1
|
Dadge SD, Yadav S, Rathaur S, Gayen JR. LC-ESI-MS/MS method validation for simultaneous quantification of FDA-approved anticancer agents futibatinib and binimetinib in rat plasma: Insights from preclinical pharmacokinetics. Biomed Chromatogr 2024; 38:e6005. [PMID: 39237854 DOI: 10.1002/bmc.6005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/14/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
This study investigates the combination of FGFR inhibitor futibatinib (FTB) and MEK inhibitor binimetinib (BTB) for KRASmt NSCLC therapy. An analytical method was developed and validated for measuring FTB and BTB concentrations in rat plasma, adhering to USFDA guidelines. Using liquid-liquid extraction on 45-μL plasma samples, a 6.5-min run time was achieved. The linear calibration curve ranged from 2 to 100 ng/mL. Intra-day and inter-day accuracy ranged between 92.06% and 100.08%. Four blank injections post high-concentration samples resolved significant carryover. Extraction recoveries averaged 92.06% to 102.37% across concentrations. No significant endogenous interference was detected in blank plasma. The LLOQ for both drugs was 2.0 ng/mL. Selectivity, matrix effects, stability, and dilution integrity met the acceptance criteria. The method assessed FTB and BTB interaction potential in combination therapy at 5 mg/kg. The findings provide essential pharmacokinetics insights for future clinical trials.
Collapse
Affiliation(s)
- Shailesh D Dadge
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shubhi Yadav
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shivam Rathaur
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Jiaur R Gayen
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| |
Collapse
|
2
|
Shi J, Fu Q, Ma Q, Wei Z, Su X, Li X. Autoimmune complications of tyrosine kinase inhibitors in cancer therapy: Clinical insights, mechanisms, and future perspectives. Medicine (Baltimore) 2024; 103:e39928. [PMID: 39465760 PMCID: PMC11460853 DOI: 10.1097/md.0000000000039928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Indexed: 10/29/2024] Open
Abstract
The tyrosine kinase inhibitors (TKIs) have emerged as a promising class of novel anticancer drugs, achieving significant success in clinical applications. However, the risk of autoimmune diseases associated with these drugs has raised widespread concerns. In this review, TKI-induced autoimmune diseases are reviewed in order to understand this complex phenomenon through clinical research and molecular mechanism exploration. Despite the relatively low incidence of autoimmune diseases, their potential severity demands heightened attention. The potential mechanisms underlying TKI-induced autoimmune diseases may involve immune system dysregulation, alterations in immune cell function, activation of inflammatory responses, and attacks on self-antigens. Various preventive strategies, including clinical monitoring, personalized treatment, optimization of therapeutic approaches, and patient education and communication, can be employed to effectively address these potential risks. Future research directions should delve into the molecular mechanisms of TKI-induced autoimmune diseases, integrate studies on genetics and immunogenetics, advance the development of novel TKIs, explore the possibilities of combining immunotherapy with TKI treatment, and propel large-scale clinical trials.
Collapse
Affiliation(s)
- Juan Shi
- Department of Pharmacy, The First People’s Hospital of Jinan, Jinan, China
| | - Qingyuan Fu
- Department of Neurology, The First People’s Hospital of Jinan, Jinan, China
| | - Quancheng Ma
- Department of Clinical Laboratory, The First People’s Hospital of Jinan, Jinan, China
| | - Zhenzhen Wei
- Department of Science and Education, The First People’s Hospital of Jinan, Jinan, China
| | - Xiaolian Su
- Department of Obstetrics and Gynecology, The First People’s Hospital of Jinan, Jinan, China
| | - Xiao Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| |
Collapse
|
3
|
Chaudhary HA, Cannon TL, Winer A. Targeting Non-V600 Mutations in BRAF: A Single Institution Retrospective Analysis and Review of the Literature. Drugs R D 2024; 24:395-403. [PMID: 39177935 PMCID: PMC11455815 DOI: 10.1007/s40268-024-00475-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND AND OBJECTIVE While successful treatment paradigms for BRAF V600 mutations have been developed, 10% of BRAF mutations are not at V600 and lack a standard treatment regimen. This study summarizes the current body of knowledge on the treatment of non-V600 mutations and reports a single institution experience. METHODS We conducted a literature review to summarize relevant preclinical and clinical published data on the response of non-V600 mutations to targeted therapies. We performed a retrospective analysis of INOVA Schar Cancer patients registered in our Molecular Tumor Board database with non-V600 BRAF mutations who were recipients of targeted therapy and assessed their time to next treatment and best response. RESULTS Published preclinical and clinical data have demonstrated limiting results in the response of non-V600 mutated cancers to targeted therapies. Response rates were variable for the major classes of BRAF mutations including class II and class III mutations as well as, BRAF fusions. Data collected from our INOVA cohort offered promising results with one patient achieving partial remission and two patients achieving stable disease. CONCLUSIONS This article reflects the current understanding of targeted therapies in non-V600 mutations. Further large-scale studies separating BRAF mutations based on their mechanism of activation will expand our understanding.
Collapse
Affiliation(s)
- Hirra A Chaudhary
- INOVA, Schar Cancer Institute, Fairfax, VA, USA.
- UVA School of Medicine, INOVA Fairfax Medical Campus, Fairfax, VA, USA.
| | | | | |
Collapse
|
4
|
Li K, Yang B, Du Y, Ding Y, Shen S, Sun Z, Liu Y, Wang Y, Cao S, Ren W, Wang X, Li M, Zhang Y, Wu J, Zheng W, Yan W, Li L. The HOXC10/NOD1/ERK axis drives osteolytic bone metastasis of pan-KRAS-mutant lung cancer. Bone Res 2024; 12:47. [PMID: 39191757 PMCID: PMC11349752 DOI: 10.1038/s41413-024-00350-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 06/09/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024] Open
Abstract
While KRAS mutation is the leading cause of low survival rates in lung cancer bone metastasis patients, effective treatments are still lacking. Here, we identified homeobox C10 (HOXC10) as a lynchpin in pan-KRAS-mutant lung cancer bone metastasis. Through RNA-seq approach and patient tissue studies, we demonstrated that HOXC10 expression was dramatically increased. Genetic depletion of HOXC10 preferentially impeded cell proliferation and migration in vitro. The bioluminescence imaging and micro-CT results demonstrated that inhibition of HOXC10 significantly reduced bone metastasis of KRAS-mutant lung cancer in vivo. Mechanistically, the transcription factor HOXC10 activated NOD1/ERK signaling pathway to reprogram epithelial-mesenchymal transition (EMT) and bone microenvironment by activating the NOD1 promoter. Strikingly, inhibition of HOXC10 in combination with STAT3 inhibitor was effective against KRAS-mutant lung cancer bone metastasis by triggering ferroptosis. Taken together, these findings reveal that HOXC10 effectively alleviates pan-KRAS-mutant lung cancer with bone metastasis in the NOD1/ERK axis-dependent manner, and support further development of an effective combinatorial strategy for this kind of disease.
Collapse
Affiliation(s)
- Kun Li
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Health Science Center, East China Normal University, Shanghai, 200241, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China
| | - Bo Yang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yingying Du
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yi Ding
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Shihui Shen
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, 200240, China
| | - Zhengwang Sun
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yun Liu
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yuhan Wang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Siyuan Cao
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wenjie Ren
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiangyu Wang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mengjuan Li
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yunpeng Zhang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Juan Wu
- Department of Pharmacy The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Wei Zheng
- Orthopaedic Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
- Department of Orthopedics, General Hospital of Western Theater Command, Chengdu, 610000, China.
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China.
| | - Wangjun Yan
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Lei Li
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China.
- School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, 200240, China.
| |
Collapse
|
5
|
Iriarte C, Yeh JE, Alloo A, Boull C, Carlberg VM, Coughlin CC, Lara-Corrales I, Levy R, Nguyen CV, Oza VS, Patel AB, Rotemberg V, Shah SD, Zheng L, Miller CH, Hlobik M, Daigneault J, Choi JN, Huang JT, Vivar KL. Mucocutaneous toxicities from MEK inhibitors: a scoping review of the literature. Support Care Cancer 2024; 32:610. [PMID: 39174797 DOI: 10.1007/s00520-024-08810-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND MEK inhibitors cause a wide spectrum of mucocutaneous toxicities which can delay or interrupt life-saving therapy. PURPOSE To summarize the morphology, incidence, and clinical presentation of mucocutaneous toxicities from MEK inhibitors via a scoping review of the literature. METHODS We conducted a scoping review of the published literature, including clinical trials, retrospective and prospective studies, reviews, and case reports and series. All included literature was analyzed by a panel of pediatric and adult oncodermatologists. RESULTS Of 1626 initial citations, 227 articles met final inclusion criteria. Our review identified follicular reactions, ocular toxicities, xerosis, eczematous dermatitis, edema, and paronychia as the most common mucocutaneous side effects from MEK inhibitor therapy. Grade 1 and 2 reactions were the most prevalent and were typically managed while continuing treatment; however, grade 3 toxicities requiring dose reductions or treatment interruptions were also reported. CONCLUSION Mucocutaneous toxicities to MEK inhibitor therapy are common and most often mild in severity. Early recognition and treatment can mitigate disruptions in oncologic therapy.
Collapse
Affiliation(s)
- Christopher Iriarte
- Department of Dermatology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Gryzmish 522, Boston, MA, 02215, USA.
- Department of Dermatology, Harvard Medical School, Boston, MA, USA.
| | - Jennifer E Yeh
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA, USA
| | - Allireza Alloo
- Department of Dermatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Christina Boull
- Department of Dermatology, University of Minnesota, Minneapolis, MN, USA
| | - Valerie M Carlberg
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI, USA
- Children's Wisconsin, Milwaukee, WI, USA
| | - Carrie C Coughlin
- Division of Dermatology, Departments of Medicine and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Irene Lara-Corrales
- Division of Dermatology, Hospital for Sick Children, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| | - Rebecca Levy
- Division of Dermatology, Hospital for Sick Children, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| | - Cuong V Nguyen
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Vikash S Oza
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, USA
| | - Anisha B Patel
- Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- University of Texas Health Science Center- Houston, Houston, TX, USA
| | - Veronica Rotemberg
- Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sonal D Shah
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Lida Zheng
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Corinne H Miller
- Galter Health Sciences Library and Learning Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Madeline Hlobik
- Dermatology Section, Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Jaclyn Daigneault
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jennifer N Choi
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Jennifer T Huang
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Dermatology Section, Division of Immunology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Karina L Vivar
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Division of Pediatric Dermatology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| |
Collapse
|
6
|
Apps JR, Gonzalez-Meljem JM, Guiho R, Pickles JC, Prince E, Schwalbe E, Joshi N, Stone TJ, Ogunbiyi O, Chalker J, Bassey A, Otto G, Davies R, Hughes D, Brandner S, Tan E, Lee V, Hayhurst C, Kline C, Castellano S, Hankinson T, Deutschbein T, Jacques TS, Martinez-Barbera JP. Recurrent adamantinomatous craniopharyngiomas show MAPK pathway activation, clonal evolution and rare TP53-loss-mediated malignant progression. Acta Neuropathol Commun 2024; 12:127. [PMID: 39127699 PMCID: PMC11316312 DOI: 10.1186/s40478-024-01838-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
The two types of craniopharyngioma, adamantinomatous (ACP) and papillary (PCP), are clinically relevant tumours in children and adults. Although the biology of primary craniopharyngioma is starting to be unravelled, little is known about the biology of recurrence. To fill this gap in knowledge, we have analysed through methylation array, RNA sequencing and pERK1/2 immunohistochemistry a cohort of paired primary and recurrent samples (32 samples from 14 cases of ACP and 4 cases of PCP). We show the presence of copy number alterations and clonal evolution across recurrence in 6 cases of ACP, and analysis of additional whole genome sequencing data from the Children's Brain Tumour Network confirms chromosomal arm copy number changes in at least 7/67 ACP cases. The activation of the MAPK/ERK pathway, a feature previously shown in primary ACP, is observed in all but one recurrent cases of ACP. The only ACP without MAPK activation is an aggressive case of recurrent malignant human craniopharyngioma harbouring a CTNNB1 mutation and loss of TP53. Providing support for a functional role of this TP53 mutation, we show that Trp53 loss in a murine model of ACP results in aggressive tumours and reduced mouse survival. Finally, we characterise the tumour immune infiltrate showing differences in the cellular composition and spatial distribution between ACP and PCP. Together, these analyses have revealed novel insights into recurrent craniopharyngioma and provided preclinical evidence supporting the evaluation of MAPK pathway inhibitors and immunomodulatory approaches in clinical trials in against recurrent ACP.
Collapse
Affiliation(s)
- John R Apps
- Institute of Cancer and Genomic Sciences, Edgbaston Campus, University of Birmingham, Birmingham, B15 2TT, UK.
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK.
| | - Jose Mario Gonzalez-Meljem
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Tecnologico de Monterrey, School of Engineering and Sciences, Mexico City, Mexico
| | - Romain Guiho
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Oniris, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, Nantes Université, 44000, Nantes, France
| | - Jessica C Pickles
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Eric Prince
- Departments of Neurosurgery and Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, USA
| | - Edward Schwalbe
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Nikhil Joshi
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Thomas J Stone
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Olumide Ogunbiyi
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Jane Chalker
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Akang Bassey
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Georg Otto
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Rosalind Davies
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | | | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Enrica Tan
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Victoria Lee
- Sheffield Children's Hospital NHS Foundation Trust, Sheffield, UK
| | | | - Cassie Kline
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sergi Castellano
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Todd Hankinson
- Departments of Neurosurgery and Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, USA
| | - Timo Deutschbein
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
- Medicover Oldenburg MVZ, Oldenburg, Germany
| | - Thomas S Jacques
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
7
|
Zahmatyar M, Kharaz L, Abiri Jahromi N, Jahanian A, Shokri P, Nejadghaderi SA. The safety and efficacy of binimetinib for lung cancer: a systematic review. BMC Pulm Med 2024; 24:379. [PMID: 39090580 PMCID: PMC11295668 DOI: 10.1186/s12890-024-03178-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Lung cancer, accounting for a significant proportion of global cancer cases and deaths, poses a considerable health burden. Non-small cell lung cancer (NSCLC) patients have a poor prognosis and limited treatment options due to late-stage diagnosis and drug resistance. Dysregulated of the mitogen-activated protein kinase (MAPK) pathway, which is implicated in NSCLC pathogenesis, underscores the potential of MEK inhibitors such as binimetinib. Despite promising results in other cancers, comprehensive studies evaluating the safety and efficacy of binimetinib in lung cancer are lacking. This systematic review aimed to investigate the safety and efficacy of binimetinib for lung cancer treatment. METHODS We searched PubMed, Scopus, Web of Science, and Google Scholar until September 2023. Clinical trials evaluating the efficacy or safety of binimetinib for lung cancer treatment were included. Studies were excluded if they included individuals with conditions unrelated to lung cancer, investigated other treatments, or had different types of designs. The quality assessment was conducted utilizing the National Institutes of Health tool. RESULTS Seven studies with 228 participants overall were included. Four had good quality judgments, and three had fair quality judgments. The majority of patients experienced all-cause adverse events, with diarrhea, fatigue, and nausea being the most commonly reported adverse events of any grade. The objective response rate (ORR) was up to 75%, and the median progression-free survival (PFS) was up to 9.3 months. The disease control rate after 24 weeks varied from 41% to 64%. Overall survival (OS) ranged between 3.0 and 18.8 months. Notably, treatment-related adverse events were observed in more than 50% of patients, including serious adverse events such as colitis, febrile neutropenia, and pulmonary infection. Some adverse events led to dose limitation and drug discontinuation in five studies. Additionally, five studies reported cases of death, mostly due to disease progression. The median duration of treatment ranged from 14.8 weeks to 8.4 months. The most common dosage of binimetinib was 30 mg or 45 mg twice daily, sometimes used in combination with other agents like encorafenib or hydroxychloroquine. CONCLUSIONS Only a few studies have shown binimetinib to be effective, in terms of improving OS, PFS, and ORR, while most of the studies found nonsignificant efficacy with increased toxicity for binimetinib compared with traditional chemotherapy in patients with lung cancer. Further large-scale randomized controlled trials are recommended.
