence for therapeutic approaches for BRAF-mutated metastatic melanoma. RECENT FINDINGS Clinical trials involving combined BRAF/MEK inhibition with either vemurafenib plus cobimetinib or dabrafenib plus trametinib have shown improved overall survival compared to monotherapy with BRAF inhibitors alone. In a subset of patients with good prognostic factors, long-term clinical benefit has been noted. Simultaneously, developments in immunotherapy have suggested long-lasting survival for some patients. In advanced BRAF-mutated melanoma, both BRAF/MEK inhibition and immunotherapy agents show improved overall survival and, in a small population of patients, prolonged and long-term benefit as compared to standard chemotherapy. Trials are currently underway evaluating sequencing of these therapies and the safety of targeted therapy plus immunotherapy combinations.
Collapse
|
809
|
Distant intracranial failure in melanoma brain metastases treated with stereotactic radiosurgery in the era of immunotherapy and targeted agents. Adv Radiat Oncol 2017; 2:572-580. [PMID: 29204524 PMCID: PMC5707419 DOI: 10.1016/j.adro.2017.07.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/12/2017] [Indexed: 12/19/2022] Open
Abstract
Purpose Stereotactic radiosurgery (SRS) in combination with immunotherapy (IMT) or targeted therapy is increasingly being used in the setting of melanoma brain metastases (MBMs). The synergistic properties of combination therapy are not well understood. We compared the distant intracranial failure rates of intact MBMs treated with SRS, SRS + IMT, and SRS + targeted therapy. Methods and materials Combination therapy was defined as delivery of SRS within 3 months of IMT (anti-CTLA-4 /anti-PD-1 therapy) or targeted therapy (BRAF/MEK inhibitors). The primary endpoint was distant intracranial failure after SRS, which was defined as any new MBM identified on brain magnetic resonance imaging. Outcomes were evaluated using the Kaplan Meier method and Cox proportional hazards. Results A total of 72 patients with melanoma with 233 MBMs were treated between April 2006 and April 2016. The number of MBMs within each treatment group was as follows: SRS: 121; SRS + IMT: 48; and SRS + targeted therapy: 64. The median follow-up was 8.9 months. One-year distant intracranial control rates for SRS, SRS + IMT, and SRS + targeted therapy were 11.5%, 60%, and 10%, respectively (P < .001). On multivariate analysis, after adjusting for steroid use and number of MBMs, SRS + IMT remained associated with a significant reduction in distant intracranial failure compared with SRS (hazard ratio [HR], 0.48; 95% confidence interval [CI], 0.29-0.80; P = .003) and compared with SRS + targeted therapy (HR, 0.41; 95% CI, 0.25-0.68; P = .001).One-year local control for SRS, SRS + IMT, and SRS + targeted therapy was 66%, 85%, and 72%, respectively (P = .044). On multivariate analysis, after adjusting for dose, SRS + IMT remained associated with a significant reduction in local failure compared with SRS alone (HR, 0.37; 95% CI, 0.14-0.95; P = .04). Conclusions SRS with immunotherapy is associated with decreased distant and local intracranial failure compared with SRS alone. Prospective studies are warranted to validate this result.
Collapse
|
810
|
Allegrezza MJ, Rutkowski MR, Stephen TL, Svoronos N, Perales-Puchalt A, Nguyen JM, Payne KK, Singhal S, Eruslanov EB, Tchou J, Conejo-Garcia JR. Trametinib Drives T-cell-Dependent Control of KRAS-Mutated Tumors by Inhibiting Pathological Myelopoiesis. Cancer Res 2017; 76:6253-6265. [PMID: 27803104 DOI: 10.1158/0008-5472.can-16-1308] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/19/2016] [Indexed: 12/11/2022]
Abstract
Targeted therapies elicit seemingly paradoxical and poorly understood effects on tumor immunity. Here, we show that the MEK inhibitor trametinib abrogates cytokine-driven expansion of monocytic myeloid-derived suppressor cells (mMDSC) from human or mouse myeloid progenitors. MEK inhibition also reduced the production of the mMDSC chemotactic factor osteopontin by tumor cells. Together, these effects reduced mMDSC accumulation in tumor-bearing hosts, limiting the outgrowth of KRas-driven breast tumors, even though trametinib largely failed to directly inhibit tumor cell proliferation. Accordingly, trametinib impeded tumor progression in vivo through a mechanism requiring CD8+ T cells, which was paradoxical given the drug's reported ability to inhibit effector lymphocytes. Confirming our observations, adoptive transfer of tumor-derived mMDSC reversed the ability of trametinib to control tumor growth. Overall, our work showed how the effects of trametinib on immune cells could partly explain its effectiveness, distinct from its activity on tumor cells themselves. More broadly, by providing a more incisive view into how MEK inhibitors may act against tumors, our findings expand their potential uses to generally block mMDSC expansion, which occurs widely in cancers to drive their growth and progression. Cancer Res; 76(21); 6253-65. ©2016 AACR.
Collapse
Affiliation(s)
- Michael J Allegrezza
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Melanie R Rutkowski
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Tom L Stephen
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Nikolaos Svoronos
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Alfredo Perales-Puchalt
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Jenny M Nguyen
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Kyle K Payne
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Evgeniy B Eruslanov
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Julia Tchou
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jose R Conejo-Garcia
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania.
| |
Collapse
|
811
|
Cohen R, Cervera P, Svrcek M, Pellat A, Dreyer C, de Gramont A, André T. BRAF-Mutated Colorectal Cancer: What Is the Optimal Strategy for Treatment? Curr Treat Options Oncol 2017; 18:9. [PMID: 28214977 DOI: 10.1007/s11864-017-0453-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OPINION STATEMENT The BRAF activating mutation, harbored by approximately 10% of colorectal cancers (CRC), confers dramatic prognosis to advanced diseases. In early-stage setting, the identification of the BRAF mutation does not impact the therapeutic decision. Yet, the BRAF mutation could be considered as a stratification factor in adjuvant trials, because of its prognostic impact after relapse. Moreover, both BRAF mutation and mismatch repair (MMR) statuses should be determined in all CRC to help identify sporadic tumors versus Lynch syndrome-related tumors. Indeed, in patients with MMR-deficient (dMMR) tumors and MLH1 loss of expression, the BRAFV600E mutation indicates a sporadic origin. In advanced BRAF-mutated CRC, the standard of care remains fluoropyrimidine-based cytotoxic regimen in combination with bevacizumab. Although a recent meta-analysis showed that there was insufficient data to justify the exclusion of anti-EGFR monoclonal antibodies, antiangiogenic agents should be preferred in the first-line setting. Despite the lack of a randomized phase 3 study dedicated to BRAF-mutated CRC, chemotherapy intensification combining a quadruple association of 5-fluorouracil, oxaliplatin, irinotecan (FOLFOXIRI), and bevacizumab seems like a valid option. Although first results with BRAF inhibitors as single agents in BRAF-mutated CRC were disappointing, their association with therapies targeting the MAPK pathway seems to overcome the primary resistance to BRAF inhibition. In the field of sporadic CRC, the BRAF mutation is strongly associated with MMR deficiency. Considering breakthrough results of immune checkpoint inhibitors in dMMR repair tumors, determination of the MMR status appears to be mandatory. Given the dramatic prognosis conferred by the BRAF mutation, patients with BRAF-mutated advanced CRC need to be systematically identified and proposed for clinical trial enrolment in order to benefit from innovative therapies.
Collapse
Affiliation(s)
- Romain Cohen
- Department of Medical Oncology, Saint-Antoine Hospital, APHP, 184 rue du Faubourg Saint-Antoine, 75012, Paris, France
| | - Pascale Cervera
- Department of Pathology, Saint-Antoine Hospital, APHP, 184 rue du Faubourg Saint-Antoine, 75012, Paris, France
| | - Magali Svrcek
- Department of Pathology, Saint-Antoine Hospital, APHP, 184 rue du Faubourg Saint-Antoine, 75012, Paris, France
| | - Anna Pellat
- Department of Medical Oncology, Saint-Antoine Hospital, APHP, 184 rue du Faubourg Saint-Antoine, 75012, Paris, France
| | - Chantal Dreyer
- Department of Medical Oncology, Saint-Antoine Hospital, APHP, 184 rue du Faubourg Saint-Antoine, 75012, Paris, France
| | - Aimery de Gramont
- Department of Medical Oncology, Institut Hospitalier Franco-Britannique, 4 rue Kléber, 92300, Levallois-Perret, France.,GERCOR, Oncology Multidisciplinary Group, 151 rue du Faubourg Saint Antoine, 75011, Paris, France
| | - Thierry André
- Department of Medical Oncology, Saint-Antoine Hospital, APHP, 184 rue du Faubourg Saint-Antoine, 75012, Paris, France. .,GERCOR, Oncology Multidisciplinary Group, 151 rue du Faubourg Saint Antoine, 75011, Paris, France.
| |
Collapse
|
812
|
Tsai AK, Khan AY, Worgo CE, Wang LL, Liang Y, Davila E. A Multikinase and DNA-PK Inhibitor Combination Immunomodulates Melanomas, Suppresses Tumor Progression, and Enhances Immunotherapies. Cancer Immunol Res 2017; 5:790-803. [PMID: 28775208 DOI: 10.1158/2326-6066.cir-17-0009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 05/17/2017] [Accepted: 07/25/2017] [Indexed: 12/19/2022]
Abstract
Combination therapies have the potential to improve outcomes in melanoma patients but have not yet been clinically efficacious. Here, we used high-throughput flow cytometry-based screening to identify and characterize candidate therapies that might synergize with and augment T-cell immunotherapy efficacy. Two lead therapies, regorafenib (Reg) and NU7441, were selected based on their ability to alter a variety of immunomodulatory proteins, including CD55, CD73, CD155, programmed death-ligand 1 (PD-L1), nerve growth factor receptor (NGFR), and HLA class I in a heterogeneous panel of melanomas. The therapies also upregulated several melanoma antigens, inhibited proliferation, and perturbed activation of oncogenic signaling pathways in melanomas. T cells treated with the therapies proliferated normally and exhibited a favorably altered phenotype, including increased CD25, CD28, inducible T-cell costimulator (ICOS), and reduced expression of coinhibitory receptors. Cytokine production was also increased in treated T cells. When administered in mice, REg suppressed melanoma progression in a CD8+ T cell-dependent manner when used alone and with various immunotherapies. Additionally, Reg altered the number, phenotype, and function of various T-cell subsets in the tumor microenvironment. These studies reveal that Reg and NU7441 influence the immunobiology of both tumor cells and T cells and enhance the efficacy of various immunotherapies. Cancer Immunol Res; 5(9); 790-803. ©2017 AACR.
