Advances in Targeted Therapy for Melanoma: a Focus on MEK Inhibition

Total Page:16

File Type:pdf, Size:1020Kb

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] . Notably, since 2011, there have of BRAF mutations in melanoma result from 10.4155/CLI.14.76 been US FDA approvals for four new agents the substitution of a glutamic acid for a valine used to treat metastatic disease, three of (V600E), though other variants exist [2]. It is which – vemurafenib (PLX4032; RG7204), now known that this change results in a con- dabrafenib (GSK2118436) and trametinib stitutively active kinase with resultant per- (GSK1120212) – target aberrant MAPK sistent downstream activation of the MAPK pathway signaling. The identification of pathway [8]. The subsequent development of 19 mutated BRAF as a major driver of oncogen- the selective BRAF inhibitors vemurafenib esis in approximately half of all melanomas, and dabrafenib represented a landmark break- and the resultant successful clinical devel- through in the treatment of metastatic mela- opment of selective BRAF inhibitors has noma [3,4]. Vemurafenib, the first molecular 2014 opened the door for novel agents that target targeted agent to receive regulatory approval this pathway [2–4]. Among these are MEK in 2011, has demonstrated an overall survival targeted therapies, and while MEK inhibi- advantage in a Phase III study when com- tors have been in development for some time, pared with chemotherapy in patients with V600E recent evidence of early clinical activity in BRAF melanoma [3,9]. Dabrafenib has many tumor types, including thyroid and also been shown to improve progression-free ovarian cancer, as well as specific subsets of survival (PFS) when compared with dacarba- melanoma has provided a platform for the zine in a randomized Phase III trial [4]. High continued expansion of possible treatment response rates coupled with the potential for options [5,6]. a relatively rapid response make these agents The RAS-RAF-MEK signaling cascade particularly useful for patients with symp- is an integral component in regulating cell tomatic or rapidly progressive disease. How- part of 10.4155/CLI.14.76 © 2014 Future Science Ltd Clin. Investig. (Lond.) (2014) 6(19), 903–913 ISSN 2041-6792 903 Review: Clinical Trial Outcomes Salama & Kim ever, like many systemic agents for the treatment of Figures 1 & 2 highlight some of the potential media- advanced cancer, limitations exist due to the emergence tors involved in this process. Additionally, treatment of resistance as well as toxicity. On average, responses with these agents is uniquely associated with the devel- with BRAF inhibitor monotherapy last 6–7 months, opment of hyperproliferative skin lesions, including and most patients relapse within a year of starting treat- squamous cell carcinomas (SCCs), largely attributed ment [4,9–11] . The development of resistance is a multi- to persistent MAPK signaling through RAS mediated factorial process, though MAPK reactivation through paradoxical activation of other RAF isoforms [22]. different mechanisms appears to play a key role [12–21] . The hypothesis that MEK inhibition in melanoma has the potential to have a meaningful clinical benefit Receptor tyrosine has now come full circle. In 2013, the MEK inhibitor kinase trametinib was approved as monotherapy for the treat- ment of patients with metastatic BRAFV600 melanoma, and the approval of its use in combination with dab- rafenib in this same population came in early 2014. Subsequent studies have continued to build upon the understanding of the molecular pathogenesis of mela- Cell membrane NRASmut RAS noma. In addition to having a role in BRAF-mutated melanoma, it is also becoming apparent that other dis- Dabrafenib tinct subsets, including NRAS mutated and uveal mela- CRAF Vemurafenib LGX818 noma may also be susceptible to targeting this pathway. This review aims to summarize the history of MEK COT MEK inhibitor development in melanoma, the current role in clinical practice as well as future therapeutic strategies. MEKmut ERK Early MEK inhibitor development As the MAPK pathway plays an integral role in reg- ulating cell growth and proliferation, there has long Sustained proliferation and been an interest in targeting this pathway for thera- suppression of apoptosis peutic