Collapse
Affiliation(s)
- Mahdi Zahmatyar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ladan Kharaz
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Ali Jahanian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pourya Shokri
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Aria Nejadghaderi
- HIV/STI Surveillance Research Center, WHO Collaborating Center for HIV Surveillance, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran.
- Systematic Review and Meta‑analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
8
|
Takano Y, Shimokata T, Urakawa H, Kikumori T, Ando Y. Long-term response to MEK inhibitor monotherapy in a patient with papillary thyroid carcinoma harboring BRAF V600E mutation. Int Cancer Conf J 2024; 13:184-188. [PMID: 38962055 PMCID: PMC11217198 DOI: 10.1007/s13691-024-00670-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/04/2024] [Indexed: 07/05/2024] Open
Abstract
Solid tumors harboring mutations in the Braf gene (BRAF) are currently treated by combination Braf/MEK inhibitor therapy, and there is an extensive literature on patient response rates. Alternatively, few studies have documented the clinical response of BRAF mutation-positive solid tumors to MEK inhibitor monotherapy. We report the case of a 57-year-old female diagnosed with papillary thyroid carcinoma and progressive lung metastases initially treated by total thyroidectomy and subsequent thyroid-stimulating hormone suppression therapy. Next-generation sequencing revealed that the tumor harbored a BRAF V600E mutation, and the patient was enrolled in a clinical study of the oral MEK1/2 inhibitor binimetinib. Shortly after starting treatment, the patient experienced pneumothorax due to rapid regression of lung metastases, and computed tomography after 6 months of binimetinib treatment revealed a partial sustained response. One year later, the dose was reduced because of an acneiform rash. After 5 years of binimetinib treatment, lung metastases had regrown, and treatment was switched to the oral multikinase inhibitor lenvatinib. This case demonstrates the potential of MEK inhibitor monotherapy as an alternative treatment for BRAF mutation-positive papillary thyroid carcinoma.
Collapse
Affiliation(s)
- Yuko Takano
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, Nagoya, Japan
| | - Tomoya Shimokata
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Hiroshi Urakawa
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Toyone Kikumori
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, Nagoya, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
9
|
Na B, Shah SR, Vasudevan HN. Past, Present, and Future Therapeutic Strategies for NF-1-Associated Tumors. Curr Oncol Rep 2024; 26:706-713. [PMID: 38709422 PMCID: PMC11169015 DOI: 10.1007/s11912-024-01527-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE OF REVIEW Neurofibromatosis type 1 (NF-1) is a cancer predisposition syndrome caused by mutations in the NF1 tumor suppressor gene that encodes the neurofibromin protein, which functions as a negative regulator of Ras signaling. We review the past, current, and future state of therapeutic strategies for tumors associated with NF-1. RECENT FINDINGS Therapeutic efforts for NF-1-associated tumors have centered around inhibiting Ras output, leading to the clinical success of downstream MEK inhibition for plexiform neurofibromas and low-grade gliomas. However, MEK inhibition and similar molecular monotherapy approaches that block Ras signaling do not work for all patients and show limited efficacy for more aggressive cancers such as malignant peripheral nerve sheath tumors and high-grade gliomas, motivating novel treatment approaches. We highlight the current therapeutic landscape for NF-1-associated tumors, broadly categorizing treatment into past strategies for serial Ras pathway blockade, current approaches targeting parallel oncogenic and tumor suppressor pathways, and future avenues of investigation leveraging biologic and technical innovations in immunotherapy, pharmacology, and gene delivery.
Collapse
Affiliation(s)
- Brian Na
- Department of Neurology, UCLA Neuro-Oncology Program, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Shilp R Shah
- Samueli School of Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Harish N Vasudevan
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA.
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
10
|
Corcoran RB, Do KT, Kim JE, Cleary JM, Parikh AR, Yeku OO, Xiong N, Weekes CD, Veneris J, Ahronian LG, Mauri G, Tian J, Norden BL, Michel AG, Van Seventer EE, Siravegna G, Camphausen K, Chi G, Fetter IJ, Brugge JS, Chen H, Takebe N, Penson RT, Juric D, Flaherty KT, Sullivan RJ, Clark JW, Heist RS, Matulonis UA, Liu JF, Shapiro GI. Phase I/II Study of Combined BCL-xL and MEK Inhibition with Navitoclax and Trametinib in KRAS or NRAS Mutant Advanced Solid Tumors. Clin Cancer Res 2024; 30:1739-1749. [PMID: 38456660 PMCID: PMC11061595 DOI: 10.1158/1078-0432.ccr-23-3135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/11/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE MEK inhibitors (MEKi) lack monotherapy efficacy in most RAS-mutant cancers. BCL-xL is an anti-apoptotic protein identified by a synthetic lethal shRNA screen as a key suppressor of apoptotic response to MEKi. PATIENTS AND METHODS We conducted a dose escalation study (NCT02079740) of the BCL-xL inhibitor navitoclax and MEKi trametinib in patients with RAS-mutant tumors with expansion cohorts for: pancreatic, gynecologic (GYN), non-small cell lung cancer (NSCLC), and other cancers harboring KRAS/NRAS mutations. Paired pretreatment and day 15 tumor biopsies and serial cell-free (cf)DNA were analyzed. RESULTS A total of 91 patients initiated treatment, with 38 in dose escalation. Fifty-eight percent had ≥3 prior therapies. A total of 15 patients (17%) had colorectal cancer, 19 (11%) pancreatic, 15 (17%) NSCLC, and 32 (35%) GYN cancers. The recommended phase II dose (RP2D) was established as trametinib 2 mg daily days 1 to 14 and navitoclax 250 mg daily days 1 to 28 of each cycle. Most common adverse events included diarrhea, thrombocytopenia, increased AST/ALT, and acneiform rash. At RP2D, 8 of 49 (16%) evaluable patients achieved partial response (PR). Disease-specific differences in efficacy were noted. In patients with GYN at the RP2D, 7 of 21 (33%) achieved a PR and median duration of response 8.2 months. No PRs occurred in patients with colorectal cancer, NSCLC, or pancreatic cancer. MAPK pathway inhibition was observed in on-treatment tumor biopsies. Reductions in KRAS/NRAS mutation levels in cfDNA correlated with clinical benefit. CONCLUSIONS Navitoclax in combination with trametinib was tolerable. Durable clinical responses were observed in patients with RAS-mutant GYN cancers, warranting further evaluation in this population.
Collapse
Affiliation(s)
- Ryan B. Corcoran
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Khanh T. Do
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jeong E. Kim
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - James M. Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Aparna R. Parikh
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Oladapo O. Yeku
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Niya Xiong
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Colin D. Weekes
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jennifer Veneris
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Leanne G. Ahronian
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Gianluca Mauri
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, and Department of Hematology Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Jun Tian
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Bryanna L. Norden
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Alexa G. Michel
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Emily E. Van Seventer
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Giulia Siravegna
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kyle Camphausen
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Gary Chi
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Isobel J. Fetter
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Joan S. Brugge
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Helen Chen
- National Institute of Health, National Cancer Institute, Division of Cancer Treatment and Diagnosis, Cancer Therapy Evaluation Program, Bethesda, Maryland
| | - Naoko Takebe
- National Institute of Health, National Cancer Institute, Division of Cancer Treatment and Diagnosis, Cancer Therapy Evaluation Program, Bethesda, Maryland
| | - Richard T. Penson
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Dejan Juric
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Keith T. Flaherty
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Ryan J. Sullivan
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey W. Clark
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Rebecca S. Heist
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Ursula A. Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joyce F. Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Geoffrey I. Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
11
|
Elez E, Cubillo A, Alfonso PG, Middleton MR, Chau I, Alkuzweny B, Alcasid A, Zhang X, Van Cutsem E. Binimetinib in combination with nivolumab or nivolumab and ipilimumab in patients with previously treated microsatellite-stable metastatic colorectal cancer with RAS mutations in an open-label phase 1b/2 study. BMC Cancer 2024; 24:446. [PMID: 38600471 PMCID: PMC11007903 DOI: 10.1186/s12885-024-12153-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 03/20/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND In patients with previously treated RAS-mutated microsatellite-stable (MSS) metastatic colorectal cancer (mCRC), a multicenter open-label phase 1b/2 trial was conducted to define the safety and efficacy of the MEK1/MEK2 inhibitor binimetinib in combination with the immune checkpoint inhibitor (ICI) nivolumab (anti-PD-1) or nivolumab and another ICI, ipilimumab (anti-CTLA4). METHODS In phase 1b, participants were randomly assigned to Arm 1A (binimetinib 45 mg twice daily [BID] plus nivolumab 480 mg once every 4 weeks [Q4W]) or Arm 1B (binimetinib 45 mg BID plus nivolumab 480 mg Q4W and ipilimumab 1 mg/kg once every 8 weeks [Q8W]) to determine the maximum tolerable dose (MTD) and recommended phase 2 dose (RP2D) of binimetinib. The MTD/RP2D was defined as the highest dosage combination that did not cause medically unacceptable dose-limiting toxicities in more than 35% of treated participants in Cycle 1. During phase 2, participants were randomly assigned to Arm 2A (binimetinib MTD/RP2D plus nivolumab) or Arm 2B (binimetinib MTD/RP2D plus nivolumab and ipilimumab) to assess the safety and clinical activity of these combinations. RESULTS In phase 1b, 21 participants were randomized to Arm 1A or Arm 1B; during phase 2, 54 participants were randomized to Arm 2A or Arm 2B. The binimetinib MTD/RP2D was determined to be 45 mg BID. In phase 2, no participants receiving binimetinib plus nivolumab achieved a response. Of the 27 participants receiving binimetinib, nivolumab, and ipilimumab, the overall response rate was 7.4% (90% CI: 1.3, 21.5). Out of 75 participants overall, 74 (98.7%) reported treatment-related adverse events (AEs), of whom 17 (22.7%) reported treatment-related serious AEs. CONCLUSIONS The RP2D binimetinib regimen had a safety profile similar to previous binimetinib studies or nivolumab and ipilimumab combination studies. There was a lack of clinical benefit with either drug combination. Therefore, these data do not support further development of binimetinib in combination with nivolumab or nivolumab and ipilimumab in RAS-mutated MSS mCRC. TRIAL REGISTRATION NCT03271047 (09/01/2017).
Collapse
Affiliation(s)
- Elena Elez
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Antonio Cubillo
- Centro Integral, Oncológico Clara Campal, HM CIOCC, Madrid, Spain
- Facultad HM Hospitales de Ciencias de La Salud UCJC, 28050, Madrid, Spain
| | - Pilar Garcia Alfonso
- Medical Oncology Service, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, Madrid, Spain
| | - Mark R Middleton
- Department of Oncology, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Ian Chau
- Gastrointestinal Unit, Royal Marsden Hospital, London & Surrey, UK
| | | | | | | | - Eric Van Cutsem
- University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium
| |
Collapse
|
12
|
Van Cutsem E, Yaeger R, Delord JP, Tabernero J, Siu LL, Ducreux M, Siena S, Elez E, Kasper S, Zander T, Steeghs N, Murphy D, Edwards M, Wainberg ZA. Phase Ib/II Study of the Efficacy and Safety of Binimetinib (MEK162) Plus Panitumumab for Mutant or Wild-Type RAS Metastatic Colorectal Cancer. Oncologist 2023; 28:e1209-e1218. [PMID: 37597246 PMCID: PMC10712701 DOI: 10.1093/oncolo/oyad210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/08/2023] [Indexed: 08/21/2023] Open
Abstract
INTRODUCTION Activating RAS gene mutations occur in approximately 55% of patients with metastatic colorectal cancer (mCRC) and are associated with poorer clinical outcomes due to epidermal growth factor receptor (EGFR) blockade resistance. Combined EGFR and mitogen-activated protein kinase (MEK) inhibition may extend response to EGFR inhibition and overcome acquired resistance. This phase Ib/II dose escalation trial evaluated the safety and activity of dual inhibition with binimetinib (MEK1/2 inhibitor) and panitumumab (EGFR inhibitor [EGFRi]) in patients with RAS mutant or BRAF wild type (WT)/RAS WT mCRC. METHODS Phase Ib dose escalation started with binimetinib 45 mg twice daily plus panitumumab 6 mg/kg administered every 2 weeks. In the phase II study, patients with measurable mCRC were enrolled into 4 groups based on previous anti-EGFR monoclonal antibody therapy and RAS mutational status. RESULTS No patients in the phase Ib portion (n = 10) had a response; 70% of patients had stable disease. In the phase II portion (n = 43), overall response rate (ORR, confirmed) was 2.3% with one partial response in the RAS WT group, DCR was 30.2%, and median progression-free survival was 1.8 months (95%CI, 1.6-3.3). All patients experienced ≥1 adverse event, with the most common being diarrhea (71.7%), vomiting (52.8%), nausea (50.9%), fatigue (49.1%), dermatitis acneiform (43.4%), and rash (41.5%). Most patients required treatment interruption or dose reduction due to difficulties tolerating treatment. CONCLUSIONS The combination of binimetinib and panitumumab had substantial toxicity and limited clinical activity for patients with mutant or WT RAS mCRC, independent of EGFRi treatment history (Trial registration: NCT01927341).
Collapse
Affiliation(s)
- Eric Van Cutsem
- Digestive Oncology, University Hospitals Gasthuisberg, Leuven and KU Leuven, Leuven, Belgium
| | - Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jean-Pierre Delord
- Medical Oncology Department, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France
| | - Josep Tabernero
- Medical Oncology Department, Vall d’Hebron Campus Hospital and Institute of Oncology (VHIO), IOB-Quiron, UVic-UICC, Barcelona, Spain
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Michel Ducreux
- Department of Medical Oncology, Gustave Roussy, Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Oncology, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Elena Elez
- Medical Oncology Department, Vall d’Hebron Campus Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Stefan Kasper
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
| | - Thomas Zander
- Department of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne, University of Cologne, Duesseldorf, Germany
- Department of Internal Medicine I, Gastrointestinal Cancer Group Cologne (GCGC), University Clinic Cologne, Cologne, Germany
| | - Neeltje Steeghs
- Department of Medical Oncology and Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Zev A Wainberg
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
13
|
Saltos AN, Creelan BC, Tanvetyanon T, Chiappori AA, Antonia SJ, Shafique MR, Ugrenovic-Petrovic M, Sansil S, Neuger A, Ozakinci H, Boyle TA, Kim J, Haura EB, Gray JE. A phase I/IB trial of binimetinib in combination with erlotinib in NSCLC harboring activating KRAS or EGFR mutations. Lung Cancer 2023; 183:107313. [PMID: 37499521 DOI: 10.1016/j.lungcan.2023.107313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Activating mutations in EGFR or KRAS are highly prevalent in NSCLC, share activation of the MAPK pathway and may be amenable to combination therapy to prevent negative feedback activation. METHODS In this phase 1/1B trial, we tested the combination of binimetinib and erlotinib in patients with advanced NSCLC with at least 1 prior line of treatment (unless with activating EGFR mutation which could be treatment-naïve). A subsequent phase 1B expansion accrued patients with either EGFR- or KRAS-mutation using the recommended phase 2 dose (RP2D) from Phase 1. The primary objective was to evaluate the safety of binimetinib plus erlotinib and establish the RP2D. RESULTS 43 patients enrolled (dose-escalation = 23; expansion = 20). 17 harbored EGFR mutation and 22 had KRAS mutation. The RP2D was erlotinib 100 mg daily and binimetinib 15 mg BID × 5 days/week. Common AEs across all doses included diarrhea (69.8%), rash (44.2%), fatigue (32.6%), and nausea (32.6%), and were primarily grade 1/2. Among KRAS mutant patients, 1 (5%) had confirmed partial response and 8 (36%) achieved stable disease as best overall response. Among EGFR mutant patients, 9 were TKI-naïve with 8 (89%) having partial response, and 8 were TKI-pretreated with no partial responses and 1 (13%) stable disease as best overall response. CONCLUSIONS Binimetinib plus erlotinib demonstrated a manageable safety profile and modest efficacy including one confirmed objective response in a KRAS mutant patient. While clinical utility of this specific combination was limited, these results support development of combinations using novel small molecule inhibitors of RAS, selective EGFR- and other MAPK pathway inhibitors, many of which have improved therapeutic indices. CLINICAL TRIAL REGISTRATION NCT01859026.
