651
|
Abstract
PURPOSE OF REVIEW BRAF/MEK inhibitor has changed the treatment landscape in patients with advanced and metastatic melanoma with prolonged overall survival and progression-free survival. Since three treatment combinations exist with similar efficacy therapy decisions are often made based on the side effect profile. Additionally, on-target side effects or class effects have to be properly managed to ensure treatment adherence. RECENT FINDINGS Sequential treatment with BRAF/MEK inhibition and immunotherapy might increase toxicity with a sepsis-like syndrome and triple therapy with concomitant BRAF/MEK inhibition and anti-PD1/PD-L1 antibody therapy induces severe side effects in the vast majority of patients. SUMMARY Toxicity of combination therapy with BRAF/MEK inhibitors is generally manageable, reversible and infrequently associated with treatment discontinuation. In case of persisting off-target effects the change to another combination therapy can resolve side effects.
Collapse
Affiliation(s)
- Alvaro Moreira
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai
- The Kimberly and Eric J. Waldman Department of Dermatology at Mount Sinai, New York, NY, USA
| | - Céleste Lebbé
- Université de Paris, AP-HP Dermatology, INSERM U976, Saint Louis Hospital, Paris, France
| | - Lucie Heinzerling
- Department of Dermatology, Universitätsklinikum München (LMU), Munich, Germany
- Department of Dermatology, Universitätsklinikum Erlangen, Germany and Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
| |
Collapse
|
652
|
Houten R, Greenhalgh J, Mahon J, Nevitt S, Beale S, Boland A, Lambe T, Dundar Y, Kotas E, McEntee J. Encorafenib with Binimetinib for the Treatment of Patients with BRAF V600 Mutation-Positive Unresectable or Metastatic Melanoma: An Evidence Review Group Perspective of a NICE Single Technology Appraisal. PHARMACOECONOMICS - OPEN 2021; 5:13-22. [PMID: 32291725 PMCID: PMC7895893 DOI: 10.1007/s41669-020-00206-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
As part of the Single Technology Appraisal process, the National Institute for Health and Care Excellence (NICE) invited Pierre Fabre to submit evidence for the clinical and cost-effectiveness of encorafenib with binimetinib (Enco + Bini) versus dabrafenib with trametinib (Dab + Tram) as a first-line treatment for advanced (unresectable or metastatic) BRAF V600 mutation-positive melanoma. The Liverpool Reviews and Implementation Group at the University of Liverpool was commissioned as the Evidence Review Group (ERG). This article summarises the ERG's review of the company's evidence submission (CS), and the Appraisal Committee's (AC's) final decision. The main clinical evidence in the CS was derived from the COLUMBUS trial and focused on the efficacy of Enco + Bini (encorafenib 450 mg per day plus binimetinib 45 mg twice daily) compared to vemurafenib. The company conducted network meta-analyses (NMAs) to indirectly estimate the relative effects of progression-free survival (PFS), overall survival (OS), adverse events (AEs) and health-related quality of life (HRQoL) for Enco + Bini versus Dab + Tram. None of the results from the NMAs demonstrated a statistically significant difference between the treatment regimens for any outcomes. The ERG advised caution when interpreting the results from the company's NMAs due to limitations relating to the methods. The ERG considered that use of the OS and PFS hazard ratios (HRs) generated by the company's NMAs to model the relative effectiveness of Enco + Bini versus Dab + Tram in the company model was inappropriate as these estimates were not statistically significantly different. The ERG amended the company's economic model to include estimates of equivalent efficacy, safety and HRQoL for Enco + Bini and Dab + Tram. The ERG considered use of different estimates of relative dose intensity to be inappropriate and used the same estimate for both drug combinations. The ERG also concluded that as only the prices of drug combinations were different, a cost comparison was an appropriate method of economic analysis. Using this approach (combined with confidential discounted drug prices for Enco + Bini and Dab + Tram), treatment with Enco + Bini was more cost effective than treatment with Dab + Tram. The AC raised concerns that an absence of evidence of a difference in outcomes between Enco + Bini and Dab + Tram did not constitute evidence of absence. However, as the numerical differences in outcomes generated by the company's networks were small, the AC did not have a preferred approach and considered that both the company's and the ERG's methods of incorporating outcome estimates into the economic model were suitable for decision making. The NICE AC recommended Enco + Bini as a first-line treatment for unresectable or metastatic melanoma with a BRAF V600 mutation.
Collapse
Affiliation(s)
- Rachel Houten
- Liverpool Reviews and Implementation Group, University of Liverpool, Whelan Building, Liverpool, L69 3GB UK
| | - Janette Greenhalgh
- Liverpool Reviews and Implementation Group, University of Liverpool, Whelan Building, Liverpool, L69 3GB UK
| | - James Mahon
- Coldingham Analytical Services, Berwick-upon-Tweed, UK
| | - Sarah Nevitt
- Liverpool Reviews and Implementation Group, University of Liverpool, Whelan Building, Liverpool, L69 3GB UK
| | - Sophie Beale
- Liverpool Reviews and Implementation Group, University of Liverpool, Whelan Building, Liverpool, L69 3GB UK
| | - Angela Boland
- Liverpool Reviews and Implementation Group, University of Liverpool, Whelan Building, Liverpool, L69 3GB UK
| | - Tosin Lambe
- Liverpool Reviews and Implementation Group, University of Liverpool, Whelan Building, Liverpool, L69 3GB UK
| | - Yenal Dundar
- Liverpool Reviews and Implementation Group, University of Liverpool, Whelan Building, Liverpool, L69 3GB UK
| | - Eleanor Kotas
- Liverpool Reviews and Implementation Group, University of Liverpool, Whelan Building, Liverpool, L69 3GB UK
| | - Joanne McEntee
- North West Medicines Information Centre, Liverpool, L69 3GF UK
| |
Collapse
|
653
|
Gregory TA, Chumbley LB, Henson JW, Theeler BJ. Adult pilocytic astrocytoma in the molecular era: a comprehensive review. CNS Oncol 2021; 10:CNS68. [PMID: 33448230 PMCID: PMC7962176 DOI: 10.2217/cns-2020-0027] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
Adult pilocytic astrocytoma (PA) is less prevalent than pediatric PA and is associated with a worse prognosis. In a literature review, we found that 88.3% of the molecular alterations in adult PA are associated with MAPK pathway dysregulation. The most common alterations are fusions of BRAF. Understanding of the mechanisms underlying this pathway has evolved substantially, heralding advancements in specific targeted therapy. Here, we review clinical and molecular features of adult PA, characteristics predicting aggressive behavior and approaches to standard and investigational therapies. We highlight epigenetic profiling and integrated diagnosis as an essential component of classifying PA.
Collapse
Affiliation(s)
- Timothy A Gregory
- Department of Medicine, Neurology, Madigan Army Medical Center, Tacoma, WA 98431, USA
| | - Lyndon B Chumbley
- University of Rochester School of Medicine & Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - John W Henson
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Medical Center, Seattle, WA 98122, USA
| | - Brett J Theeler
- Department of Neurology, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
- John P Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
- NIH/NCI Neuro-Oncology Branch, Bethesda, MD 20892-8202, USA
| |
Collapse
|
654
|
Hicks HM, McKenna LR, Espinoza VL, Pozdeyev N, Pike LA, Sams SB, LaBarbera D, Reigan P, Raeburn CD, E Schweppe R. Inhibition of BRAF and ERK1/2 has synergistic effects on thyroid cancer growth in vitro and in vivo. Mol Carcinog 2021; 60:201-212. [PMID: 33595872 DOI: 10.1002/mc.23284] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 11/09/2022]
Abstract
Mutations in the BRAF gene are highly prevalent in thyroid cancer. However, the response rate of thyroid tumors to BRAF-directed therapies has been mixed. Increasingly, combination therapies inhibiting the MAPK pathway at multiple nodes have shown promise. Recently developed ERK1/2 inhibitors are of interest for use in combination therapies as they have the advantage of inhibiting the most downstream node of the MAPK pathway, therefore preventing pathway reactivation. Here, we examined the effect of combined BRAF inhibition (dabrafenib) and ERK1/2 inhibition (SCH772984) on the growth and survival of a panel of BRAF-mutant thyroid cancer cell lines using in vitro and in vivo approaches. We found that resistance due to MAPK pathway reactivation occurs quickly with single-agent BRAF inhibition, but can be prevented with combined BRAF and ERK1/2 inhibition. Combined inhibition also results in synergistic growth inhibition, decreased clonogenic survival, and enhanced induction of apoptosis in a subset of BRAF-mutant thyroid cancer cells. Finally, combined inhibition of BRAF and ERK1/2 results in enhanced inhibition of tumor growth in an anaplastic thyroid cancer in vivo model. These results provide key rationale to pursue combined BRAF and ERK1/2 inhibition as an alternative therapeutic strategy for BRAF-mutant advanced thyroid cancer patients.
Collapse
Affiliation(s)
- Hannah M Hicks
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Logan R McKenna
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Surgery, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Veronica L Espinoza
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Nikita Pozdeyev
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Bioinformatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laura A Pike
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sharon B Sams
- Department of Pathology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Daniel LaBarbera
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Philip Reigan
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Christopher D Raeburn
- Department of Surgery, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rebecca E Schweppe
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pathology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
655
|
Halle BR, Johnson DB. Defining and Targeting BRAF Mutations in Solid Tumors. Curr Treat Options Oncol 2021; 22:30. [PMID: 33641072 DOI: 10.1007/s11864-021-00827-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2021] [Indexed: 12/23/2022]
Abstract
OPINION STATEMENT BRAF mutations are present in up to 8% of human cancers, and comprise a viable therapeutic target in many patients harboring these mutations. Specific BRAF-targeted therapies, such as vemurafenib, dabrafenib, and encorafenib, have transformed treatment of many BRAF-mutated cancers, producing meaningful clinical benefit with more tolerable safety profiles compared to prior standard-of-care treatments. BRAF inhibitors were first approved for use in metastatic melanoma, although resistance almost always limited their long-term effectiveness. Combination therapy with BRAF and MEK inhibitors has proven effective in delaying the onset of resistance, and produces additional clinical benefit across cancers. Although not promising initially in treatment of BRAF-mutated colorectal carcinoma, BRAF inhibitors in colorectal cancer were successfully combined with EGFR inhibitors, resulting in significant treatment response. Refining the use of BRAF and MEK inhibitors in less common tumor types (and for non-V600 mutations) and delaying the development of resistance remain pertinent future considerations in treating BRAF-mutated cancers. In this review, we will discuss the prevalence of BRAF mutations across human cancers and evidence on the efficacy and safety of current management strategies for various BRAF-mutant solid tumors.
Collapse
Affiliation(s)
- Briana R Halle
- Vanderbilt University School of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, 777 PRB, 2220 Pierce Ave., Nashville, TN, 37232, USA.
| |
Collapse
|
656
|
Sioufi K, Das S, Say EAT. A Case of Extracellular Signal-Regulated Kinase Inhibitor-Associated Retinopathy. JAMA Ophthalmol 2021; 138:1002-1004. [PMID: 32729890 DOI: 10.1001/jamaophthalmol.2020.2716] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Kareem Sioufi
- Retina Service, Storm Eye Institute, Medical University of South Carolina, Charleston
| | - Sudeep Das
- Retina Service, Storm Eye Institute, Medical University of South Carolina, Charleston
| | - Emil Anthony T Say
- Retina Service, Storm Eye Institute, Medical University of South Carolina, Charleston
| |
Collapse
|
657
|
Chen X, Luo L, Shen C, Ding P, Luo J. An In Silico Method for Predicting Drug Synergy Based on Multitask Learning. Interdiscip Sci 2021; 13:299-311. [PMID: 33611781 DOI: 10.1007/s12539-021-00422-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/29/2021] [Accepted: 02/07/2021] [Indexed: 12/20/2022]
Abstract
To make better use of all kinds of knowledge to predict drug synergy, it is crucial to successfully establish a drug synergy prediction model and leverage the reconstruction of sparse known drug targets. Therefore, we present an in silico method that predicts the synergy scores of drug pairs based on multitask learning (DSML) that could fuse drug targets, protein-protein interactions, anatomical therapeutic chemical codes, a priori knowledge of drug combinations. To simultaneously reconstruct drug-target protein interactions and synergistic drug combinations, DSML benefits indirectly from the associations with relation through proteins. In cross-validation experiments, DSML improved the ability to predict drug synergy. Moreover, the reconstruction of drug-target interactions and the incorporation of multisource knowledge significantly improved drug combination predictions by a large margin. The potential drug combinations predicted by DSML demonstrate its ability to predict drug synergy.