benefit. One of the first MEK inhibitors to enter Figure 1. Potential mechanisms of resistance to clinical testing included the non-ATP-competitive selective BRAF inhibition that are dependent on MAPK MEK 1/2 inhibitor CI-1040. CI-1040 is highly selec- pathway activation/MEK signaling. tive for MEK, and acts to form a bond at a hydropho- Receptor tyrosine bic binding pocket on MEK 1/2. This induces a con- kinase formational change in unphosphorylated MEK, and essentially locks the protein in its inactive state [23] An initial dose escalation study that included patients with a variety of solid tumors, including melanoma, estab- lished that CI-1040 could be safely administered, but subsequent studies failed to demonstrate clinical effi- Cell membrane NRASmut RAS cacy [23,24]. PD-0325901 is a second-generation MEK inhibitor that had a more favorable pharmacokinetic Dabrafenib profile than CI-1040, and demonstrated promising CRAF Vemurafenib preclinical activity [25,26]. In early phase testing, there LGX818 was some signal of potential clinical activity with a Selumetinib CI-1040 COT few partial responses (PRs) and stable disease seen in Trametinib PD-0325901 Cobimetinib MEK162 some of the melanoma patients that were treated [27,28]. Pimasertib However, there were a number of concerning toxicities MEKmut ERK noted, including neurologic effects as well as retinal vein occlusion. With this concern, development of this agent was subsequently suspended. Growth inhibition Selumetinib Figure 2. MEK targeted strategies provide a mechanism Selumetinib (ARRY-142886; AZD-6244) is a second- to overcome selected mediators of resistance. generation, highly selective MEK 1/2 inhibitor that 904 Clin. Investig. (Lond.) (2014) 6(19) future science group Advances in targeted therapy for melanoma: a focus on MEK inhibition Review: Clinical Trial Outcomes has recently shown some promising results in later generation MEK inhibitors, but it is unique in that it phase clinical trials for patients with metastatic mel- has a favorable mean peak:trough ratio, making once anoma, particularly with a newer formulation of the daily dosing feasible [34,35]. Initial clinical trials with drug. Initial preclinical work and molecular model- trametinib yielded promising results, and established ing data showed that selumetinib was able to bind the 2 mg daily as the recommended dose to move forward allosteric inhibitor site on MEK 1/2, rendering it cata- in subsequent studies on the basis of pharmacokinetic, lytically inactive. Early phase testing of the free base pharmacodymic, long-term safety and preliminary formulation of selumetinib established 100 mg b.i.d. efficacy data although the MTD administered was 3 as the dose to pursue in subsequent studies, largely due mg daily [35]. Multiple tumor types were treated in this to cutaneous toxicity seen at higher doses [29]. How- Phase I study, with an overall response rate of approxi- ever, in a Phase II study using the initial formulation mately 10%, however, BRAF mutant melanoma of the drug in which patients with advanced melanoma appeared to be a particularly sensitive subset. A total of were randomized to receive either selumetinib or temo- 97 patients with melanoma were enrolled, 36 of which zolomide, response rates were similar and there was no had BRAF mutant tumors [36]. In a separate analysis of improvement in PFS in the selumetinib arm [30]. In the cohort of melanoma patients, the 30 patients with this study, median progression free survival (PFS) was BRAF mutant disease that had never received BRAF only 78 days and the response rate was 5.8% for those inhibitor therapy had an overall unconfirmed response who received selumetinib treatment, and even among rate (complete response (CR) + PR) of 40%, with a patients with BRAF mutant melanoma, the response median PFS of 5.7 months [36]. Additionally, of the six rate was 11%. Subsequent studies using a hydrogen sul- patients with BRAF mutant melanoma that had previ- fate formulation, which appears to have a more favor- ously received a BRAF inhibitor, one had a PR and able pharmacokinetic profile, have demonstrated more four had stable disease. Overall, trametinib appeared promising results [31] . Phase I studies have established to be well tolerated, with the most common adverse 75 mg b.i.d.