Collapse
Affiliation(s)
- Alexander K Tsai
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| | - Asra Y Khan
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| | - Christina E Worgo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| | - Lucy L Wang
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| | - Yuanyuan Liang
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, Maryland
| | - Eduardo Davila
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland. .,Department of Microbiology and Immunology, University of Maryland, Baltimore, Maryland
| |
Collapse
|
813
|
Betof AS, Nipp RD, Giobbie-Hurder A, Johnpulle RAN, Rubin K, Rubinstein SM, Flaherty KT, Lawrence DP, Johnson DB, Sullivan RJ. Impact of Age on Outcomes with Immunotherapy for Patients with Melanoma. Oncologist 2017; 22:963-971. [PMID: 28476944 PMCID: PMC5553960 DOI: 10.1634/theoncologist.2016-0450] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 03/03/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Monoclonal antibodies (mAb) targeting PD-1/PD-L1 have revolutionized melanoma treatment, yet data regarding effectiveness and tolerability across age groups is limited. We sought to determine the impact of age on overall survival (OS), progression-free survival (PFS), and rates of immune-mediated toxicities in patients treated with anti-PD-1/anti-PD-L1 mAb at two academic medical centers. METHODS We retrospectively collected data on all patients with metastatic melanoma treated with anti-PD-1/PD-L1 mAb between May 2009 and April 2015. We used Kaplan-Meier and Cox regression analyses to assess OS and PFS and identify factors associated with these outcomes. We also compared rates of autoimmune toxicity across age groups. RESULTS Of 254 patients, 57 (22.4%) were <50 years old, 85 (33.5%) were age 50-64, 65 (25.6%) were age 65-74, and 47 (18.5%) were ≥75 years. Across age groups, no differences existed in median OS (age <50: 22.9 months, age 50-64: 25.3 months, age 65-74: 22.0 months, age ≥75: 24.3 months) or PFS (age <50: 4.1 months, age 50-64: 6.5 months, age 65-74: 5.4 months, age ≥75: 7.9 months). The presence of liver metastases and elevated pre-treatment lactate dehydrogenase (LDH) were associated with reduced OS. Presence of liver metastasis, pretreatment LDH, BRAF mutation, and type of melanoma correlated with PFS. Overall, 110 patients (43.3%) experienced immune-mediated toxicities; 25 (9.8%) had colitis and 26 (10.2%) had endocrine toxicity. Rates of colitis, hepatitis, and pneumonitis did not differ across age groups. CONCLUSION We demonstrated that patients could safely tolerate anti-PD1/PDL-1 mAb therapy and achieve similar outcomes regardless of their age. IMPLICATIONS FOR PRACTICE Immunotherapy has revolutionized treatment for patients with metastatic melanoma, yet data are lacking regarding the effectiveness and tolerability of these treatments for older patients. In this study, we demonstrated that patients with melanoma safely tolerate immunotherapy and achieve similar outcomes regardless of their age. Specifically, we utilized data from two academic cancer centers and found no significant difference in overall survival, progression free survival, or immune-related toxicities, other than arthritis, across age groups. As the population ages, studies such as this will become critical to help us understand how best to treat older adults with cancer.
Collapse
Affiliation(s)
- Allison S Betof
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| | - Ryan D Nipp
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| | - Anita Giobbie-Hurder
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Krista Rubin
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| | | | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| | - Donald P Lawrence
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| | | | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center & Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
814
|
Koelblinger P, Dornbierer J, Dummer R. A review of binimetinib for the treatment of mutant cutaneous melanoma. Future Oncol 2017; 13:1755-1766. [PMID: 28587477 DOI: 10.2217/fon-2017-0170] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although significant progress has been made in the treatment of unresectable or metastatic melanoma, at least half of all advanced melanoma patients eventually progress and pass away due to their disease. In particular, patients with NRAS-mutated melanoma still face limited therapeutic options, with immunotherapy being the current treatment type of choice. Binimetinib is a selective inhibitor of MEK, a central kinase in the tumor-promoting MAPK pathway. The results of a recent Phase III trial rendered binimetinib the first targeted therapy agent to significantly improve progression-free survival in NRAS-mutated melanoma. This review will summarize the development and clinical data of binimetinib in melanoma in general and also explore the potential future role of this substance as single agent or combination therapy.
Collapse
Affiliation(s)
- Peter Koelblinger
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Department of Dermatology, Paracelsus Medical University, Salzburg, Austria
| | - Joelle Dornbierer
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
815
|
Utter K, Goldman C, Weiss SA, Shapiro RL, Berman RS, Wilson MA, Pavlick AC, Osman I. Treatment Outcomes for Metastatic Melanoma of Unknown Primary in the New Era: A Single-Institution Study and Review of the Literature. Oncology 2017; 93:249-258. [PMID: 28746931 DOI: 10.1159/000478050] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/28/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Metastatic melanoma of unknown primary (MUP) is uncommon, biologically ill defined, and clinically understudied. MUP outcomes are seldom reported in clinical trials. In this study, we analyze responses of MUP patients treated with systemic therapy in an attempt to inform treatment guidelines for this unique population. METHODS New York University (NYU)'s prospective melanoma database was searched for MUP patients treated with systemic therapy. PubMed and Google Scholar were searched for MUP patients treated with immunotherapy or targeted therapy reported in the literature, and their response and survival data were compared to the MUP patient data from NYU. Both groups' response data were compared to those reported for melanoma of known primary (MKP). RESULTS The MUP patients treated at NYU had better outcomes on immunotherapy but worse on targeted therapy than the MUP patients in the literature. The NYU MUP patients and those in the literature had worse outcomes than the majority-MKP populations in 10 clinical trial reports. CONCLUSIONS Our study suggests that MUP patients might have poorer outcomes on systemic therapy as compared to MKP patients. Our cohort was small and limited data were available, highlighting the need for increased reporting of MUP outcomes and multi-institutional efforts to understand the mechanism behind the observed differences.
Collapse
Affiliation(s)
- Kierstin Utter
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
816
|
Reddy SM, Reuben A, Wargo JA. Influences of BRAF Inhibitors on the Immune Microenvironment and the Rationale for Combined Molecular and Immune Targeted Therapy. Curr Oncol Rep 2017; 18:42. [PMID: 27215436 DOI: 10.1007/s11912-016-0531-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The identification of key driver mutations in melanoma has led to the development of targeted therapies aimed at BRAF and MEK, but responses are often limited in duration. There is growing evidence that MAPK pathway activation impairs antitumor immunity and that targeting this pathway may enhance responses to immunotherapies. There is also evidence that immune mechanisms of resistance to targeted therapy exist, providing the rationale for combining targeted therapy with immunotherapy. Preclinical studies have demonstrated synergy in combining these strategies, and combination clinical trials are ongoing. It is, however, becoming clear that additional translational studies are needed to better understand toxicity, proper timing, and sequence of therapy, as well as the utility of multidrug regimens and effects of other targeted agents on antitumor immunity. Insights gained through translational research in preclinical models and clinical studies will provide mechanistic insight into therapeutic response and resistance and help devise rational strategies to enhance therapeutic responses.
Collapse
Affiliation(s)
- Sangeetha M Reddy
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Unit 463, Houston, TX, 77030, USA
| | - Alexandre Reuben
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
817
|
Brinker TJ, Schadendorf D, Klode J, Cosgarea I, Rösch A, Jansen P, Stoffels I, Izar B. Photoaging Mobile Apps as a Novel Opportunity for Melanoma Prevention: Pilot Study. JMIR Mhealth Uhealth 2017; 5:e101. [PMID: 28747297 PMCID: PMC5550737 DOI: 10.2196/mhealth.8231] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 07/03/2017] [Accepted: 07/12/2017] [Indexed: 12/13/2022] Open
Abstract
Background Around 90% of melanomas are caused by ultraviolet (UV) exposure and are therefore eminently preventable. Unhealthy tanning behavior is mostly initiated in early adolescence, often with the belief that it increases attractiveness; the problems related to skin atrophy and malignant melanoma are too far in the future to fathom. Photoaging desktop programs, in which an image is altered to predict future appearance, have been successful in positively influencing behavior in adiposity or tobacco prevention settings. Objective To develop and test a photoaging app designed for melanoma prevention. Methods We harnessed the widespread availability of mobile phones and adolescents’ interest in appearance to develop a free mobile app called Sunface. This app has the user take a self-portrait (ie, a selfie), and then photoages the image based on Fitzpatrick skin type and individual UV protection behavior. Afterward, the app explains the visual results and aims at increasing self-competence on skin cancer prevention by providing guideline recommendations on sun protection and the ABCDE rule for melanoma self-detection. The underlying aging algorithms are based on publications showing UV-induced skin damage by outdoor as well as indoor tanning. To get a first impression on how well the app would be received in a young target group, we included a total sample of 25 students in our cross-sectional pilot study with a median age of 22 (range 19-25) years of both sexes (11/25, 44% female; 14/25, 56% male) attending the University of Essen in Germany. Results The majority of enrolled students stated that they would download the app (22/25, 88%), that the intervention had the potential to motivate them to use sun protection (23/25, 92%) and that they thought such an app could change their perceptions that tanning makes you attractive (19/25, 76%). Only a minority of students disagreed or fully disagreed that they would download such an app (2/25, 8%) or that such an app could change their perceptions on tanning and attractiveness (4/25, 16%). Conclusions Based on previous studies and the initial study results presented here, it is reasonable to speculate that the app may induce behavioral change in the target population. Further work is required to implement and examine the effectiveness of app-based photoaging interventions within risk groups from various cultural backgrounds.
Collapse
Affiliation(s)
- Titus Josef Brinker
- Department of Dermatology, Venerology and Allergology, University-Hospital Essen, University of Duisburg-Essen, Essen, Germany.,West German Cancer Center, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, Venerology and Allergology, University-Hospital Essen, University of Duisburg-Essen, Essen, Germany.,West German Cancer Center, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Joachim Klode
- Department of Dermatology, Venerology and Allergology, University-Hospital Essen, University of Duisburg-Essen, Essen, Germany.,West German Cancer Center, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ioana Cosgarea
- Department of Dermatology, Venerology and Allergology, University-Hospital Essen, University of Duisburg-Essen, Essen, Germany.,West German Cancer Center, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Alexander Rösch
- Department of Dermatology, Venerology and Allergology, University-Hospital Essen, University of Duisburg-Essen, Essen, Germany.,West German Cancer Center, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Philipp Jansen
- Department of Dermatology, Venerology and Allergology, University-Hospital Essen, University of Duisburg-Essen, Essen, Germany.,West German Cancer Center, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ingo Stoffels
- Department of Dermatology, Venerology and Allergology, University-Hospital Essen, University of Duisburg-Essen, Essen, Germany.,West German Cancer Center, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Benjamin Izar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States.,Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
818
|
Dermit M, Dokal A, Cutillas PR. Approaches to identify kinase dependencies in cancer signalling networks. FEBS Lett 2017; 591:2577-2592. [DOI: 10.1002/1873-3468.12748] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 06/27/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Maria Dermit
- Cell Signalling & Proteomics Group; Barts Cancer Institute (CRUK Centre); Queen Mary University of London; UK
| | - Arran Dokal
- Cell Signalling & Proteomics Group; Barts Cancer Institute (CRUK Centre); Queen Mary University of London; UK
| | - Pedro R. Cutillas
- Cell Signalling & Proteomics Group; Barts Cancer Institute (CRUK Centre); Queen Mary University of London; UK
| |
Collapse
|
819
|
Quéreux G, Herbreteau G, Knol AC, Vallée A, Khammari A, Théoleyre S, Saint-Jean M, Dréno B, Denis MG. Efficient treatment of a metastatic melanoma patient with a combination of BRAF and MEK inhibitors based on circulating tumor DNA analysis: a case report. BMC Res Notes 2017; 10:320. [PMID: 28743309 PMCID: PMC5526247 DOI: 10.1186/s13104-017-2650-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 07/21/2017] [Indexed: 12/21/2022] Open
Abstract
Background Fixed tissues are the standard samples used in routine practice for molecular testing. But sometimes tissues are lacking or difficult to obtain. In these cases, circulating tumor DNA released from tumor cells can be used as an alternative source of tumor DNA. Case presentation We present the case of a 63-year-old Caucasian woman with a metastatic melanoma and a very poor performance status. A plasma sample was tested and the BRAF p.V600E mutation was detected. Based on this result, a treatment combining a BRAF inhibitor and a MEK inhibitor was immediately started. This patient achieved a complete response. In addition, by repeating the plasma test, we could obtain a precise kinetic of release of mutated BRAF DNA in plasma. Conclusions We report here for the first time the efficient treatment of a metastatic melanoma patient on the basis of circulating tumor DNA analysis. This urgent treatment provided a dramatic response in a patient with a very poor initial condition. The kinetic data most likely reflect treatment efficacy.