Collapse
Affiliation(s)
- Xin Chen
- School of Computer Science, University of South China, Hengyang, 421001, Hunan, China
| | - Lingyun Luo
- School of Computer Science, University of South China, Hengyang, 421001, Hunan, China.,Hunan Medical Big Data International Sci.&Tech. Innovation Cooperation Base, Hengyang, 421000, Hunan, China
| | - Cong Shen
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China
| | - Pingjian Ding
- School of Computer Science, University of South China, Hengyang, 421001, Hunan, China. .,Hunan Medical Big Data International Sci.&Tech. Innovation Cooperation Base, Hengyang, 421000, Hunan, China.
| | - Jiawei Luo
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, Hunan, China
| |
Collapse
|
658
|
Mukai K, Kamata M, Miyazaki M, Nagata M, Fukaya S, Hayashi K, Fukuyasu A, Ishikawa T, Ohnishi T, Tada Y, Tanaka T. Edoxaban prevented adverse effects including pyrexia and elevation of D-dimer caused by the combination of BRAF and MEK inhibitors in a patient with BRAF-mutant melanoma. J Dermatol 2021; 48:707-709. [PMID: 33600004 DOI: 10.1111/1346-8138.15813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/28/2021] [Indexed: 11/29/2022]
Abstract
The combination of BRAF inhibitor and MEK inhibitor is one of the first-line treatments for unresectable BRAF-mutant melanoma or as an adjuvant therapy. However, some patients who received the combination of dabrafenib and trametinib (CombiDT) or the combination of encorafenib and binimetinib (CombiEB) had adverse events (AEs) including pyrexia. We herein report a patient with BRAF-mutated melanoma who repeatedly developed elevated levels of D-dimer and pyrexia after CombiDT and CombiEB treatments. Moreover, concomitant edoxaban prevented these AEs, enabling the patient to continue receiving CombiEB.
Collapse
Affiliation(s)
- Kei Mukai
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Masahiro Kamata
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Mirei Miyazaki
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Mayumi Nagata
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Saki Fukaya
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Kotaro Hayashi
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Atsuko Fukuyasu
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Takeko Ishikawa
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Takamitsu Ohnishi
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Yayoi Tada
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Takamitsu Tanaka
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
659
|
Zhang H, Wang Y, Zheng Q, Tang K, Fang R, Wang Y, Sun Q. Research Interest and Public Interest in Melanoma: A Bibliometric and Google Trends Analysis. Front Oncol 2021; 11:629687. [PMID: 33680968 PMCID: PMC7930473 DOI: 10.3389/fonc.2021.629687] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 01/04/2021] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Melanoma is a severe skin cancer that metastasizes quickly. Bibliometric analysis can quantify hotspots of research interest. Google Trends can provide information to address public concerns. METHODS The top 15 most frequently cited articles on melanoma each year from 2015 to 2019, according to annual citations, were retrieved from the Web of Science database. Original articles, reviews, and research letters were included in this research. For the Google Trends analysis, the topic "Melanoma" was selected as the keyword. Online search data from 2004 to 2019 were collected. Four countries (New Zealand, Australia, the United States and the United Kingdom) were selected for seasonal analysis. Annual trends in relative search volume and seasonal variation were analyzed, and the top related topics and rising related topics were also selected and analyzed. RESULTS The top 15 most frequently cited articles each year were all original articles that focused on immunotherapy (n=8), omics (n=5), and the microbiome (n=2). The average relative search volume remained relatively stable across the years. The seasonal variation analysis revealed that the peak appeared in summer, and the valley appeared in winter. The diseases associated with or manifestations of melanoma, treatment options, risk factors, diagnostic tools, and prognosis were the topics in which the public was most interested. Most of the topics revealed by bibliometric and Google Trends analyses were consistent, with the exception of issues related to the molecular biology of melanoma. CONCLUSION This study revealed the trends in research interest and public interest in melanoma, which may pave the way for further research.
Collapse
|
660
|
Wilson MA, Fecher LA. The Role of Systemic Therapy in Advanced Cutaneous Melanoma of the Head and Neck. Otolaryngol Clin North Am 2021; 54:329-342. [PMID: 33602512 DOI: 10.1016/j.otc.2020.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The treatment of advanced melanoma has changed dramatically over the last decade. With the discovery of activating BRAF mutations and the development of targeted therapies and checkpoint inhibitors, the overall survival of patients with advanced melanoma has improved. This article provides an overview of systemic therapies, including the pivotal agents that have led to these advances.
Collapse
Affiliation(s)
- Melissa A Wilson
- Sidney Kimmel Cancer Center, Thomas Jefferson University, 1025 Walnut Street, Suite 700, Philadelphia, PA 19107, USA
| | - Leslie A Fecher
- University of Michigan, Rogel Cancer Center, C343 MIB, 1500 East Medical Center Drive, SPC 5848, Ann Arbor, MI 48109-5848, USA.
| |
Collapse
|
661
|
Pediatric Glioma: An Update of Diagnosis, Biology, and Treatment. Cancers (Basel) 2021; 13:cancers13040758. [PMID: 33673070 PMCID: PMC7918156 DOI: 10.3390/cancers13040758] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 01/03/2023] Open
Abstract
Simple Summary Recent research has enhanced our understanding of the diverse biological processes that occur in pediatric gliomas; and molecular genetic analysis has become essential to diagnose and treat these conditions. Because targetable molecular aberrations can be detected in pediatric gliomas, identifying these aberrations is very important. This review provides an overview of pediatric gliomas, and describes recent developments made in strategies for their diagnosis and treatment. Additionally, it presents a current picture of pediatric gliomas in light of advances in molecular genetics, and describes the current scientific progress in gliomas’ treatment using information from recently completed and ongoing clinical trials. The era of incorporating molecular genetic analysis into clinical practice is emerging. Abstract Recent research has promoted elucidation of the diverse biological processes that occur in pediatric central nervous system (CNS) tumors. Molecular genetic analysis is essential not only for proper classification, but also for monitoring biological behavior and clinical management of tumors. Ever since the 2016 World Health Organization classification of CNS tumors, molecular profiling has become an indispensable step in the diagnosis, prediction of prognosis, and treatment of pediatric as well as adult CNS tumors. These molecular data are changing diagnosis, leading to new guidelines, and offering novel molecular targeted therapies. The Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy (cIMPACT-NOW) makes practical recommendations using recent advances in CNS tumor classification, particularly in molecular discernment of these neoplasms as morphology-based classification of tumors is being replaced by molecular-based classification. In this article, we summarize recent knowledge to provide an overview of pediatric gliomas, which are major pediatric CNS tumors, and describe recent developments in strategies employed for their diagnosis and treatment.
Collapse
|
662
|
Váraljai R, Horn S, Sucker A, Piercianek D, Schmitt V, Carpinteiro A, Becker KA, Reifenberger J, Roesch A, Felsberg J, Reifenberger G, Sure U, Schadendorf D, Helfrich I. Integrative Genomic Analyses of Patient-Matched Intracranial and Extracranial Metastases Reveal a Novel Brain-Specific Landscape of Genetic Variants in Driver Genes of Malignant Melanoma. Cancers (Basel) 2021; 13:cancers13040731. [PMID: 33578810 PMCID: PMC7916600 DOI: 10.3390/cancers13040731] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/28/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Development of brain metastases in advanced melanoma patients is a frequent event that limits patients' quality of life and survival. Despite recent insights into melanoma genetics, systematic analyses of genetic alterations in melanoma brain metastasis formation are lacking. Moreover, whether brain metastases harbor distinct genetic alterations beyond those observed at different anatomic sites of the same patient remains unknown. EXPERIMENTAL DESIGN AND RESULTS In our study, 54 intracranial and 18 corresponding extracranial melanoma metastases were analyzed for mutations using targeted next generation sequencing of 29 recurrently mutated driver genes in melanoma. In 11 of 16 paired samples, we detected nucleotide modifications in brain metastases that were absent in matched metastases at extracranial sites. Moreover, we identified novel genetic variants in ARID1A, ARID2, SMARCA4 and BAP1, genes that have not been linked to brain metastases before; albeit most frequent mutations were found in ARID1A, ARID2 and BRAF. Conclusion: Our data provide new insights into the genetic landscape of intracranial melanoma metastases supporting a branched evolution model of metastasis formation.
Collapse
Affiliation(s)
- Renáta Váraljai
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
| | - Susanne Horn
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
- Faculty Rudolf-Schönheimer-Institute for Biochemistry, University of Leipzig, 04103 Leipzig, Germany
| | - Antje Sucker
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
| | - Daniela Piercianek
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
- Department of Neurosurgery, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany
| | - Verena Schmitt
- Institute of Anatomy, Medical Faculty, University Duisburg-Essen, 45147 Essen, Germany;
| | - Alexander Carpinteiro
- Department of Molecular Biology, Medical Faculty, University Duisburg-Essen, 45147 Essen, Germany; (A.C.); (K.A.B.)
| | - Katrin Anne Becker
- Department of Molecular Biology, Medical Faculty, University Duisburg-Essen, 45147 Essen, Germany; (A.C.); (K.A.B.)
| | - Julia Reifenberger
- Department of Dermatology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany;
| | - Alexander Roesch
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
| | - Jörg Felsberg
- Institute of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany;
| | - Guido Reifenberger
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
- Institute of Neuropathology, Heinrich Heine University, 40225 Düsseldorf, Germany;
| | - Ulrich Sure
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
- Department of Neurosurgery, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany
| | - Dirk Schadendorf
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
| | - Iris Helfrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany; (R.V.); (S.H.); (A.S.); (A.R.); (D.S.)
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, 45147 Essen, Germany; (D.P.); (G.R.); (U.S.)
- Correspondence: ; Tel.: +49-201-723-1648; Fax: +49-201-723-5525
| |
Collapse
|
663
|
Funck-Brentano E, Malissen N, Roger A, Lebbé C, Deilhes F, Frénard C, Dréno B, Meyer N, Grob JJ, Tétu P, Saiag P. Which adjuvant treatment for patients with BRAF V600-mutant cutaneous melanoma? Ann Dermatol Venereol 2021; 148:145-155. [PMID: 33579557 DOI: 10.1016/j.annder.2020.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/22/2020] [Accepted: 11/05/2020] [Indexed: 12/21/2022]
Abstract
Treatment of patients with melanoma has considerably improved over the past decade and more recently with adjuvant therapies for patients with American Joint Committee on Cancer (AJCC) stage III (loco-regional metastases) or IV (distant metastases) totally resected melanoma, in order to prevent recurrence. In the adjuvant setting, two options are available to patients with BRAFV600-mutant AJCC stage III totally resected melanoma: anti-PD-1 blockers (nivolumab or pembrolizumab) or BRAF plus MEK inhibitors (dabrafenib plus trametinib). In the absence of comparative studies, it is difficult to determine which of these options is best. Our aim was to review published studies focusing on the management of patients with BRAFV600-mutant melanoma in the adjuvant setting. We also reviewed the main clinical trials of BRAF plus MEK inhibitors and immunotherapy in advanced (i.e. unresectable metastatic) BRAF-mutant melanoma in an attempt to identify results potentially affecting the management of patients on adjuvants. More adverse events are observed with targeted therapy, but all resolve rapidly upon drug discontinuation, whereas with immune checkpoint blockers some adverse events may persist. New therapeutic strategies are emerging, notably neoadjuvant therapies for stage III patients and adjuvant therapies for stage II patients; the place of the adjuvant strategy amidst all these options will soon be re-evaluated. The choice of adjuvant treatment could influence the choice of subsequent treatments in neo-adjuvant or metastatic settings. This review will lead clinicians to a better understanding of the different adjuvant treatments available for patients with totally resected AJCC stage III and IV BRAFV600-mutant melanoma before considering subsequent treatment strategies.