Collapse
Affiliation(s)
- Andreas N Saltos
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA.
| | - Ben C Creelan
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Tawee Tanvetyanon
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Alberto A Chiappori
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Scott J Antonia
- Center for Cancer Immunotherapy, Duke Cancer Institute, 20 Duke Medicine Cir., Durham, NC 27710, USA
| | - Michael R Shafique
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | | | - Samer Sansil
- Cancer Pharmacokinetics & Pharmacodynamics Core, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Anthony Neuger
- Cancer Pharmacokinetics & Pharmacodynamics Core, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Hilal Ozakinci
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Theresa A Boyle
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Eric B Haura
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Jhanelle E Gray
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| |
Collapse
|
14
|
Shabir G, Saeed A, Zahid W, Naseer F, Riaz Z, Khalil N, Muneeba, Albericio F. Chemistry and Pharmacology of Fluorinated Drugs Approved by the FDA (2016-2022). Pharmaceuticals (Basel) 2023; 16:1162. [PMID: 37631077 PMCID: PMC10458641 DOI: 10.3390/ph16081162] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Fluorine is characterized by high electronegativity and small atomic size, which provide this molecule with the unique property of augmenting the potency, selectivity, metabolic stability, and pharmacokinetics of drugs. Fluorine (F) substitution has been extensively explored in drug research as a means of improving biological activity and enhancing chemical or metabolic stability. Selective F substitution onto a therapeutic or diagnostic drug candidate can enhance several pharmacokinetic and physicochemical properties such as metabolic stability and membrane permeation. The increased binding ability of fluorinated drug target proteins has also been reported in some cases. An emerging line of research on F substitution has been addressed by using 18F as a radiolabel tracer atom in the extremely sensitive methodology of positron emission tomography (PET) imaging. This review aims to report on the fluorinated drugs approved by the US Food and Drug Administration (FDA) from 2016 to 2022. It cites selected examples from a variety of therapeutic and diagnostic drugs. FDA-approved drugs in this period have a variety of heterocyclic cores, including pyrrole, pyrazole, imidazole, triazole, pyridine, pyridone, pyridazine, pyrazine, pyrimidine, triazine, purine, indole, benzimidazole, isoquinoline, and quinoline appended with either F-18 or F-19. Some fluorinated oligonucleotides were also authorized by the FDA between 2019 and 2022.
Collapse
Affiliation(s)
- Ghulam Shabir
- Department of Chemistry, Quaid-I-Azam University, Islamabad 45320, Pakistan;
| | - Aamer Saeed
- Department of Chemistry, Quaid-I-Azam University, Islamabad 45320, Pakistan;
| | - Wajeeha Zahid
- Department of Chemistry, Government Graduate College Toba Tek Singh, Punjab 36050, Pakistan; (W.Z.); (F.N.); (Z.R.); (N.K.); (M.)
| | - Fatima Naseer
- Department of Chemistry, Government Graduate College Toba Tek Singh, Punjab 36050, Pakistan; (W.Z.); (F.N.); (Z.R.); (N.K.); (M.)
| | - Zainab Riaz
- Department of Chemistry, Government Graduate College Toba Tek Singh, Punjab 36050, Pakistan; (W.Z.); (F.N.); (Z.R.); (N.K.); (M.)
| | - Nafeesa Khalil
- Department of Chemistry, Government Graduate College Toba Tek Singh, Punjab 36050, Pakistan; (W.Z.); (F.N.); (Z.R.); (N.K.); (M.)
| | - Muneeba
- Department of Chemistry, Government Graduate College Toba Tek Singh, Punjab 36050, Pakistan; (W.Z.); (F.N.); (Z.R.); (N.K.); (M.)
| | - Fernando Albericio
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
15
|
Rodon Ahnert J, Tan DSW, Garrido-Laguna I, Harb W, Bessudo A, Beck JT, Rottey S, Bahary N, Kotecki N, Zhu Z, Deng S, Kowalski K, Wei C, Pathan N, Laliberte RJ, Messersmith WA. Avelumab or talazoparib in combination with binimetinib in metastatic pancreatic ductal adenocarcinoma: dose-finding results from phase Ib of the JAVELIN PARP MEKi trial. ESMO Open 2023; 8:101584. [PMID: 37379764 PMCID: PMC10515283 DOI: 10.1016/j.esmoop.2023.101584] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/07/2023] [Accepted: 05/15/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Combinations of avelumab [anti-programmed death-ligand 1 (anti-PD-L1)] or talazoparib [poly(adenosine diphosphate ribose) polymerase (PARP) inhibitor] with binimetinib (MEK inhibitor) were expected to result in additive or synergistic antitumor activity relative to each drug administered alone. Here, we report phase Ib results from JAVELIN PARP MEKi, which investigated avelumab or talazoparib combined with binimetinib in metastatic pancreatic ductal adenocarcinoma (mPDAC). PATIENTS AND METHODS Patients with mPDAC that had progressed with prior treatment received avelumab 800 mg every 2 weeks plus binimetinib 45 mg or 30 mg two times daily (continuous), or talazoparib 0.75 mg daily plus binimetinib 45 mg or 30 mg two times daily (7 days on/7 days off). The primary endpoint was dose-limiting toxicity (DLT). RESULTS A total of 22 patients received avelumab plus binimetinib 45 mg (n = 12) or 30 mg (n = 10). Among DLT-evaluable patients, DLT occurred in five of 11 patients (45.5%) at the 45-mg dose, necessitating de-escalation to 30 mg; DLT occurred in three of 10 patients (30.0%) at the 30-mg dose. Among patients treated at the 45-mg dose, one (8.3%) had a best overall response of partial response. Thirteen patients received talazoparib plus binimetinib 45 mg (n = 6) or 30 mg (n = 7). Among DLT-evaluable patients, DLT occurred in two of five patients (40.0%) at the 45-mg dose, necessitating de-escalation to 30 mg; DLT occurred in two of six patients (33.3%) at the 30-mg dose. No objective responses were observed. CONCLUSIONS Combinations of avelumab or talazoparib plus binimetinib resulted in higher-than-expected DLT rates. However, most DLTs were single occurrences, and the overall safety profiles were generally consistent with those reported for the single agents. CLINICAL TRIAL REGISTRATION ClinicalTrials.govNCT03637491; https://clinicaltrials.gov/ct2/show/NCT03637491.
Collapse
Affiliation(s)
- J Rodon Ahnert
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA.
| | - D S-W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - I Garrido-Laguna
- Division of Oncology, University of Utah Huntsman Cancer Institute, Salt Lake City, USA
| | - W Harb
- Syneos Health, Morrisville, USA
| | - A Bessudo
- California Cancer Associates for Research and Excellence, San Diego, USA
| | - J T Beck
- Highlands Oncology, Springdale, USA
| | - S Rottey
- Department of Medical Oncology, UZ Gent, Gent, Belgium
| | - N Bahary
- AHN Cancer Institute, Allegheny Health Network, Pittsburgh, USA
| | - N Kotecki
- Department of Medical Oncology, Jules Bordet Institute, Brussels, Belgium
| | | | | | | | | | | | | | - W A Messersmith
- Division of Medical Oncology, University of Colorado Cancer Center, Aurora, USA
| |
Collapse
|
16
|
Bauer S, Larkin J, Hodi FS, Stephen F, Kapiteijn EHW, Schwartz GK, Calvo E, Yerramilli-Rao P, Piperno-Neumann S, Carvajal RD. A phase Ib trial of combined PKC and MEK inhibition with sotrastaurin and binimetinib in patients with metastatic uveal melanoma. Front Oncol 2023; 12:975642. [PMID: 37359242 PMCID: PMC10288853 DOI: 10.3389/fonc.2022.975642] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/28/2022] [Indexed: 06/28/2023] Open
Abstract
Background Uveal melanoma is a disease characterized by constitutive activation of the G alpha pathway and downstream signaling of protein kinase C (PKC) and the mitogen-activated protein kinase (MAPK) pathway. While limited clinical activity has been observed in patients with metastatic disease with inhibition of PKC or MEK alone, preclinical data has demonstrated synergistic antitumor effects with concurrent inhibition of PKC and MEK. Method We conducted a phase Ib study of the PKC inhibitor sotrastaurin in combination with the MEK inhibitor binimetinib in patients with metastatic uveal melanoma using a Bayesian logistic regression model guided by the escalation with overdose control principle (NCT01801358). Serial blood samples and paired tumor samples were collected for pharmacokinetic (PK) and pharmacodynamic analysis. Results Thirty-eight patients were treated across six dose levels. Eleven patients experienced DLTs across the five highest dose levels tested, most commonly including vomiting (n=3), diarrhea (n=3), nausea (n=2), fatigue (n=2) and rash (n=2). Common treatment related adverse events included diarrhea (94.7%), nausea (78.9%), vomiting (71.1%), fatigue (52.6%), rash (39.5%), and elevated blood creating phosphokinase (36.8%). Two dose combinations satisfying criteria for the maximum tolerated dose (MTD) were identified: (1) sotrastaurin 300 mg and binimetinib 30 mg; and, (2) sotrastaurin 200 mg and binimetinib 45 mg. Exposure to both drugs in combination was consistent with single-agent data for either drug, indicating no PK interaction between sotrastaurin and binimetinib. Stable disease was observed in 60.5% of patients treated. No patient achieved a radiographic response per RECIST v1.1. Conclusions Concurrent administration of sotrastaurin and binimetinib is feasible but associated with substantial gastrointestinal toxicity. Given the limited clinical activity achieved with this regimen, accrual to the phase II portion of the trial was not initiated.
Collapse
Affiliation(s)
- Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Duisburg-Essen, Medical School, Essen, Germany
| | - James Larkin
- Department of Medical Oncology and Hematology, The Royal Marsden Hospital, London, United Kingdom
| | - F. Stephen Hodi
- Melanoma Center and Center for Immuno-Oncology, Dana−Farber Cancer Institute, Boston, MD, United States
| | - Frank Stephen
- Hebrew University Hadassah Medical School, The Sharett Institute of Oncology, Jerusalem, Israel
- Jacob Schachter, Sheba Medical Center at Tel Hashomer, Tel-Aviv University Medical School, Tel Aviv, Israel
| | - Ellen H. W. Kapiteijn
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, Netherlands
| | - Gary K. Schwartz
- Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, United States
| | - Emilano Calvo
- Early Phase Clinical Drug Development in Oncology, START Madrid-CIOCC, Centro Integral Oncológico Clara Campa, Madrid, Spain
| | - Padmaja Yerramilli-Rao
- Translational Clinical Oncology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States
| | | | - Richard D. Carvajal
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, Netherlands
| |
Collapse
|
17
|
Zeng W, Mao R, Zhang Z, Chen X. Combination Therapies for Advanced Biliary Tract Cancer. J Clin Transl Hepatol 2023; 11:490-501. [PMID: 36643047 PMCID: PMC9817051 DOI: 10.14218/jcth.2022.00277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/09/2022] [Accepted: 08/18/2022] [Indexed: 01/18/2023] Open
Abstract
Biliary tract cancers (BTCs) are a group of malignant neoplasms that have recently increased in incidence and have a poor prognosis. Surgery is the only curative therapy. However, most patients are only indicated for palliative therapy because of advanced-stage disease at diagnosis and rapid progression. The current first-line treatment for advanced BTC is gemcitabine and cisplatin chemotherapy. Nonetheless, many patients develop resistance to this regimen. Over the years, few chemotherapy regimens have managed to improve the overall survival of patients. Accordingly, novel therapies such as targeted therapy have been introduced to treat this patient population. Extensive research on tumorigenesis and the genetic profiling of BTC have revealed the heterogenicity and potential target pathways, such as EGFR, VEGF, MEK/ERK, PI3K and mTOR. Moreover, mutational analysis has documented the presence of IDH1, FGFR2, HER2, PRKACA, PRKACB, BRAF, and KRAS gene aberrations. The emergence of immunotherapy in recent years has expanded the treatment landscape for this group of malignancies. Cancer vaccines, adoptive cell transfer, and immune checkpoint inhibitors have been extensively investigated in trials of BTC. Therefore, patient stratification and a combination of various therapies have become a reasonable and important clinical strategy to improve patient outcomes. This review elaborates the literature on combined treatment strategies for advanced BTC from the past few years and ongoing clinical trials to provide new inspiration for the treatment of advanced BTC.
Collapse
Affiliation(s)
- Weifeng Zeng
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, Hubei, China
- Hubei key laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruiqi Mao
- Clinic Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, Hubei, China
- Hubei key laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Correspondence to: Zhanguo Zhang and Xiaoping Chen, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Huazhong University of Science and Technology, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, 1095 Jiefang Avenue, Wuhan, Hubei 430030, China. ORCID: https://orcid.org/0000-0002-4527-4975 (ZZ). Tel: +86-27-83663400, Fax: +86-27-83662851, E-mail: (ZZ) and (XC)
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, Hubei, China
- Hubei key laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Correspondence to: Zhanguo Zhang and Xiaoping Chen, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Huazhong University of Science and Technology, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, 1095 Jiefang Avenue, Wuhan, Hubei 430030, China. ORCID: https://orcid.org/0000-0002-4527-4975 (ZZ). Tel: +86-27-83663400, Fax: +86-27-83662851, E-mail: (ZZ) and (XC)
| |
Collapse
|
18
|
Liu Y, Ma L, Li M, Tian Z, Yang M, Wu X, Wang X, Shang G, Xie M, Chen Y, Liu X, Jiang L, Wu W, Xu C, Xia L, Li G, Dai S, Chen Z. Structures of human TR4LBD-JAZF1 and TR4DBD-DNA complexes reveal the molecular basis of transcriptional regulation. Nucleic Acids Res 2023; 51:1443-1457. [PMID: 36651297 PMCID: PMC9943680 DOI: 10.1093/nar/gkac1259] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/30/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Testicular nuclear receptor 4 (TR4) modulates the transcriptional activation of genes and plays important roles in many diseases. The regulation of TR4 on target genes involves direct interactions with DNA molecules via the DNA-binding domain (DBD) and recruitment of coregulators by the ligand-binding domain (LBD). However, their regulatory mechanisms are unclear. Here, we report high-resolution crystal structures of TR4DBD, TR4DBD-DNA complexes and the TR4LBD-JAZF1 complex. For DNA recognition, multiple factors come into play, and a specific mutual selectivity between TR4 and target genes is found. The coactivators SRC-1 and CREBBP can bind at the interface of TR4 originally occupied by the TR4 activation function region 2 (AF-2); however, JAZF1 suppresses the binding through a novel mechanism. JAZF1 binds to an unidentified surface of TR4 and stabilizes an α13 helix never reported in the nuclear receptor family. Moreover, the cancer-associated mutations affect the interactions and the transcriptional activation of TR4 in vitro and in vivo, respectively. Overall, our results highlight the crucial role of DNA recognition and a novel mechanism of how JAZF1 reinforces the autorepressed conformation and influences the transcriptional activation of TR4, laying out important structural bases for drug design for a variety of diseases, including diabetes and cancers.