Collapse
Affiliation(s)
- Gaelle Quéreux
- Department of Dermatology, INSERM CIC1413, Nantes University Hospital, Nantes, France.,CRCINA INSERM U1232, Nantes, France.,Immuno-Dermatology Laboratory, Nantes University Hospital, Nantes, France
| | - Guillaume Herbreteau
- CRCINA INSERM U1232, Nantes, France.,Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, 44093, Nantes Cedex 01, France
| | - Anne-Chantal Knol
- CRCINA INSERM U1232, Nantes, France.,Immuno-Dermatology Laboratory, Nantes University Hospital, Nantes, France
| | - Audrey Vallée
- CRCINA INSERM U1232, Nantes, France.,Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, 44093, Nantes Cedex 01, France
| | - Amir Khammari
- Department of Dermatology, INSERM CIC1413, Nantes University Hospital, Nantes, France.,CRCINA INSERM U1232, Nantes, France.,Immuno-Dermatology Laboratory, Nantes University Hospital, Nantes, France
| | - Sandrine Théoleyre
- CRCINA INSERM U1232, Nantes, France.,Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, 44093, Nantes Cedex 01, France
| | - Mélanie Saint-Jean
- Department of Dermatology, INSERM CIC1413, Nantes University Hospital, Nantes, France.,CRCINA INSERM U1232, Nantes, France.,Immuno-Dermatology Laboratory, Nantes University Hospital, Nantes, France
| | - Brigitte Dréno
- Department of Dermatology, INSERM CIC1413, Nantes University Hospital, Nantes, France.,CRCINA INSERM U1232, Nantes, France.,Immuno-Dermatology Laboratory, Nantes University Hospital, Nantes, France
| | - Marc G Denis
- CRCINA INSERM U1232, Nantes, France. .,Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, 44093, Nantes Cedex 01, France.
| |
Collapse
|
820
|
Grätz V, Lüttmann N, Haase O, Langan EA, Kemmling A, Zillikens D, Terheyden P. Meningeal melanomatosis following discontinuation of dabrafenib: implications for the maintenance of long-term complete remission. Melanoma Res 2017; 27:503-506. [PMID: 28723725 DOI: 10.1097/cmr.0000000000000373] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A subset of 10-20% of patients under continuous BRAF inhibitor monotherapy achieve long-term progression-free and overall survival. Definitive criteria for the safe cessation of BRAF inhibitor monotherapy in treatment-responsive melanoma patients are lacking. We report a patient who remained in complete remission (CR) for 5 years under dabrafenib. The treatment was withdrawn because of concerns about cardiac toxicity. Four months thereafter the patient developed neurological symptoms, including diplopia and bilateral visual loss. Meningeal melanomatosis and parenchymal brain metastases were diagnosed. Extracerebral metastases were excluded. Reinduction of dabrafenib, combined with trametinib, led to the rapid relief of the neurological symptoms, and a partial remission was confirmed radiologically. Unfortunately, the response was not maintained and the patient died 9 months later. This observation demonstrates that discontinuation of BRAF inhibition can result in loss of disease control. On the basis of this observation, we suggest that BRAF-targeted therapy should be withdrawn only when the risks of continued treatment exceed the risk for disease relapse. However, future studies are urgently required to confirm and quantify the risk for rapid disease relapse following withdrawal of BRAF inhibitor monotherapy.
Collapse
Affiliation(s)
- Victoria Grätz
- Department of Dermatology, University of Luebeck, Luebeck, Germany
| | | | | | | | | | | | | |
Collapse
|
821
|
PD-L1 Expression and Immune Escape in Melanoma Resistance to MAPK Inhibitors. Clin Cancer Res 2017; 23:6054-6061. [DOI: 10.1158/1078-0432.ccr-16-1688] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 05/11/2017] [Accepted: 07/14/2017] [Indexed: 11/16/2022]
|
822
|
Niessner H, Sinnberg T, Kosnopfel C, Smalley KSM, Beck D, Praetorius C, Mai M, Beissert S, Kulms D, Schaller M, Garbe C, Flaherty KT, Westphal D, Wanke I, Meier F. BRAF Inhibitors Amplify the Proapoptotic Activity of MEK Inhibitors by Inducing ER Stress in NRAS-Mutant Melanoma. Clin Cancer Res 2017; 23:6203-6214. [PMID: 28724666 DOI: 10.1158/1078-0432.ccr-17-0098] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/25/2017] [Accepted: 07/11/2017] [Indexed: 12/14/2022]
Abstract
Purpose: NRAS mutations in malignant melanoma are associated with aggressive disease requiring rapid antitumor intervention, but there is no approved targeted therapy for this subset of patients. In clinical trials, the MEK inhibitor (MEKi) binimetinib displayed modest antitumor activity, making combinations a requisite. In a previous study, the BRAF inhibitor (BRAFi) vemurafenib was shown to induce endoplasmic reticulum (ER) stress that together with inhibition of the RAF-MEK-ERK (MAPK) pathway amplified its proapoptotic activity in BRAF-mutant melanoma. The present study investigated whether this effect might extent to NRAS-mutant melanoma, in which MAPK activation would be expected.Experimental Design and Results: BRAFi increased pERK, but also significantly increased growth inhibition and apoptosis induced by the MEKi in monolayer, spheroids, organotypic, and patient-derived tissue slice cultures of NRAS-mutant melanoma. BRAFi such as encorafenib induced an ER stress response via the PERK pathway, as detected by phosphorylation of eIF2α and upregulation of the ER stress-related factors ATF4, CHOP, and NUPR1 and the proapoptotic protein PUMA. MEKi such as binimetinib induced the expression of the proapoptotic protein BIM and activation of the mitochondrial pathway of apoptosis, the latter of which was enhanced by combination with encorafenib. The increased apoptotic rates caused by the combination treatment were significantly reduced through siRNA knockdown of ATF4 and BIM, confirming its critical roles in this process.Conclusions: The data presented herein encourage further advanced in vivo and clinical studies to evaluate MEKi in combination with ER stress inducing BRAFi as a strategy to treat rapidly progressing NRAS-mutant melanoma. Clin Cancer Res; 23(20); 6203-14. ©2017 AACR.
Collapse
Affiliation(s)
- Heike Niessner
- Department of Dermatology, Oncology, University Medical Center, Tübingen, Germany.
| | - Tobias Sinnberg
- Department of Dermatology, Oncology, University Medical Center, Tübingen, Germany
| | - Corinna Kosnopfel
- Department of Dermatology, Oncology, University Medical Center, Tübingen, Germany
| | - Keiran S M Smalley
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Daniela Beck
- Department of Dermatology, Oncology, University Medical Center, Tübingen, Germany
| | - Christian Praetorius
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, TU Dresden, Germany
| | - Marion Mai
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, Dresden, Germany
| | - Stefan Beissert
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, Dresden, Germany
| | - Dagmar Kulms
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, TU Dresden, Germany
| | - Martin Schaller
- Department of Dermatology, Oncology, University Medical Center, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, Oncology, University Medical Center, Tübingen, Germany
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Dana Westphal
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, TU Dresden, Germany
| | - Ines Wanke
- Department of Dermatology, Oncology, University Medical Center, Tübingen, Germany
| | - Friedegund Meier
- Department of Dermatology, Oncology, University Medical Center, Tübingen, Germany.,Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
| |
Collapse
|
823
|
Cavalieri S, Di Guardo L, Cossa M, Cimminiello C, Del Vecchio M. Unusual Skin Carcinomas Induced by BRAF Inhibitor for Metastatic Melanoma: A Case Report. J Clin Diagn Res 2017; 11:XD06-XD08. [PMID: 28893027 DOI: 10.7860/jcdr/2017/26881.10200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/19/2017] [Indexed: 11/24/2022]
Abstract
The most frequently reported skin tumours during treatment with targeted therapies for BRAF (B type Rapidly Accelerated Fibrosarcoma kinase) mutated metastatic melanoma are squamous cell carcinomas (SCCs). Basal cell carcinomas (BCCs) have been described in such setting, but no cases of multiple and recurring tumours have been reported so far. A patient with a history of chronic sun exposure and more than 10 BCCs removed since 1998 started treatment with vemurafenib for BRAF mutated metastatic melanoma. Therapy was complicated by sporadic episodes of atrial fibrillation and by the development of recurrent, multiple and diffuse BCCs. So, vemurafenib was discontinued and dabrafenib and trametinib were started. Since then, only four BCCs occurred in the patient. Histopathological re-examination showed that most BCCs occurred under vemurafenib presented with squamous features. Such characteristic was significantly less evident before therapy start and in lesions removed under treatment with dabrafenib and trametinib. BRAF inhibition (BRAFi) without MEK inhibition induces mitogen activated kinases overactivation, with consequent skin toxicity and acquired drug resistance. The BCCs removed from our patient showed squamous features, more evident during vemurafenib monotherapy. Both the switch from vemurafenib to dabrafenib and the addition of MEK inhibitor (MEKi) might have reduced the incidence of BCCs and their squamous differentiation.