Collapse
Affiliation(s)
- E Funck-Brentano
- Department of General and Oncologic Dermatology, Ambroise-Paré hospital, AP-HP, Boulogne-Billancourt, France; Research unit EA4340 "Biomarkers and clinical trials in oncology and onco-hematology", Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, France.
| | - N Malissen
- Department of Dermatology and Skin Cancer, Aix-Marseille University, AP-HM, Hôpital Timone, Marseille, France
| | - A Roger
- Department of General and Oncologic Dermatology, Ambroise-Paré hospital, AP-HP, Boulogne-Billancourt, France; Research unit EA4340 "Biomarkers and clinical trials in oncology and onco-hematology", Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, France
| | - C Lebbé
- Inserm U976, Department of Dermatology, Dermatology, Paris University, Hôpital Saint-Louis, AP-HP, Paris, France
| | - F Deilhes
- Dermatology Department, CHU de Toulouse, Toulouse, France
| | - C Frénard
- Department of Dermatology, CRCINA, CIC1413, CHU de Nantes, université de Nantes, Nantes, France
| | - B Dréno
- Department of Dermatology, CRCINA, CIC1413, CHU de Nantes, université de Nantes, Nantes, France
| | - N Meyer
- Dermatology Department, CHU de Toulouse, Toulouse, France
| | - J-J Grob
- Department of Dermatology and Skin Cancer, Aix-Marseille University, AP-HM, Hôpital Timone, Marseille, France
| | - P Tétu
- Department of Dermatology, CRCINA, CIC1413, CHU de Nantes, université de Nantes, Nantes, France
| | - P Saiag
- Department of General and Oncologic Dermatology, Ambroise-Paré hospital, AP-HP, Boulogne-Billancourt, France; Research unit EA4340 "Biomarkers and clinical trials in oncology and onco-hematology", Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, France
| |
Collapse
|
664
|
Marinova AM, Reilly JL, Wong V, Weiss S, Olszanski AJ. Metastatic Melanoma With Leptomeningeal Disease. J Adv Pract Oncol 2021; 12:79-83. [PMID: 33552663 PMCID: PMC7844196 DOI: 10.6004/jadpro.2021.12.1.6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Leptomeningeal disease in patients with melanoma historically portends a grim prognosis, with median survival measured in weeks to months. The advent of effective immunotherapy and targeted agents may modify the outcome of such patients. This case report describes a 43-year-old patient diagnosed with stage IIIa BRAF-positive cutaneous melanoma in 2012 who subsequently developed leptomeningeal involvement as her sole site of melanotic metastasis. She received multiple systemic therapies and radiotherapy and survived 2.5 years after her diagnosis with central nervous system involvement. This case report highlights the importance of a multidisciplinary team and the advent of effective agents, which offers the potential for significantly improved outcomes for patients with metastatic melanoma involving the central nervous system.
Collapse
|
665
|
Gassenmaier M, Lenders MM, Forschner A, Leiter U, Weide B, Garbe C, Eigentler TK, Wagner NB. Serum S100B and LDH at Baseline and During Therapy Predict the Outcome of Metastatic Melanoma Patients Treated with BRAF Inhibitors. Target Oncol 2021; 16:197-205. [PMID: 33555543 PMCID: PMC7935737 DOI: 10.1007/s11523-021-00792-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Despite impressive response rates, most patients with advanced melanoma ultimately progress following therapy with B-Raf proto-oncogene (BRAF) inhibitors (BRAFi). Therefore, frequent radiologic assessments are necessary, and reliable serum biomarkers would be beneficial for disease monitoring. OBJECTIVE This study investigated the ability of lactate dehydrogenase (LDH) and S100 calcium-binding protein B (S100B) to detect response and disease progression during treatment with BRAFi. PATIENTS AND METHODS Baseline levels of LDH and S100B and repeated measurements during therapy were recorded retrospectively in 191 patients with metastatic melanoma. LDH and S100B levels were compared between distinct time points (baseline, first follow-up visit [FV], best objective response [BR], and progressive disease [PD]). The prognostic ability of the serum biomarkers in relation to disease-specific survival (DSS) was assessed with univariable and multivariable Cox regression analysis. RESULTS Elevated baseline LDH and S100B correlated with impaired DSS. In contrast with LDH (P = 0.12), S100B levels at FV correlated with response (P = 0.0030). Both markers significantly decreased during the first weeks of BRAFi treatment (LDH, P = 0.00034; S100B, P < 0.0001) and increased between BR and PD (LDH, P = 0.016; S100B, P < 0.0001). Patients with elevated S100B (P = 0.00062) but not with elevated LDH (P = 0.067) at the time point of radiologically confirmed PD showed significantly impaired DSS after PD. Interestingly, DSS after PD differed significantly according to S100B levels determined as early as 8 weeks (median) before PD (P = 0.0024). CONCLUSIONS LDH and S100B are suitable serum biomarkers during therapy with BRAFi. S100B shows stronger correlation with response and exhibits more accuracy in predicting PD. Close biomarker monitoring with S100B is recommended during treatment with BRAFi to detect PD early.
Collapse
Affiliation(s)
| | - Max M Lenders
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Andrea Forschner
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Ulrike Leiter
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Benjamin Weide
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Thomas K Eigentler
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Nikolaus B Wagner
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany. .,Department of Dermatology and Allergology, Kantonsspital St. Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland.
| |
Collapse
|
666
|
Keller BA, Laight BJ, Varette O, Broom A, Wedge MÈ, McSweeney B, Cemeus C, Petryk J, Lo B, Burns B, Nessim C, Ong M, Chica RA, Atkins HL, Diallo JS, Ilkow CS, Bell JC. Personalized oncology and BRAF K601N melanoma: model development, drug discovery, and clinical correlation. J Cancer Res Clin Oncol 2021; 147:1365-1378. [PMID: 33555379 DOI: 10.1007/s00432-021-03545-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 01/25/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE Mutations in BRAF are the most prominent activating mutations in melanoma and are increasingly recognized in other cancers. There is currently no accepted treatment regimen for patients with mutant BRAFK601N melanoma, and the study of melanoma driven by BRAF mutations at the 601 locus is lacking due to a paucity of cellular model systems. Therefore, we sought to better understand the treatment and clinical approach to patients with mutant BRAFK601N melanoma and subsequently develop a novel personalized oncology platform for rare or treatment-refractory cancers. METHODS We developed and characterized the first patient-derived, naturally occurring BRAFK601N melanoma model, described herein as OHRI-MEL-13, and assessed efficacy using the Prestwick Chemical Library and select targeted therapeutics. RESULTS OHRI-MEL-13 exhibits loss of heterozygosity of BRAF, closely mimics the original tumor's gene expression profile, is tumorigenic in immune-deficient murine models, and is available for public accession through American Type Culture Collection. We present in silico modeling data, which illustrates the therapeutic failure of BRAFV600E-targeted therapies in BRAFK601N mutants. Our platform elucidated a unique role for MEK inhibition with cobimetinib, which resulted in short-term clinical success by reducing the metastatic burden. CONCLUSION Our model of BRAFK601N-activated melanoma was developed, thoroughly characterized, and made available for public accession. This model served to demonstrate the feasibility of a novel personalized oncology platform that could be optimized at an institutional level for rare variant or treatment-refractory cancers. We also demonstrate the clinical utility of monotherapy MEK inhibition in a case of BRAFK601N melanoma.
Collapse
Affiliation(s)
- Brian A Keller
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada.
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, K1H 8M5, Canada.
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, 501 Smyth Road, Ottawa, K1H 8L6, Canada.
| | - Brian J Laight
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | - Oliver Varette
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, K1H 8M5, Canada
| | - Aron Broom
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie-Curie Private, Ottawa, K1N 6N5, Canada
| | - Marie-Ève Wedge
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, K1H 8M5, Canada
| | - Benjamin McSweeney
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | - Catia Cemeus
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | - Julia Petryk
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | - Bryan Lo
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, 501 Smyth Road, Ottawa, K1H 8L6, Canada
- Molecular Oncology Diagnostics Laboratory, The Ottawa Hospital, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | - Bruce Burns
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | - Carolyn Nessim
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
- Division of General Surgery, The Ottawa Hospital, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | - Michael Ong
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
- Division of Medical Oncology, The Ottawa Hospital, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | - Roberto A Chica
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie-Curie Private, Ottawa, K1N 6N5, Canada
| | - Harold L Atkins
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, K1H 8M5, Canada
- The Ottawa Hospital Blood and Marrow Transplant Program, The Ottawa Hospital, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | - Jean-Simon Diallo
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, K1H 8M5, Canada
| | - Carolina S Ilkow
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, K1H 8M5, Canada
| | - John C Bell
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, K1H 8M5, Canada
| |
Collapse
|
667
|
The ERK mitogen-activated protein kinase signaling network: the final frontier in RAS signal transduction. Biochem Soc Trans 2021; 49:253-267. [PMID: 33544118 DOI: 10.1042/bst20200507] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/30/2020] [Accepted: 01/08/2021] [Indexed: 12/11/2022]
Abstract
The RAF-MEK-ERK mitogen-activated protein kinase (MAPK) cascade is aberrantly activated in a diverse set of human cancers and the RASopathy group of genetic developmental disorders. This protein kinase cascade is one of the most intensely studied cellular signaling networks and has been frequently targeted by the pharmaceutical industry, with more than 30 inhibitors either approved or under clinical evaluation. The ERK-MAPK cascade was originally depicted as a serial and linear, unidirectional pathway that relays extracellular signals, such as mitogenic stimuli, through the cytoplasm to the nucleus. However, we now appreciate that this three-tiered protein kinase cascade is a central core of a complex network with dynamic signaling inputs and outputs and autoregulatory loops. Despite our considerable advances in understanding the ERK-MAPK network, the ability of cancer cells to adapt to the inhibition of key nodes reveals a level of complexity that remains to be fully understood. In this review, we summarize important developments in our understanding of the ERK-MAPK network and identify unresolved issues for ongoing and future study.
Collapse
|
668
|
Malignant Melanoma of the Gastrointestinal Tract: Symptoms, Diagnosis, and Current Treatment Options. Cells 2021; 10:cells10020327. [PMID: 33562484 PMCID: PMC7915313 DOI: 10.3390/cells10020327] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/21/2022] Open
Abstract
Malignant melanoma (MM) has become the fifth most frequent cancer in the UK. It is the most common carcinoma to metastasize to the gastrointestinal (GI) tract. MM particularly has an affinity to spread to the small bowel, which is followed by the involvement of the stomach and large intestine. Excellent endoscopic options including video capsule endoscopy and enteroscopy are available for a precise diagnosis of GI involvement by a metastatic MM. The complete surgical resection of GI metastatic MM in carefully selected patients not only provides symptom control, but has also been associated with an increase in overall survival. The approval of BRAF-targeted therapies and immune checkpoint inhibitors has transformed therapeutic approaches for patients with metastatic MM over the past decade. Currently, the overall survival of patients with advanced metastatic MM who have been treated with a combination of immunotherapeutic agents reaches 52% at five years. The role of surgery for patients with the metastatic involvement of the GI tract with MM is evolving in the era of effective systemic treatments.
Collapse
|
669
|
Tan JY, Wijesinghe IVS, Alfarizal Kamarudin MN, Parhar I. Paediatric Gliomas: BRAF and Histone H3 as Biomarkers, Therapy and Perspective of Liquid Biopsies. Cancers (Basel) 2021; 13:cancers13040607. [PMID: 33557011 PMCID: PMC7913734 DOI: 10.3390/cancers13040607] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Gliomas are major causes of worldwide cancer-associated deaths in children. Generally, paediatric gliomas can be classified into low-grade and high-grade gliomas. They differ significantly from adult gliomas in terms of prevalence, molecular alterations, molecular mechanisms and predominant histological types. The aims of this review article are: (i) to discuss the current updates of biomarkers in paediatric low-grade and high-grade gliomas including their diagnostic and prognostic values, and (ii) to discuss potential targeted therapies in treating paediatric low-grade and high-grade gliomas. Our findings revealed that liquid biopsy is less invasive than tissue biopsy in obtaining the samples for biomarker detections in children. In addition, future clinical trials should consider blood-brain barrier (BBB) penetration of therapeutic drugs in paediatric population. Abstract Paediatric gliomas categorised as low- or high-grade vary markedly from their adult counterparts, and denoted as the second most prevalent childhood cancers after leukaemia. As compared to adult gliomas, the studies of diagnostic and prognostic biomarkers, as well as the development of therapy in paediatric gliomas, are still in their infancy. A body of evidence demonstrates that B-Raf Proto-Oncogene or V-Raf Murine Sarcoma Viral Oncogene Homolog B (BRAF) and histone H3 mutations are valuable biomarkers for paediatric low-grade gliomas (pLGGs) and high-grade gliomas (pHGGs). Various diagnostic methods involving fluorescence in situ hybridisation, whole-genomic sequencing, PCR, next-generation sequencing and NanoString are currently used for detecting BRAF and histone H3 mutations. Additionally, liquid biopsies are gaining popularity as an alternative to tumour materials in detecting these biomarkers, but still, they cannot fully replace solid biopsies due to several limitations. Although histone H3 mutations are reliable prognosis biomarkers in pHGGs, children with these mutations have a dismal prognosis. Conversely, the role of BRAF alterations as prognostic biomarkers in pLGGs is still in doubt due to contradictory findings. The BRAF V600E mutation is seen in the majority of pLGGs (as seen in pleomorphic xanthoastrocytoma and gangliomas). By contrast, the H3K27M mutation is found in the majority of paediatric diffuse intrinsic pontine glioma and other midline gliomas in pHGGs. pLGG patients with a BRAF V600E mutation often have a lower progression-free survival rate in comparison to wild-type pLGGs when treated with conventional therapies. BRAF inhibitors (Dabrafenib and Vemurafenib), however, show higher overall survival and tumour response in BRAF V600E mutated pLGGs than conventional therapies in some studies. To date, targeted therapy and precision medicine are promising avenues for paediatric gliomas with BRAF V600E and diffuse intrinsic pontine glioma with the H3K27M mutations. Given these shortcomings in the current treatments of paediatric gliomas, there is a dire need for novel therapies that yield a better therapeutic response. The present review discusses the diagnostic tools and the perspective of liquid biopsies in the detection of BRAF V600E and H3K27M mutations. An in-depth understanding of these biomarkers and the therapeutics associated with the respective challenges will bridge the gap between paediatric glioma patients and the development of effective therapies.
