210496Orig1s000

Total Page:16

File Type:pdf, Size:1020Kb

210496Orig1s000 CENTER FOR DRUG EVALUATION AND RESEARCH APPLICATION NUMBER: 210496Orig1s000 MULTI-DISCIPLINE REVIEW Summary Review Office Director Cross Discipline Team Leader Review Clinical Review Non-Clinical Review Statistical Review Clinical Pharmacology Review Multi-Disciplinary Review and Evaluation NDA 210496 BRAFTOVI™ (encorafenib) NDA Multi-Disciplinary Review and Evaluation Application Type NDA (Original-1) Application Number 210496 Priority or Standard Standard Submit Date June 30, 2017 Received Date June 30, 2017 PDUFA Goal Date June 30, 2018 Division/Office DOP2/OHOP Established Name Encorafenib (Proposed) Trade Name BRAFTOVI™ Pharmacologic Class Kinase inhibitor Code Name LGX818 Applicant Array BioPharma Inc. Formulation 50 mg and 75 mg capsules Dosing Regimen 450 mg orally once daily in combination with binimetinib Applicant Proposed In combination with binimetinib, for the treatment of patients Indication/Population with unresectable or metastatic melanoma with a BRAF V600E or V600K mutation as detected by an FDA-approved test Recommendation on Regular approval Regulatory Action Recommended In combination with binimetinib, for the treatment of patients Indication/Population with unresectable or metastatic melanoma with a BRAF V600E or V600K mutation, as detected by an FDA-approved test 1 Reference ID: 4282494 Multi-Disciplinary Review and Evaluation NDA 210496 BRAFTOVI™ (encorafenib) Table of Contents Reviewers of Multi-Disciplinary Review and Evaluation...............................................................14 Additional Reviewers of Application ............................................................................................15 Glossary ........................................................................................................................................16 1 Executive Summary...............................................................................................................19 1.1. Product Introduction......................................................................................................19 1.2. Conclusions on the Substantial Evidence of Effectiveness.............................................19 1.3. Benefit-Risk Assessment ................................................................................................21 1.4. Patient Experience Data.................................................................................................24 2 Therapeutic Context..............................................................................................................25 2.1. Analysis of Condition......................................................................................................25 2.2. Analysis of Current Treatment Options .........................................................................25 3 Regulatory Background .........................................................................................................29 3.1. U.S. Regulatory Actions and Marketing History.............................................................29 3.2. Summary of Presubmission/Submission Regulatory Activity ........................................29 4 Significant Issues from Other Review Disciplines Pertinent to Clinical Conclusions on Efficacy and Safety ................................................................................................................32 4.1. Office of Scientific Investigations (OSI) ..........................................................................32 4.2. Product Quality ..............................................................................................................32 4.3. Clinical Microbiology......................................................................................................33 4.4. Devices and Companion Diagnostic Issues ....................................................................33 5 Nonclinical Pharmacology/Toxicology ..................................................................................34 5.1. Executive Summary........................................................................................................34 5.2. Referenced NDAs, BLAs, DMFs ......................................................................................36 5.3. Pharmacology ................................................................................................................36 5.4. ADME/PK........................................................................................................................45 5.5. Toxicology ......................................................................................................................47 5.5.1. General Toxicology .................................................................................................47 5.5.2. Genetic Toxicology..................................................................................................52 5.5.3. Carcinogenicity .......................................................................................................53 2 Reference ID: 4282494 Multi-Disciplinary Review and Evaluation NDA 210496 BRAFTOVI™ (encorafenib) 5.5.4. Reproductive and Developmental Toxicology ........................................................54 5.5.5. Other Toxicology Studies ........................................................................................63 6 Clinical Pharmacology ...........................................................................................................64 6.1. Executive Summary........................................................................................................64 6.2. Summary of Clinical Pharmacology Assessment............................................................67 6.2.1. Pharmacology and Clinical Pharmacokinetics.........................................................67 6.2.2. General Dosing and Therapeutic Individualization.................................................68 6.3. Comprehensive Clinical Pharmacology Review..............................................................71 6.3.1. General Pharmacology and Pharmacokinetic Characteristics ................................71 6.3.2. Clinical Pharmacology Questions............................................................................75 7 Sources of Clinical Data and Review Strategy .......................................................................87 7.1. Table of Clinical Studies .................................................................................................87 7.2. Review Strategy .............................................................................................................91 8 Statistical and Clinical and Evaluation...................................................................................92 8.1. Review of Relevant Individual Trials Used to Support Efficacy ......................................92 8.1.1. COLUMBUS – Trial Design.......................................................................................93 8.1.2. LOGIC2 – Trial Design............................................................................................103 8.1.3. COLUMBUS – Study Results..................................................................................105 8.1.4. LOGIC2 – Study Results.........................................................................................129 8.1.5. Assessment of Efficacy Across Trials.....................................................................133 8.1.6. Integrated Assessment of Effectiveness...............................................................137 8.2. Review of Safety...........................................................................................................139 8.2.1. Safety Review Approach .......................................................................................139 8.2.2. Adequacy of Applicant’s Clinical Safety Assessments...........................................144 8.2.3. Safety Results........................................................................................................146 8.2.4. Analysis of Submission-Specific Safety Issues.......................................................173 8.2.5. Clinical Outcome Assessment (COA) Analyses Informing Safety/Tolerability ......203 8.2.6. Safety Analyses by Demographic Subgroups........................................................203 8.2.7. Specific Safety Studies/Clinical Trials....................................................................205 8.2.8. Additional Safety Explorations..............................................................................205 8.2.9. Safety in the Postmarket Setting ..........................................................................206 8.2.10. Integrated Assessment of Safety...................................................................206 3 Reference ID: 4282494 Multi-Disciplinary Review and Evaluation NDA 210496 BRAFTOVI™ (encorafenib) SUMMARY AND CONCLUSIONS..................................................................................................208 8.3. Statistical Issues ...........................................................................................................208 8.4. Conclusions and Recommendations ............................................................................208 9 Advisory Committee Meeting and Other External Consultations .......................................210 10 Pediatrics.............................................................................................................................211