Collapse
Affiliation(s)
- Yunlong Liu
- State Key Laboratory of Agrobiotechnology and Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Lulu Ma
- State Key Laboratory of Agrobiotechnology and Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Min Li
- National Protein Science Facility, Tsinghua University, Beijing 100084, China
| | - Zizi Tian
- State Key Laboratory of Agrobiotechnology and Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Meiting Yang
- State Key Laboratory of Agrobiotechnology and Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xi Wu
- State Key Laboratory of Agrobiotechnology and Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xue Wang
- State Key Laboratory of Agrobiotechnology and Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Guohui Shang
- State Key Laboratory of Agrobiotechnology and Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Mengjia Xie
- State Key Laboratory of Agrobiotechnology and Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yiyun Chen
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
| | - Xin Liu
- State Key Laboratory of Agrobiotechnology and Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Lun Jiang
- State Key Laboratory of Agrobiotechnology and Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Wei Wu
- State Key Laboratory of Agrobiotechnology and Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Chaoqun Xu
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicines, Ministry of Education, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Liqun Xia
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Gonghui Li
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Shaodong Dai
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Zhongzhou Chen
- To whom correspondence should be addressed. Tel: +86 10 62734078; Fax: +86 10 62734078;
| |
Collapse
|
19
|
Wang X, Luo Z, Chen J, Chen Y, Ji D, Fan L, Chen L, Zhao Q, Hu P, Sun P, Jia Z, Guo J, Si L. First-in-human phase I dose-escalation and dose-expansion trial of the selective MEK inhibitor HL-085 in patients with advanced melanoma harboring NRAS mutations. BMC Med 2023; 21:2. [PMID: 36600247 PMCID: PMC9814429 DOI: 10.1186/s12916-022-02669-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 11/18/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND HL-085 is a selective, orally administered MEK1/2 inhibitor. We aimed to evaluate the safety and efficacy of HL-085 in patients with advanced melanoma harboring NRAS mutations. METHODS This was a multicenter phase 1 study. HL-085 was administered twice daily in a standard 3 + 3 dose-escalation design (10 dose cohorts; 0.5-18 mg twice daily), followed by dose expansion at the recommended phase II dose (RP2D). The primary endpoints included tolerability, dose-limiting toxicity (DLT), maximum tolerated dose (MTD) and RP2D. RESULTS Between September 13, 2017, and January 18, 2021, 42 patients were enrolled (dose escalation phase: n = 30; dose expansion phase: n = 12). No DLT was reported during dose escalation and MTD was not reached with HL-085 doses up to 18 mg twice daily. The RP2D was 12 mg twice daily. The most common all-grade drug-related adverse events (AEs) across all dose levels were rash (61.9%), increased creatine phosphokinase (CK, 59.5%), face edema (50.0%), increased aspartate aminotransferase (47.6%), peripheral edema (40.5%), diarrhea (33.3%), alanine aminotransferase (33.3%), and paronychia (19.0%), most of which were grade 1 and 2. Most frequency of grade ≥ 3 AEs were CK (14.2%), asthenia (7.1%), peripheral edema (4.8%), and acneiform dermatitis (4.8%). In the cohort of 12 mg twice daily dose (15 patients), confirmed objective response rate was 26.7%; disease control rate was 86.7%; median duration of response was 2.9 months; median progression-free survival was 3.6 months. CONCLUSIONS The HL-085 showed acceptable tolerability and substantial clinical activity in patients with advanced melanoma harboring NRAS mutations. TRIAL REGISTRATION Trial registration ClinicalTrials.gov number: NCT03973151.
Collapse
Affiliation(s)
- Xuan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Research Institute, Beijing, China
| | - Zhiguo Luo
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jing Chen
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Chen
- Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Dongmei Ji
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Li Fan
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Chen
- Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Qian Zhao
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Pei Hu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Peng Sun
- Department of Clinical Research and Development, Shanghai Kechow Pharma, Inc., Shanghai, China
| | - Zhongwei Jia
- Department of Clinical Research and Development, Shanghai Kechow Pharma, Inc., Shanghai, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Research Institute, Beijing, China.
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Research Institute, Beijing, China.
| |
Collapse
|
20
|
Garutti M, Bergnach M, Polesel J, Palmero L, Pizzichetta MA, Puglisi F. BRAF and MEK Inhibitors and Their Toxicities: A Meta-Analysis. Cancers (Basel) 2022; 15:cancers15010141. [PMID: 36612138 PMCID: PMC9818023 DOI: 10.3390/cancers15010141] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
PURPOSE This meta-analysis summarizes the incidence of treatment-related adverse events (AE) of BRAFi and MEKi. METHODS A systematic search of Medline/PubMed was conducted to identify suitable articles published in English up to 31 December 2021. The primary outcomes were profiles for all-grade and grade 3 or higher treatment-related AEs, and the analysis of single side effects belonging to both categories. RESULTS The overall incidence of treatment-related all-grade Aes was 99% for Encorafenib (95% CI: 0.97-1.00) and 97% for Trametinib (95% CI: 0.92-0.99; I2 = 66%) and Binimetinib (95% CI: 0.94-0.99; I2 = 0%). In combined therapies, the rate was 98% for both Vemurafenib + Cobimetinib (95% CI: 0.96-0.99; I2 = 77%) and Encorafenib + Binimetinib (95% CI: 0.96-1.00). Grade 3 or higher adverse events were reported in 69% of cases for Binimetinib (95% CI: 0.50-0.84; I2 = 71%), 68% for Encorafenib (95% CI: 0.61-0.74), and 72% for Vemurafenib + Cobimetinib (95% CI: 0.65-0.79; I2 = 84%). The most common grade 1-2 AEs were pyrexia (43%) and fatigue (28%) for Dabrafenib + Trametinib and diarrhea for both Vemurafenib + Cobimetinib (52%) and Encorafenib + Binimetinib (34%). The most common AEs of grade 3 or higher were pyrexia, rash, and hypertension for Dabrafenib + Trametinib (6%), rash and hypertension for Encorafenib + Binimetinib (6%), and increased AST and ALT for Vemurafenib + Cobimetinib (10%). CONCLUSIONS Our study provides comprehensive data on treatment-related adverse events of BRAFi and MEKi combination therapies, showing related toxicity profiles to offer a helpful tool for clinicians in the choice of therapy.
Collapse
Affiliation(s)
- Mattia Garutti
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Correspondence: ; Tel.: +39-04-3465-9092
| | | | - Jerry Polesel
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Lorenza Palmero
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Maria Antonietta Pizzichetta
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Department of Dermatology, University of Trieste, 34123 Trieste, Italy
| | - Fabio Puglisi
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Department of Medicine, University of Udine, 33100 Udine, Italy
| |
Collapse
|
21
|
Barteselli G, Goodman GR, Patel Y, Caro I, Xue C, McCallum S. Characterization of Serous Retinopathy Associated with Cobimetinib: Integrated Safety Analysis of Four Studies. Drug Saf 2022; 45:1491-1499. [PMID: 36310331 PMCID: PMC9700562 DOI: 10.1007/s40264-022-01248-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2022] [Indexed: 12/20/2022]
Abstract
INTRODUCTION AND OBJECTIVE Serous retinopathy can be associated with MEK inhibitors, including cobimetinib. We present results of an integrated safety analysis to further characterize ocular functional and structural changes due to serous retinopathy. METHODS Four studies evaluating cobimetinib at the approved dose and schedule in combination with other oncology drugs were included. Study CO39721 incorporated standardized ophthalmologic assessments to fully characterize serous retinopathy events over time and was the primary study for analysis. Supporting information was provided by studies GO28141, WO29479, and GO30182. RESULTS In total, 655 patients received one or more doses of cobimetinib and comprised the safety-evaluable population. Overall, 117 patients (17.9%) had one or more serous retinopathy events, 24 (3.7%) had two or more events, and four (0.6%) had three or more events. Grade 3 events occurred in < 2.5% of patients. In CO39721, the median time to onset was 15 days (range 7-111); median time to resolution of first occurrence was 26 days (range 6-591 + days). Twelve of 25 patients (48.0%) recovered without a dose modification and 4/25 (16.0%) were recovered/recovering following a dose modification. The most frequent presentation of serous retinopathy was focal subretinal fluid on optical coherence tomography (62.8% of cases); in some instances (25.7% of cases), subretinal fluid was multifocal. There was no loss of visual function or visual acuity at serous retinopathy onset or resolution. CONCLUSIONS Results from this integrated safety analysis indicate that cobimetinib-associated serous retinopathy can be managed with or without a dose modification of cobimetinib at the discretion of the treating physician. No visual loss or permanent retinal damage was identified on comprehensive ophthalmologic assessments. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov identifiers: NCT03178851, NCT01689519, NCT02322814, and NCT02788279.
Collapse
Affiliation(s)
- Giulio Barteselli
- Genentech, Inc. (a Member of the Roche Group), 1 DNA Way, South San Francisco, CA, USA
| | - Grant R Goodman
- Genentech, Inc. (a Member of the Roche Group), 1 DNA Way, South San Francisco, CA, USA
| | - Yogesh Patel
- F. Hoffmann-La Roche Ltd, Welwyn Garden City, UK
| | - Ivor Caro
- Genentech, Inc. (a Member of the Roche Group), 1 DNA Way, South San Francisco, CA, USA
| | - Cloris Xue
- Hoffmann-La Roche Ltd, Mississauga, ON, Canada
| | - Samuel McCallum
- Genentech, Inc. (a Member of the Roche Group), 1 DNA Way, South San Francisco, CA, USA.
| |
Collapse
|
22
|
Wei XL, Zhang Y, Zhao HY, Fang WF, Luo HY, Qiu MZ, He MM, Zou BY, Xie J, Jin CL, Zhou XF, Wang F, Wang FH, Li YH, Wang ZQ, Xu RH. Safety and Clinical Activity of SHR7390 Monotherapy or Combined With Camrelizumab for Advanced Solid Tumor: Results From Two Phase I Trials. Oncologist 2022; 28:e36-e44. [PMID: 36398872 PMCID: PMC9847543 DOI: 10.1093/oncolo/oyac225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/16/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND SHR7390 is a novel, selective MEK1/2 inhibitor. Here, we report results from two phase I trials conducted to evaluate the tolerability, safety and antitumor activity of SHR7390 monotherapy for advanced solid tumors and SHR7390 plus camrelizumab for treatment-refractory advanced or metastatic colorectal cancer (CRC). PATIENTS AND METHODS Patients received SHR7390 alone or combined with fixed-dose camrelizumab (200 mg every 2 weeks) in an accelerated titration scheme to determine the maximum tolerated dose (MTD). A recommended dose for expansion was determined based on the safety and tolerability of the dose-escalation stage. The primary endpoints were dose limiting toxicity (DLT) and MTD. RESULTS In the SHR7390 monotherapy trial, 16 patients were enrolled. DLTs were reported in the 1.0 mg cohort, and the MTD was 0.75 mg. Grade ≥3 treatment-related adverse events (TRAEs) were recorded in 4 patients (25.0%). No patients achieved objective response. In the SHR7390 combination trial, 22 patients with CRC were enrolled. One DLT was reported in the 0.5 mg cohort and the MTD was not reached. Grade ≥3 TRAEs were observed in 8 patients (36.4%), with the most common being rash (n=4). One grade 5 TRAE (increased intracranial pressure) occurred. Five patients (22.7%) achieved partial response, including one of 3 patients with MSS/MSI-L and BRAF mutant tumors, one of 15 patients with MSS/MSI-L and BRAF wild type tumors, and all 3 patients with MSI-H tumors. CONCLUSIONS SHR7390 0.5 mg plus camrelizumab showed a manageable safety profile. Preliminary clinical activity was reported regardless of MSI and BRAF status.
Collapse
Affiliation(s)
- Xiao-Li Wei
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Yang Zhang
- Department of Clinical Research Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Hong-Yun Zhao
- Department of Clinical Research Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Wen-Feng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Hui-Yan Luo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Miao-Zhen Qiu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Ming-Ming He
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Ben-Yan Zou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Jie Xie
- Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd. (formerly Jiangsu Hengrui Medicine Co., Ltd.), Shanghai, People’s Republic of China
| | - Chun-Lei Jin
- Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd. (formerly Jiangsu Hengrui Medicine Co., Ltd.), Shanghai, People’s Republic of China
| | - Xian-Feng Zhou
- Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd. (formerly Jiangsu Hengrui Medicine Co., Ltd.), Shanghai, People’s Republic of China
| | - Feng Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Feng-Hua Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Yu-Hong Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Zhi-Qiang Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Rui-Hua Xu
- Corresponding author: Rui-Hua Xu, MD, Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Science; 651 Dong Feng Road East, Guangzhou 510060, Guangdong Province, People’s Republic of China. Tel: +86 20 8734 3468; Fax: +86 20 8734 3468;
| |
Collapse
|
23
|
de Blank PMK, Gross AM, Akshintala S, Blakeley JO, Bollag G, Cannon A, Dombi E, Fangusaro J, Gelb BD, Hargrave D, Kim A, Klesse LJ, Loh M, Martin S, Moertel C, Packer R, Payne JM, Rauen KA, Rios JJ, Robison N, Schorry EK, Shannon K, Stevenson DA, Stieglitz E, Ullrich NJ, Walsh KS, Weiss BD, Wolters PL, Yohay K, Yohe ME, Widemann BC, Fisher MJ. MEK inhibitors for neurofibromatosis type 1 manifestations: Clinical evidence and consensus. Neuro Oncol 2022; 24:1845-1856. [PMID: 35788692 PMCID: PMC9629420 DOI: 10.1093/neuonc/noac165] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The wide variety of clinical manifestations of the genetic syndrome neurofibromatosis type 1 (NF1) are driven by overactivation of the RAS pathway. Mitogen-activated protein kinase kinase inhibitors (MEKi) block downstream targets of RAS. The recent regulatory approvals of the MEKi selumetinib for inoperable symptomatic plexiform neurofibromas in children with NF1 have made it the first medical therapy approved for this indication in the United States, the European Union, and elsewhere. Several recently published and ongoing clinical trials have demonstrated that MEKi may have potential benefits for a variety of other NF1 manifestations, and there is broad interest in the field regarding the appropriate clinical use of these agents. In this review, we present the current evidence regarding the use of existing MEKi for a variety of NF1-related manifestations, including tumor (neurofibromas, malignant peripheral nerve sheath tumors, low-grade glioma, and juvenile myelomonocytic leukemia) and non-tumor (bone, pain, and neurocognitive) manifestations. We discuss the potential utility of MEKi in related genetic conditions characterized by overactivation of the RAS pathway (RASopathies). In addition, we review practical treatment considerations for the use of MEKi as well as provide consensus recommendations regarding their clinical use from a panel of experts.