Collapse
Affiliation(s)
| | | | | | - Ishwar Parhar
- Correspondence: ; Tel.: +603-5514-6304; Fax: +603-5515-6341
| |
Collapse
|
670
|
Garcia-Sampedro A, Gaggia G, Ney A, Mahamed I, Acedo P. The State-of-the-Art of Phase II/III Clinical Trials for Targeted Pancreatic Cancer Therapies. J Clin Med 2021; 10:566. [PMID: 33546207 PMCID: PMC7913382 DOI: 10.3390/jcm10040566] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a devastating disease with very poor prognosis. Currently, surgery followed by adjuvant chemotherapy represents the only curative option which, unfortunately, is only available for a small group of patients. The majority of pancreatic cancer cases are diagnosed at advanced or metastatic stage when surgical resection is not possible and treatment options are limited. Thus, novel and more effective therapeutic strategies are urgently needed. Molecular profiling together with targeted therapies against key hallmarks of pancreatic cancer appear as a promising approach that could overcome the limitations of conventional chemo- and radio-therapy. In this review, we focus on the latest personalised and multimodal targeted therapies currently undergoing phase II or III clinical trials. We discuss the most promising findings of agents targeting surface receptors, angiogenesis, DNA damage and cell cycle arrest, key signalling pathways, immunotherapies, and the tumour microenvironment.
Collapse
Affiliation(s)
| | | | | | | | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, London NW3 2QG, UK; (A.G.-S.); (G.G.); (A.N.); (I.M.)
| |
Collapse
|
671
|
Danesi R, Fogli S, Indraccolo S, Del Re M, Dei Tos AP, Leoncini L, Antonuzzo L, Bonanno L, Guarneri V, Pierini A, Amunni G, Conte P. Druggable targets meet oncogenic drivers: opportunities and limitations of target-based classification of tumors and the role of Molecular Tumor Boards. ESMO Open 2021; 6:100040. [PMID: 33540286 PMCID: PMC7859305 DOI: 10.1016/j.esmoop.2020.100040] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/15/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
The therapeutic landscape of cancer is changing rapidly due to the growing number of approved drugs capable of targeting specific genetic alterations. This aspect, together with the development of noninvasive methods for the assessment of somatic mutations in the peripheral blood of patients, generated a growing interest toward a new tumor-agnostic classification system based on ‘predictive’ biomarkers. The current review article discusses this emerging alternative approach to the classification of cancer and its implications for the selection of treatments. It is suggested that different types of cancers sharing the same molecular profiles could benefit from the same targeted drugs. Although recent clinical trials have demonstrated that this approach cannot be generalized, there are also specific examples that demonstrate the clinical utility of this alternative vision. In this rapidly evolving scenario, a multidisciplinary approach managed by institutional Molecular Tumor Boards is fundamental to interpret the biological and clinical relevance of genetic alterations and the complexity of their relationship with treatment response. The identification of oncogenic drivers offers the opportunity to develop target-specific drugs. The inhibition of crucial pathways realizes the principle of druggable target to exploit cancer vulnerability. The approval of new anticancer agents based on target-based concept represents a paradigm shift in cancer therapy. However, only few drugs have been approved so far on an agnostic basis and the concept of biomarker cannot be generalized. Tumor Molecular Boards will have an increasing role in the identification of new therapeutic options in selected patients.
Collapse
Affiliation(s)
- R Danesi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - S Fogli
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - S Indraccolo
- Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - M Del Re
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - A P Dei Tos
- Department of Medicine, School of Medicine, University of Padua, Padua, Italy
| | - L Leoncini
- Department of Medical Biotechnology, Anatomic Pathology Division, University of Siena, Siena, Italy
| | - L Antonuzzo
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - L Bonanno
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - V Guarneri
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - A Pierini
- Integrated Access, Roche, Monza, Italy
| | - G Amunni
- Institute for the Study, Prevention and Oncology Network (ISPRO), Florence, Italy.
| | - P Conte
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| |
Collapse
|
672
|
Botella-Estrada R, Boada-García A, Carrera-Álvarez C, Fernández-Figueras M, González-Cao M, Moreno-Ramírez D, Nagore E, Ríos-Buceta L, Rodríguez-Peralto JL, Samaniego-González E, Tejera-Vaquerizo A, Vílchez-Márquez F, Descalzo-Gallego MA, García-Doval I. Clinical Practice Guideline on Melanoma From the Spanish Academy of Dermatology and Venereology (AEDV). ACTAS DERMO-SIFILIOGRAFICAS 2021; 112:142-152. [PMID: 32721390 DOI: 10.1016/j.ad.2020.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 07/04/2020] [Indexed: 10/23/2022] Open
Abstract
Specialist approaches to the diagnosis and treatment of melanoma have undergone many changes. This guideline aims to provide Spanish dermatologists with evidence-based information for resolving the most common doubts that arise in clinical practice. Members of the Spanish Oncologic Dermatology and Surgery Group (GEDOC) with experience treating melanoma were invited to participate in drafting the guideline. The group developed a new guideline on the basis of existing ones, using the ADAPTE collaboration process, first summarizing the care process and posing relevant clinical questions, then selecting guidelines with the best scores according to the AGREE II (Appraisal of Guidelines for Research and Evaluation) tool. Finally, the group searched the selected guidelines for answers to the clinical questions, drafted recommendations, and sent them for external review. The guideline is structured around 21 clinical questions chosen for their relevance to issues that make clinical decisions about the management of melanoma difficult. Evidence from existing guidelines was used to answer the questions. A limitation of this guide derives from the scarce evidence available for answering some questions. Moreover, some areas are changing rapidly, so recommendations must be updated often. The present guideline offers answers to clinical questions about the routine management of melanoma in clinical practice and provides dermatologists with a reference to guide decisions, taking into consideration the resources available and patient preferences.
Collapse
Affiliation(s)
- R Botella-Estrada
- Servicio de Dermatología, Hospital Universitario La Fe, Valencia, España; Departamento de Medicina, Universitat de València, Valencia, España.
| | - A Boada-García
- Servicio de Dermatología, Hospital Universitario Germans Trias i Pujol, Badalona, Barcelona, España
| | | | - M Fernández-Figueras
- Área de Anatomía Patológica, Hospital Universitario General de Cataluña-Quirón Salud, Barcelona, España
| | - M González-Cao
- Translational Cancer Research Unit, Instituto Oncológico Dr. Rosell, Hospital Universitario Dexeus, Barcelona, España
| | - D Moreno-Ramírez
- Servicio de Dermatología, Hospital Universitario Virgen Macarena, Sevilla, España
| | - E Nagore
- Servicio de Dermatología, Instituto Valenciano de Oncología, Valencia, España
| | - L Ríos-Buceta
- Servicio de Dermatología, Hospital Universitario Ramón y Cajal, Madrid, España
| | - J L Rodríguez-Peralto
- Servicio de Anatomía Patológica, Hospital Universitario 12 de Octubre, Madrid, España
| | - E Samaniego-González
- Servicio de Dermatología, Complejo Asistencial Universitario de León, León, España
| | | | - F Vílchez-Márquez
- Servicio de Dermatología, Hospital Universitario Virgen de las Nieves, Granada, España
| | - M A Descalzo-Gallego
- Unidad de Investigación, Fundación Piel Sana, Academia Española de Dermatología y Venereología, Madrid, España
| | - I García-Doval
- Unidad de Investigación, Fundación Piel Sana, Academia Española de Dermatología y Venereología, Madrid, España
| |
Collapse
|
673
|
Nawwar AA, Searle J, Lyburn ID. Pembrolizumab-Induced Thyroiditis and Colitis-Presentation and Resolution on Serial FDG PET/CT. Clin Nucl Med 2021; 46:e121-e122. [PMID: 32969909 DOI: 10.1097/rlu.0000000000003306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
ABSTRACT A 58-year-old man with previous melanoma of the left leg underwent whole-body 18F-FDG PET/CT to stage metastatic disease prior to commencing pembrolizumab. Follow-up FDG PET/CT after 3 months of treatment showed partial metabolic response of soft tissue and nodal metastases and diffuse increased thyroid and colonic uptake, suggestive of thyroiditis and colitis. Pembrolizumab was ceased, and a repeat FDG PET/CT scan showed regression of uptake in the thyroid gland and colon, in keeping with resolution of inflammatory change. Immune-related adverse events induced by Immune checkpoint inhibitors, such as pembrolizumab, should be recognized-cessation of treatment often leads to resolution.
Collapse
|
674
|
Botella-Estrada R, Boada-García A, Carrera-Álvarez C, Fernández-Figueras M, González-Cao M, Moreno-Ramírez D, Nagore E, Ríos-Buceta L, Rodríguez-Peralto J, Samaniego-González E, Tejera-Vaquerizo A, Vílchez-Márquez F, Descalzo-Gallego M, García-Doval I. Clinical Practice Guideline on Melanoma From the Spanish Academy of Dermatology and Venereology (AEDV). ACTAS DERMO-SIFILIOGRAFICAS 2021. [DOI: 10.1016/j.adengl.2020.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
675
|
Dimitriou F, Urner-Bloch U, Eggenschwiler C, Mitsakakis N, Mangana J, Dummer R, Urner M. The association between immune checkpoint or BRAF/MEK inhibitor therapy and uveitis in patients with advanced cutaneous melanoma. Eur J Cancer 2021; 144:215-223. [PMID: 33373866 DOI: 10.1016/j.ejca.2020.11.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/20/2020] [Accepted: 11/15/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Treatment with immune checkpoint and BRAF/MEK inhibitors has significantly improved the survival of patients with advanced cutaneous melanoma and other metastatic malignancies. Therapy-related uveitis is a rare ocular adverse event, which may potentially lead to legal blindness. The epidemiology of treatment-related uveitis is currently insufficiently known. PATIENTS AND METHODS In this cohort study, we asked whether exposure to either immune checkpoint or BRAF/MEK inhibitors was associated with a higher risk of developing uveitis compared with the general population. Based on a Bayesian framework, we estimated the probability of developing uveitis with a right-censored, exponential survival model using data from the Zurich Melanoma Registry. The registry included all adult patients treated for advanced cutaneous melanoma between January 2008 and December 2018 at the University Hospital of Zurich, Switzerland. RESULTS In total, 304 patients (64%) were treated with immune checkpoint and 186 patients (38%) with BRAF/MEK inhibitors. Median follow-up time was 74 days (interquartile range: 57-233 days). Eleven patients developed uveitis and 30 patients died. We estimated the probability of developing uveitis per year in the general population as 0.05% (95% credibility interval [CrI]: 0.02%-0.1%). Corresponding posterior probabilities of treatment-related uveitis were 3.48% (95% CrI: 0.93%-7.49%) and 5.04% (95% CrI: 2.07%-9.19%) for immune checkpoint or BRAF/MEK inhibitors (posterior probability for difference: 76%). CONCLUSIONS Immune checkpoint and particularly BRAF/MEK inhibitor therapies are associated with an increase in the risk of developing uveitis. Treatment-related uveitis is not associated with systemic adverse events of immune checkpoint or BRAF/MEK inhibitors.