Collapse
Affiliation(s)
- Stefano Cavalieri
- Faculty, Department of Medical Oncology, Fondazione Istituto Nazionale dei Tumori, Milan, Italy
| | - Lorenza Di Guardo
- Faculty, Department of Medical Oncology, Fondazione Istituto Nazionale dei Tumori, Milan, Italy
| | - Mara Cossa
- Faculty, Department of Pathology, Fondazione Istituto Nazionale dei Tumori, Milan, Italy
| | - Carolina Cimminiello
- Faculty, Department of Medical Oncology, Fondazione Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Del Vecchio
- Faculty, Department of Medical Oncology, Fondazione Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
824
|
Forsyth PA, Smalley KSM, Sondak VK. BRAF-MEK inhibition in melanoma brain metastases: a new hope. Lancet Oncol 2017; 18:836-837. [DOI: 10.1016/s1470-2045(17)30449-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 05/26/2017] [Indexed: 11/29/2022]
|
825
|
Luo C, Shen J. Research progress in advanced melanoma. Cancer Lett 2017; 397:120-126. [DOI: 10.1016/j.canlet.2017.03.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 12/12/2022]
|
826
|
Hélias-Rodzewicz Z, Funck-Brentano E, Terrones N, Beauchet A, Zimmermann U, Marin C, Saiag P, Emile JF. Variation of mutant allele frequency in NRAS Q61 mutated melanomas. BMC DERMATOLOGY 2017; 17:9. [PMID: 28668077 PMCID: PMC5494128 DOI: 10.1186/s12895-017-0061-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/21/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND Somatic mutations of BRAF or NRAS activating the MAP kinase cell signaling pathway are present in 70% of cutaneous melanomas. The mutant allele frequency of BRAF V600E (M%BRAF) was recently shown to be highly heterogeneous in melanomas. The present study focuses on the NRAS Q61 mutant allele frequency (M%NRAS). METHODS Retrospective quantitative analyze of 104 NRAS mutated melanomas was performed using pyrosequencing. Mechanisms of M%NRAS imbalance were studied by fluorescence in situ hybridization (FISH) and microsatellite analysis. RESULTS M%NRAS was increased in 27.9% of cases. FISH revealed that chromosome 1 instability was the predominant mechanism of M%NRAS increase, with chromosome 1 polysomy observed in 28.6% of cases and intra-tumor cellular heterogeneity with copy number variations of chromosome 1/NRAS in 23.8%. Acquired copy-neutral loss of heterozygosity (LOH) was less frequent (19%). However, most samples with high M%NRAS had only one copy of NRAS locus surrounding regions suggesting a WT allele loss. Clinical characteristics and survival of patients with either <60% or ≥60% of M%NRAS were not different. CONCLUSION As recently shown for M%BRAF, M%NRAS is highly heterogeneous. The clinical impacts of high M%NRAS should be investigated in a larger series of patients.
Collapse
Affiliation(s)
- Zofia Hélias-Rodzewicz
- Research Unit EA4340 Biomarkers in Cancerology and Hemato Oncology, Versailles SQY University, Paris-Saclay University, 9, Avenue Charles de Gaulle, 92104 Boulogne-Billancourt, France
- Department of Pathology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Elisa Funck-Brentano
- Research Unit EA4340 Biomarkers in Cancerology and Hemato Oncology, Versailles SQY University, Paris-Saclay University, 9, Avenue Charles de Gaulle, 92104 Boulogne-Billancourt, France
- Department of Dermatology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Nathalie Terrones
- Research Unit EA4340 Biomarkers in Cancerology and Hemato Oncology, Versailles SQY University, Paris-Saclay University, 9, Avenue Charles de Gaulle, 92104 Boulogne-Billancourt, France
| | - Alain Beauchet
- Department of Public Health, Ambroise Paré Hospital Ap-HP, Boulogne-Billancourt, France
| | - Ute Zimmermann
- Research Unit EA4340 Biomarkers in Cancerology and Hemato Oncology, Versailles SQY University, Paris-Saclay University, 9, Avenue Charles de Gaulle, 92104 Boulogne-Billancourt, France
- Department of Pathology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Cristi Marin
- Research Unit EA4340 Biomarkers in Cancerology and Hemato Oncology, Versailles SQY University, Paris-Saclay University, 9, Avenue Charles de Gaulle, 92104 Boulogne-Billancourt, France
- Department of Pathology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Philippe Saiag
- Research Unit EA4340 Biomarkers in Cancerology and Hemato Oncology, Versailles SQY University, Paris-Saclay University, 9, Avenue Charles de Gaulle, 92104 Boulogne-Billancourt, France
- Department of Dermatology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Jean-François Emile
- Research Unit EA4340 Biomarkers in Cancerology and Hemato Oncology, Versailles SQY University, Paris-Saclay University, 9, Avenue Charles de Gaulle, 92104 Boulogne-Billancourt, France
- Department of Pathology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| |
Collapse
|
827
|
Davies MA, Saiag P, Robert C, Grob JJ, Flaherty KT, Arance A, Chiarion-Sileni V, Thomas L, Lesimple T, Mortier L, Moschos SJ, Hogg D, Márquez-Rodas I, Del Vecchio M, Lebbé C, Meyer N, Zhang Y, Huang Y, Mookerjee B, Long GV. Dabrafenib plus trametinib in patients with BRAF V600-mutant melanoma brain metastases (COMBI-MB): a multicentre, multicohort, open-label, phase 2 trial. Lancet Oncol 2017; 18:863-873. [PMID: 28592387 PMCID: PMC5991615 DOI: 10.1016/s1470-2045(17)30429-1] [Citation(s) in RCA: 541] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Dabrafenib plus trametinib improves clinical outcomes in BRAFV600-mutant metastatic melanoma without brain metastases; however, the activity of dabrafenib plus trametinib has not been studied in active melanoma brain metastases. Here, we report results from the phase 2 COMBI-MB trial. Our aim was to build on the current body of evidence of targeted therapy in melanoma brain metastases through an evaluation of dabrafenib plus trametinib in patients with BRAFV600-mutant melanoma brain metastases. METHODS This ongoing, multicentre, multicohort, open-label, phase 2 study evaluated oral dabrafenib (150 mg twice per day) plus oral trametinib (2 mg once per day) in four patient cohorts with melanoma brain metastases enrolled from 32 hospitals and institutions in Europe, North America, and Australia: (A) BRAFV600E-positive, asymptomatic melanoma brain metastases, with no previous local brain therapy, and an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1; (B) BRAFV600E-positive, asymptomatic melanoma brain metastases, with previous local brain therapy, and an ECOG performance status of 0 or 1; (C) BRAFV600D/K/R-positive, asymptomatic melanoma brain metastases, with or without previous local brain therapy, and an ECOG performance status of 0 or 1; and (D) BRAFV600D/E/K/R-positive, symptomatic melanoma brain metastases, with or without previous local brain therapy, and an ECOG performance status of 0, 1, or 2. The primary endpoint was investigator-assessed intracranial response in cohort A in the all-treated-patients population. Secondary endpoints included intracranial response in cohorts B, C, and D. This study is registered with ClinicalTrials.gov, number NCT02039947. FINDINGS Between Feb 28, 2014, and Aug 5, 2016, 125 patients were enrolled in the study: 76 patients in cohort A; 16 patients in cohort B; 16 patients in cohort C; and 17 patients in cohort D. At the data cutoff (Nov 28, 2016) after a median follow-up of 8·5 months (IQR 5·5-14·0), 44 (58%; 95% CI 46-69) of 76 patients in cohort A achieved an intracranial response. Intracranial response by investigator assessment was also achieved in nine (56%; 95% CI 30-80) of 16 patients in cohort B, seven (44%; 20-70) of 16 patients in cohort C, and ten (59%; 33-82) of 17 patients in cohort D. The most common serious adverse events related to study treatment were pyrexia for dabrafenib (eight [6%] of 125 patients) and decreased ejection fraction (five [4%]) for trametinib. The most common grade 3 or worse adverse events, regardless of study drug relationship, were pyrexia (four [3%] of 125) and headache (three [2%]). INTERPRETATION Dabrafenib plus trametinib was active with a manageable safety profile in this melanoma population that was consistent with previous dabrafenib plus trametinib studies in patients with BRAFV600-mutant melanoma without brain metastases, but the median duration of response was relatively short. These results provide evidence of clinical benefit with dabrafenib plus trametinib and support the need for additional research to further improve outcomes in patients with melanoma brain metastases. FUNDING Novartis.
Collapse
Affiliation(s)
- Michael A Davies
- Melanoma Medical Oncology and Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Philippe Saiag
- Service de Dermatologie Générale et Oncologique, Hôpital A Paré, Assistance Publique-Hôpitaux de Paris, Boulogne Billancourt, France; EA 4340, Université Versailles Saint-Quentin-en-Yvelines, Boulogne Billancourt, France
| | - Caroline Robert
- Gustave Roussy, Département de Médecine Oncologique, Service de Dermatologie et Université Paris-Sud, Faculté de Médecine, Villejuif, France
| | - Jean-Jacques Grob
- Service de Dermatologie, Centre Hospitalo-Universitaire Timone, Aix Marseille University, Marseille, France
| | - Keith T Flaherty
- Developmental Therapeutics and Melanoma Programs, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Ana Arance
- Department of Medical Oncology, Hospital Clinic of Barcelona, Carrer de Villarroel, Barcelona, Spain
| | - Vanna Chiarion-Sileni
- Melanoma and Oesophageal Oncology Unit, Veneto Oncology Institute-IRCCS, Padova, Italy
| | - Luc Thomas
- Service de Dermatologie, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | | | - Laurent Mortier
- Clinique de Dermatologie, Unité d'Onco-Dermatologie, Le Centre Hospitalier Régional Universitaire de Lille, University Lille 2, Lille, France
| | - Stergios J Moschos
- Melanoma Program, Medical Oncology, UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - David Hogg
- Clinical Cancer Research Unit, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Iván Márquez-Rodas
- Servicio de Oncología Médica; Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | | | - Céleste Lebbé
- APHP Dermatology and CIC Departments, INSERM U976, University Paris Diderot, Hôpital Saint Louis Paris, Paris, France
| | - Nicolas Meyer
- Medical Oncology, Institut Universitaire du Cancer Toulouse Oncopole, Toulouse, France
| | - Ying Zhang
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Yingjie Huang
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| |
Collapse
|
828
|
Baik CS, Myall NJ, Wakelee HA. Targeting BRAF-Mutant Non-Small Cell Lung Cancer: From Molecular Profiling to Rationally Designed Therapy. Oncologist 2017; 22:786-796. [PMID: 28487464 PMCID: PMC5507646 DOI: 10.1634/theoncologist.2016-0458] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/06/2017] [Indexed: 12/28/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related deaths globally. However, the identification of oncogenic driver alterations involved in the initiation and maintenance of NSCLC, such as epidermal growth factor receptor mutations and anaplastic lymphoma kinase translocation, has led to the development of novel therapies that directly target mutant proteins and associated signaling pathways, resulting in improved clinical outcomes. As sequencing techniques have improved, the molecular heterogeneity of NSCLC has become apparent, leading to the identification of a number of potentially actionable oncogenic driver mutations. Of these, one of the most promising therapeutic targets is B-Raf proto-oncogene, serine/threonine kinase (BRAF). Mutations in BRAF, observed in 2%-4% of NSCLCs, typically lead to constitutive activation of the protein and, as a consequence, lead to activation of the mitogen-activated protein kinase signaling pathway. Direct inhibition of mutant BRAF and/or the downstream mitogen-activated protein kinase kinase (MEK) has led to prolonged survival in patients with BRAF-mutant metastatic melanoma. This comprehensive review will discuss the clinical characteristics and prognostic implications of BRAF-mutant NSCLC, the clinical development of BRAF and MEK inhibitors from melanoma to NSCLC, and practical considerations for clinicians involving BRAF mutation screening and the choice of targeted therapy. IMPLICATIONS FOR PRACTICE Personalized medicine has begun to provide substantial benefit to patients with oncogene-driven non-small cell lung cancer (NSCLC). However, treatment options for patients with oncogenic driver mutations lacking targeted treatment strategies remain limited. Direct inhibition of mutant B-Raf proto-oncogene, serine/threonine kinase (BRAF) and/or downstream mitogen-activated protein kinase kinase (MEK) has the potential to change the course of the disease for patients with BRAF-mutant NSCLC, as it has in BRAF-mutant melanoma. Optimization of screening strategies for rare mutations and the choice of appropriate agents on an individual basis will be key to providing timely and successful intervention.