Collapse
Affiliation(s)
- Peter M K de Blank
- Department of Pediatrics, University of Cincinnati and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Andrea M Gross
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Jaishri O Blakeley
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Ashley Cannon
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Eva Dombi
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Jason Fangusaro
- Children's Hospital of Atlanta, Emory University and the Aflac Cancer Center, Atlanta, Georgia, USA
| | - Bruce D Gelb
- Department of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Darren Hargrave
- Department of Oncology, Great Ormond Street Hospital for Children, London, UK
| | - AeRang Kim
- Center for Neuroscience and Behavioral Medicine and Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - Laura J Klesse
- Department of Pediatrics, Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Mignon Loh
- Benioff Children's Hospital, University of California San Francisco, San Francisco, California, USA
| | - Staci Martin
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Christopher Moertel
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Roger Packer
- Center for Neuroscience and Behavioral Medicine and Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - Jonathan M Payne
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Katherine A Rauen
- Department of Pediatrics, University of California Davis, Sacramento, California, USA
| | - Jonathan J Rios
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, Texas, USA
| | - Nathan Robison
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Elizabeth K Schorry
- Department of Pediatrics, University of Cincinnati and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kevin Shannon
- Benioff Children's Hospital, University of California San Francisco, San Francisco, California, USA
| | - David A Stevenson
- Department of Pediatrics, Division of Medical Genetics, Stanford University, Stanford, California, USA
| | - Elliot Stieglitz
- Benioff Children's Hospital, University of California San Francisco, San Francisco, California, USA
| | - Nicole J Ullrich
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Karin S Walsh
- Center for Neuroscience and Behavioral Medicine and Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - Brian D Weiss
- Department of Pediatrics, University of Cincinnati and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Pamela L Wolters
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Kaleb Yohay
- Department of Neurology and Pediatrics, New York University Grossman School of Medicine, New York, New York, USA
| | - Marielle E Yohe
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Brigitte C Widemann
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Michael J Fisher
- Division of Oncology, The Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
24
|
Zhao Q, Wang T, Wang H, Cui C, Zhong W, Fu D, Xi W, Si L, Guo J, Cheng Y, Tian H, Hu P. Phase I pharmacokinetic study of an oral, small-molecule MEK inhibitor tunlametinib in patients with advanced NRAS mutant melanoma. Front Pharmacol 2022; 13:1039416. [PMID: 36386136 PMCID: PMC9663925 DOI: 10.3389/fphar.2022.1039416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
Background: Malignant melanoma is an aggressive disease. Tunlametinib (HL-085) is a potent, selective, and orally bioavailable MEK1/2 inhibitor. The objective of this study was to determine the pharmacokinetics (PK) of tunlametinib and its main metabolite M8 in patients with NRAS-mutant melanoma following a single dose and multiple doses in a phase I safety and PK study. Methods: A multiple-center phase I study was performed in patients with melanoma including dose-escalation phase and dose-expansion phase. PK following a single oral dose and multiple doses of 0.5–18 mg twice daily was assessed. Results: A total of 30 participants were included in the dose escalation phase and then 11 patients were included in the dose-expansion phase (12 mg twice daily). Tunlametinib plasma concentration rapidly increased after dosing, with a Tmax of 0.5–1 h. Mean elimination half-life (t1/2) was dose-independent and had a range from 21.84 to 34.41 h. Mean apparent clearance (CL/F) and distribution volume (V/F) were 28.44–51.93 L/h and 1199.36–2009.26 L, respectively. The average accumulation ratios of AUC and Cmax after the multiple administration of tunlametinib were 1.64–2.73 and 0.82–2.49, respectively. Tunlametinib was rapidly transformed into the main metabolite M8 and M8 reached the peak concentration about 1 h after administration. Mean t1/2 of M8 was 6.1–33.54 h. The body exposure of M8 in plasma was 36%–67% of that of tunlametinib. There were general dose-proportional increases in maximum concentration (Cmax) and area under the curve (AUC) of tunlametinib and M8 both in the single dose phase and in the multiple doses phase. Conclusion: Tunlametinib was absorbed rapidly and eliminated at a medium speed after drug withdrawal. Pharmacokinetic body exposure increased in general dose-proportional manner from 0.5 mg up to 18 mg. Slight accumulation was found after multiple oral doses. The pharmacokinetics of tunlametinib and its metabolite suggest that twice daily dosing is appropriate for tunlametinib.
Collapse
Affiliation(s)
- Qian Zhao
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Teng Wang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Huanhuan Wang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Cheng Cui
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Wen Zhong
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Diyi Fu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Wanlin Xi
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Research Institute, Beijing, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Research Institute, Beijing, China
| | - Ying Cheng
- Shanghai KeChow Pharma, Inc., Shanghai, China
| | - Hongqi Tian
- Shanghai KeChow Pharma, Inc., Shanghai, China
| | - Pei Hu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
- *Correspondence: Pei Hu,
| |
Collapse
|
25
|
Schuler M, Zimmer L, Kim KB, Sosman JA, Ascierto PA, Postow MA, De Vos FY, van Herpen CM, Carlino MS, Johnson DB, Berking C, Reddy MB, Harney AS, Berlin JD, Amaria RN. Phase Ib/II Trial of Ribociclib in Combination with Binimetinib in Patients with NRAS-mutant Melanoma. Clin Cancer Res 2022; 28:3002-3010. [PMID: 35294522 PMCID: PMC9365377 DOI: 10.1158/1078-0432.ccr-21-3872] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/22/2022] [Accepted: 03/10/2022] [Indexed: 01/26/2023]
Abstract
PURPOSE Enhanced MAPK pathway signaling and cell-cycle checkpoint dysregulation are frequent in NRAS-mutant melanoma and, as such, the regimen of the MEK inhibitor binimetinib and the selective CDK4/6 inhibitor ribociclib is a rational combination. PATIENTS AND METHODS This is a phase Ib/II, open-label study of ribociclib + binimetinib in patients with NRAS-mutant melanoma (NCT01781572). Primary objectives were to estimate the MTD/recommended phase II dose (RP2D) of the combination (phase Ib) and to characterize combination antitumor activity at the RP2D (phase II). Tumor genomic characterization and pharmacokinetics/pharmacodynamics were also evaluated. RESULTS Ten patients (16.4%) experienced dose-limiting toxicities in cycle 1 of phase Ib. Overall response rate in the phase II cohort (n = 41) for the selected RP2D (binimetinib 45 mg twice daily + ribociclib 200 mg once daily, 21 days on/7 days off) was 19.5% [8/41; 95% confidence interval (CI), 8.8-34.9]. The response rate was 32.5% (13/40; 95% CI, 20.1-48.0) in patients with NRAS mutation with concurrent alterations of CDKN2A, CDK4, or CCND1. Median progression-free survival was 3.7 months (95% CI, 3.5-5.6) and median overall survival was 11.3 months (95% CI, 9.3-14.2) for all patients. Common treatment-related toxicities included creatine phosphokinase elevation, rash, edema, anemia, nausea, diarrhea, and fatigue. Pharmacokinetics and safety were consistent with single-agent data, supporting a lack of drug-drug interaction. CONCLUSIONS Ribociclib + binimetinib can be safely administered and is clinically active in patients with NRAS-mutant melanoma. Co-mutations of cell-cycle genes may define a population with greater likelihood of treatment benefit. See related commentary by Moschos, p. 2977.
Collapse
Affiliation(s)
- Martin Schuler
- West German Cancer Center Essen, Department of Medical Oncology, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK), partner site University Hospital Essen, Essen, Germany
| | - Lisa Zimmer
- West German Cancer Center Essen, Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Kevin B. Kim
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Jeffrey A. Sosman
- Robert H. Lurie Cancer Center, Northwestern Medical Group, Chicago, Illinois
| | - Paolo A. Ascierto
- Melanoma Unit, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Michael A. Postow
- Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Filip Y.F.L. De Vos
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | - Matteo S. Carlino
- Westmead Hospital, Crown Princess, Mary Cancer Centre, Corner of Hawkesbury and Darcy Roads, Westmead, New South Wales, Australia.,Australia Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Douglas B. Johnson
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, The Vanderbilt Clinic, Nashville, Tennessee
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | | | | | - Jordan D. Berlin
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, The Vanderbilt Clinic, Nashville, Tennessee
| | - Rodabe N. Amaria
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Corresponding Author: Rodabe N. Amaria, Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030. Phone: 713-745-5530; E-mail:
| |
Collapse
|
26
|
Rajkumar S, Berry D, Heney KA, Strong C, Ramsay L, Lajoie M, Alkallas R, Nguyen TT, Thomson C, Ahanfeshar-Adams M, Dankner M, Petrella T, Rose AAN, Siegel PM, Watson IR. Melanomas with concurrent BRAF non-p.V600 and NF1 loss-of-function mutations are targetable by BRAF/MEK inhibitor combination therapy. Cell Rep 2022; 39:110634. [PMID: 35385748 DOI: 10.1016/j.celrep.2022.110634] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/15/2021] [Accepted: 03/16/2022] [Indexed: 02/08/2023] Open
Abstract
Although combination BRAF/MEK inhibition has produced significant survival benefits for BRAF p.V600 mutant melanomas, targeted therapies approved for BRAF non-p.V600 mutant melanomas remain limited. Through the analysis of 772 cutaneous melanoma exomes, we reveal that BRAF non-p.V600 mutations co-occurs more frequently with NF1 loss, but not with oncogenic NRAS mutations, than expected by chance. We present cell signaling data, which demonstrate that BRAF non-p.V600 mutants can signal as monomers and dimers within an NF1 loss context. Concordantly, BRAF inhibitors that inhibit both monomeric and dimeric BRAF synergize with MEK inhibition to significantly reduce cell viability in vitro and tumor growth in vivo in BRAF non-p.V600 mutant melanomas with co-occurring NF1 loss-of-function mutations. Our data suggest that patients harboring BRAF non-p.V600 mutant melanomas may benefit from current FDA-approved BRAF/MEK inhibitor combination therapy currently reserved for BRAF p.V600 mutant patients.
Collapse
Affiliation(s)
- Shivshankari Rajkumar
- Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Diana Berry
- Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Kayla A Heney
- Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Colton Strong
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - LeeAnn Ramsay
- Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada
| | - Mathieu Lajoie
- Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada
| | - Rached Alkallas
- Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Human Genetics, McGill University, Montréal, QC H3A 0C7, Canada
| | - Tan-Trieu Nguyen
- Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada
| | - Cameron Thomson
- University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | | | - Matthew Dankner
- Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Teresa Petrella
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
| | - April A N Rose
- Department of Oncology, McGill University, Montréal, QC H4A 3T2, Canada; Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, Montréal, QC H3T 1E2, Canada
| | - Peter M Siegel
- Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada; Department of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Ian R Watson
- Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada; Research Institute of the McGill University Health Centre, Montréal, QC H3H 2R9, Canada.
| |
Collapse
|
27
|
Rajput N, Soni F, Sahu AK, Jadav T, Sharma S, Sengupta P. Degradation kinetics and characterization of major degradants of binimetinib employing liquid chromatography-high resolution mass spectrometry. J Pharm Biomed Anal 2022; 215:114753. [DOI: 10.1016/j.jpba.2022.114753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/04/2022] [Accepted: 04/02/2022] [Indexed: 10/18/2022]
|
28
|
MEK inhibitors for pre-treated, NRAS-mutated metastatic melanoma: A multi-centre, retrospective study. Eur J Cancer 2022; 166:24-32. [PMID: 35272084 DOI: 10.1016/j.ejca.2022.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND MEK inhibitors (MEKi) have shown clinical efficacy for NRAS-mutated, metastasized melanoma in randomised controlled trials, yet their clinical use is currently restricted to advanced, pre-treated patients, which is a different situation compared to previous trials. Data on their efficacy in the current real-world use are scarce. METHODS In this retrospective, multi-centre study, we evaluated the clinical course of disease of patients treated with MEKi with at least one previous treatment line in five German cancer centres. RESULTS Thirty-three patients were included, 19 males (58%) and 14 females (42%), with a median age of 64 years. Ninety-one percent of patients were pre-treated with immune checkpoint inhibitors, 90% of patients had elevated serum lactate dehydrogenase (LDH) levels at treatment initiation, 33% suffered from cerebral metastases and 30% had an Eastern Cooperative Oncology Group performance status of 2 or higher. The response rate was 18.2%; the disease control rate was 48.5%. Median progression-free survival was 2.8 months (95% confidence interval (CI): 1.6-3.9 months), and median overall survival was 7.1 months (95% CI: 5.8-8.3 months). In subgroup analysis, clinical efficacy was similar also in patients with high LDH levels and cerebral metastases, and there was a better outcome in males and in patients treated with trametinib vs. other MEKi, which may be based on selection bias. Overall, the clinical efficacy was similar compared to previous clinical trials in earlier treatment lines. CONCLUSIONS MEKi fulfil the need for an in-between treatment to stabilise the course of disease in advanced NRAS-mutated melanoma, but expectations regarding ongoing tumour response should be tempered.
Collapse
|
29
|
Zimmer L, Livingstone E, Krackhardt A, Schultz ES, Göppner D, Assaf C, Trebing D, Stelter K, Windemuth-Kieselbach C, Ugurel S, Schadendorf D. Encorafenib, binimetinib plus pembrolizumab triplet therapy in patients with advanced BRAF V600 mutant melanoma: safety and tolerability results from the phase I IMMU-TARGET trial. Eur J Cancer 2021; 158:72-84. [PMID: 34655839 DOI: 10.1016/j.ejca.2021.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Combination of immune checkpoint inhibitors and mitogen-activated protein kinase (MAPK) pathway inhibitors (MAPKi) has been proposed to enhance the durability of anti-tumour responses induced by MAPKi. Here, we present phase I safety results from an open-label, phase I/II study of pembrolizumab (PEM), encorafenib (ENC) and binimetinib (BIN) triplet therapy in advanced, B-Raf proto-oncogene serine/threonine kinase (BRAF)V600-mutated melanoma (IMMU-TARGET, NCT02902042). METHODS The dose finding phase I part used a 3 + 3 design, starting with the approved doses of PEM (200 mg every three weeks), ENC (450 mg once daily [QD]) and BIN (45 mg twice daily [BID]) as dose level (DL) 0. Reduction of the ENC and BIN doses (300 mg QD and 30 mg BID at DL-1 and 200 mg QD and 30 mg BID at DL-2) was preplanned in case of ≥2 dose-limiting toxicities (DLTs). Primary objectives were to estimate the recommended phase II dose of the triplet combination, DLT and safety. As per the sponsor's decision, the study was terminated after the phase I part, as the clinical efficacy of the combination is currently being investigated in a pivotal, placebo-controlled (PEM mono), double-blinded phase III trial (STARBOARD,NCT04657991). RESULTS Fifteen patients were enrolled. DLTs of DL0 were creatine phosphokinase (CPK) elevation plus cytokine release syndrome (n = 1) and gamma glutamyl transferase (GGT) increase (n = 1). No DLT was observed in further 3 + 3 patients at DL-1. One (isolated GGT elevations) DLT of DL0 was questionable, as the patient had further episodes of isolated GGT elevations after treatment discontinuation. Hence, further 6 patients were enrolled at DL0: here, no DLT occurred. In total, 13 of 15 patients (87%) experienced a treatment-related adverse event (TRAE) and 8 patients (53%), a grade ≥III TRAE; there were no TRAE-related deaths. Increases in aspartate aminotransferases, GGT (6/15 patients) and CPK elevations (4/15) were the most common grade III-IV TRAE. In median, patients received triplet therapy for 24 weeks (interquartile range [IQR], 12-45). Of the 14 patients evaluable for efficacy, the overall response rate was 64% (95% confidence interval [CI], 35-87). At a median follow-up of 25 months (IQR, 9-28), progression-free survival at 12 months was 41% (95% CI, 13-68). CONCLUSIONS Triplet therapy with PEM, ENC and BIN as used in the study was feasible and safe and led to clinically meaningful disease control.
Collapse
Affiliation(s)
- Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen, Germany.