Collapse
Affiliation(s)
- Florentia Dimitriou
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Ursula Urner-Bloch
- Private Ophthalmic Practice in Cooperation with the Skin Cancer Unit, University Hospital of Zurich, Zurich, Switzerland
| | | | - Nicholas Mitsakakis
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada
| | - Joanna Mangana
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.
| | - Martin Urner
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
676
|
van Breeschoten J, Wouters MWJM, de Wreede LC, Hilarius DH, Haanen JB, Blank CU, Aarts MJB, van den Berkmortel FWPJ, de Groot JWB, Hospers GAP, Kapiteijn E, Piersma D, van Rijn RS, Suijkerbuijk KPM, Blokx WAM, Ten Tije AJ, van der Veldt AAM, Vreugdenhil G, Boers MJ, van den Eertwegh AJM. Nationwide Outcomes of Advanced Melanoma According to BRAFV600 Status. Am J Clin Oncol 2021; 44:82-89. [PMID: 33332931 DOI: 10.1097/coc.0000000000000786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE The aim of this study was to evaluate treatment patterns and overall survival (OS) of patients with BRAFV600 wild-type and BRAFV600-mutant advanced melanoma in the Netherlands. METHODS We selected patients of 18 years and over, diagnosed between 2016 and 2017 with unresectable stage IIIC or IV melanoma, registered in the Dutch Melanoma Treatment Registry. To assess the association of BRAFV600-mutation status with OS we used the Cox proportional-hazards model. RESULTS A total of 642 BRAFV600 wild-type and 853 mutant patients were included in the analysis. Median OS did not differ significantly between both groups, 15.2 months (95% confidence interval [CI]: 13.2-19.2) versus 20.6 months (95% CI: 18.3-25.0). Survival rates at 6 and 12 months were significantly lower for BRAFV600 wild-type patients compared with BRAFV600-mutant patients, 72.0% (95% CI: 68.6-75.6) and 56.0% (95% CI: 52.2-60.0) versus 83.4% (95% CI: 80.9-85.9) and 65.7% (95% CI: 62.6-69.0). Two-year survival was not significantly different between both groups, 41.1% (95% CI: 37.2-45.3) versus 47.0% (95% CI: 43.6-60.6). Between 0 and 10 months, BRAFV600 wild-type patients had a decreased survival with a hazard ratio for OS of 2.00 (95% CI: 1.62-2.46) but this effect disappeared after 10 months. At 12 months, BRAFV600-mutant patients had started with second-line systemic treatment more often compared with BRAFV600 wild-type patients (50% vs. 19%). CONCLUSION These results suggest that advanced BRAFV600 wild-type melanoma patients have worse survival than BRAFV600-mutated patients during the first 10 months after diagnosis because of less available treatment options.
Collapse
Affiliation(s)
- Jesper van Breeschoten
- Dutch Institute for Clinical Auditing
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam
| | | | | | | | | | - Christian U Blank
- Medical Oncology and Immunology
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam
| | - Maureen J B Aarts
- Department of Medical Oncology, Maastricht University Medical Centre, Maastricht
| | | | | | - Geke A P Hospers
- Department of Medical Oncology, University Medical Centre Groningen, Groningen
| | | | - Djura Piersma
- Department of Internal Medicine, Medisch Spectrum Twente, Enschede
| | | | | | | | | | | | | | - Marye J Boers
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | |
Collapse
|
677
|
Santiappillai NT, Abuhammad S, Slater A, Kirby L, McArthur GA, Sheppard KE, Smith LK. CDK4/6 Inhibition Reprograms Mitochondrial Metabolism in BRAF V600 Melanoma via a p53 Dependent Pathway. Cancers (Basel) 2021; 13:cancers13030524. [PMID: 33572972 PMCID: PMC7866416 DOI: 10.3390/cancers13030524] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors are being tested in numerous clinical trials and are currently employed successfully in the clinic for the treatment of breast cancers. Understanding their mechanism of action and interaction with other therapies is vital in their clinical development. CDK4/6 regulate the cell cycle via phosphorylation and inhibition of the tumour suppressor RB, and in addition can phosphorylate many cellular proteins and modulate numerous cellular functions including cell metabolism. Metabolic reprogramming is observed in melanoma following standard-of-care BRAF/MEK inhibition and is involved in both therapeutic response and resistance. In preclinical models, CDK4/6 inhibitors overcome BRAF/MEK inhibitor resistance, leading to sustained tumour regression; however, the metabolic response to this combination has not been explored. Here, we investigate how CDK4/6 inhibition reprograms metabolism and if this alters metabolic reprogramming observed upon BRAF/MEK inhibition. Although CDK4/6 inhibition has no substantial effect on the metabolic phenotype following BRAF/MEK targeted therapy in melanoma, CDK4/6 inhibition alone significantly enhances mitochondrial metabolism. The increase in mitochondrial metabolism in melanoma cells following CDK4/6 inhibition is fuelled in part by both glutamine metabolism and fatty acid oxidation pathways and is partially dependent on p53. Collectively, our findings identify new p53-dependent metabolic vulnerabilities that may be targeted to improve response to CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Nancy T. Santiappillai
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne 3052, Australia; (N.T.S.); (S.A.); (A.S.); (L.K.); (G.A.M.)
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne 3052, Australia
| | - Shatha Abuhammad
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne 3052, Australia; (N.T.S.); (S.A.); (A.S.); (L.K.); (G.A.M.)
| | - Alison Slater
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne 3052, Australia; (N.T.S.); (S.A.); (A.S.); (L.K.); (G.A.M.)
| | - Laura Kirby
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne 3052, Australia; (N.T.S.); (S.A.); (A.S.); (L.K.); (G.A.M.)
| | - Grant A. McArthur
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne 3052, Australia; (N.T.S.); (S.A.); (A.S.); (L.K.); (G.A.M.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3052, Australia
| | - Karen E. Sheppard
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne 3052, Australia; (N.T.S.); (S.A.); (A.S.); (L.K.); (G.A.M.)
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne 3052, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3052, Australia
- Correspondence: (K.E.S.); (L.K.S.)
| | - Lorey K. Smith
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne 3052, Australia; (N.T.S.); (S.A.); (A.S.); (L.K.); (G.A.M.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3052, Australia
- Correspondence: (K.E.S.); (L.K.S.)
| |
Collapse
|
678
|
van Breeschoten J, Wouters MWJM, Hilarius DL, Haanen JB, Blank CU, Aarts MJB, van den Berkmortel FWPJ, de Groot JWB, Hospers GAP, Kapiteijn E, Piersma D, van Rijn RS, Suijkerbuijk KPM, Blokx WAM, Tije BJJT, Veldt AAMVD, Vreugdenhil A, Boers-Sonderen MJ, van den Eertwegh AJM. First-line BRAF/MEK inhibitors versus anti-PD-1 monotherapy in BRAF V600-mutant advanced melanoma patients: a propensity-matched survival analysis. Br J Cancer 2021; 124:1222-1230. [PMID: 33495600 DOI: 10.1038/s41416-020-01229-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/30/2020] [Accepted: 12/10/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Anti-PD-1 antibodies and BRAF/MEK inhibitors are the two main groups of systemic therapy in the treatment of BRAFV600-mutant advanced melanoma. Until now, data are inconclusive on which therapy to use as first-line treatment. The aim of this study was to use propensity score matching to compare first-line anti-PD-1 monotherapy vs. BRAF/MEK inhibitors in advanced BRAFV600-mutant melanoma patients. METHODS We selected patients diagnosed between 2014 and 2017 with advanced melanoma and a known BRAFV600-mutation treated with first-line BRAF/MEK inhibitors or anti-PD-1 antibodies, registered in the Dutch Melanoma Treatment Registry. Patients were matched based on their propensity scores using the nearest neighbour and the optimal matching method. RESULTS Between 2014 and 2017, a total of 330 and 254 advanced melanoma patients received BRAF/MEK inhibitors and anti-PD-1 monotherapy as first-line systemic therapy. In the matched cohort, patients receiving anti-PD-1 antibodies as a first-line treatment had a higher median and 2-year overall survival compared to patients treated with first-line BRAF/MEK inhibitors, 42.3 months (95% CI: 37.3-NE) vs. 19.8 months (95% CI: 16.7-24.3) and 65.4% (95% CI: 58.1-73.6) vs. 41.7% (95% CI: 34.2-51.0). CONCLUSIONS Our data suggest that in the matched BRAFV600-mutant advanced melanoma patients, anti-PD-1 monotherapy is the preferred first-line treatment in patients with relatively favourable patient and tumour characteristics.
Collapse
Affiliation(s)
- Jesper van Breeschoten
- Dutch Institute for Clinical Auditing, Rijnsburgerweg 10, Leiden, 2333AA, The Netherlands.,Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1118, Amsterdam, 1081HZ, The Netherlands
| | - Michel W J M Wouters
- Dutch Institute for Clinical Auditing, Rijnsburgerweg 10, Leiden, 2333AA, The Netherlands.,Department of Surgical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands
| | - Doranne L Hilarius
- Department of Pharmacy, Rode Kruis Ziekenhuis, Vondellaan 13, Beverwijk, 1942LE, The Netherlands
| | - John B Haanen
- Department of Medical Oncology and Immunology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands
| | - Christian U Blank
- Department of Medical Oncology and Immunology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands.,Department of Molecular Oncology & Immunology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066CX, The Netherlands
| | - Maureen J B Aarts
- Department of Medical Oncology, Maastricht University Medical Centre, P. Debyelaan 25, Maastricht, 6229 HX, The Netherlands
| | | | | | - Geke A P Hospers
- Department of Medical Oncology, University Medical Centre Groningen, Hanzeplein 1, Groningen, 9713GZ, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Centre, Albinusdreef 2, Leiden, 2333ZA, The Netherlands
| | - Djura Piersma
- Department of Internal Medicine, Medisch Spectrum Twente, Koningsplein 1, Enschede, 7512KZ, The Netherlands
| | - Roos S van Rijn
- Department of Internal Medicine, Medical Centre Leeuwarden, Henri Dunantweg 2, Leeuwarden, 8934AD, The Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Centre Utrecht, Heidelberglaan 100, Utrecht, 3584CX, The Netherlands
| | - Willeke A M Blokx
- Department of Pathology, Division of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht. Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Bert-Jan J Ten Tije
- Department of Internal Medicine, Amphia Hospital, Molengracht 21, Breda, 4818CK, The Netherlands
| | - Astrid A M van der Veldt
- Departments of Medical Oncology and Radiology & Nuclear Medicine, Erasmus Medical Centre, 's-Gravendijkwal 230, Rotterdam, 3015CE, The Netherlands
| | - Art Vreugdenhil
- Department of Internal Medicine, Maxima Medical Centre, De Run 4600, Eindhoven, 5504DB, The Netherlands
| | - Marye J Boers-Sonderen
- Department of Medical Oncology, Radboud University Medical Centre, Geert Grooteplein Zuid 10, Nijmegen, 6525GA, The Netherlands
| | - Alfonsus J M van den Eertwegh
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1118, Amsterdam, 1081HZ, The Netherlands.
| |
Collapse
|
679
|
Weilandt J, Diehl K, Schaarschmidt ML, Kiecker F, Sasama B, Pronk M, Ohletz J, Könnecke A, Müller V, Utikal J, Hillen U, Harth W, Peitsch WK. Patientenpräferenzen für die Therapie fortgeschrittener Melanome: Einfluss von Komorbidität. J Dtsch Dermatol Ges 2021; 19:58-72. [PMID: 33491889 DOI: 10.1111/ddg.14293_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Juliane Weilandt
- Klinik für Dermatologie und Phlebologie, Vivantes Klinikum im Friedrichshain, Berlin
| | - Katharina Diehl
- Mannheimer Institut für Public Health, Sozial- und Präventivmedizin, Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim
| | - Marthe-Lisa Schaarschmidt
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Mannheim, Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim
| | - Felix Kiecker
- Klinik für Dermatologie, Venerologie und Allergologie, Charité Universitätsmedizin Berlin, Berlin.,Klinik für Dermatologie und Venerologie, Vivantes Klinikum Neukölln, Berlin
| | - Bianca Sasama
- Klinik für Dermatologie und Phlebologie, Vivantes Klinikum im Friedrichshain, Berlin
| | - Melanie Pronk
- Klinik für Dermatologie und Allergologie, Vivantes Klinikum Spandau, Berlin
| | - Jan Ohletz
- Klinik für Dermatologie und Allergologie, Vivantes Klinikum Spandau, Berlin
| | - Andreas Könnecke
- Klinik für Dermatologie und Venerologie, Vivantes Klinikum Neukölln, Berlin
| | - Verena Müller
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Mannheim, Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim.,Klinische Kooperationseinheit Dermato-Onkologie, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Deutschland
| | - Jochen Utikal
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Mannheim, Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim.,Klinische Kooperationseinheit Dermato-Onkologie, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Deutschland
| | - Uwe Hillen
- Klinik für Dermatologie und Venerologie, Vivantes Klinikum Neukölln, Berlin
| | - Wolfgang Harth
- Klinik für Dermatologie und Allergologie, Vivantes Klinikum Spandau, Berlin
| | - Wiebke K Peitsch
- Klinik für Dermatologie und Phlebologie, Vivantes Klinikum im Friedrichshain, Berlin
| |
Collapse
|
680
|
Fujisawa Y, Ito T, Kato H, Irie H, Kaji T, Maekawa T, Asai J, Yamamoto Y, Fujimura T, Nakai Y, Yasuda M, Matsuyama K, Muto I, Matsushita S, Uchi H, Nakamura Y, Uehara J, Yoshino K. Outcome of combination therapy using BRAF and MEK inhibitors among Asian patients with advanced melanoma: An analysis of 112 cases. Eur J Cancer 2021; 145:210-220. [PMID: 33503528 DOI: 10.1016/j.ejca.2020.12.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/20/2020] [Accepted: 12/14/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND As most clinical trials evaluating BRAF and MEK inhibitor combination therapy (B + Minh) have been conducted in Western countries, little is known about the effect of B + Minh among East Asian populations. MATERIAL AND METHODS Data from patients with advanced melanoma treated using B + Minh (either dabrafenib + trametinib or encorafenib + binimetinib) were retrospectively collected from 16 institutes in Japan. Response rates, adverse events, patterns of failure and survival were analysed. RESULTS We analysed 112 of 144 collected patient records and, of these, 14 had acral/mucosal melanoma. The response rate for the entire cohort was 75.0%. There were no statistical differences in response rates between acral/mucosal and cutaneous melanomas (64.3% versus 76.5%), whereas previous treatment using immune checkpoint inhibitors (ICIs) did not affect response (72.7% versus 73.9%) to B + Minh, response to ICI after B + Minh was only 20%. Patients who achieved complete response had the best overall survival rates at 24 months (94.7%). Elevated serum lactate dehydrogenase levels and 3 or more metastatic sites were independently associated with survival. The most common relapse site was the brain (17.9%). More than half of the patients (58.8%) experienced grade III/IV pyrexia. CONCLUSION B + Minh was effective among Japanese patients with melanoma, including those with acral/mucosal melanoma. Factors associated with survival were similar to previous Western studies. B + Minh response was not affected by the previous use of ICI; however, vigilance against brain metastasis during B + Minh therapy is required as the brain was our most commonly encountered relapse site.