Collapse
Affiliation(s)
- Christina S Baik
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington, USA
| | | | | |
Collapse
|
829
|
Zippel D, Markel G, Shapira-Frommer R, Ben-Betzalel G, Goitein D, Ben-Ami E, Nissan A, Schachter J, Schneebaum S. Perioperative BRAF inhibitors in locally advanced stage III melanoma. J Surg Oncol 2017. [PMID: 28650570 DOI: 10.1002/jso.24744] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND OBJECTIVES Stage III malignant melanoma is a heterogeneous disease where those cases deemed marginally resectable or irresecatble are frequently incurable by surgery alone. Targeted therapy takes advantage of the high incidence of BRAF mutations in melanomas, most notably the V600E mutation. These agents have rarely been used in a neoadjuvant setting prior to surgery. METHODS Thirteen consecutive patients with confirmed BRAFV600E regionally advanced melanoma deemed marginally resectable or irrresectable, were treated with BRAF inhibiting agents, prior to undergoing surgery. The primary outcome measures were a successful resection and pathological response. Disease-free survival was a secondary outcome measure. RESULTS Overall, 12/13 patients showed a marked clinical responsiveness to medical treatment, enabling a macroscopically successful resection in all cases. Four patients had a complete pathological response with no viable tumor evident in the resected specimens and eight patients showed evidence of minimally residual tumor with extensive tumoral necrosis and fibrosis. One patient progressed and died before surgery. At a median follow up of 20 months, 10 patients remain free of disease. CONCLUSIONS Perioperative treatment with BRAF inhibiting agents in BRAFV600E mutated Stage III melanoma patients facilitates surgical resection and affords satisfactory disease free survival.
Collapse
Affiliation(s)
- Douglas Zippel
- Department of Surgery C, Chaim Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gal Markel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Oncology, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Roni Shapira-Frommer
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Oncology, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Guy Ben-Betzalel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Oncology, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - David Goitein
- Department of Surgery C, Chaim Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eytan Ben-Ami
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Oncology, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Aviram Nissan
- Department of Surgery C, Chaim Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jacob Schachter
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Oncology, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Schlomo Schneebaum
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
830
|
de la Cruz-Merino L, Di Guardo L, Grob JJ, Venosa A, Larkin J, McArthur GA, Ribas A, Ascierto PA, Evans JTR, Gomez-Escobar A, Barteselli G, Eng S, Hsu JJ, Uyei A, Dréno B. Clinical features of serous retinopathy observed with cobimetinib in patients with BRAF-mutated melanoma treated in the randomized coBRIM study. J Transl Med 2017; 15:146. [PMID: 28646893 PMCID: PMC5483259 DOI: 10.1186/s12967-017-1246-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/15/2017] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Serous chorioretinopathy has been associated with MEK inhibitors, including cobimetinib. We describe the clinical features of serous retinopathy observed with cobimetinib in patients with BRAF V600-mutated melanoma treated in the Phase III coBRIM study. METHODS In the coBRIM study, 493 patients were treated in two randomly assigned treatment groups: cobimetinib and vemurafenib (n = 247) or vemurafenib (n = 246). All patients underwent prospective ophthalmic examinations at screening, at regular intervals during the study, and whenever ocular symptoms developed. Patients with serous retinopathy were identified in the study database using a group of relevant and synonymous adverse event terms. RESULTS Eighty-six serous retinopathy events were reported in 70 patients (79 events in 63 cobimetinib and vemurafenib-treated patients vs seven events in seven vemurafenib-treated patients). Most patients with serous retinopathy identified by ophthalmic examination had no symptoms or had mild symptoms, among them reduced visual acuity, blurred vision, dyschromatopsia, and photophobia. Serous retinopathy usually occurred early during cobimetinib and vemurafenib treatment; median time to onset was 1.0 month. Most events were managed by observation and continuation of cobimetinib without dose modification and resolved or were resolving by the data cutoff date (19 Sept 2014). CONCLUSIONS Cobimetinib treatment was associated with serous retinopathy in patients with BRAF V600-mutated melanoma. Retinopathy was generally asymptomatic or mild. Periodic ophthalmologic evaluations at regular intervals and at the manifestation of any visual disturbance are recommended to facilitate early detection and resolution of serous retinopathy while patients are taking cobimetinib. Trial Registration Clinicaltrials.gov (NCT01689519). First received: September 18, 2012.
Collapse
Affiliation(s)
- Luis de la Cruz-Merino
- Servicio de Oncología Médica, Hospital Universitario Virgen Macarena, Avenida Doctor Fedriani 3, 41071 Seville, Spain
| | | | | | | | | | - Grant A. McArthur
- Peter MacCallum Cancer Centre, East Melbourne, VIC Australia
- University of Melbourne, Parkville, VIC Australia
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA USA
| | | | | | - Antonio Gomez-Escobar
- Servicio de Oncología Médica, Hospital Universitario Virgen Macarena, Avenida Doctor Fedriani 3, 41071 Seville, Spain
| | | | - Susan Eng
- Genentech, Inc., South San Francisco, CA USA
| | | | - Anne Uyei
- Genentech, Inc., South San Francisco, CA USA
| | | |
Collapse
|
831
|
Keller HR, Zhang X, Li L, Schaider H, Wells JW. Overcoming resistance to targeted therapy with immunotherapy and combination therapy for metastatic melanoma. Oncotarget 2017; 8:75675-75686. [PMID: 29088901 PMCID: PMC5650456 DOI: 10.18632/oncotarget.18523] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/05/2017] [Indexed: 02/07/2023] Open
Abstract
Resistance to targeted therapy is an ongoing problem for the successful treatment of Stage IV metastatic melanoma. For many patients, the use of targeted therapies, such as BRAF kinase inhibitors, were initially promising yet resistance inevitably occurred. Even after combining BRAF kinase inhibitors with MEK pathway inhibitors to offset re-activation of the MAP kinase pathway, resistance is still documented. Similarly, outcomes with immune checkpoint inhibitors as monotherapy were optimistic for some patients without relapse or progression, yet the majority of patients undergoing monotherapy have progressive disease. Will immunotherapy and combination therapy trials overcome resistance in metastatic melanoma? In an effort to treat resistant disease, new clinical trials evaluating the combination of immunotherapy with other therapies, such as kinase inhibitors, adoptive cell therapy, chimeric CD40 ligand to boost costimulation, or a tumor-specific oncolytic virus enhancing granulocyte macrophage colony-stimulating factor (GM-CSF) expression, are currently underway. Updated studies on the mechanisms of resistance, immune escape and options to reinvigorate immune cells support the continued discovery of new and improved forms of therapy.
Collapse
Affiliation(s)
- Hilary R Keller
- The University of Queensland School of Medicine, Ochsner Clinical School, Brisbane, QLD, Australia.,The University of Queensland School of Medicine, Ochsner Clinical School, New Orleans, LA, USA.,The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia.,Laboratory of Translational Cancer Research, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - Xin Zhang
- Laboratory of Translational Cancer Research, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - Li Li
- Laboratory of Translational Cancer Research, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - Helmut Schaider
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - James W Wells
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
832
|
Delord JP, Robert C, Nyakas M, McArthur GA, Kudchakar R, Mahipal A, Yamada Y, Sullivan R, Arance A, Kefford RF, Carlino MS, Hidalgo M, Gomez-Roca C, Michel D, Seroutou A, Aslanis V, Caponigro G, Stuart DD, Moutouh-de Parseval L, Demuth T, Dummer R. Phase I Dose-Escalation and -Expansion Study of the BRAF Inhibitor Encorafenib (LGX818) in Metastatic BRAF-Mutant Melanoma. Clin Cancer Res 2017; 23:5339-5348. [PMID: 28611198 DOI: 10.1158/1078-0432.ccr-16-2923] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 04/07/2017] [Accepted: 06/05/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Encorafenib, a selective BRAF inhibitor (BRAFi), has a pharmacologic profile that is distinct from that of other clinically active BRAFis. We evaluated encorafenib in a phase I study in patients with BRAFi treatment-naïve and pretreated BRAF-mutant melanoma.Experimental Design: The pharmacologic activity of encorafenib was first characterized preclinically. Encorafenib monotherapy was then tested across a range of once-daily (50-700 mg) or twice-daily (75-150 mg) regimens in a phase I, open-label, dose-escalation and -expansion study in adult patients with histologically confirmed advanced/metastatic BRAF-mutant melanoma. Study objectives were to determine the maximum tolerated dose (MTD) and/or recommended phase II dose (RP2D), characterize the safety and tolerability and pharmacokinetic profile, and assess the preliminary antitumor activity of encorafenib.Results: Preclinical data demonstrated that encorafenib inhibited BRAF V600E kinase activity with a prolonged off-rate and suppressed proliferation and tumor growth of BRAF V600E-mutant melanoma models. In the dose-escalation phase, 54 patients (29 BRAFi-pretreated and 25 BRAFi-naïve) were enrolled. Seven patients in the dose-determining set experienced dose-limiting toxicities. Encorafenib at a dose of 300 mg once daily was declared the RP2D. In the expansion phase, the most common all-cause adverse events were nausea (66%), myalgia (63%), and palmar-plantar erythrodysesthesia (54%). In BRAFi-naïve patients, the overall response rate (ORR) and median progression-free survival (mPFS) were 60% and 12.4 months [95% confidence interval (CI), 7.4-not reached (NR)]. In BRAFi-pretreated patients, the ORR and mPFS were 22% and 1.9 months (95% CI, 0.9-3.7).Conclusions: Once-daily dosing of single-agent encorafenib had a distinct tolerability profile and showed varying antitumor activity across BRAFi-pretreated and BRAFi-naïve patients with advanced/metastatic melanoma. Clin Cancer Res; 23(18); 5339-48. ©2017 AACR.