Recommended publications
  • Transcriptomic Analysis of Native Versus Cultured Human and Mouse Dorsal Root Ganglia Focused on Pharmacological Targets Short
    bioRxiv preprint doi: https://doi.org/10.1101/766865; this version posted September 12, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. Transcriptomic analysis of native versus cultured human and mouse dorsal root ganglia focused on pharmacological targets Short title: Comparative transcriptomics of acutely dissected versus cultured DRGs Andi Wangzhou1, Lisa A. McIlvried2, Candler Paige1, Paulino Barragan-Iglesias1, Carolyn A. Guzman1, Gregory Dussor1, Pradipta R. Ray1,#, Robert W. Gereau IV2, # and Theodore J. Price1, # 1The University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson, TX, 75080, USA 2Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine # corresponding authors [email protected], [email protected] and [email protected] Funding: NIH grants T32DA007261 (LM); NS065926 and NS102161 (TJP); NS106953 and NS042595 (RWG). The authors declare no conflicts of interest Author Contributions Conceived of the Project: PRR, RWG IV and TJP Performed Experiments: AW, LAM, CP, PB-I Supervised Experiments: GD, RWG IV, TJP Analyzed Data: AW, LAM, CP, CAG, PRR Supervised Bioinformatics Analysis: PRR Drew Figures: AW, PRR Wrote and Edited Manuscript: AW, LAM, CP, GD, PRR, RWG IV, TJP All authors approved the final version of the manuscript. 1 bioRxiv preprint doi: https://doi.org/10.1101/766865; this version posted September 12, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • Human Kinome Profiling Identifies a Requirement for AMP-Activated
    Human kinome profiling identifies a requirement for AMP-activated protein kinase during human cytomegalovirus infection Laura J. Terrya, Livia Vastagb,1, Joshua D. Rabinowitzb, and Thomas Shenka,2 aDepartment of Molecular Biology and bDepartment of Chemistry and the Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544 Contributed by Thomas Shenk, January 11, 2012 (sent for review December 29, 2011) Human cytomegalovirus (HCMV) modulates numerous cellular (7). Thus, the connections between AMPK activity and metabolic signaling pathways. Alterations in signaling are evident from the changes during HCMV infection have remained unclear. broad changes in cellular phosphorylation that occur during HCMV We confirmed the requirement for AMPK during infection, infection and from the altered activity of multiple kinases. Here we and we show that an AMPK antagonist, compound C, blocks report a comprehensive RNAi screen, which predicts that 106 cellular HCMV-induced changes to glycolysis and inhibits viral gene kinases influence growth of the virus, most of which were not expression. These studies argue that AMPK or a related, com- previously linked to HCMV replication. Multiple elements of the pound C-sensitive kinase is an essential contributor to metabolic AMP-activated protein kinase (AMPK) pathway scored in the screen. changes initiated by HCMV and provide unique insight into As a regulator of carbon and nucleotide metabolism, AMPK is poised potential antiviral strategies. to activate many of the metabolic pathways induced by HCMV infection. An AMPK inhibitor, compound C, blocked a substantial Results portion of HCMV-induced metabolic changes, inhibited the accumu- HumanKinomeScreenIdentifies Putative Effectors of HCMV Replication. lation of all HCMV proteins tested, and markedly reduced the We conducted an siRNA screen of the human kinome to perform an production of infectious progeny.
    [Show full text]
  • Support Info
    Electronic Supplementary Material (ESI) for RSC Advances. This journal is © The Royal Society of Chemistry 2014 Supporting Information Design and synthesis of pyrrole–5-(2,6-dichlorobenzyl)sulfonylindolin-2-ones with C- 3’ side chains as potent Met kinase inhibitors Chia-Wei Liu,a Chun-Liang Lai,a Yu-Hsiang Lin,a Li-Wei Teng,a Sheng-chuan Yang,a Win-Yin Wei,a Shu Fu Lin,a Ju-Ying Yang,a Hung-Jyun Huang,a Ru-Wen Wang,a Chao-Cheng Chiang,a Mei-Hui Lee,a Yu- Chuan Wang,b Shih-Hsien Chuang,a Jia-Ming Chang,a Ying-Shuan E. Lee,a and Jiann-Jyh Huang*a,b aDevelopment Center for Biotechnology, No. 101, Lane 169, Kangning St., Xizhi District, New Taipei City 22180, Taiwan bDepartment of Applied Chemistry, National Chiayi University, No. 300, Syuefu Rd., Chiayi City 60004, Taiwan *Corresponding Author. Tel.: +886 5 271 7959; Fax: +886 5 271 7901. E-mail address: [email protected] (J.