Collapse
Affiliation(s)
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Hiroshi Kato
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Hiroyuki Irie
- Department of Dermatology, Kyoto University Graduate School of Medicine, Japan
| | - Tatsuya Kaji
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan
| | - Takeo Maekawa
- Department of Dermatology, Jichi Medical University, Japan
| | - Jun Asai
- Department of Dermatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Yuki Yamamoto
- Department of Dermatology, Wakayama Medical University, Japan
| | - Taku Fujimura
- Department of Dermatology,Tohoku University Graduate School of Medicine, Japan
| | - Yasuo Nakai
- Department of Dermatology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | | | | | - Ikko Muto
- Department of Dermatology, Kurume University School of Medicine, Japan
| | - Shigeto Matsushita
- Department of Dermato-Oncology / Dermatology, National Hospital Organization Kagoshima Medical Center, Kagoshima, Japan
| | - Hiroshi Uchi
- Department Dermato-Oncology, National Hospital Organization Kyushu Cancer Center, Japan
| | | | - Jiro Uehara
- Department of Dermatologic Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Japan
| | - Koji Yoshino
- Department of Dermatologic Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Japan
| |
Collapse
|
681
|
Phadke M, Ozgun A, Eroglu Z, Smalley KSM. Melanoma brain metastases: Biological basis and novel therapeutic strategies. Exp Dermatol 2021; 31:31-42. [PMID: 33455008 DOI: 10.1111/exd.14286] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/21/2020] [Accepted: 01/13/2021] [Indexed: 01/09/2023]
Abstract
The development of brain metastases is the deadliest complication of advanced melanoma and has long been associated with a dismal prognosis. The recent years have seen incredible progress in the development of therapies for melanoma brain metastases (MBM), with both targeted therapies (the BRAF-MEK inhibitor combination) and immune checkpoint inhibitors (the anti-CTLA-4, anti-PD-1 combination) showing impressive levels of activity. Despite this, durations of response for these therapies remain lower at intracranial sites of metastasis compared to extracranial metastases and it has been suggested that there are unique features of the brain microenvironment that contribute to therapeutic escape. In this review, we outline the latest research into the biology and pathophysiology of melanoma brain metastasis development and progression. We then discuss the current status of clinical trial that are open to patients with MBM and end by describing the ongoing challenges for the field.
Collapse
Affiliation(s)
- Manali Phadke
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Alpaslan Ozgun
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Zeynep Eroglu
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Keiran S M Smalley
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, Tampa, FL, USA.,The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
682
|
Hernández-Lemus E, Martínez-García M. Pathway-Based Drug-Repurposing Schemes in Cancer: The Role of Translational Bioinformatics. Front Oncol 2021; 10:605680. [PMID: 33520715 PMCID: PMC7841291 DOI: 10.3389/fonc.2020.605680] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer is a set of complex pathologies that has been recognized as a major public health problem worldwide for decades. A myriad of therapeutic strategies is indeed available. However, the wide variability in tumor physiology, response to therapy, added to multi-drug resistance poses enormous challenges in clinical oncology. The last years have witnessed a fast-paced development of novel experimental and translational approaches to therapeutics, that supplemented with computational and theoretical advances are opening promising avenues to cope with cancer defiances. At the core of these advances, there is a strong conceptual shift from gene-centric emphasis on driver mutations in specific oncogenes and tumor suppressors-let us call that the silver bullet approach to cancer therapeutics-to a systemic, semi-mechanistic approach based on pathway perturbations and global molecular and physiological regulatory patterns-we will call this the shrapnel approach. The silver bullet approach is still the best one to follow when clonal mutations in driver genes are present in the patient, and when there are targeted therapies to tackle those. Unfortunately, due to the heterogeneous nature of tumors this is not the common case. The wide molecular variability in the mutational level often is reduced to a much smaller set of pathway-based dysfunctions as evidenced by the well-known hallmarks of cancer. In such cases "shrapnel gunshots" may become more effective than "silver bullets". Here, we will briefly present both approaches and will abound on the discussion on the state of the art of pathway-based therapeutic designs from a translational bioinformatics and computational oncology perspective. Further development of these approaches depends on building collaborative, multidisciplinary teams to resort to the expertise of clinical oncologists, oncological surgeons, and molecular oncologists, but also of cancer cell biologists and pharmacologists, as well as bioinformaticians, computational biologists and data scientists. These teams will be capable of engaging on a cycle of analyzing high-throughput experiments, mining databases, researching on clinical data, validating the findings, and improving clinical outcomes for the benefits of the oncological patients.
Collapse
Affiliation(s)
- Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mireya Martínez-García
- Sociomedical Research Unit, National Institute of Cardiology “Ignacio Chávez”, Mexico City, Mexico
| |
Collapse
|
683
|
Response to BRAF and MEK1/2 inhibition in a young adult with BRAF V600E mutant epithelioid glioblastoma multiforme: A Case Report and Literature Review. Curr Probl Cancer 2021; 45:100701. [PMID: 33461766 DOI: 10.1016/j.currproblcancer.2020.100701] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/15/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022]
Abstract
Epithelioid glioblastoma multiforme (eGBM) is a rare and aggressive variant of glioblastoma multiforme (GBM) that predominantly affects younger patients and can be difficult to distinguish from other gliomas. Data on how patients with eGBM might be best treated are limited, although genomic analyses have shown that almost half of tumours harbour activating BRAF gene mutations. Here we present the case of a young female with BRAF V600E-mutant eGBM who had a prolonged response to targeted therapy with the BRAF and MEK1/2 inhibitors dabrafenib and trametinib. We review current knowledge about eGBM, including the emerging role for BRAF- ± MEK1/2- targeted therapy.
Collapse
|
684
|
Murciano-Goroff YR, Drilon A, Stadler ZK. The NCI-MATCH: A National, Collaborative Precision Oncology Trial for Diverse Tumor Histologies. Cancer Cell 2021; 39:22-24. [PMID: 33434511 PMCID: PMC10640715 DOI: 10.1016/j.ccell.2020.12.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The NCI-MATCH is a national master protocol trial, published in the Journal of Clinical Oncology, in which diverse tumors are sequenced and patients assigned to treatment. The trial demonstrates the feasibility of identifying rare and common actionable genetic alterations and underscores the strength of academic/community partnerships for improving trial access.
Collapse
Affiliation(s)
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
685
|
Ahmed R, Muralidharan R, Srivastava A, Johnston SE, Zhao YD, Ekmekcioglu S, Munshi A, Ramesh R. Molecular Targeting of HuR Oncoprotein Suppresses MITF and Induces Apoptosis in Melanoma Cells. Cancers (Basel) 2021; 13:cancers13020166. [PMID: 33418925 PMCID: PMC7825065 DOI: 10.3390/cancers13020166] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 01/14/2023] Open
Abstract
Simple Summary The human antigen R (HuR) protein regulates the expression of hundreds of proteins in a cell that support tumor growth, drug resistance, and metastases. HuR is overexpressed in several human cancers, including melanoma, and is a molecular target for cancer therapy. Our study objective, therefore, was to develop HuR-targeted therapy for melanoma. We identified that HuR regulates the microphthalmia-associated transcription factor (MITF) that has been implicated in both intrinsic and acquired drug resistance in melanoma and is a putative therapeutic target in melanoma. Using a gene therapeutic approach, we demonstrated silencing of HuR reduced MITF protein expression and inhibited the growth of melanoma cells but not normal melanocytes. However, combining HuR-targeted therapy with a small molecule MEK inhibitor suppressed MITF and produced a synergistic antitumor activity against melanoma cells. Our study results demonstrate that HuR is a promising target for melanoma treatment and offers new combinatorial treatment strategies for overriding MITF-mediated drug resistance. Abstract Background: Treatment of metastatic melanoma possesses challenges due to drug resistance and metastases. Recent advances in targeted therapy and immunotherapy have shown clinical benefits in melanoma patients with increased survival. However, a subset of patients who initially respond to targeted therapy relapse and succumb to the disease. Therefore, efforts to identify new therapeutic targets are underway. Due to its role in stabilizing several oncoproteins’ mRNA, the human antigen R (HuR) has been shown as a promising molecular target for cancer therapy. However, little is known about its potential role in melanoma treatment. Methods: In this study, we tested the impact of siRNA-mediated gene silencing of HuR in human melanoma (MeWo, A375) and normal melanocyte cells in vitro. Cells were treated with HuR siRNA encapsulated in a lipid nanoparticle (NP) either alone or in combination with MEK inhibitor (U0126) and subjected to cell viability, cell-cycle, apoptosis, Western blotting, and cell migration and invasion assays. Cells that were untreated or treated with control siRNA-NP (C-NP) were included as controls. Results: HuR-NP treatment significantly reduced the expression of HuR and HuR-regulated oncoproteins, induced G1 cell cycle arrest, activated apoptosis signaling cascade, and mitigated melanoma cells’ aggressiveness while sparing normal melanocytes. Furthermore, we demonstrated that HuR-NP treatment significantly reduced the expression of the microphthalmia-associated transcription factor (MITF) in both MeWo and MITF-overexpressing MeWo cells (p < 0.05). Finally, combining HuR-NP with U0126 resulted in synergistic antitumor activity against MeWo cells (p < 0.01). Conclusion: HuR-NP exhibited antitumor activity in melanoma cells independent of their oncogenic B-RAF mutational status. Additionally, combinatorial therapy incorporating MEK inhibitor holds promise in overriding MITF-mediated drug resistance in melanoma.
Collapse
Affiliation(s)
- Rebaz Ahmed
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.A.); (R.M.); (A.S.)
- Graduate Program in Biomedical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ranganayaki Muralidharan
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.A.); (R.M.); (A.S.)
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (Y.D.Z.); (A.M.)
| | - Akhil Srivastava
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.A.); (R.M.); (A.S.)
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (Y.D.Z.); (A.M.)
| | - Sarah E. Johnston
- Department of Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Yan D. Zhao
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (Y.D.Z.); (A.M.)
- Department of Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Suhendan Ekmekcioglu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Anupama Munshi
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (Y.D.Z.); (A.M.)
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Rajagopal Ramesh
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.A.); (R.M.); (A.S.)
- Graduate Program in Biomedical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (Y.D.Z.); (A.M.)
- Correspondence: ; Tel.: +1-405-271-6101
| |
Collapse
|
686
|
Han J, Liu Y, Yang S, Wu X, Li H, Wang Q. MEK inhibitors for the treatment of non-small cell lung cancer. J Hematol Oncol 2021; 14:1. [PMID: 33402199 PMCID: PMC7786519 DOI: 10.1186/s13045-020-01025-7] [Citation(s) in RCA: 197] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
BRAF and KRAS are two key oncogenes in the RAS/RAF/MEK/MAPK signaling pathway. Concomitant mutations in both KRAS and BRAF genes have been identified in non-small cell lung cancer (NSCLC). They lead to the proliferation, differentiation, and apoptosis of tumor cells by activating the RAS/RAF/MEK/ERK signaling pathway. To date, agents that target RAS/RAF/MEK/ERK signaling pathway have been investigated in NSCLC patients harboring BRAF mutations. BRAF and MEK inhibitors have gained approval for the treatment of patients with NSCLC. According to the reported findings, the combination of MEK inhibitors with chemotherapy, immune checkpoint inhibitors, epidermal growth factor receptor-tyrosine kinase inhibitors or BRAF inhibitors is highly significant for improving clinical efficacy and causing delay in the occurrence of drug resistance. This review summarized the existing experimental results and presented ongoing clinical studies as well. However, further researches need to be conducted to indicate how we can combine other drugs with MEK inhibitors to significantly increase therapeutic effects on patients with lung cancer.