Collapse
Affiliation(s)
| | | | | | - Grant A McArthur
- Peter MacCallum Cancer Centre and the University of Melbourne, Australia
| | | | - Amit Mahipal
- Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | | | - Ryan Sullivan
- Massachusetts General Hospital, Boston, Massachusetts
| | - Ana Arance
- Department of Medical Oncology and Translational Genomics and Targeted Therapeutics in Solid Tumors, Hospital Clínic, Barcelona, Spain
| | - Richard F Kefford
- Crown Princess Mary Cancer Centre Westmead Hospital, Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Macquarie University, Sydney, New South Wales, Australia
| | - Matteo S Carlino
- Crown Princess Mary Cancer Centre Westmead Hospital, Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
| | - Manuel Hidalgo
- Spanish National Cancer Research Centre (CNIO) and Centro Integral Oncologico Clara Campal, Madrid, Spain
| | | | | | | | | | | | - Darrin D Stuart
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | | | | |
Collapse
|
833
|
Eigentler TK, Mühlenbein C, Follmann M, Schadendorf D, Garbe C. S3-Leitlinie Diagnostik, Therapie und Nachsorge des Melanoms - Update 2015/2016, Kurzversion 2.0. J Dtsch Dermatol Ges 2017; 15:e1-e41. [DOI: 10.1111/ddg.13247] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
834
|
Precision medicine for hepatocellular carcinoma: driver mutations and targeted therapy. Oncotarget 2017; 8:55715-55730. [PMID: 28903454 PMCID: PMC5589693 DOI: 10.18632/oncotarget.18382] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/10/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most frequent cause of tumor-related mortality and there are an estimated approximately 850,000 new cases annually. Most HCC patients are diagnosed at middle or advanced stage, losing the opportunity of surgery. The development of HCC is promoted by accumulated diverse genetic mutations, which confer selective growth advantages to tumor cells and are called "driver mutations". The discovery of driver mutations provides a novel precision medicine strategy for late stage HCC, called targeted therapy. In this review, we summarized currently discovered driver mutations and corresponding signaling pathways, made an overview of identification methods of driver mutations and genes, and classified targeted drugs for HCC. The knowledge of mutational landscape deepen our understanding of carcinogenesis and promise future precision medicine for HCC patients.
Collapse
|
835
|
Kirouac DC, Schaefer G, Chan J, Merchant M, Orr C, Huang SMA, Moffat J, Liu L, Gadkar K, Ramanujan S. Clinical responses to ERK inhibition in BRAFV600E-mutant colorectal cancer predicted using a computational model. NPJ Syst Biol Appl 2017; 3:14. [PMID: 28649441 PMCID: PMC5460205 DOI: 10.1038/s41540-017-0016-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/18/2017] [Accepted: 05/04/2017] [Indexed: 12/11/2022] Open
Abstract
Approximately 10% of colorectal cancers harbor BRAFV600E mutations, which constitutively activate the MAPK signaling pathway. We sought to determine whether ERK inhibitor (GDC-0994)-containing regimens may be of clinical benefit to these patients based on data from in vitro (cell line) and in vivo (cell- and patient-derived xenograft) studies of cetuximab (EGFR), vemurafenib (BRAF), cobimetinib (MEK), and GDC-0994 (ERK) combinations. Preclinical data was used to develop a mechanism-based computational model linking cell surface receptor (EGFR) activation, the MAPK signaling pathway, and tumor growth. Clinical predictions of anti-tumor activity were enabled by the use of tumor response data from three Phase 1 clinical trials testing combinations of EGFR, BRAF, and MEK inhibitors. Simulated responses to GDC-0994 monotherapy (overall response rate = 17%) accurately predicted results from a Phase 1 clinical trial regarding the number of responding patients (2/18) and the distribution of tumor size changes ("waterfall plot"). Prospective simulations were then used to evaluate potential drug combinations and predictive biomarkers for increasing responsiveness to MEK/ERK inhibitors in these patients.
Collapse
Affiliation(s)
- Daniel C. Kirouac
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Gabriele Schaefer
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Jocelyn Chan
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Mark Merchant
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Christine Orr
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Shih-Min A. Huang
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - John Moffat
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Lichuan Liu
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Kapil Gadkar
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Saroja Ramanujan
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| |
Collapse
|
836
|
Alqahtani S, Alhefdhi AY, Almalik O, Anwar I, Mahmood R, Mahasin Z, Al-Tweigeri T. Primary oral malignant melanoma metastasis to the brain and breast: A case report and literature review. Oncol Lett 2017; 14:1275-1280. [PMID: 28789341 PMCID: PMC5529946 DOI: 10.3892/ol.2017.6304] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/30/2017] [Indexed: 12/30/2022] Open
Abstract
Primary oral malignant melanoma is a rare tumor, which is estimated to comprise 0.2–8.0% of all melanoma cases. This type of cancer is fairly uncommon, its prognosis is dismal, and it frequently exhibits a biologically aggressive behavior. The common location of primary oral malignant melanoma is the hard palate and maxillary alveolus. In ~85% of cases, the melanoma will metastasize to the liver, lung, bone and brain early in the course of the disease. The present study reports the case of a 50-year-old premenopausal woman who presented with primary oral malignant spindle cell melanoma (T3bN2aM0) and underwent complete surgical resection followed by an adjuvant course of radiation therapy. After 1 year, the patient presented with sudden onset slurred speech, and upon examination, was found to have left-sided hemiparesis and a hard left breast mass. Workup confirmed breast and brain metastasis. The patient developed lung metastasis 4 weeks later and was referred for palliative care.
Collapse
Affiliation(s)
- Saad Alqahtani
- Department of Surgery, College of Medicine, Al Majmaah University, Academic City, Al Majmaah 15341, Kingdom of Saudi Arabia
| | - Amal Y Alhefdhi
- Department of General Surgery, King Faisal Specialist Hospital and Research Centre, Alfaisal University, Riyadh 11211, Kingdom of Saudi Arabia
| | - Osama Almalik
- Department of General Surgery, King Faisal Specialist Hospital and Research Centre, Alfaisal University, Riyadh 11211, Kingdom of Saudi Arabia
| | - Ihab Anwar
- Department of General Surgery, King Faisal Specialist Hospital and Research Centre, Alfaisal University, Riyadh 11211, Kingdom of Saudi Arabia
| | - Rana Mahmood
- Department of Radiation Oncology, Oncology Center, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Kingdom of Saudi Arabia
| | - Zeyad Mahasin
- Department of Otolaryngology, Head and Neck Surgery and Communication, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Kingdom of Saudi Arabia
| | - Taher Al-Tweigeri
- Department of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Kingdom of Saudi Arabia
| |
Collapse
|
837
|
Li D, Li H, Shi H. [Clinical features and prognosis of Langerhans cell histiocytosis in children: an analysis of 34 cases]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:627-631. [PMID: 28606227 PMCID: PMC7390292 DOI: 10.7499/j.issn.1008-8830.2017.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/21/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To investigate the clinical features and prognosis of children with Langerhans cell histiocytosis (LCH). METHODS A retrospective analysis was performed for the clinical data of 34 children with newly diagnosed LCH. RESULTS The 34 children had a median age of 14.5 months (range: 22 d to 60 months). Of all 34 children, 23 were aged 0-2 years and 11 were aged >2 years. There were 17 children in the high-risk group and 17 in the low-risk group. Thirty children received chemotherapy, and the 6-week and 12-month overall response rates were 67% (20/30) and 87% (26/30), respectively. The 3-year overall survival (OS) rate was 86%±6% and the 3-year event-free survival (EFS) rate was 64%±9%. Compared with the low-risk group, the high-risk group had significantly lower 6-week chemotherapy response rate (47% vs 87%; P<0.05), 3-year OS rate (72%±12% vs 100%; P<0.05), and 3-year EFS rate (46%±13% vs 82%±9%; P<0.05). There was no significant difference in the 12-month chemotherapy response rate between the high-risk and low-risk groups (80% vs 93; P>0.05). The high-risk group had a recurrence rate of 27% and a mortality rate of 27%. There were no recurrence or deaths in the low-risk group. CONCLUSIONS Children with LCH have a high overall survival rate, but the high-risk patient has a low 6-week response rate of induction chemotherapy and poor long-term prognosis.
Collapse
Affiliation(s)
- Dan Li
- Department of Pediatric Hematology and Oncology, Chengdu Women and Children's Central Hospital, Chengdu 610091, China.
| | | | | |
Collapse
|
838
|
Zhao DY, Lim KH. Current biologics for treatment of biliary tract cancers. J Gastrointest Oncol 2017; 8:430-440. [PMID: 28736630 PMCID: PMC5506280 DOI: 10.21037/jgo.2017.05.04] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 04/26/2017] [Indexed: 12/16/2022] Open
Abstract
Biliary tract cancers (BTC) is a group of malignancies that arise from the epithelial cells of the biliary tree. These cancers are typically classified by anatomic site of origin: intrahepatic cholangiocarcinoma (IHCC) and extrahepatic cholangiocarcinoma (EHCC), and gallbladder cancer (GBC). To date, complete surgical resection remains the mainstay of treatment especially for earlier stage disease. Unfortunately, most patients present with advanced or metastatic disease, when systemic chemotherapy is the only treatment option. Due to the paucity of effective treatments, BTCs have a dismal prognosis. There is a tremendous need to better understand the disease biology, discover new therapies, and improve clinical outcomes for this challenging disease. Next-generation sequencing has produced a more accurate and detailed picture of the molecular signatures in BTCs. The three BTC histologic subtypes are, in fact, quite molecularly distinct. IHCC commonly contain FGFR2 fusions and IDH 1 and 2 mutations, whereas EHCC and GBC tend to carry mutations in EGFR, HER2, and MAPK pathway. In light of this emerging knowledge, clinical trials have become more biomarker-driven, which allows capturing of subsets of patients that are most likely to respond to certain therapies. Many new and promising targeted therapeutics are currently in the pipeline. Here we review the genetic landscape of BTCs while focusing on new molecular targets and targeted therapeutics currently being investigated in biomarker-driven clinical trials.
Collapse
Affiliation(s)
- Diana Y. Zhao
- Medical Scientist Training Program, Department of Internal Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
839
|
Packer RJ, Pfister S, Bouffet E, Avery R, Bandopadhayay P, Bornhorst M, Bowers DC, Ellison D, Fangusaro J, Foreman N, Fouladi M, Gajjar A, Haas-Kogan D, Hawkins C, Ho CY, Hwang E, Jabado N, Kilburn LB, Lassaletta A, Ligon KL, Massimino M, Meeteren SV, Mueller S, Nicolaides T, Perilongo G, Tabori U, Vezina G, Warren K, Witt O, Zhu Y, Jones DT, Kieran M. Pediatric low-grade gliomas: implications of the biologic era. Neuro Oncol 2017; 19:750-761. [PMID: 27683733 PMCID: PMC5464436 DOI: 10.1093/neuonc/now209] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
For the past decade, it has been recognized that pediatric low-grade gliomas (LGGs) and glial-neuronal tumors carry distinct molecular alterations with resultant aberrant intracellular signaling in the Ras-mitogen-activated protein kinase pathway. The conclusions and recommendations of a consensus conference of how best to integrate the growing body of molecular genetic information into tumor classifications and, more importantly, for future treatment of pediatric LGGs are summarized here. There is uniform agreement that molecular characterization must be incorporated into classification and is increasingly critical for appropriate management. Molecular-targeted therapies should be integrated expeditiously, but also carefully into the management of these tumors and success measured not only by radiographic responses or stability, but also by functional outcomes. These trials need to be carried out with the caveat that the long-term impact of molecularly targeted therapy on the developing nervous system, especially with long duration treatment, is essentially unknown.