Recommended publications
  • 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]
  • 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]
  • 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]
  • Targeting Epha2 in Cancer Ta Xiao1, Yuhang Xiao2, Wenxiang Wang3,4, Yan Yan Tang4, Zhiqiang Xiao2* and Min Su3,4*
    Xiao et al. Journal of Hematology & Oncology (2020) 13:114 https://doi.org/10.1186/s13045-020-00944-9 REVIEW Open Access Targeting EphA2 in cancer Ta Xiao1, Yuhang Xiao2, Wenxiang Wang3,4, Yan Yan Tang4, Zhiqiang Xiao2* and Min Su3,4* Abstract Eph receptors and the corresponding Eph receptor-interacting (ephrin) ligands jointly constitute a critical cell signaling network that has multiple functions. The tyrosine kinase EphA2, which belongs to the family of Eph receptors, is highly produced in tumor tissues, while found at relatively low levels in most normal adult tissues, indicating its potential application in cancer treatment. After 30 years of investigation, a large amount of data regarding EphA2 functions have been compiled. Meanwhile, several compounds targeting EphA2 have been evaluated and tested in clinical studies, albeit with limited clinical success. The present review briefly describes the contribution of EphA2-ephrin A1 signaling axis to carcinogenesis. In addition, the roles of EphA2 in resistance to molecular-targeted agents were examined. In particular, we focused on EphA2’s potential as a target for cancer treatment to provide insights into the application of EphA2 targeting in anticancer strategies. Overall, EphA2 represents a potential target for treating malignant tumors. Keywords: EphA2 receptor, Ephrin A1, Cancer, Therapy, Target Introduction ligand-binding and intrinsic enzymatic activities, respect- Ephrin receptors (Eph) represent the most important ively [7, 8]. Eph receptors are grouped into A and B class of receptor tyrosine kinases (RTKs) [1]. EphA1, the categories according to their extracellular domains, firstly described Eph receptor, was identified in liver which determine the binding affinity for ligands (Eph cancer cells while screening for RTKs in 1987 [2].
    [Show full text]
  • BRAF Gene and Melanoma: Back to the Future
    International Journal of Molecular Sciences Review BRAF Gene and Melanoma: Back to the Future Margaret Ottaviano 1,2,3,*,† , Emilio Francesco Giunta 4,† , Marianna Tortora 3 , Marcello Curvietto 5, Laura Attademo 2, Davide Bosso 2, Cinzia Cardalesi 2, Mario Rosanova 2, Pietro De Placido 1, Erica Pietroluongo 1 , Vittorio Riccio 1, Brigitta Mucci 1, Sara Parola 1, Maria Grazia Vitale 5, Giovannella Palmieri 3 , Bruno Daniele 2 , Ester Simeone 5 and on behalf of SCITO YOUTH ‡ 1 Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; [email protected] (P.D.P.); [email protected] (E.P.); [email protected] (V.R.); [email protected] (B.M.); [email protected] (S.P.) 2 Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; [email protected] (L.A.); [email protected] (D.B.); [email protected] (C.C.); [email protected] (M.R.); [email protected] (B.D.) 3 CRCTR Coordinating Rare Tumors Reference Center of Campania Region, 80131 Naples, Italy; [email protected] (M.T.); [email protected] (G.P.) 4 Department of Precision Medicine, Università Degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy; [email protected] 5 Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, 80131 Naples, Italy; [email protected] (M.C.); [email protected] (M.G.V.); [email protected] (E.S.) * Correspondence: [email protected] † These authors contributed equally to this work. ‡ Membership of the SCITO YOUTH is provided in the Acknowledgments. Citation: Ottaviano, M.; Giunta, E.F.; Tortora, M.; Curvietto, M.; Attademo, Abstract: As widely acknowledged, 40–50% of all melanoma patients harbour an activating BRAF L.; Bosso, D.; Cardalesi, C.; Rosanova, mutation (mostly BRAF V600E).
    [Show full text]
  • MEKINIST (Trametinib) Label Snda 204114 S-001
    HIGHLIGHTS OF PRESCRIBING INFORMATION • Cardiomyopathy: Assess LVEF before treatment, after one month of These highlights do not include all the information needed to use treatment, then every 2 to 3 months thereafter. (5.4, 2.3) MEKINIST safely and effectively. See full prescribing information for • Ocular Toxicities: Perform ophthalmologic evaluation for any visual MEKINIST. disturbances. For Retinal Vein Occlusion (RVO), permanently MEKINIST (trametinib) tablets, for oral use discontinue MEKINIST. (5.5, 2.3). Initial U.S. Approval: 2013 • Interstitial Lung Disease (ILD): Withhold MEKINIST for new or progressive unexplained pulmonary symptoms. Permanently discontinue ------------------------- RECENT MAJOR CHANGES --------------------------- MEKINIST for treatment-related ILD or pneumonitis. (5.6, 2.3) Indications and Usage (1) 01/2014 • Serious Febrile Reactions can occur when MEKINIST is used in Dosage and Administration (2.2-2.3) 01/2014 combination with dabrafenib. (5.7, 2.3) Warnings and Precautions (5-5.10) 01/2014 • Serious Skin Toxicity: Monitor for skin toxicities and for secondary infections. Discontinue for intolerable Grade 2, or Grade 3 or 4 rash not -------------------------- INDICATIONS AND USAGE ---------------------------- improving within 3 weeks despite interruption of MEKINIST. (5.8, 2.3) MEKINIST is a kinase inhibitor indicated as a single agent and in • Hyperglycemia: Monitor serum glucose levels in patients with pre- combination with dabrafenib for the treatment of patients with unresectable or existing diabetes or hyperglycemia. (5.9, 2.3) metastatic melanoma with BRAF V600E or V600K mutations as detected by • Embryofetal Toxicity: Can cause fetal harm. Advise females of an FDA-approved test. The use in combination is based on the demonstration reproductive potential of potential risk to the fetus.