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen, Germany.
| | - Angela Krackhardt
- Technische Universität München, School of Medicine, Klinik und Poliklinik Für Innere Medizin III, Klinikum Rechts der Isar, Ismaningerstr. 22, Munich 81675, Germany; German Cancer Consortium (DKTK), Technische Universität München, Partner Site Munich, Germany.
| | - Erwin S Schultz
- Department of Dermatology, University Hospital of the Paracelsus Medical Private University, Nuremberg, Germany.
| | - Daniela Göppner
- Clinic for Dermatology and Allergology, Justus-Liebig-University, Gießen, Germany.
| | - Chalid Assaf
- Department of Dermatology, Helios-Klinikum Krefeld, Germany.
| | - Dietrich Trebing
- Department of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Brandenburg Medical School Theodor Fontane, Dessau, Germany.
| | - Kai Stelter
- Department of Biostatistics, Alcedis GmbH, Giessen, Germany.
| | | | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen, Germany.
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen, Germany.
| |
Collapse
|
30
|
Seervai RNH, Cho WC, Chu EY, Marques-Piubelli ML, Ledesma DA, Richards K, Heberton MM, Nelson KC, Nagarajan P, Torres-Cabala CA, Prieto VG, Curry JL. Diverse landscape of dermatologic toxicities from small-molecule inhibitor cancer therapy. J Cutan Pathol 2021; 49:61-81. [PMID: 34622477 DOI: 10.1111/cup.14145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/29/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Advances in molecular biology and genetics have contributed to breakthrough treatments directed at specific pathways associated with the development of cancer. Small-molecule inhibitors (Nibs) aimed at a variety of cellular pathways have been efficacious; however, they are associated with significant dermatologic toxicities. METHODS We conducted a comprehensive review of dermatologic toxicities associated with Nibs categorized into the following five groups: (a) mitogen-activated protein kinase; (b) growth factor/multi-tyrosine kinase; (c) cell division/DNA repair; (d) signaling associated with myeloproliferative neoplasms; and (e) other signaling pathways. Prospective phase I, II, or III clinical trials, retrospective literature reviews, systematic reviews/meta-analyses, and case reviews/reports were included for analysis. RESULTS Dermatologic toxicities reviewed were associated with every class of Nibs and ranged from mild to severe or life-threatening adverse skin reactions. Inflammatory reactions manifesting as maculopapular, papulopustular/acneiform, and eczematous lesions were frequent types of dermatologic toxicities seen with Nibs. Squamous cell carcinoma with keratoacanthoma-like features was associated with a subset of Nibs. Substantial overlap in dermatologic toxicities was found between Nibs. CONCLUSIONS Dermatologic toxicities from Nibs are diverse and may overlap between classes of Nibs. Recognition of the various types of toxicities from Nibs is critical for patient care in the era of "oncodermatology/dermatopathology."
Collapse
Affiliation(s)
- Riyad N H Seervai
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.,Department of Dermatology, Baylor College of Medicine, Houston, Texas, USA
| | - Woo Cheal Cho
- Department of Pathology, Section of Dermatopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Emily Y Chu
- Department of Dermatology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mario L Marques-Piubelli
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Debora A Ledesma
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kristen Richards
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Meghan M Heberton
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kelly C Nelson
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Priyadharsini Nagarajan
- Department of Pathology, Section of Dermatopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Carlos A Torres-Cabala
- Department of Pathology, Section of Dermatopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Victor G Prieto
- Department of Pathology, Section of Dermatopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jonathan L Curry
- Department of Pathology, Section of Dermatopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
31
|
Teo MYM, Fong JY, Lim WM, In LLA. Current Advances and Trends in KRAS Targeted Therapies for Colorectal Cancer. Mol Cancer Res 2021; 20:30-44. [PMID: 34462329 DOI: 10.1158/1541-7786.mcr-21-0248] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/25/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022]
Abstract
Kirsten Rat Sarcoma (KRAS) gene somatic point mutations is one of the most prominently mutated proto-oncogenes known to date, and accounts for approximately 60% of all colorectal cancer cases. One of the most exciting drug development areas against colorectal cancer is the targeting of undruggable kinases and kinase-substrate molecules, although whether and how they can be integrated with other therapies remains a question. Current clinical trial data have provided supporting evidence on the use of combination treatment involving MEK inhibitors and either one of the PI3K inhibitors for patients with metastatic colorectal cancer to avoid the development of resistance and provide effective therapeutic outcome rather than using a single agent alone. Many clinical trials are also ongoing to evaluate different combinations of these pathway inhibitors in combination with immunotherapy for patients with colorectal cancer whose current palliative treatment options are limited. Nevertheless, continued assessment of these targeted cancer therapies will eventually allow patients with colorectal cancer to be treated using a personalized medicine approach. In this review, the most recent scientific approaches and clinical trials targeting KRAS mutations directly or indirectly for the management of colorectal cancer are discussed.
Collapse
Affiliation(s)
- Michelle Yee Mun Teo
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Jung Yin Fong
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Wan Ming Lim
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia.
| |
Collapse
|
32
|
Selection of Oral Therapeutics in China for the Treatment of Colorectal Cancer. Curr Treat Options Oncol 2021; 22:55. [PMID: 34097129 DOI: 10.1007/s11864-021-00852-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 12/24/2022]
Abstract
OPINION STATEMENT Intravenous administration of fluoropyrimidine-based chemotherapy has been the backbone of treatment in colorectal cancer (CRC) for decades. The availability of oral capecitabine has improved the tolerability and simplified combination schedules. In addition to capecitabine, several other oral drugs have proven efficacy, particularly in palliative treatment lines. Clinical guidelines describe several available third-line treatment options for metastatic CRC (mCRC), but few insights are provided to guide the selection and sequence. In this review, we describe the available evidence and most recent data concerning oral drugs with proven efficacy in CRC, including antiangiogenetic tyrosine kinase inhibitors (VEGFR TKIs), inhibitors blocking EGFR/Raf/MEK/ERK signaling pathway and modified fluoropyrimidine, and share recommendations and insights on selecting third-line oral therapies for mCRC in China. In general, third-line treatment options for mCRC are mainly regorafenib, fruquintinib, and chemo/targeted therapy reintroduction, while FTD/TPI was rarely used in China probably due to poor accessibility. Fruquintinib is preferred in patients with poor performance status (PS), elder age, and severe organ dysfunction, compared to regorafenib. New drugs of clinical trials were more recommended for the patients with BRAF mutant tumor, and those with good previous treatment efficacy tended to be recommended for chemo/targeted therapy reintroduction. The management of mCRC is evolving, and it must be emphasized that the consideration and recommendations presented here reflect current treatment practices in China and thus might change according to new clinical data as well as the availability of new oral drugs.
Collapse
|
33
|
Cleary JM, Wang V, Heist RS, Kopetz ES, Mitchell EP, Zwiebel JA, Kapner KS, Chen HX, Li S, Gray RJ, McShane LM, Rubinstein LV, Patton DR, Meric-Bernstam F, Dillmon MS, Williams PM, Hamilton SR, Conley BA, Aguirre AJ, O'Dwyer PJ, Harris LN, Arteaga CL, Chen AP, Flaherty KT. Differential Outcomes in Codon 12/13 and Codon 61 NRAS-Mutated Cancers in the Phase II NCI-MATCH Trial of Binimetinib in Patients with NRAS-Mutated Tumors. Clin Cancer Res 2021; 27:2996-3004. [PMID: 33637626 PMCID: PMC8542423 DOI: 10.1158/1078-0432.ccr-21-0066] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/11/2021] [Accepted: 02/18/2021] [Indexed: 01/02/2023]
Abstract
PURPOSE Preclinical and clinical data suggest that downstream inhibition with an MEK inhibitor, such as binimetinib, might be efficacious for NRAS-mutated cancers. PATIENTS AND METHODS Patients enrolled in the NCI-MATCH trial master protocol underwent tumor biopsy and molecular profiling by targeted next-generation sequencing. Patients with NRAS-mutated tumors, except melanoma, were enrolled in subprotocol Z1A, a single-arm study evaluating binimetinib 45 mg twice daily. The primary endpoint was objective response rate (ORR). Secondary endpoints included progression-free survival (PFS) and overall survival (OS). A post hoc analysis examined the association of NRAS mutation type with outcome. RESULTS In total, 47 eligible patients with a refractory solid tumor harboring a codon 12, 13, or 61 NRAS mutation were treated. Observed toxicity was moderate, and 30% of patients discontinued treatment because of binimetinib-associated toxicity. The ORR was 2.1% (1/47 patients). A patient with malignant ameloblastoma harboring a codon 61 NRAS mutation achieved a durable partial response (PR). A patient with NRAS codon 61-mutated colorectal cancer had an unconfirmed PR, and two other patients with NRAS codon 61-mutated colorectal had stable disease for at least 12 months. In an exploratory analysis, patients with colorectal cancer bearing a NRAS codon 61 mutation (n = 8) had a significantly longer OS (P = 0.03) and PFS (P = 0.007) than those with codon 12 or 13 mutations (n = 16). CONCLUSIONS Single-agent binimetinib did not show promising efficacy in NRAS-mutated cancers. The observation of increased OS and PFS in patients with codon 61 NRAS-mutated colorectal cancer merits further investigation.
Collapse
Affiliation(s)
- James M Cleary
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | | | - Rebecca S Heist
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - E Scott Kopetz
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Edith P Mitchell
- Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - James A Zwiebel
- Investigational Drug Branch, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Kevin S Kapner
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Helen X Chen
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Shuli Li
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Lisa M McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Larry V Rubinstein
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - David R Patton
- Center for Biomedical Informatics and Information Technology, NCI, Bethesda, Maryland
| | - Funda Meric-Bernstam
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | | | - P Mickey Williams
- Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | - Barbara A Conley
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Andrew J Aguirre
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Lyndsay N Harris
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | | | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
34
|
Fung AS, Graham DM, Chen EX, Stockley TL, Zhang T, Le LW, Albaba H, Pisters KM, Bradbury PA, Trinkaus M, Chan M, Arif S, Zurawska U, Rothenstein J, Zawisza D, Effendi S, Gill S, Sawczak M, Law JH, Leighl NB. A phase I study of binimetinib (MEK 162), a MEK inhibitor, plus carboplatin and pemetrexed chemotherapy in non-squamous non-small cell lung cancer. Lung Cancer 2021; 157:21-29. [PMID: 34052705 DOI: 10.1016/j.lungcan.2021.05.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION MEK inhibition is a potential therapeutic strategy in non-small cell lung cancer (NSCLC). This phase I study evaluates the MEK inhibitor binimetinib plus carboplatin and pemetrexed in stage IV non-squamous NSCLC patients (NCT02185690). METHODS A standard 3 + 3 dose-escalation design was used. Binimetinib 30 mg BID (dose level 1 [DL1]) or 45 mg BID (dose level 2 [DL2]) was given with standard doses of carboplatin and pemetrexed using an intermittent dosing schedule. The primary outcome was determination of the recommended phase II dose (RP2D) and safety of binimetinib. Secondary outcomes included efficacy, pharmacokinetics, and an exploratory analysis of response based on mutation subtype. RESULTS Thirteen patients (6 DL1, 7 DL2) were enrolled: 7 KRAS, 5 EGFR, and 1 NRAS mutation. The RP2D was binimetinib 30 mg BID. Eight patients (61.5%) had grade 3/4 adverse events, with dose limiting toxicities in 2 patients at DL2. Twelve patients were evaluated for response, with an investigator-assessed objective response rate (ORR) of 50% (95% CI 21.1%-78.9%; ORR 33.3% by independent-review, IR), and disease control rate 83.3% (95% CI 51.6%-97.9%). Median progression free survival (PFS) was 4.5 months (95% CI 2.6 months-NA), with a 6-month and 12-month PFS rate of 38.5% (95% CI 19.3%-76.5%) and 25.6% (95% CI 8.9%-73.6%), respectively. In an exploratory analysis, KRAS/NRAS-mutated patients had an ORR of 62.5% (ORR 37.5% by IR) vs. 25% in KRAS/NRAS wild-type patients. In MAP2K1-mutated patients, the ORR was 42.8%. CONCLUSION The addition of binimetinib to carboplatin and pemetrexed appears to have manageable toxicity with evidence of activity in advanced non-squamous NSCLC.
Collapse
Affiliation(s)
- A S Fung
- Department of Oncology, Queen's University, Canada; Princess Margaret Cancer Centre, University Health Network, Canada
| | - D M Graham
- Princess Margaret Cancer Centre, University Health Network, Canada; Division of Medical Oncology, University of Toronto, Canada; The Christie NHSFoundation Trust, Manchester, UK
| | - E X Chen
- Princess Margaret Cancer Centre, University Health Network, Canada; Division of Medical Oncology, University of Toronto, Canada
| | - T L Stockley
- Division of Clinical Laboratory Genetics, University Health Network, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada; Advanced Molecular Diagnostics Laboratory, University Health Network, Canada
| | - T Zhang
- Division of Clinical Laboratory Genetics, University Health Network, Canada; Advanced Molecular Diagnostics Laboratory, University Health Network, Canada
| | - L W Le
- Princess Margaret Cancer Centre, University Health Network, Canada
| | - H Albaba
- Princess Margaret Cancer Centre, University Health Network, Canada
| | - K M Pisters
- Princess Margaret Cancer Centre, University Health Network, Canada; MD Anderson Cancer Centre, Houston, TX, United States
| | - P A Bradbury
- Princess Margaret Cancer Centre, University Health Network, Canada; Division of Medical Oncology, University of Toronto, Canada
| | - M Trinkaus
- Division of Medical Oncology, University of Toronto, Canada; Markham Stouffville Hospital, Markham, Canada
| | - M Chan
- Division of Medical Oncology, University of Toronto, Canada; Trillium Health Partners, Mississauga, Canada
| | - S Arif
- Division of Medical Oncology, University of Toronto, Canada; Trillium Health Partners, Mississauga, Canada
| | - U Zurawska
- Division of Medical Oncology, University of Toronto, Canada; St. Joseph's Health Centre, Toronto, Canada
| | - J Rothenstein
- Division of Medical Oncology, University of Toronto, Canada; RS McLaughlin Durham Cancer Centre, Oshawa, Canada
| | - D Zawisza
- Princess Margaret Cancer Centre, University Health Network, Canada
| | - S Effendi
- Princess Margaret Cancer Centre, University Health Network, Canada
| | - S Gill
- Princess Margaret Cancer Centre, University Health Network, Canada
| | - M Sawczak
- Princess Margaret Cancer Centre, University Health Network, Canada
| | - J H Law
- Princess Margaret Cancer Centre, University Health Network, Canada
| | - N B Leighl
- Princess Margaret Cancer Centre, University Health Network, Canada; Division of Medical Oncology, University of Toronto, Canada.
| |
Collapse
|
35
|
Trametinib-associated Hyponatremia in a Child With Low-grade Glioma is Not Seen Following Treatment With Alternative MEK Inhibitor. J Pediatr Hematol Oncol 2021; 43:e550-e553. [PMID: 32520842 DOI: 10.1097/mph.0000000000001859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/05/2020] [Indexed: 01/22/2023]
Abstract
Molecularly targeted therapy with MEK inhibitors has been increasingly incorporated into the treatment of pediatric low-grade gliomas, but this promising therapy is associated with distinctive and specific toxicities. Understanding life-threatening MEK inhibitor toxicities and their management is critical to MEK inhibitor safety, especially among young children. This report describes severe hyponatremia associated with trametinib in an infant with progressive low-grade glioma without underlying endocrine dysfunction, which recurred despite significant dose reduction. Therapy with an alternative MEK inhibitor, binimetinib, provided excellent tumor response without hyponatremia, suggesting that some toxicities may be avoided by changing MEK inhibitor agents within the same class.