Collapse
Affiliation(s)
- Jing Han
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Sen Yang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Xuan Wu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Hongle Li
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China.
| | - Qiming Wang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China.
| |
Collapse
|
687
|
Abstract
About half of all cutaneous melanomas harbor activating mutations in the BRAF oncogene. Dependence on this pathway makes the tumors vulnerable to BRAF (and downstream MEK) inhibition, and three drug combinations are approved to target this vulnerability in advanced melanomas with BRAFV600 mutations. Responses to BRAF/MEK inhibitors are usually fast, but durability of response can be limited. Five-year data from BRAF/MEK inhibitors show long-term survival benefit for a third of the patients. There is a wide variety of known mechanisms of resistance to BRAF/MEK inhibition, such as mitogen-activated protein kinase reactivation, activation of parallel pathways, alterations in cell-cycle regulation, and non-genetic resistance mechanisms. Strategies that have been explored to overcome these mechanisms include alternative dosing regimens, addition of another kinase inhibitor, and use of anti-PD-1 immunotherapy either in combination or post-relapse on BRAF/MEK inhibitor therapies.
Collapse
|
688
|
Rutkowski P, Mandalà M. New Therapies in Advanced Cutaneous Malignancies: Conclusions. NEW THERAPIES IN ADVANCED CUTANEOUS MALIGNANCIES 2021:441-448. [DOI: 10.1007/978-3-030-64009-5_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
689
|
Fusco MJ, Piña Y, Macaulay RJ, Sahebjam S, Forsyth PA, Peguero E, Walko CM. Durable Progression-Free Survival With the Use of BRAF and MEK Inhibitors in Four Cases With BRAF V600E-Mutated Gliomas. Cancer Control 2021; 28:10732748211040013. [PMID: 34620004 PMCID: PMC8506147 DOI: 10.1177/10732748211040013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION BRAF V600 E mutations have been identified in a subset of patients with primary brain tumors. Combination therapy with BRAF and Mitogen-activated protein kinase (MEK) inhibitors (BRAF/MEKi) targeting sequential steps in the MAPK pathway has replaced BRAFi monotherapy as the standard of care in multiple tumors with BRAF V600 E mutations, and clinical evidence for this strategy continues to grow in primary brain tumors. CASE SERIES We describe four patients with BRAF V600 E mutated gliomas, including a 21-year-old woman with a ganglioglioma WHO grade I, a 19-year-old man with a pleomorphic xanthoastrocytoma WHO grade III, and 21-year-old and 33-year-old women with epithelioid GBM WHO grade IV, who achieved durable progression-free survival with combination BRAF/MEKi. CONCLUSION Combination of BRAF/MEK inhibition can be a novel, promising approach as targeted therapy in gliomas with BRAF V600 E mutations, especially those that are resistant to standard therapy. Our cases, along with other early reports utilizing dabrafenib/trametinib, highlight the importance of somatic next-generation sequencing, particularly in younger patients. Interim results from clinical trials utilizing dabrafenib/trametinib have been promising thus far, and our case series suggests that durable clinical benefit is possible, even in the setting of glioblastoma, WHO grade IV.
Collapse
Affiliation(s)
- Michael J. Fusco
- Department of Individualized Cancer Medicine, Moffitt Cancer Center, Tampa, FL, USA
| | - Yolanda Piña
- Department of Neuro-Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Robert J. Macaulay
- Department of Neuro-Oncology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Anatomic Pathology, Moffitt Cancer Center, Tampa, FL, USA
| | - Solmaz Sahebjam
- Department of Neuro-Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Peter A. Forsyth
- Department of Neuro-Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Edwin Peguero
- Department of Neuro-Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Christine M. Walko
- Department of Individualized Cancer Medicine, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
690
|
Höppener DJ, Grünhagen DJ, Eggermont AMM, van der Veldt AAM, Verhoef C. An Overview of Liver Directed Locoregional Therapies. Surg Oncol Clin N Am 2021; 30:103-123. [PMID: 33220800 DOI: 10.1016/j.soc.2020.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
An overview of all liver-directed locoregional therapies, including surgical resection for melanoma liver metastases (MLMs), is provided. MLM patients are divided by their primary melanoma location; cutaneous, uvea (eye), and mucosal melanoma. If patients with isolated cutaneous MLMs are considered for surgical resection, treatment with systemic therapy should be part of the treatment course. For uveal MLMs, complete surgical or ablative treatment of all MLMs suggests superior results compared with other liver-directed or systemic therapies, based on current evidence, no recommendations for any liver-directed regional therapy in the treatment of mucosal MLMs can be made.
Collapse
Affiliation(s)
- Diederik J Höppener
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Dirk J Grünhagen
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Alexander M M Eggermont
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands.
| |
Collapse
|
691
|
Wahler S, Müller A, Koll C, Seyed-Abbaszadeh P, Von Der Schulenburg JM. Economic evaluation of adverse events of dabrafenib plus trametinib versus nivolumab in patients with advanced BRAF-mutant cutaneous melanoma for adjuvant therapy in Germany. JOURNAL OF MARKET ACCESS & HEALTH POLICY 2020; 9:1861804. [PMID: 33456727 PMCID: PMC7781974 DOI: 10.1080/20016689.2020.1861804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 05/30/2023]
Abstract
Background: Adjuvant treatment options have become the standard therapy for stage III and IV resectable cutaneous melanoma. Two recent studies led to the registration of dabrafenib and trametinib as targeted therapies for BRAF-mutated melanoma, and of immunotherapy with nivolumab irrespective of BRAF-mutation status. Both therapies have different spectrums of adverse events. Objective: To estimate the financial impact of side effects from the perspective of the German statutory sick funds to compare both therapeutic options and to relate the burden to the overall costs of the treatment. STUDY DESIGN AND SETTING Thirty-six adverse event categories for the combination of dabrafenib and trametinib ('combi treatment') and for nivolumab were extracted from the original publications of the studies named COMBI-AD and CheckMate 238. PATIENTS AND INTERVENTION For all event categories a diagnosis and therapy recommendation were determined according to current national or international guidelines or from leading German textbooks. MAIN OUTCOME MEASURE The resulting diagnostic steps, treatments, and therapies were evaluated with unit costs based on the German fee schedule for ambulatory physicians, the German G-DRG scheme, and the German drug price list. RESULTS The number of events with nivolumab per one hundred treatments amounted to 3.8 mandatory hospitalizations, 3.5 emergency care events and 0.8 life-threatening events. For the combi treatment, the respective number of events per one hundred treatments was 2.7, 1.8, and 0.5. The overall cost burden was calculated as €899 for nivolumab and €861 for combi-treatment. CONCLUSION The treatment of adverse events resulting from adjuvant melanoma therapy showed comparable costs for both therapies.
Collapse
Affiliation(s)
- S Wahler
- St. Bernward GmbH, Hamburg, Germany
| | - A Müller
- Analytics Services GmbH, Munich, Germany
| | - C Koll
- Diabetes Praxis Blankenese, Hamburg, Germany
| | | | | |
Collapse
|
692
|
Ali SR, Dobbs TD, Slade R, Whitaker IS. Multidimensional indicators of scholarly impact in the skin oncology literature: is there a correlation between bibliometric and altmetric profiles? J Plast Surg Hand Surg 2020; 55:232-241. [PMID: 33356756 DOI: 10.1080/2000656x.2020.1858842] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Bibliometric and altmetric analyses are used to identify landmark publications in their respective research field. We hypothesised that highly cited skin oncology articles correlate positively with the Oxford Evidence Based Medicine scoring level, altmetric score (AS) and rank within the top 100 manuscripts.Methods: Thomson Reuter's Web of Science citation indexing database was searched to identify all English-language skin oncology full-text articles in the last 75 years. The top 100 articles with the highest citation count were analysed by subject matter, publishing journal, author, year, institution, individual and five-year impact factor, AS and Oxford EBM level. Results: 180,132 articles were identified. The most cited article (Hodi et al.) demonstrated improved survival with ipilimumab in patients with metastatic melanoma (7894 citations). The article with the highest AS was Esteva et al. (AS = 576.7, 'dermatologist-level classification of skin cancer with deep neural networks'). No difference was found between evidence level and citation count (r = -0.1239, p = 0.2291), but a significant difference was seen for AS (r = -0.3024, p = 0.0028). AS scores increased over time, whereas bibliometrics did not. Conclusion: This work highlights the most influential work in the skin oncology field in the last 75 years. We have identified a differential relationship between commonly used metrics and evidence level in the field of skin oncology. As the digitalisation of research output and consumption increases, both bibliometric and altmetric analyses need to be considered when an article's impact is being assessed.
Collapse
Affiliation(s)
- Stephen R Ali
- Reconstructive Surgery and Regenerative Medicine Research Group, Institute of Life Sciences, Swansea University Medical School, Swansea, UK.,Welsh Centre for Burns and Plastic Surgery Morriston Hospital, Swansea, UK
| | - Thomas D Dobbs
- Reconstructive Surgery and Regenerative Medicine Research Group, Institute of Life Sciences, Swansea University Medical School, Swansea, UK.,Welsh Centre for Burns and Plastic Surgery Morriston Hospital, Swansea, UK
| | - Robert Slade
- Welsh Centre for Burns and Plastic Surgery Morriston Hospital, Swansea, UK
| | - Iain S Whitaker
- Reconstructive Surgery and Regenerative Medicine Research Group, Institute of Life Sciences, Swansea University Medical School, Swansea, UK.,Welsh Centre for Burns and Plastic Surgery Morriston Hospital, Swansea, UK
| |
Collapse
|
693
|
Seethapathy H, Lee MD, Strohbehn IA, Efe O, Rusibamayila N, Chute DF, Colvin RB, Rosales IA, Fadden RM, Reynolds KL, Sullivan RJ, Kaufman HL, Jhaveri KD, Sise ME. Clinical features of acute kidney injury in patients receiving dabrafenib and trametinib. Nephrol Dial Transplant 2020; 37:507-514. [PMID: 33355659 DOI: 10.1093/ndt/gfaa372] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To characterize the incidence, risk factors, and clinical features of acute kidney injury (AKI) in patients receiving dabrafenib and trametinib. METHODS We performed a retrospective cohort study examining the kidney outcomes of patients in a large healthcare system who received dabrafenib/trametinib between 2010 and 2019. The primary outcome was AKI, defined as a 1.5-fold increase in serum creatinine from baseline within a 12-month study period. AKI severity and etiology was determined for each case by chart review. Logistic regression was used to evaluate baseline predictors of AKI. RESULTS 199 patients who received dabrafenib in our healthcare system from 2010-2019 were included in the analysis. Forty-two patients (21%) experienced AKI within 12 months; 10 patients (5% of total cohort, 24% of AKI) experienced AKI occurring during a dabrafenib/trametinib-induced febrile syndrome characterized by fever, chills, gastrointestinal symptoms, and elevated liver enzymes. Pre-existing liver disease was the only significant predictor of AKI in the cohort. One patient had biopsy-proven granulomatous acute interstitial nephritis that resolved with corticosteroids. CONCLUSIONS Oncologists and nephrologists should be aware that AKI is common after dabrafenib/trametinib and a substantial number of cases occur in the setting of treatment-induced pyrexia.