Collapse
Affiliation(s)
- Roger J Packer
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
| | - Stephan Pfister
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Eric Bouffet
- Paediatric Neuro-Oncology Program, Research Institute and The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Robert Avery
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
| | - Pratiti Bandopadhayay
- Department of Pediatrics, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
| | - Miriam Bornhorst
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - Daniel C Bowers
- Department of Pediatrics, UT Southwestern Medical School, Dallas, Texas, USA
| | - David Ellison
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee. USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jason Fangusaro
- Ann and Robert H. Lurie Children's Hospital of Chicago Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, University of Colorado, Aurora, Colorado, USA
| | - Nicholas Foreman
- Northwestern Feinberg School of Medicine, Chicago, Illinois; Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Maryam Fouladi
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
| | - Amar Gajjar
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
| | - Cynthia Hawkins
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee. USA
| | - Cheng-Ying Ho
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Eugene Hwang
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - Nada Jabado
- Ann and Robert H. Lurie Children's Hospital of Chicago Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, University of Colorado, Aurora, Colorado, USA
| | - Lindsay B Kilburn
- Brain Tumor Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - Alvaro Lassaletta
- Northwestern Feinberg School of Medicine, Chicago, Illinois; Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Keith L Ligon
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
| | - Maura Massimino
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
| | | | - Sabine Mueller
- Department of Neurology, Pediatrics and Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | - Theo Nicolaides
- Department of Neurology, Pediatrics and Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | - Giorgio Perilongo
- Department of Woman's and Child's Health, University of Padua, Padua, Italy
| | - Uri Tabori
- Division of Haematology/Oncology, Research Institute and The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Gilbert Vezina
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
- Division of Neuroradiology, Washington, District of Columbia, USA
| | - Katherine Warren
- National Cancer Institute, Pediatric Oncology and Neuro-Oncology Branches, Bethesda, Maryland, USA
| | - Olaf Witt
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yuan Zhu
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - David T Jones
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mark Kieran
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
| |
Collapse
|
840
|
Du L, Che Z, Wang-Gillam A. Promising therapeutics of gastrointestinal cancers in clinical trials. J Gastrointest Oncol 2017; 8:524-533. [PMID: 28736639 DOI: 10.21037/jgo.2017.01.08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Many novel therapeutics are being developed for patients with cancers along the gastrointestinal (GI) tract. These emerging agents are frequently classified by their biological targets such as tumor growth pathways, tumor metabolism, microenvironment, etc. Some agents targeting cancer growth pathways are based on existing clinically validated therapeutic targets, such as regorafenib for hepatocellular carcinoma (HCC), while other agents focus on newly identified targets, such as FGFR fusions in cholangiocarcinoma. Drugs modifying the immunosuppressive tumor microenvironment have emerged as an attractive area of clinical investigation. Moreover, drugs targeting the stem-cell like qualities of cancer and the tight junction protein claudin 18.2 have generated quite a lot of excitement in the field. In this paper, we will systemically review the recent promising agents and therapeutic strategies in GI cancers.
Collapse
Affiliation(s)
- Lingling Du
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | | | - Andrea Wang-Gillam
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
841
|
Griewank KG, Schilling B. Next-Generation Sequencing to Guide Treatment of Advanced Melanoma. Am J Clin Dermatol 2017; 18:303-310. [PMID: 28229402 DOI: 10.1007/s40257-017-0260-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Next-generation sequencing (NGS) has provided significant insights into the pathogenesis of human malignancies. In advanced melanoma, two therapeutic avenues have appeared and have immediately become the standard of care, i.e. targeted therapy with small molecule inhibitors, and immune checkpoint blockade. Sequencing has always been essential for determining which patients may benefit from targeted therapies (e.g. the presence of BRAF mutations). While sequencing does not currently help recognize which patients might benefit from immune checkpoint blockade, recent data suggest that this may change. Multiple studies have identified tumor mutation profiles associated with patients benefiting from immune checkpoint blockade therapy. These findings suggest comprehensive tumor sequencing may become a critical step for predicting therapy responses to all systemic therapies. In this review, the current and potential future impact of NGS on treatment decisions in advanced melanoma will be summarized and discussed.
Collapse
Affiliation(s)
- Klaus G Griewank
- Department of Dermatology, University Hospital, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Hufelandstr. 55, Essen, 45147, Germany.
- Dermatopathologie bei Mainz, Bahnhofstr. 2b, Nieder-Olm, 55268, Germany.
| | - Bastian Schilling
- Department of Dermatology, University Hospital, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Hufelandstr. 55, Essen, 45147, Germany.
- Department of Dermatology, Venerology and Allergology, University Hospital Würzburg, Josef-Schneider Straße 2, Würzburg, 97080, Germany.
| |
Collapse
|
842
|
Chatterjee S, Huang EHB, Christie I, Burns TF. Reactivation of the p90RSK-CDC25C Pathway Leads to Bypass of the Ganetespib-Induced G 2-M Arrest and Mediates Acquired Resistance to Ganetespib in KRAS-Mutant NSCLC. Mol Cancer Ther 2017; 16:1658-1668. [PMID: 28566436 DOI: 10.1158/1535-7163.mct-17-0114] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/08/2017] [Accepted: 05/12/2017] [Indexed: 11/16/2022]
Abstract
A subset of non-small cell lung cancers (NSCLC) are dependent upon oncogenic driver mutations, including the most frequently observed driver mutant KRAS, which is associated with a poor prognosis. As direct RAS targeting in the clinic has been unsuccessful to date, use of Hsp90 inhibitors appeared to be a promising therapy for KRAS-mutant NSCLC; however, limited clinical efficacy was observed due to rapid resistance. Furthermore, the combination of the Hsp90 inhibitor (Hsp90i), ganetespib, and docetaxel was tested in a phase III clinical trial and failed to demonstrate benefit. Here, we investigated the mechanism(s) of resistance to ganetespib and explored why the combination with docetaxel failed in the clinic. We have not only identified a critical role for the bypass of the G2-M cell-cycle checkpoint as a mechanism of ganetespib resistance (GR) but have also found that GR leads to cross-resistance to docetaxel. Reactivation of p90RSK and its downstream target, CDC25C, was critical for GR and mediated the bypass of a G2-M arrest. Overexpression of either p90RSK or CDC25C lead to bypass of G2-M arrest and induced ganetespib resistance in vitro and in vivo Moreover, resistance was dependent on p90RSK/CDC25C signaling, as synthetic lethality to ERK1/2, p90RSK, or CDC25C inhibitors was observed. Importantly, the combination of ganetespib and p90RSK or CDC25C inhibitors was highly efficacious in parental cells. These studies provide a way forward for Hsp90 inhibitors through the development of novel rationally designed Hsp90 inhibitor combinations that may prevent or overcome resistance to Hsp90i. Mol Cancer Ther; 16(8); 1658-68. ©2017 AACR.
Collapse
Affiliation(s)
- Suman Chatterjee
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Eric H-B Huang
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Ian Christie
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Timothy F Burns
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.
| |
Collapse
|
843
|
Kim S, Kim HT, Suh HS. Combination therapy of BRAF inhibitors for advanced melanoma with BRAF V600 mutation: a systematic review and meta-analysis. J DERMATOL TREAT 2017; 29:314-321. [PMID: 28504036 DOI: 10.1080/09546634.2017.1330530] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Although BRAF inhibitors have been used to treat advanced melanoma with BRAF mutation, combination strategies are suggested due to acquired resistance to BRAF inhibitors. OBJECTIVE To assess the efficacy of BRAF inhibitor-based combination therapy for the treatment of advanced melanoma with BRAF mutation. METHODS We conducted a systematic review and meta-analysis of studies that compared BRAF inhibitor-based combination therapy with BRAF inhibitor monotherapy. We searched MEDLINE, EMBASE, the Cochrane Library and relevant conference proceedings. The random-effects inverse variance and Mantel-Haenszel methods were used to pool the results. RESULTS Four randomized controlled trials and one cohort study were identified. A combination therapy with BRAF inhibitors and MEK inhibitors was used in all studies. The combined hazard ratios of overall survival (OS) and progression-free survival (PFS) comparing combination therapy with monotherapy were 0.70 [95% confidence interval (CI) 0.62-0.78] and 0.59 (95% CI 0.55-0.63), respectively. The combined risk ratio of objective response rate (ORR) was 1.30 (95% CI 1.20-1.40), which meant more patients achieved complete/partial responses in combination therapy group than those in the monotherapy group. CONCLUSIONS Combination therapy with BRAF inhibitors and MEK inhibitors significantly improved OS, PFS, and ORR in patients with advanced melanoma with BRAF mutation.
Collapse
Affiliation(s)
- Siin Kim
- a College of Pharmacy , Pusan National University , Busandaehak-ro 63 beon-gil, Geumjeong-gu , Busan , South Korea
| | - Hyung Tae Kim
- a College of Pharmacy , Pusan National University , Busandaehak-ro 63 beon-gil, Geumjeong-gu , Busan , South Korea
| | - Hae Sun Suh
- a College of Pharmacy , Pusan National University , Busandaehak-ro 63 beon-gil, Geumjeong-gu , Busan , South Korea
| |
Collapse
|
844
|
Xue G, Kohler R, Tang F, Hynx D, Wang Y, Orso F, Prêtre V, Ritschard R, Hirschmann P, Cron P, Roloff T, Dummer R, Mandalà M, Bichet S, Genoud C, Meyer AG, Muraro MG, Spagnoli GC, Taverna D, Rüegg C, Merghoub T, Massi D, Tang H, Levesque MP, Dirnhofer S, Zippelius A, Hemmings BA, Wicki A. mTORC1/autophagy-regulated MerTK in mutant BRAFV600 melanoma with acquired resistance to BRAF inhibition. Oncotarget 2017; 8:69204-69218. [PMID: 29050198 PMCID: PMC5642473 DOI: 10.18632/oncotarget.18213] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 05/17/2017] [Indexed: 12/19/2022] Open
Abstract
BRAF inhibitors (BRAFi) and the combination therapy of BRAF and MEK inhibitors (MEKi) were recently approved for therapy of metastatic melanomas harbouring the oncogenic BRAFV600 mutation. Although these therapies have shown pronounced therapeutic efficacy, the limited durability of the response indicates an acquired drug resistance that still remains mechanistically poorly understood at the molecular level. We conducted transcriptome gene profiling in BRAFi-treated melanoma cells and identified that Mer tyrosine kinase (MerTK) is specifically upregulated. MerTK overexpression was demonstrated not only in melanomas resistant to BRAFi monotherapy (5 out of 10 samples from melanoma patients) but also in melanoma resistant to BRAFi+MEKi (1 out of 3), although MEKi alone does not affect MerTK. Mechanistically, BRAFi-induced activation of Zeb2 stimulates MerTK in BRAFV600 melanoma through mTORC1-triggered activation of autophagy. Co-targeting MerTK and BRAFV600 significantly reduced tumour burden in xenografted mice, which was pheno-copied by co-inhibition of autophagy and mutant BRAFV600.