Collapse
|
36
|
LoRusso PM, Sekulic A, Sosman JA, Liang WS, Carpten J, Craig DW, Solit DB, Bryce AH, Kiefer JA, Aldrich J, Nasser S, Halperin R, Byron SA, Pilat MJ, Boerner SA, Durecki D, Hendricks WPD, Enriquez D, Izatt T, Keats J, Legendre C, Markovic SN, Weise A, Naveed F, Schmidt J, Basu GD, Sekar S, Adkins J, Tassone E, Sivaprakasam K, Zismann V, Calvert VS, Petricoin EF, Fecher LA, Lao C, Eder JP, Vogelzang NJ, Perlmutter J, Gorman M, Manica B, Fox L, Schork N, Zelterman D, DeVeaux M, Joseph RW, Cowey CL, Trent JM. Identifying treatment options for BRAFV600 wild-type metastatic melanoma: A SU2C/MRA genomics-enabled clinical trial. PLoS One 2021; 16:e0248097. [PMID: 33826614 PMCID: PMC8026051 DOI: 10.1371/journal.pone.0248097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/09/2021] [Indexed: 12/15/2022] Open
Abstract
Although combination BRAF and MEK inhibitors are highly effective for the 40-50% of cutaneous metastatic melanomas harboring BRAFV600 mutations, targeted agents have been ineffective for BRAFV600wild-type (wt) metastatic melanomas. The SU2C Genomics-Enabled Medicine for Melanoma Trial utilized a Simon two-stage optimal design to assess whether comprehensive genomic profiling improves selection of molecular-based therapies for BRAFV600wt metastatic melanoma patients who had progressed on standard-of-care therapy, which may include immunotherapy. Of the response-evaluable patients, binimetinib was selected for 20 patients randomized to the genomics-enabled arm, and nine were treated on the alternate treatment arm. Response rates for 27 patients treated with targeted recommendations included one (4%) partial response, 18 (67%) with stable disease, and eight (30%) with progressive disease. Post-trial genomic and protein pathway activation mapping identified additional drug classes that may be considered for future studies. Our results highlight the complexity and heterogeneity of metastatic melanomas, as well as how the lack of response in this trial may be associated with limitations including monotherapy drug selection and the dearth of available single and combination molecularly-driven therapies to treat BRAFV600wt metastatic melanomas.
Collapse
Affiliation(s)
- Patricia M. LoRusso
- Yale Cancer Center, Yale University, New Haven, CT, United States of America
| | - Aleksandar Sekulic
- Mayo Clinic, Scottsdale, AZ, United States of America
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Jeffrey A. Sosman
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL, United States of America
| | - Winnie S. Liang
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - John Carpten
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - David W. Craig
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - David B. Solit
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Alan H. Bryce
- Mayo Clinic, Scottsdale, AZ, United States of America
| | - Jeffrey A. Kiefer
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Jessica Aldrich
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Sara Nasser
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Rebecca Halperin
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Sara A. Byron
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Mary Jo Pilat
- Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, United States of America
| | - Scott A. Boerner
- Yale Cancer Center, Yale University, New Haven, CT, United States of America
| | - Diane Durecki
- Yale Cancer Center, Yale University, New Haven, CT, United States of America
| | | | - Daniel Enriquez
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Tyler Izatt
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Jonathan Keats
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Christophe Legendre
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | | | - Amy Weise
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States of America
| | - Fatima Naveed
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | | | - Gargi D. Basu
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Shobana Sekar
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Jonathan Adkins
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Erica Tassone
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | | | - Victoria Zismann
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Valerie S. Calvert
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, United States of America
| | - Emanuel F. Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, United States of America
| | - Leslie Anne Fecher
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, United States of America
| | - Christopher Lao
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, United States of America
| | - J. Paul Eder
- Yale Cancer Center, Yale University, New Haven, CT, United States of America
| | | | | | | | - Barbara Manica
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States of America
| | - Lisa Fox
- Yale Cancer Center, Yale University, New Haven, CT, United States of America
| | - Nicholas Schork
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Daniel Zelterman
- Yale Cancer Center, Yale University, New Haven, CT, United States of America
| | - Michelle DeVeaux
- Yale Cancer Center, Yale University, New Haven, CT, United States of America
- Regeneron Pharmaceuticals, Tarrytown, NY, United States of America
| | | | - C. Lance Cowey
- Charles A. Sammons Cancer Center/Baylor University Medical Center, Dallas, TX, United States of America
| | - Jeffrey M. Trent
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| |
Collapse
|
37
|
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
|
38
|
Forschner A, Sinnberg T, Mroz G, Schroeder C, Reinert CP, Gatidis S, Bitzer M, Eigentler T, Garbe C, Niessner H, Röcken M, Roggia C, Armeanu-Ebinger S, Riess O, Mattern S, Nann D, Bonzheim I. Case Report: Combined CDK4/6 and MEK Inhibition in Refractory CDKN2A and NRAS Mutant Melanoma. Front Oncol 2021; 11:643156. [PMID: 33732653 PMCID: PMC7959243 DOI: 10.3389/fonc.2021.643156] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/25/2021] [Indexed: 01/14/2023] Open
Abstract
There are only limited treatment options for metastatic NRAS mutant melanoma patients with resistance to immune checkpoint inhibitors. Besides activation of the mitogen-activated protein (MAP) kinase pathway, they often have additional disturbances in cell cycle regulation. However, unlike BRAF mutant melanoma, no targeted therapy has yet been approved for NRAS mutant melanoma so far. Here we present a NRAS mutant melanoma patient with response to combined binimetinib and ribociclib therapy following characterization of the molecular defects of the tumor by panel sequencing. Next generation sequencing (708 cancer genes) of a soft tissue metastasis revealed a homozygous deletion of CDKN2A in addition to the previously known NRAS mutation, as well as amplification of CCNE1 and CDK6. Immunohistochemical staining of the altered cell cycle genes confirmed loss of p16, reduced expression of p21 and high expression of CDK6 and cyclin D1. As the patient had been progressive on combined immunotherapy, targeted therapy with combined MEK and CDK4/6 inhibition was initiated as recommended by the molecular tumor board. Response to treatment was monitored with PET/CT and liquid biopsy, serum LDH, and S100. In addition, a patient-derived xenograft (PDX) was used to prove the efficacy of the two drugs in combination. Furthermore, senescence-associated beta-galactosidase staining showed that more cells were senescent under the combination treatment of binimetinib and ribociclib. Our case demonstrates how an individualized, molecular-based therapeutic approach could be found based on next-generation sequencing results. Furthermore our report highlights the fruitful and efficient collaboration of dermatooncologists, human geneticists, molecular pathologists, biochemists, radiologists, and nuclear physicians. Further studies are urgently needed to expand the very limited therapeutic landscape of NRAS mutated melanoma.
Collapse
Affiliation(s)
- Andrea Forschner
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Tobias Sinnberg
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany.,iFIT Cluster of Excellence (EXC 2180), University of Tübingen, Tübingen, Germany
| | - Gabi Mroz
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Christopher Schroeder
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Christian Philipp Reinert
- Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Sergios Gatidis
- Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Michael Bitzer
- Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Thomas Eigentler
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Heike Niessner
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Martin Röcken
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany.,iFIT Cluster of Excellence (EXC 2180), University of Tübingen, Tübingen, Germany
| | - Cristiana Roggia
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Sorin Armeanu-Ebinger
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Olaf Riess
- iFIT Cluster of Excellence (EXC 2180), University of Tübingen, Tübingen, Germany.,German Deutsche Forschungsgemeinschaft (DFG) Next Generation Sequencing (NGS) Competence Center, NGS Competence Center Tübingen (NCCT), Tübingen, Germany
| | - Sven Mattern
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Dominik Nann
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Irina Bonzheim
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
39
|
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: 23] [Impact Index Per Article: 7.7] [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
|
40
|
Yang W, Sun Y. Promising Molecular Targets for the Targeted Therapy of Biliary Tract Cancers: An Overview. Onco Targets Ther 2021; 14:1341-1366. [PMID: 33658799 PMCID: PMC7920611 DOI: 10.2147/ott.s297643] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Biliary tract cancer (BTC) is a leading cause of cancer-related death, due to the limited benefits of current systematic therapies and the heterogeneity of the tumor itself. High heterogeneity means that the clinical and molecular features vary between different subtypes of BTC, while the underlying molecular mechanisms remain unclear. Targeted therapy, where inhibitors are developed to selectively combine with targeted molecules in order to block abnormal signaling pathways in BTC, has shown promise as an emerging form of treatment for various types of cancer. In this article, a comprehensive review is conducted to examine potential molecular targets for BTC targeted therapy and their mechanisms. Furthermore, preliminary data published from clinical trials is utilized to analyze the main drugs used to combat BTC. The collective information presented in this article has provided useful insights into the current understanding of BTC.
Collapse
Affiliation(s)
- Wenwei Yang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Yongkun Sun
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| |
Collapse
|
41
|
McClure E, Patel A, Carr MJ, Sun J, Zager JS. The combination of encorafenib and binimetinib for the treatment of patients with BRAF-mutated advanced, unresectable, or metastatic melanoma: an update. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1847639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Erin McClure
- University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Ayushi Patel
- University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Michael J. Carr
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - James Sun
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
- Department of Surgery, University Hospitals, Cleveland Medical Center, Cleveland, Ohio, USA
| | - Jonathan S. Zager
- University of South Florida Morsani College of Medicine, Tampa, Florida, USA
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
42
|
Ruffinelli JC, Santos Vivas C, Sanz-Pamplona R, Moreno V. New advances in the clinical management of RAS and BRAF mutant colorectal cancer patients. Expert Rev Gastroenterol Hepatol 2021; 15:65-79. [PMID: 32946312 DOI: 10.1080/17474124.2021.1826305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION In colorectal carcinogenesis, genetic alterations in RAS and BRAF oncogenes play an important role for cancer initiation and/or progression and represent a key focus in the search for targeted therapies. Despite many years of research and a great amount of studies, until very recently this pathway was considered extremely hard to downregulate to obtain a significant clinical impact in colorectal cancer patients. But better times are coming with the advent of new promising drugs and combinations strategies. AREAS COVERED In this review, we go over the biological characteristics of the MAPK pathway in colorectal tumors, while illustrating the clinical correlation of RAS and BRAF mutations, particularly its prognostic and predictive value. We also present newly data about recent improvements in the treatment strategy for patients harboring these types of tumors. EXPERT COMMENTARY With great advances in the knowledge of molecular basis of RAS and BRAF mutant colorectal cancer in conjunction with biotechnology development and the constant effort for improvement, in the near future many new therapeutic options would be available for the management of this group of patient with dismal prognosis.
Collapse
Affiliation(s)
- Jose Carlos Ruffinelli
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet De Llobregat , Barcelona, Spain.,Colorectal Cancer Group, ONCOBELL Program, Institut De Recerca Biomedica De Bellvitge (IDIBELL) , Barcelona, Spain
| | - Cristina Santos Vivas
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet De Llobregat , Barcelona, Spain.,Colorectal Cancer Group, ONCOBELL Program, Institut De Recerca Biomedica De Bellvitge (IDIBELL) , Barcelona, Spain.,Consortium for Biomedical Research in Oncology (CIBERONC) , Barcelona, Spain.,Department of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona , Barcelona, Spain
| | - Rebeca Sanz-Pamplona
- Colorectal Cancer Group, ONCOBELL Program, Institut De Recerca Biomedica De Bellvitge (IDIBELL) , Barcelona, Spain.,Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP, Catalan Institute of Oncology (ICO), L'Hospitalet De Llobregat , Barcelona, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP) , Barcelona, Spain
| | - Victor Moreno
- Colorectal Cancer Group, ONCOBELL Program, Institut De Recerca Biomedica De Bellvitge (IDIBELL) , Barcelona, Spain.,Department of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona , Barcelona, Spain.,Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP, Catalan Institute of Oncology (ICO), L'Hospitalet De Llobregat , Barcelona, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP) , Barcelona, Spain
| |
Collapse
|
43
|
Ramanathan RK, Von Hoff DD, Eskens F, Blumenschein G, Richards D, Genvresse I, Reschke S, Granvil C, Skubala A, Peña C, Mross K. Phase Ib Trial of the PI3K Inhibitor Copanlisib Combined with the Allosteric MEK Inhibitor Refametinib in Patients with Advanced Cancer. Target Oncol 2020; 15:163-174. [PMID: 32314268 PMCID: PMC7591420 DOI: 10.1007/s11523-020-00714-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Dual inhibition of PI3K and MAPK signaling is conceptually a promising anticancer therapy. Objective This phase 1b trial investigated the safety, maximum tolerated dose (MTD), recommended phase II dose, pharmacokinetics, tumor response, fluorodeoxyglucose positron emission tomography (FDG-PET) pharmacodynamics, and biomarker explorations for the combination of pan-PI3K inhibitor copanlisib and allosteric MEK inhibitor refametinib in patients with advanced solid tumors. Patients and methods This was an adaptive trial with eight dose cohorts combining dose escalation and varying schedules in repeated 28-day cycles. Patients received copanlisib (0.2–0.8 mg/kg intravenously) intermittently (days 1, 8, 15) or weekly (days 1, 8, 15, 22) each cycle, and refametinib (30–50 mg twice daily orally) continuously or 4 days on/3 days off. Patients with KRAS, NRAS, BRAF, or PI3KCA mutations were eligible for the expansion cohort. Results In the dose-escalation (n = 49) and expansion (n = 15) cohorts, the most common treatment-emergent adverse events included diarrhea (59.4%), nausea, acneiform rash, and fatigue (51.6% each). Dose-limiting toxicities included oral mucositis (n = 4), increased alanine aminotransferase/aspartate aminotransferase (n = 3), acneiform rash, hypertension (n = 2 each), and diarrhea (n = 1). MTD was copanlisib 0.4 mg/kg weekly and refametinib 30 mg twice daily. No pharmacokinetic interactions were identified. Decreased tumor FDG uptake and MEK-ERK signaling inhibition were demonstrated during treatment. Best response was stable disease (n = 21); median treatment duration was 6 weeks. Conclusions Despite sound rationale and demonstrable pharmacodynamic tumor activity in relevant tumor populations, a dose and schedule could not be identified for this drug combination that were both tolerable and offered clear efficacy in the population assessed. Clinicaltrials.gov identifier NCT01392521. Electronic supplementary material The online version of this article (10.1007/s11523-020-00714-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Ferry Eskens
- Erasmus MC Cancer Institute, PO Box 2040, 3015 GD, Rotterdam, The Netherlands
| | - George Blumenschein
- The University of Texas MD Anderson Cancer Center, Unit 432, PO Box 301402, Houston, TX, 77030, USA
| | - Donald Richards
- US Oncology Research, Texas Oncology, 910 E. Houston St., Suite 100, Tyler, TX, 75702, USA
| | - Isabelle Genvresse
- Pharmaceutical Division, Bayer AG, Müllerstraße 178, 13353, Berlin, Germany
| | - Susanne Reschke
- Pharmaceutical Division, Bayer AG, Müllerstraße 178, 13353, Berlin, Germany
| | - Camille Granvil
- Bayer HealthCare Pharmaceuticals, Inc., 100 Bayer Blvd, Whippany, NJ, 07981, USA
| | - Adam Skubala
- Chrestos Concept GmbH & Co. KG, Girardetstr. 1-5, 45131, Essen, Germany
| | - Carol Peña
- Bayer HealthCare Pharmaceuticals, Inc., 100 Bayer Blvd, Whippany, NJ, 07981, USA
| | - Klaus Mross
- KTB Klinik für Tumorbiologie, Breisacher Str. 117, 79106, Freiburg im Breisgau, Baden-Württemberg, Germany
| |
Collapse
|
44
|
Cimino SK, Eng C. Up-and-Coming Experimental Drug Options for Metastatic Colorectal Cancer. J Exp Pharmacol 2020; 12:475-485. [PMID: 33204182 PMCID: PMC7667584 DOI: 10.2147/jep.s259287] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer is one of the top causes of cancer and cancer-related deaths worldwide. The prognosis of metastatic colorectal cancer is poor and treatment options are limited. Many patients will run out of treatment options before they become medically unfit for therapy. As such, there is a need to expand upon the current understanding of disease biology as well as drug resistance mechanisms in order to create new approaches for therapy. In this review article, we will discuss the mechanistic rationale and clinical data for new drugs and therapeutic combinations under development for metastatic colorectal cancer.