Collapse
Affiliation(s)
- Harish Seethapathy
- Division of Nephrology, Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Meghan D Lee
- Division of Nephrology, Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Ian A Strohbehn
- Division of Nephrology, Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Orhan Efe
- Division of Nephrology, Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Nifasha Rusibamayila
- Division of Nephrology, Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Donald F Chute
- Division of Nephrology, Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Robert B Colvin
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Ivy A Rosales
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Riley M Fadden
- Division of Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kerry L Reynolds
- Division of Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryan J Sullivan
- Division of Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Howard L Kaufman
- Division of Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kenar D Jhaveri
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Division of Kidney Diseases and Hypertension, Great Neck, NY, USA
| | - Meghan E Sise
- Division of Nephrology, Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
694
|
Clinical Implications of Acquired BRAF Inhibitors Resistance in Melanoma. Int J Mol Sci 2020; 21:ijms21249730. [PMID: 33419275 PMCID: PMC7766699 DOI: 10.3390/ijms21249730] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Understanding the role of mitogen-activated protein kinase (MAPK) pathway-activating mutations in the development and progression of melanoma and their possible use as therapeutic targets has substantially changed the management of this neoplasm, which, until a few years ago, was burdened by severe mortality. However, the presence of numerous intrinsic and extrinsic mechanisms of resistance to BRAF inhibitors compromises the treatment responses’ effectiveness and durability. The strategy of overcoming these resistances by combination therapy has proved successful, with the additional benefit of reducing side effects derived from paradoxical activation of the MAPK pathway. Furthermore, the use of other highly specific inhibitors, intermittent dosing schedules and the association of combination therapy with immune checkpoint inhibitors are promising new therapeutic strategies. However, numerous issues related to dose, tolerability and administration sequence still need to be clarified, as is to be expected from currently ongoing trials. In this review, we describe the clinical results of using BRAF inhibitors in advanced melanoma, with a keen interest in strategies aimed at overcoming resistance.
Collapse
|
695
|
Kun E, Tsang YTM, Ng CW, Gershenson DM, Wong KK. MEK inhibitor resistance mechanisms and recent developments in combination trials. Cancer Treat Rev 2020; 92:102137. [PMID: 33340965 DOI: 10.1016/j.ctrv.2020.102137] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/26/2020] [Accepted: 11/29/2020] [Indexed: 02/07/2023]
Abstract
The mitogen-activated protein kinase (MAPK) pathway plays a vital role in cellular processes such as gene expression, cell proliferation, cell survival, and apoptosis. Also known as the RAS-RAF-MEK-ERK pathway, the MAPK pathway has been implicated in approximately one-third of all cancers. Mutations in RAS or RAF genes such as KRAS and BRAF are common, and these mutations typically promote malignancies by over-activating MEK and ERK downstream, which drives sustained cell proliferation and uninhibited cell growth. Development of drugs targeting this pathway has been a research area of great interest, especially drugs targeting the inhibition of MEK. In vitro and clinical studies have shown promise for certain MEK inhibitors (MEKi) , and MEKi have become the first treatment option for certain cancers. Despite promising results, not all patients have a response to MEKi, and mechanisms of resistance typically arise in patients who do have a positive initial response. This paper summarizes recent developments regarding MEKi, the mechanisms of adaptive resistance to MEKi, and the potential solutions to the issue of adaptive MEKi resistance.
Collapse
Affiliation(s)
- E Kun
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Y T M Tsang
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - C W Ng
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - D M Gershenson
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - K K Wong
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
696
|
PD-L1 blockade in combination with inhibition of MAPK oncogenic signaling in patients with advanced melanoma. Nat Commun 2020; 11:6262. [PMID: 33288749 PMCID: PMC7721806 DOI: 10.1038/s41467-020-19810-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
Combining PD-L1 blockade with inhibition of oncogenic mitogen-activated protein kinase (MAPK) signaling may result in long-lasting responses in patients with advanced melanoma. This phase 1, open-label, dose-escalation and -expansion study (NCT02027961) investigated safety, tolerability and preliminary efficacy of durvalumab (anti–PD-L1) combined with dabrafenib (BRAF inhibitor) and trametinib (MEK inhibitor) for patients with BRAF-mutated melanoma (cohort A, n = 26), or durvalumab and trametinib given concomitantly (cohort B, n = 20) or sequentially (cohort C, n = 22) for patients with BRAF-wild type melanoma. Adverse events and treatment discontinuation rates were more common than previously reported for these agents given as monotherapy. Objective responses were observed in 69.2% (cohort A), 20.0% (cohort B) and 31.8% (cohort C) of patients, with evidence of improved tumor immune infiltration and durable responses in a subset of patients with available biopsy samples. In conclusion, combined MAPK inhibition and anti–PD-L1 therapy may provide treatment options for patients with advanced melanoma. Immune checkpoints inhibitors or MAPK inhibitors are currently used as standard of care therapies for patients with advanced melanoma. Here the authors report a phase 1 clinical trial testing the anti-PD-L1 antibody durvalumab in combination with the BRAF inhibitor dafrafenib and the MEK inhibitor trametinib in patients with BRAFV600-mutant melanoma.
Collapse
|
697
|
Lee S, Suh HB, Choi SJ, Kang J, Kang JW, Kwon EJ, Kim HJ, Kim YH, Shin K. Identification of prognostic mRNAs in metastatic cutaneous melanoma. Melanoma Res 2020; 30:543-547. [PMID: 33003118 DOI: 10.1097/cmr.0000000000000697] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cutaneous melanoma is the most common cause of skin cancer-related deaths worldwide. There is an urgent need to identify prognostic biomarkers to facilitate decision-making for treatment of metastatic cutaneous melanoma. Gene expression microarrays and RNA-seq technology have recently improved or changed current prognostic and therapeutic strategies for several cancers. However, according to the current melanoma staging system, prognosis is almost entirely dependent on clinicopathological features. To identify novel prognostic biomarkers, we investigated gene expression and clinical data for patients with cutaneous melanoma from three cohorts of The Cancer Genome Atlas and Gene Expression Omnibus. Kaplan-Meier survival analysis using median values of each gene as cutoff value revealed that nine genes (ABCC3, CAPS2, CCR6, CDCA8, CLU, DPF1, PTK2B, SATB1, and SYNE1) were statistically significant prognostic biomarkers of metastatic cutaneous melanoma in all three independent cohorts. Low expression of two genes (CDCA8 and DPF1) and high expression of seven genes (ABCC3, CAPS2, CCR6, CLU, PTK2B, SATB1, and SYNE) were significantly associated with positive metastatic cutaneous melanoma prognoses. In conclusion, we suggest nine novel prognostic biomarkers for cutaneous metastatic melanoma.
Collapse
Affiliation(s)
| | - Hie Bum Suh
- Department of Radiology, School of Medicine, Pusan National University
- Biomedical Research Institute, Pusan National University Hospital
| | | | | | | | | | | | - Yun Hak Kim
- Department of Biomedical Informatics
- Department of Anatomy, School of Medicine, Pusan National University
| | - Kihyuk Shin
- Biomedical Research Institute, Pusan National University Hospital
- Department of Dermatology
- Department of Dermatology, Pusan National University Yangsan Hospital
- Research Institute for Convergence of Biomedical Science and Technology, Yangsan, Korea
| |
Collapse
|
698
|
Braig ZV, Tagliero AJ, Rose PS, Elhassan BT, Barlow JD, Wagner ER, Sanchez-Sotelo J, Houdek MT. Humeral stress shielding following cemented endoprosthetic reconstruction: An under-reported complication? J Surg Oncol 2020; 123:505-509. [PMID: 33259663 DOI: 10.1002/jso.26300] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 01/20/2023]
Abstract
INTRODUCTION The proximal humerus is a common location for primary and non-primary tumors. Reconstruction of the proximal humerus is commonly performed with an endoprosthesis with low rates of structural failure. The incidence and risk factors for stress shielding are under reported. METHODS Thirty-nine (19 male, 20 female) patients underwent resection of the proximal humerus and reconstruction with a cemented modular endoprosthesis between 2000 and 2018. The mean resection length was 12 ± 4 cm and was most commonly performed for metastatic disease (n = 26, 67%). RESULTS Stress shielding was observed in 9 (23%) patients at a mean of 29 (6-132) months postoperatively. Patients with stress shielding were noted to have shorter intramedullary stem length (87 vs. 107 mm, p < .001), longer extramedullary implant length (16 vs. 14 cm, p = .01) and a higher extramedullary implant to stem length ratio (2.1 vs. 1.1, p < .001). The incidence of stress shielding was higher (p = .003) in patients reconstructed with 75 mm stem (n = 6, 67%) lengths. CONCLUSION Stress shielding of the humerus was associated with the use of shorter stems and long extramedullary implants. The long-term ramifications of stress shielding on implant stability, complications at the time of revision surgery, and overall patient outcomes remain unknown.
Collapse
Affiliation(s)
- Zachary V Braig
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Adam J Tagliero
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter S Rose
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Bassem T Elhassan
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Jonathan D Barlow
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Eric R Wagner
- Department of Orthopedic Surgery, Emory University, Atlanta, Georgia, USA
| | | | - Matthew T Houdek
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
699
|
LeBlanc RE, Lefferts JA, Baker ML, Linos KD. Novel LRRFIP2-RAF1 fusion identified in an acral melanoma: A review of the literature on melanocytic proliferations with RAF1 fusions and the potential therapeutic implications. J Cutan Pathol 2020; 47:1181-1186. [PMID: 32700768 DOI: 10.1111/cup.13817] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 11/29/2022]
Abstract
A small subset of cutaneous melanomas harbor oncogenic gene fusions, which could potentially serve as therapeutic targets for patients with advanced disease as novel therapies are developed. Fusions involving RAF1 are exceedingly rare in melanocytic neoplasms, occurring in less than 1% of melanomas, and usually arise in tumors that are wild type for BRAF, NRAS, and NF1. We describe herein a case of acral melanoma with two satellite metastases and sentinel lymph node involvement. The melanoma had a concomitant KIT variant and LRRFIP2-RAF1 fusion. This constellation of molecular findings has not been reported previously in melanoma. We review the existing literature on melanocytic neoplasms with RAF1 fusions and discuss the potential clinical implications of this genetic event.
Collapse
Affiliation(s)
- Robert E LeBlanc
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Joel A Lefferts
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Michael L Baker
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Konstantinos D Linos
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| |
Collapse
|
700
|
Pratt EC, Isaac E, Stater EP, Yang G, Ouerfelli O, Pillarsetty N, Grimm J. Synthesis of the PET Tracer 124I-Trametinib for MAPK/ERK Kinase Distribution and Resistance Monitoring. J Nucl Med 2020; 61:1845-1850. [PMID: 32444378 PMCID: PMC9364901 DOI: 10.2967/jnumed.120.241901] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/16/2020] [Indexed: 01/19/2023] Open
Abstract
Trametinib is an extremely potent allosteric inhibitor of mitogen-activated protein kinase (MAPK)/extracellular-signal-regulated kinase (ERK) (MEK) 1/2, which has been approved for treatment of metastatic melanoma and anaplastic thyroid cancer in patients with confirmed BRAFV600E/K mutations. Though trametinib is highly efficacious, adverse side effects, including skin, gastrointestinal, and hepatic toxicity, are dose-limiting and can lead to treatment termination. Development of a noninvasive tool to visualize and quantify the delivery and distribution of trametinib (either as a single agent or in combination with other therapeutics) to tumors and organs would be helpful in assessing therapeutic index, personalizing individual dose, and potentially predicting resistance to therapy. Methods: To address these issues, we have developed a radiolabeled trametinib and evaluated the in vitro and in vivo properties. 123I-, 124I-, and 131I-trametinib, pure tracer analogs to trametinib, were synthesized in more than 95% purity, with an average yield of 69.7% and more than 100 GBq/μmol specific activity. Results: Overall, 124I-trametinib uptake in a panel of cancer cell lines can be blocked with cold trametinib, confirming specificity of the radiotracer in vitro and in vivo. 124I-trametinib was taken up at higher rates in KRAS and BRAF mutant cell lines than in wild-type KRAS cancer cell lines. In vivo, biodistribution revealed high uptake in the liver 2 h after injection, followed by clearance through the gastrointestinal tract over 4 d. Importantly, uptake higher than expected was observed in the lung and heart for up to 24 h. Peak uptake in the skin and gastrointestinal tract was observed between 6 and 24 h, whereas in B16F10 melanoma-bearing mice peak tumor concentrations were achieved between 24 and 48 h. Tumor uptake relative to muscle and skin was relatively low, peaking at 3.4- to 8.1-fold by 72 h, respectively. The biodistribution of 124I-trametinib was significantly reduced in mice on trametinib therapy, providing a quantitative method to observe MEK inhibition in vivo. Conclusion:124I-trametinib serves as an in vivo tool to personalize the dose instead of using the current single-fixed-dose scheme and, when combined with radiomic data, to monitor the emergence of therapy resistance. In addition, the production of iodinated trametinib affords researchers the ability to measure drug distribution for improved drug delivery studies.
Collapse
Affiliation(s)
- Edwin C. Pratt
- Department of Pharmacology, Weill Cornell Graduate School, New York, New York,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elizabeth Isaac
- Department of Pharmacology, Weill Cornell Graduate School, New York, New York,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Evan P. Stater
- Department of Pharmacology, Weill Cornell Graduate School, New York, New York,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Guangbin Yang
- Organic Synthesis Core, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Ouathek Ouerfelli
- Organic Synthesis Core, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Nagavarakishore Pillarsetty
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jan Grimm
- Department of Pharmacology, Weill Cornell Graduate School, New York, New York,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|