    [Show full text]
  • Selumetinib (Koselugo™) EOCCO POLICY
    selumetinib (Koselugo™) EOCCO POLICY Policy Type: PA/SP Pharmacy Coverage Policy: EOCCO193 Description Selumetinib (Koselugo) is a mitogen-activated protein kinase (MEK) inhibitor for both MEK 1 and 2 that inhibits the phosphorylation of extracellular signal related kinase (ERK) and reducing neurofibroma numbers, volume, and proliferation. Length of Authorization Initial: 12 months Renewal: 12 months Quantity Limits Product Name Dosage Form Indication Qu antity Limit 10 mg capsules selumetinib Neurofibromatosis type 1 120 capsules/30 days (Koselugo) 25 mg capsules (NF1) Initial Evaluation I. Selumetinib (Koselugo) may be considered medically necessary when the following criteria are met: A. Member is between two and 18 years of age; AND B. Medication is prescribed by, or in consultation with, a neurosurgeon or neurologist; AND C. Documentation of baseline comprehensive ophthalmic assessments; AND D. Documentation of baseline assessment of left ventricular ejection fraction (LVEF); AND E. Member has NOT experienced disease progression (increase in tumor size or tumor spread) while on a MEK inhibitor [e.g., binimetinib (Mektovi®), cobimetinib (Cotellic®), trametinib (Mekinist®)]; AND F. A diagnosis of Neurofibromatosis type 1 (NF1) when the following are met: 1. Member has inoperable and symptomatic plexiform neurofibromas (PN); AND 2. Symptoms affect quality of life (e.g. pain, impaired physical function, compression of vital organs, respiratory impairment, visual dysfunction, and neurological dysfunction); AND 3. Diagnosis confirmed by genetic testing; OR i. Member meets at least one criterion: a. Six or more light brown spots (café-au-lait macule – CALMs) equal to, or greater than, 5 mm in longest diameter in prepubertal 1 selumetinib (Koselugo™) EOCCO POLICY patients and 15 mm in longest diameter in post pubertal patient; OR b.
    [Show full text]
  • MEK Inhibitors for the Treatment of Non-Small Cell Lung Cancer
    Han et al. J Hematol Oncol (2021) 14:1 https://doi.org/10.1186/s13045-020-01025-7 REVIEW Open Access MEK inhibitors for the treatment of non-small cell lung cancer Jing Han1†, Yang Liu2†, Sen Yang1, Xuan Wu1, Hongle Li3* and Qiming Wang1* 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 identifed in non-small cell lung cancer (NSCLC). They lead to the prolifera- tion, diferentiation, 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 fndings, the combination of MEK inhibitors with chemotherapy, immune checkpoint inhibitors, epidermal growth factor receptor-tyrosine kinase inhibitors or BRAF inhibitors is highly signifcant for improving clinical efcacy 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 signifcantly increase therapeutic efects on patients with lung cancer. Keywords: Non-small cell lung cancer, MEK inhibitors, Targeted therapy, RAS, RAF, MEK, ERK signaling pathway Introduction as squamous cell carcinoma, adenocarcinoma, large cell Lung cancer is the most common cause of cancer-related or undiferentiated carcinoma. Non-squamous carci- death worldwide, with over 1.8 million lung cancer deaths noma (70–75%) and squamous cell carcinoma (25–30%) annually [1].
    [Show full text]