-J. Huang) Table of Contents: Page Supporting Figure. Ligplot diagrams of the ATP binding site of Met S2 complexed with compounds 2 and 20. Supporting Table. Kinase profiling data of compound 20. S3 References S10 - S1 - Supporting Figure. Ligplot diagrams1 of the ATP binding site of Met complexed with compounds 2 and 20: (A) Met with 2, and (B) Met with 20. - S2 - Supporting Table. Kinase profiling data of 20. Ambit KinomeScan Kinase Profiling (1.0 μM test concentration): Percentage of Percentage of Ambit Gene Symbol control (%) Ambit Gene Symbol control (%) 20 20 AAK1 68 ARK5 27 ABL1(E255K)-phosphorylated 85 ASK1 100 ABL1(F317I)-nonphosphorylated 78 ASK2 67
    [Show full text]
  • BRAF Inhibitors Amplify the Proapoptotic Activity of MEK
    Published OnlineFirst July 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0098 Cancer Therapy: Preclinical Clinical Cancer Research BRAF Inhibitors Amplify the Proapoptotic Activity of MEK Inhibitors by Inducing ER Stress in NRAS-Mutant Melanoma Heike Niessner1,Tobias Sinnberg1, Corinna Kosnopfel1, Keiran S.M. Smalley2, Daniela Beck1, Christian Praetorius3,4, Marion Mai3, Stefan Beissert3, Dagmar Kulms3,4, Martin Schaller1, Claus Garbe1, Keith T. Flaherty5, Dana Westphal3,4, Ines Wanke1, and Friedegund Meier1,3,6 Abstract Purpose: NRAS mutations in malignant melanoma are asso- anoma. BRAFi such as encorafenib induced an ER stress ciated with aggressive disease requiring rapid antitumor interven- response via the PERK pathway, as detected by phosphorylation tion, but there is no approved targeted therapy for this subset of of eIF2a and upregulation of the ER stress–related factors ATF4, patients. In clinical trials, the MEK inhibitor (MEKi) binimetinib CHOP, and NUPR1 and the proapoptotic protein PUMA. MEKi displayed modest antitumor activity, making combinations a such as binimetinib induced the expression of the proapoptotic requisite. In a previous study, the BRAF inhibitor (BRAFi) vemur- protein BIM and activation of the mitochondrial pathway of afenib was shown to induce endoplasmic reticulum (ER) stress apoptosis, the latter of which was enhanced by combination that together with inhibition of the RAF–MEK–ERK (MAPK) with encorafenib. The increased apoptotic rates caused by the pathway amplified its proapoptotic activity in BRAF-mutant mel- combination treatment were significantly reduced through anoma. The present study investigated whether this effect might siRNA knockdown of ATF4 and BIM, confirming its critical extent to NRAS-mutant melanoma, in which MAPK activation roles in this process.
    [Show full text]
  • Combined BRAF and MEK Inhibition with Vemurafenib and Cobimetinib for Patients with Advanced Melanoma
    Review Melanoma Combined BRAF and MEK Inhibition with Vemurafenib and Cobimetinib for Patients with Advanced Melanoma Antonio M Grimaldi, Ester Simeone, Lucia Festino, Vito Vanella and Paolo A Ascierto Melanoma, Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”, Napoli, Italy cquired resistance is the most common cause of BRAF inhibitor monotherapy treatment failure, with the majority of patients experiencing disease progression with a median progression-free survival of 6-8 months. As such, there has been considerable A focus on combined therapy with dual BRAF and MEK inhibition as a means to improve outcomes compared with monotherapy. In the COMBI-d and COMBI-v trials, combined dabrafenib and trametinib was associated with significant improvements in outcomes compared with dabrafenib or vemurafenib monotherapy, in patients with BRAF-mutant metastatic melanoma. The combination of vemurafenib and cobimetinib has also been investigated. In the phase III CoBRIM study in patients with unresectable stage III-IV BRAF-mutant melanoma, treatment with vemurafenib and cobimetinib resulted in significantly longer progression-free survival and overall survival (OS) compared with vemurafenib alone. One-year OS was 74.5% in the vemurafenib and cobimetinib group and 63.8% in the vemurafenib group, while 2-year OS rates were 48.3% and 38.0%, respectively. The combination was also well tolerated, with a lower incidence of cutaneous squamous-cell carcinoma and keratoacanthoma compared with monotherapy. Dual inhibition of both MEK and BRAF appears to provide a more potent and durable anti-tumour effect than BRAF monotherapy, helping to prevent acquired resistance as well as decreasing adverse events related to BRAF inhibitor-induced activation of the MAPK-pathway.