Collapse
Affiliation(s)
- Gongda Xue
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Reto Kohler
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Fengyuan Tang
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Debby Hynx
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Yuhua Wang
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Francesca Orso
- Molecular Biotechnology Center and Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Vincent Prêtre
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Reto Ritschard
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | | | - Peter Cron
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Tim Roloff
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Mario Mandalà
- Unit of Clinical and Translational Research, Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Sandrine Bichet
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Christel Genoud
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Alexandra G Meyer
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Manuele G Muraro
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Giulio C Spagnoli
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Daniela Taverna
- Molecular Biotechnology Center and Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Curzio Rüegg
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Taha Merghoub
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Daniela Massi
- Division of Pathological Anatomy, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Huifang Tang
- Department of Pharmacology, Zhejiang University, School of Basic Medical Sciences, Hangzhou, China
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | | | - Alfred Zippelius
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Brian A Hemmings
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Andreas Wicki
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
845
|
Ma J, Guo W, Li C. Ubiquitination in melanoma pathogenesis and treatment. Cancer Med 2017; 6:1362-1377. [PMID: 28544818 PMCID: PMC5463089 DOI: 10.1002/cam4.1069] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 12/13/2022] Open
Abstract
Melanoma is one of the most aggressive skin cancers with fiercely increasing incidence and mortality. Since the progressive understanding of the mutational landscape and immunologic pathogenic factors in melanoma, the targeted therapy and immunotherapy have been recently established and gained unprecedented improvements for melanoma treatment. However, the prognosis of melanoma patients remains unoptimistic mainly due to the resistance and nonresponse to current available drugs. Ubiquitination is a posttranslational modification which plays crucial roles in diverse cellular biological activities and participates in the pathogenesis of various cancers, including melanoma. Through the regulation of multiple tumor promoters and suppressors, ubiquitination is emerging as the key contributor and therefore a potential therapeutic target for melanoma. Herein, we summarize the current understanding of ubiquitination in melanoma, from mechanistic insights to clinical progress, and discuss the prospect of ubiquitination modification in melanoma treatment.
Collapse
Affiliation(s)
- Jinyuan Ma
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weinan Guo
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
846
|
Rose AAN, Biondini M, Curiel R, Siegel PM. Targeting GPNMB with glembatumumab vedotin: Current developments and future opportunities for the treatment of cancer. Pharmacol Ther 2017; 179:127-141. [PMID: 28546082 DOI: 10.1016/j.pharmthera.2017.05.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
GPNMB has emerged as an immunomodulator and an important positive mediator of tumor progression and metastasis in numerous solid cancers. Tumor intrinsic GPNMB-mediated effects on cellular signaling, coupled with the ability of GPNMB to influence the primary tumor and metastatic microenvironments in a non-cell autonomous fashion, combine to augment malignant cancer phenotypes. In addition, GPNMB is often overexpressed in a variety of cancers, making it an attractive therapeutic target. In this regard, glembatumumab vedotin, an antibody-drug conjugate (ADC) that targets GPNMB, is currently in clinical trials as a single agent in multiple cancers. In this review, we will describe the physiological functions of GPNMB in normal tissues and summarize the processes through which GPNMB augments tumor growth and metastasis. We will review the pre-clinical and clinical development of glembatumumab vedotin, evaluate on-going clinical trials, explore emerging opportunities for this agent in new disease indications and discuss exciting possibilities for this ADC in the context of combination therapies.
Collapse
Affiliation(s)
- April A N Rose
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada; Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Marco Biondini
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada; Department of Medicine, McGill University, Montréal, Québec, Canada
| | | | - Peter M Siegel
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada; Department of Medicine, McGill University, Montréal, Québec, Canada; Department of Biochemistry, McGill University, Montréal, Québec, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada; Department of Oncology, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
847
|
Lim SY, Menzies AM, Rizos H. Mechanisms and strategies to overcome resistance to molecularly targeted therapy for melanoma. Cancer 2017; 123:2118-2129. [DOI: 10.1002/cncr.30435] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 09/22/2016] [Accepted: 09/22/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Su Yin Lim
- Faculty of Medicine and Health Sciences; Macquarie University; Sydney New South Wales Australia
- Melanoma Institute Australia; Sydney New South Wales Australia
| | - Alexander M. Menzies
- Melanoma Institute Australia; Sydney New South Wales Australia
- Sydney Medical School; University of Sydney; Sydney New South Wales Australia
- Royal North Shore Hospital; Sydney New South Wales Australia
| | - Helen Rizos
- Faculty of Medicine and Health Sciences; Macquarie University; Sydney New South Wales Australia
- Melanoma Institute Australia; Sydney New South Wales Australia
| |
Collapse
|
848
|
Daud A, Tsai K. Management of Treatment-Related Adverse Events with Agents Targeting the MAPK Pathway in Patients with Metastatic Melanoma. Oncologist 2017; 22:823-833. [PMID: 28526719 DOI: 10.1634/theoncologist.2016-0456] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 03/08/2017] [Indexed: 01/25/2023] Open
Abstract
Tremendous progress has been made in the clinical landscape of advanced-stage BRAF V600-mutant melanoma treatment over the past 5 years. Targeted therapies that inhibit specific steps of the mitogen-activated protein kinase pathway have been shown to provide significant overall treatment benefit in patients with this difficult-to-treat disease. Combination therapy with BRAF and MEK inhibitors (dabrafenib plus trametinib or vemurafenib plus cobimetinib, respectively) has become standard of care. These agents are administered until disease progression or unacceptable toxicity occurs; thus, some patients may remain on maintenance therapy for an extended period of time, while toxicities may result in early discontinuation in other patients. Because the goal of treatment is to prolong survival with minimal impairment of quality of life, drug-related adverse events (AEs) require prompt management to ensure that patients derive the best possible benefit from therapy. Proper management depends on an understanding of which AEs are most likely BRAF or MEK inhibitor associated, thus providing a rationale for dose modification of the appropriate drug. Additionally, the unique safety profile of the chosen regimen may influence patient selection and monitoring. This review discusses the toxicity profiles of these agents, with a focus on the most commonly reported and serious AEs. Here, we offer practical guidance derived from our clinical experience for the optimal management of key drug-related AEs. IMPLICATIONS FOR PRACTICE Targeted therapy with BRAF plus MEK inhibitors has become the standard of care for patients with advanced-stage BRAF V600-mutant metastatic melanoma. To provide optimal therapeutic benefit to patients, clinicians need a keen understanding of the toxicity profiles of these drugs. Prompt identification and an understanding of which adverse events are most likely BRAF or MEK inhibitor associated provide a rationale for appropriate therapy adjustments. Practical recommendations derived from clinical experience are provided for management of key drug-related toxicities.
Collapse
Affiliation(s)
- Adil Daud
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Katy Tsai
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
849
|
Ross KC, Andrews AJ, Marion CD, Yen TJ, Bhattacharjee V. Identification of the Serine Biosynthesis Pathway as a Critical Component of BRAF Inhibitor Resistance of Melanoma, Pancreatic, and Non-Small Cell Lung Cancer Cells. Mol Cancer Ther 2017; 16:1596-1609. [PMID: 28500236 DOI: 10.1158/1535-7163.mct-16-0798] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 04/04/2017] [Accepted: 04/24/2017] [Indexed: 12/12/2022]
Abstract
Metastatic melanoma cells commonly acquire resistance to BRAF V600E inhibitors (BRAFi). In this study, we identified serine biosynthesis as a critical mechanism of resistance. Proteomic assays revealed differential protein expression of serine biosynthetic enzymes PHGDH, PSPH, and PSAT1 following vemurafenib (BRAFi) treatment in sensitive versus acquired resistant melanoma cells. Ablation of PHGDH via siRNA sensitized acquired resistant cells to vemurafenib. Inhibiting the folate cycle, directly downstream of serine synthesis, with methotrexate also displayed similar sensitization. Using the DNA-damaging drug gemcitabine, we show that gemcitabine pretreatment sensitized resistant melanoma cells to BRAFis vemurafenib and dabrafenib. We extended our findings to BRAF WT tumor cell lines that are intrinsically resistant to vemurafenib and dabrafenib. Pretreatment of pancreatic cancer and non-small cell lung cancer cell lines with sublethal doses of 50 and 5 nmol/L of gemcitabine, respectively, enhanced killing by both vemurafenib and dabrafenib. The novel aspects of this study are the direct identification of serine biosynthesis as a critical mechanism of BRAF V600E inhibitor resistance and the first successful example of using gemcitabine + BRAFis in combination to kill previously drug-resistant cancer cells, creating the translational potential of pretreatment with gemcitabine prior to BRAFi treatment of tumor cells to reverse resistance within the mutational profile and the WT. Mol Cancer Ther; 16(8); 1596-609. ©2017 AACR.
Collapse
Affiliation(s)
| | - Andrew J Andrews
- Evol Science, Philadelphia, Pennsylvania.,Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | | | | |
Collapse
|
850
|
Kis O, Kaedbey R, Chow S, Danesh A, Dowar M, Li T, Li Z, Liu J, Mansour M, Masih-Khan E, Zhang T, Bratman SV, Oza AM, Kamel-Reid S, Trudel S, Pugh TJ. Circulating tumour DNA sequence analysis as an alternative to multiple myeloma bone marrow aspirates. Nat Commun 2017; 8:15086. [PMID: 28492226 PMCID: PMC5437268 DOI: 10.1038/ncomms15086] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 02/28/2017] [Indexed: 12/24/2022] Open
Abstract
The requirement for bone-marrow aspirates for genomic profiling of multiple myeloma poses an obstacle to enrolment and retention of patients in clinical trials. We evaluated whether circulating cell-free DNA (cfDNA) analysis is comparable to molecular profiling of myeloma using bone-marrow tumour cells. We report here a hybrid-capture-based Liquid Biopsy Sequencing (LB-Seq) method used to sequence all protein-coding exons of KRAS, NRAS, BRAF, EGFR and PIK3CA in 64 cfDNA specimens from 53 myeloma patients to >20,000 × median coverage. This method includes a variant filtering algorithm that enables detection of tumour-derived fragments present in cfDNA at allele frequencies as low as 0.25% (median 3.2%, range 0.25–46%). Using LB-Seq analysis of 48 cfDNA specimens with matched bone-marrow data, we detect 49/51 likely somatic mutations, with subclonal hierarchies reflecting tumour profiling (96% concordance), and four additional mutations likely missed by bone-marrow testing (>98% specificity). Overall, LB-Seq is a high fidelity adjunct to genetic profiling of bone-marrow in multiple myeloma. Genetic profiling of multiple myeloma requires painful bone marrow biopsies. Here, the authors develop an alternative non-invasive method for sequencing of five oncogenes in circulating cell-free DNA from myeloma patients, demonstrating 96% concordance with bone marrow tumour profiling results.
Collapse
Affiliation(s)
- Olena Kis
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Rayan Kaedbey
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Signy Chow
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Arnavaz Danesh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Mark Dowar
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Tiantian Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Zhihua Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Jessica Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Mark Mansour
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Esther Masih-Khan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Tong Zhang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Scott V Bratman
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Amit M Oza
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Suzanne Kamel-Reid
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Suzanne Trudel
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|