Collapse
Affiliation(s)
- Sarah K Cimino
- Department of Pharmacy, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cathy Eng
- Department of Medicine: Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
45
|
Delord JP, Italiano A, Awada A, Aftimos P, Houédé N, Lebbé C, Pages C, Lesimple T, Dinulescu M, Schellens JHM, Leijen S, Rottey S, Kruse V, Kefford R, Faivre S, Gomez-Roca C, Scheuler A, Massimini G, Raymond E. Selective Oral MEK1/2 Inhibitor Pimasertib: A Phase I Trial in Patients with Advanced Solid Tumors. Target Oncol 2020; 16:37-46. [PMID: 33170484 DOI: 10.1007/s11523-020-00768-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The Ras/Raf/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (Ras/Raf/MEK/ERK) signaling cascade is frequently constitutively activated in human cancers. Pimasertib is a selective and potent adenosine triphosphate non-competitive MEK1/2 inhibitor. OBJECTIVE Our objectives were to describe the results of a phase I, first-in-human, dose-escalation trial of pimasertib that investigated the maximum tolerated dose, recommended phase II dose, and safety, as well as other endpoints. PATIENTS AND METHODS Four dosing schedules of pimasertib (once daily [qd], 5 days on, 2 days off; qd, 15 days on, 6 days off; continuous qd; continuous twice daily [bid]) were evaluated in patients with advanced solid tumors. Each treatment cycle lasted 21 days. The primary objective was to determine the maximum tolerated dose based on dose-limiting toxicities (DLTs) evaluated during cycle 1, and the recommended phase II dose (RP2D). Secondary objectives included safety, pharmacokinetics, pharmacodynamics, and antitumor activity. RESULTS Overall, 180 patients received pimasertib (dose range 1-255 mg/day). DLTs were mainly observed at doses ≥ 120 mg/day and included skin rash/acneiform dermatitis and ocular events, such as serous retinal detachment. The most common drug-related adverse events were consistent with class effects, including diarrhea, skin disorders, ocular disorders, asthenia/fatigue, and peripheral edema. The median time to maximum pimasertib concentration was 1.5 h across dosing schedules, and the apparent terminal half-life was 5 h across qd dosing schedules. Pimasertib decreased ERK phosphorylation within 2 h of administration, which was maintained for up to 8 h at higher doses and prolonged with bid dosing. CONCLUSIONS Based on the safety profile and efficacy signals, a continuous bid regimen was the preferred dosing schedule and the RP2D was defined as 60 mg bid. TRIAL REGISTRATION ClinicalTrials.gov, NCT00982865.
Collapse
Affiliation(s)
- Jean-Pierre Delord
- Clinical Research Unit, Institut Universitaire du Cancer, Oncopole, Toulouse, France.
| | - Antoine Italiano
- Early Phase Trials and Sarcoma Units, Institut Bergonie, Bordeaux, France.,University of Bordeaux, Bordeaux, France
| | - Ahmad Awada
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Philippe Aftimos
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Nadine Houédé
- Medical Oncology, Institut de Cancérologie du Gard, CHU Caremeau, Nîmes, France
| | - Céleste Lebbé
- APHP Oncodermatology Unit, INSERM U976, CIC Hôpital Saint Louis University Paris Diderot, Paris, France
| | - Celine Pages
- APHP Oncodermatology Unit, INSERM U976, CIC Hôpital Saint Louis University Paris Diderot, Paris, France
| | - Thierry Lesimple
- Medical Oncology Department, Comprehensive Cancer Center Eugène Marquis, Rennes, France
| | - Monica Dinulescu
- Dermatology Department, Rennes University Hospital, Rennes, France
| | - Jan H M Schellens
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht, The Netherlands
| | - Suzanne Leijen
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sylvie Rottey
- Department of Medical Oncology, Ghent University Hospital and Heymans Institute of Pharmacology, Ghent University, Gent, Belgium
| | - Vibeke Kruse
- Department of Medical Oncology, Ghent University Hospital and Heymans Institute of Pharmacology, Ghent University, Gent, Belgium
| | - Richard Kefford
- Faculty of Medicine and Health Sciences, Crown Princess Mary Cancer Centre Westmead Hospital, Macquarie University, and Melanoma Institute Australia, Sydney, NSW, Australia
| | - Sandrine Faivre
- Medical Oncology, Beaujon University Hospital, Clichy, France
| | - Carlos Gomez-Roca
- Clinical Research Unit, Institut Universitaire du Cancer, Oncopole, Toulouse, France
| | - Armin Scheuler
- Global Biostatistics and Epidemiology, EMD Serono Research and Development Institute, Inc. (an affiliate of Merck KGaA, Darmstadt, Germany), Billerica, MA, USA
| | - Giorgio Massimini
- Early Clinical Oncology Global Clinical Development Biopharma, Merck KGaA, Darmstadt, Germany
| | - Eric Raymond
- Paris Diderot University Hospital, Clichy, France
| |
Collapse
|
46
|
Osumi H, Muroi A, Sakahara M, Kawachi H, Okamoto T, Natsume Y, Yamanaka H, Takano H, Kusama D, Shinozaki E, Ooki A, Yamaguchi K, Ueno M, Takeuchi K, Noda T, Nagayama S, Koshikawa N, Yao R. Evaluation of the RAS signaling network in response to MEK inhibition using organoids derived from a familial adenomatous polyposis patient. Sci Rep 2020; 10:17455. [PMID: 33060766 PMCID: PMC7567075 DOI: 10.1038/s41598-020-74530-x] [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] [Received: 05/19/2020] [Accepted: 09/30/2020] [Indexed: 01/07/2023] Open
Abstract
RAS signaling is a promising target for colorectal cancer (CRC) therapy, and a variety of selective inhibitors have been developed. However, their use has often failed to demonstrate a significant benefit in CRC patients. Here, we used patient-derived organoids (PDOs) derived from a familial adenomatous polyposis (FAP) patient to analyze the response to chemotherapeutic agents targeting EGFR, BRAF and MEK. We found that PDOs carrying KRAS mutations were resistant to MEK inhibition, while those harboring the BRAF class 3 mutation were hypersensitive. We used a systematic approach to examine the phosphorylation of RAS effectors using reverse-phase protein array (RPPA) and found increased phosphorylation of MEK induced by binimetinib. A high basal level of ERK phosphorylation and its rebound activation after MEK inhibition were detected in KRAS-mutant PDOs. Notably, the phosphorylation of EGFR and AKT was more closely correlated with that of MEK than that of ERK. Transcriptome analysis identified MYC-mediated transcription and IFN signaling as significantly correlated gene sets in MEK inhibition. Our experiments demonstrated that RPPA analysis of PDOs, in combination with the genome and transcriptome, is a useful preclinical research platform to understand RAS signaling and provides clues for the development of chemotherapeutic strategies.
Collapse
Affiliation(s)
- Hiroki Osumi
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.,Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.,Director's Office, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Atsushi Muroi
- Division of Cancer Cell Research, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Yokohama, Kanagawa, 241-8515, Japan
| | - Mizuho Sakahara
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Hiroshi Kawachi
- Division of Pathology, Cancer Institute Hospital, Department of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Takuya Okamoto
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Yasuko Natsume
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Hitomi Yamanaka
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Hiroshi Takano
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Daisuke Kusama
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Eiji Shinozaki
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Akira Ooki
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Masashi Ueno
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Kengo Takeuchi
- Division of Pathology, Cancer Institute Hospital, Department of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Tetsuo Noda
- Director's Office, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Satoshi Nagayama
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Naohiko Koshikawa
- Division of Cancer Cell Research, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Yokohama, Kanagawa, 241-8515, Japan
| | - Ryoji Yao
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| |
Collapse
|
47
|
Abstract
PURPOSE Mitogen-activates protein kinase (MAPK) inhibitors, particularly MEK inhibitors, have shifted the treatment paradigm for metastatic BRAF-mutant cutaneous melanoma; however, oncologists, ophthalmologists, and patients have noticed different toxicities of variable importance. This review aims to provide an update of the ocular adverse events (OAEs), especially retinal toxicity, associated with the use of MEK inhibitors. METHODS We conducted a scientific literature search using the PubMed database up to July 2018 with the terms "MEK inhibitors" with a "review" filter and "MEK inhibitors" with a "clinical trials" filter. Phase I-III experimental studies and reviews were selected. Current principles and techniques for diagnosing and managing MEK inhibitor retinopathy and other OAEs are discussed. RESULTS In patients treated with MEK inhibitors, including asymptomatic patients, OAEs occur with an incidence of up to 90%. Mild to severe ophthalmic toxicities are described, including visual disturbances, a 2-line decrease in Snellen visual acuity, dry eye symptoms, ocular adnexal abnormalities, visual field defects, panuveitis, and retinal toxicities, such as different degrees of MEK-associated retinopathy, vascular injury, and retinal vein occlusion. CONCLUSION MEK inhibitors can lead to different degrees of retinal, uveal, and adnexal OAE, causing visual disturbances or discomfort. One of the most relevant OAE of MEK therapy is MEK inhibitor-associated retinopathy (MEKAR), which is usually mild, self-limited, and may subside after continuous use of the drug for weeks or months, or discontinuation, thereby restoring the normal visual function of the retina, with some exceptions. Ocular adverse events are often associated with other systemic adverse effects that can modify the dosage of treatment, so the communication with the oncologist is fundamental.
Collapse
|
48
|
Huijberts SC, van Geel RM, Bernards R, Beijnen JH, Steeghs N. Encorafenib, binimetinib and cetuximab combined therapy for patients with BRAFV600E mutant metastatic colorectal cancer. Future Oncol 2020; 16:161-173. [PMID: 32027186 DOI: 10.2217/fon-2019-0748] [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: 01/10/2023] Open
Abstract
Approximately 10-15% of colorectal cancers (CRCs) harbor an activating BRAF mutation, leading to tumor growth promotion by activation of the mitogen-activated protein kinases pathway. BRAFV600E mutations are prognostic for treatment failure after first-line systemic therapy in the metastatic setting. In contrast to the efficacy of combined BRAF and MEK inhibition in melanoma, BRAFV600E mutant CRC is intrinsically unresponsive due to upregulation of HER/EGFR. However, combining the EGFR inhibitor cetuximab, the BRAF inhibitor encorafenib and the MEK inhibitor binimetinib improves overall survival. This review discusses the current treatment field for patients with BRAFV600E mutant metastatic CRC and summarizes the pharmacology, efficacy and safety of the novel doublet and triplet therapies consisting of encorafenib and cetuximab with or without binimetinib.
Collapse
Affiliation(s)
- Sanne Cfa Huijberts
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, 1066 CX, The Netherlands
| | - Robin Mjm van Geel
- Department of Clinical Pharmacy & Toxicology, Maastricht University Medical Centre, Maastricht, 6229 HX, The Netherlands.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Rene Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, 1066 CX, The Netherlands.,Utrecht University, Utrecht, 3508 TC, The Netherlands
| | - Jos H Beijnen
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, 1066 CX, The Netherlands.,Utrecht University, Utrecht, 3508 TC, The Netherlands.,Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, 1066 CX, The Netherlands
| | - Neeltje Steeghs
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, 1066 CX, The Netherlands.,Division of Medical Oncology, The Netherlands Cancer institute, Amsterdam, 1066 CX, The Netherlands
| |
Collapse
|
49
|
Xie C, McGrath NA, Monge Bonilla C, Fu J. Systemic treatment options for advanced biliary tract carcinoma. J Gastroenterol 2020; 55:944-957. [PMID: 32748173 PMCID: PMC7519922 DOI: 10.1007/s00535-020-01712-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/25/2020] [Indexed: 02/04/2023]
Abstract
Advanced biliary tract cancers (BTC) include a diverse collection of rare and heterogenous tumors with poor prognosis. The combination of gemcitabine and cisplatin is the established first-line therapy for advanced BTC. There are no accepted standard treatments in the second line setting, though there are several ongoing clinical trials that implement chemotherapy as a therapeutic strategy. The understanding of the molecular landscape of BTC has offered hope of targeted therapies to the identified actionable genomic aberrations, such as FGFR2 gene fusions, mutations of IDH1/2, HER2, BRAC1/2 and BRAF. Pembigatinib has become the first approved targeted therapy for BTC with FGFR2 fusion or other rearrangements. Recent immunotherapy has opened new therapy avenues in BTC with pembrolizumab approved for either microsatellite instability high (MSI-H) or DNA mismatch repair deficient (dMMR) advanced solid tumors, including BTC. The combination of immunotherapy with other modalities is currently being evaluated in different clinical trials, since single agent immunotherapy appears to provide modest benefits in advanced BTC. In this review, we summarize the current status of treatment options, including systemic chemotherapy, targeted therapy, immunotherapy, and various combinations in advanced BTC.
Collapse
Affiliation(s)
- Changqing Xie
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Nicole A McGrath
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cecilia Monge Bonilla
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jianyang Fu
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
50
|
ERK phosphorylation as a marker of RAS activity and its prognostic value in non-small cell lung cancer. Lung Cancer 2020; 149:10-16. [PMID: 32947221 DOI: 10.1016/j.lungcan.2020.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/26/2020] [Accepted: 09/01/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Deregulated signal transduction pathways play a key role in development, progression and therapeutic resistance of non-small cell lung cancers (NSCLC). The purpose of this study is to assess the downstream markers of two well-characterized pathways and to correlate them with clinical outcome. DESIGN 670 patients with metastatic NSCLC were prospectively enrolled in a comprehensive biomarker profiling program at a single center from 2012 to 2016. Phosphorylation of extracellular signal-regulated kinase (p-ERK), and protein kinase B (p-AKT) was assessed by standardized immunohistochemistry. Product of scores for quantity and quality of staining were calculated (immunoreactive score, 0-9). Somatic mutations of Kirsten rat sarcoma viral oncogene homolog [KRAS], epithelial growth factor receptor [EGFR], v-Raf murine sarcoma viral oncogene homolog B [BRAF] and phosphatidylinositol 3-kinase [PIK3CA]) were detected by Sanger (2012-03/2015) and amplicon NGS (04/2015-02/2016). Patients enrolled during the first year (2012) were used as discovery cohort. Patients enrolled from 2013 to 02/2016 were used as validation cohort. Clinical data were retrieved from the electronic medical records and were analyzed retrospectively. RESULTS Using a discovery cohort, we identified an immunoreactive score of p-ERK ≥3 to be prognostically relevant. The validation cohort confirmed that higher levels of p-ERK correlated with worse overall survival (OS) and higher proportion of RAS mutations. Multivariate analysis including established risk factors such EGFR, ALK or ROS mutations and metastatic disease showed a trend of a detrimental effect of high p-ERK on OS (HR 1.23, CI 0.94-1.59, p = 0.131 for p-ERK immunoreactive score ≥3) and time to treatment failure after first-line therapy in the validation cohort. Phosphorylated AKT did not correlate with clinical outcome. CONCLUSION While serving as a prognosticator in univariate analysis, highly phosphorylated ERK does not convey a significant prognostic effect for OS in the presence of other prognostic factors. Phosphorylated ERK indicates a higher activity of RAS in advanced NSCLC.
Collapse
|