    [Show full text]
  • Safety of BRAF+MEK Inhibitor Combinations: Severe Adverse Event Evaluation
    cancers Article Safety of BRAF+MEK Inhibitor Combinations: Severe Adverse Event Evaluation Tomer Meirson 1,2 , Nethanel Asher 1 , David Bomze 3 and Gal Markel 1,4,* 1 Ella Lemelbaum Institute for Immuno-oncology, Sheba Medical Center, Ramat-Gan 526260, Israel; [email protected] (T.M.); [email protected] (N.A.) 2 Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel 3 Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; [email protected] 4 Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel * Correspondence: [email protected] Received: 31 May 2020; Accepted: 15 June 2020; Published: 22 June 2020 Abstract: Aim: The selective BRAF and MEK inhibitors (BRAFi+MEKi) have substantially improved the survival of melanoma patients with BRAF V600 mutations. However, BRAFi+MEKi can also cause severe or fatal outcomes. We aimed to identify and compare serious adverse events (sAEs) that are significantly associated with BRAFi+MEKi. Methods: In this pharmacovigilance study, we reviewed FDA Adverse Event Reporting System (FAERS) data in order to detect sAE reporting in patients treated with the combination therapies vemurafenib+cobimetinib (V+C), dabrafenib+trametinib (D+T) and encorafenib+binimetinib (E+B). We evaluated the disproportionate reporting of BRAFi+MEKi-associated sAEs. Significant associations were further analyzed to identify combination-specific safety signals among BRAFi+MEKi. Results: From January 2018 through June 2019, we identified 11,721 sAE reports in patients receiving BRAFi+MEKi. Comparison of BRAFi+MEKi combinations demonstrates that skin toxicities, including Stevens–Johnson syndrome, were disproportionally reported using V+C, with an age-adjusted reporting odds ratio (adj.
    [Show full text]
  • Targeting Cyclin-Dependent Kinase 9 and Myeloid Cell Leukaemia 1 in MYC-Driven B-Cell Lymphoma
    Targeting cyclin-dependent kinase 9 and myeloid cell leukaemia 1 in MYC-driven B-cell lymphoma Gareth Peter Gregory ORCID ID: 0000-0002-4170-0682 Thesis for Doctor of Philosophy September 2016 Sir Peter MacCallum Department of Oncology The University of Melbourne Doctor of Philosophy Submitted in total fulfilment of the degree of Abstract Aggressive B-cell lymphomas include diffuse large B-cell lymphoma, Burkitt lymphoma and intermediate forms. Despite high response rates to conventional immuno-chemotherapeutic approaches, an unmet need for novel therapeutic by resistance to chemotherapy and radiotherapy. The proto-oncogene MYC is strategies is required in the setting of relapsed and refractory disease, typified frequently dysregulated in the aggressive B-cell lymphomas, however, it has proven an elusive direct therapeutic target. MYC-dysregulated disease maintains a ‘transcriptionally-addicted’ state, whereby perturbation of A significant body of evidence is accumulating to suggest that RNA polymerase II activity may indirectly antagonise MYC activity. Furthermore, very recent studies implicate anti-apoptotic myeloid cell leukaemia 1 (MCL-1) as a critical survival determinant of MYC-driven lymphoma. This thesis utilises pharmacologic and genetic techniques in MYC-driven models of aggressive B-cell lymphoma to demonstrate that cyclin-dependent kinase 9 (CDK9) and MCL-1 are oncogenic dependencies of this subset of disease. The cyclin-dependent kinase inhibitor, dinaciclib, and more selective CDK9 inhibitors downregulation of MCL1 are used
    [Show full text]
  • Mekinist Tablets
    HIGHLIGHTS OF PRESCRIBING INFORMATION Venous Thromboembolism: Deep vein thrombosis and pulmonary These highlights do not include all the information needed to use embolism (PE) can occur in patients receiving MEKINIST. (5.4, 2.7) MEKINIST safely and effectively. See full prescribing information for Cardiomyopathy: Assess left ventricular ejection fraction (LVEF) before MEKINIST. treatment, after one month of treatment, then every 2 to 3 months thereafter. (5.5, 2.7) MEKINIST® (trametinib) tablets, for oral use Ocular Toxicities: Perform ophthalmologic evaluation for any visual Initial U.S. Approval: 2013 disturbances. For Retinal Vein Occlusion (RVO), permanently ------------------------------RECENT MAJOR CHANGES------------------------ discontinue MEKINIST. (5.6, 2.7) Warnings and Precautions (5.8) 6/2020 Interstitial Lung Disease (ILD): Withhold MEKINIST for new or progressive unexplained pulmonary symptoms. Permanently discontinue ------------------------------INDICATIONS AND USAGE------------------------- MEKINIST for treatment-related ILD or pneumonitis. (5.7, 2.7) MEKINIST is a kinase inhibitor indicated as a single agent for the treatment Serious Febrile Reactions, can occur when MEKINIST is used with of BRAF-inhibitor treatment-naïve patients with unresectable or metastatic dabrafenib. (5.8, 2.7) melanoma with BRAF V600E or V600K mutations as detected by an FDA- Serious Skin Toxicities: Monitor for skin toxicities and for secondary approved test. (1.1, 2.1) infections. Permanently discontinue MEKINIST for intolerable Grade 2, MEKINIST is indicated, in combination with dabrafenib, for: or Grade 3 or 4 rash not improving within 3 weeks despite interruption the treatment of patients with unresectable or metastatic melanoma with of MEKINIST. Permanently discontinue for severe cutaneous adverse BRAF V600E or V600K mutations as detected by an FDA-approved reactions (SCARs).
    [Show full text]
  • BRAF-MEK Inhibitor Combo Approved for Adjuvant Melanoma Therapy
    Community Translations BRAF-MEK inhibitor combo approved for adjuvant melanoma therapy n April 30, 2018, the US Food and Drug Administration expanded the indication for the What’s new, what’s important combined use of dabrafenib and trametinib to The expanded approval of the dabrafenib-trametinib combina- include adjuvant treatment of BRAF-mutant melanoma tion for BRAF-mutant melanoma after complete resection is a wel- O come option for these patients who often face recurrence. The following complete surgical resection. Dabrafenib is an inhibitor of the BRAF kinase, and trametinib is an inhibi- approval was based on data from the COMBI-AD trial in which tor of the MEK kinase, both of which are components of 870 patients with stage III melanoma and BRAF V600E/K muta- the mitogen-activated protein kinase (MAPK) signaling tions and lymph-node involvement were randomised either to pathway. The 2 drugs are already approved as both single dabrafenib 150 mg twice daily in combination with trametinib agents and in combination for the treatment of BRAF- 2 mg once daily, or to 2 matched placebos. Randomization was mutated metastatic melanoma. stratified according toBRAF mutation status and disease stage. The current approval was based on data from a phase 3, The primary endpoint was RFS, and secondary endpoints international, multicenter, randomized, double-blind, pla- included OS, DMFS, FFR, and safety. As of the data cut-off, the cebo-controlled trial. The COMBI-AD trial was carried dabrafenib–trametinib combination reduced the risk of disease out from January 2013 through December 2014 at 169 recurrence or death by 53% compared with placebo (HR, 0.47; sites in 26 countries.
    [Show full text]
  • RASA1/NF1 Mutant Lung Cancer: Racing to the Clinic? Shunsuke Kitajima1 and David A
    Author Manuscript Published OnlineFirst on January 17, 2018; DOI: 10.1158/1078-0432.CCR-17-3597 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. RASA1/NF1 mutant lung cancer: Racing to the clinic? Shunsuke Kitajima1 and David A. Barbie1* 1 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA. *To whom correspondence should be addressed: David Barbie, M.D., 450 Brookline Ave, LC4115, Boston, MA 02215. Email: [email protected], Phone: (617) 632-6049, Fax: (617) 632-5786. Running Title: Targeting RASA1/NF1 mutant non-small cell lung cancer Funding: This work was supported by NIH R01CA190394 (D.A.B). Summary: Although mutation of NF1 has been described in non-small cell lung cancer (NSCLC), co-mutation with RASA1, another Ras-GTPase activating protein (RasGAP), defines a novel genetically defined subclass of NSCLC. RASA1/NF1 mutant cell lines are highly sensitive to MEK inhibitors, warranting clinical evaluation of MAPK inhibition in this subclass of patients. Text: In this issue of Clinical Cancer Research, Hayashi and colleagues report that loss-of-function mutation of RASA1, a Ras-GTPase activating protein (RasGAP), is significantly enriched in NF1 mutated non-small cell lung cancer (NSCLC) patients (1). Co-occurring mutation of RASA1 and NF1 shows mutual exclusivity with other oncogenic drivers such as KRAS and EGFR, unveiling RASA1/NF1 co-mutation as new potentially targetable subclass of NSCLC. Approximately two-thirds of NSCLC patients harbor genetic alterations that activate receptor tyrosine kinase/RAS pathway mediated downstream mitogenic signaling, including the MAPK and PI3K pathways.
    [Show full text]
  • Advances in Targeted Therapy for Melanoma: a Focus on MEK Inhibition
    Review: Clinical Trial Outcomes Salama & Kim Advances in targeted therapy for melanoma: a focus on MEK inhibition 6 Review: Clinical Trial Outcomes Advances in targeted therapy for melanoma: a focus on MEK inhibition Clin. Investig. (Lond.) Increased knowledge of the role that dysregulated MAPK signaling plays in melanoma April KS Salama*,1 & Kevin B has opened the door for novel therapeutic strategies, among them MEK-targeted Kim2 approaches. With the recent approval of the selective MEK inhibitor trametinib, both 1Division of Medical Oncology, Duke University Medical Center, DUMC 3198, as a single agent and in combination with dabrafenib for patients with BRAF-mutant Durham, NC 27710, USA metastatic melanoma, MEK-based therapy now has a place as a standard of care 2Department of Melanoma Medical option for the treatment of this disease. Early data suggest a benefit of MEK inhibition Oncology, The University of Texas MD in other melanoma subtypes as well, including NRAS-mutant and uveal melanoma. Anderson Cancer Center, 1515 Holcombe This review summarizes the clinical development and the currently available data Blvd, Houston, TX 77030, USA *Author for correspondence: regarding the role of MEK inhibitor therapy in the treatment of advanced melanoma. Tel.: +1 919 681 6932 Fax: +1 919 684 5163 Keywords: BRAF • MAPK pathway • MEK • melanoma • NRAS • targeted therapy • uveal [email protected] melanoma While there have been numerous advances growth and proliferation, and represents an in systemic therapy for the treatment of attractive target for therapeutic intervention metastatic melanoma in recent years, for [7]. First identified in a screen of cell lines and most patients it remains an incurable dis- melanoma specimens in 2002, the majority ease [1] .
    [Show full text]
  • Response To: 'Efficacy and Improved Tolerability of Combination Therapy
    Ann Rheum Dis: first published as 10.1136/annrheumdis-2019-216755 on 20 January 2020. Downloaded from Correspondence response Response to: ‘Efficacy and improved tolerability (42%) underwent an improvement of clinical and/or metabolic disease, in particular at osseous sites, with an objective partial of combination therapy with interleukin-1 or complete response on bones hypermetabolisms in 8 (33%). blockade and MAPK pathway inhibitors for the Six patients (25%) were stable, whereas 8 (33%) experienced treatment of Erdheim- Chester disease’ by a progression of their disease while under anakinra, including a progression or occurrence of CNS (n=5), cardiac (n=3, Campochiaro et al with tamponade in 2), lung (n=3) and/or skin (n=2) disease. After receiving anakinra, 11 patients (35%) were treated with 1 We read with interest the letter by Campochiaro et al that targeted therapy, including a BRAF inhibitor in eight cases and/ reported the efficacy of a combination regimen including anak- or a MEK inhibitor in seven cases. The targeted therapy was inra and targeted therapy for the treatment of Erdheim- Chester efficacious in all cases. Among the whole cohort of 262 patients, disease (ECD), a rare multisystem inflammatory histiocytosis. 117 patients (45%) were treated with targeted therapies (vemu- Based on the involvement of inflammatory cytokines, including rafenib or dabrafenib for BRAF inhibition, and trametinib or tumour necrosis factor- alpha and interleukin-1, in the patho- cobimetinib for MEK inhibition). Although these patients had physiology and the clinical manifestations of ECD, previous more cardiac and CNS involvements, they had a better survival reports demonstrated a variable efficacy with a good tolerance than the patients who were not treated with targeted therapies of daily subcutaneous anakinra and infliximab for the treatment (HR 0.6350, 95% CI 0.4170 to 0.9945, p=0.04).
    [Show full text]