1
|
Timofeev O, Giron P, Lawo S, Pichler M, Noeparast M. ERK pathway agonism for cancer therapy: evidence, insights, and a target discovery framework. NPJ Precis Oncol 2024; 8:70. [PMID: 38485987 PMCID: PMC10940698 DOI: 10.1038/s41698-024-00554-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/16/2024] [Indexed: 03/18/2024] Open
Abstract
At least 40% of human cancers are associated with aberrant ERK pathway activity (ERKp). Inhibitors targeting various effectors within the ERKp have been developed and explored for over two decades. Conversely, a substantial body of evidence suggests that both normal human cells and, notably to a greater extent, cancer cells exhibit susceptibility to hyperactivation of ERKp. However, this vulnerability of cancer cells remains relatively unexplored. In this review, we reexamine the evidence on the selective lethality of highly elevated ERKp activity in human cancer cells of varying backgrounds. We synthesize the insights proposed for harnessing this vulnerability of ERK-associated cancers for therapeutical approaches and contextualize these insights within established pharmacological cancer-targeting models. Moreover, we compile the intriguing preclinical findings of ERK pathway agonism in diverse cancer models. Lastly, we present a conceptual framework for target discovery regarding ERKp agonism, emphasizing the utilization of mutual exclusivity among oncogenes to develop novel targeted therapies for precision oncology.
Collapse
Affiliation(s)
- Oleg Timofeev
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35043, Marburg, Germany
| | - Philippe Giron
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research group Genetics, Reproduction and Development, Centre for Medical Genetics, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Steffen Lawo
- CRISPR Screening Core Facility, Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Martin Pichler
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany
| | - Maxim Noeparast
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany.
| |
Collapse
|
2
|
Dayimu A, Gupta A, Matin RN, Nobes J, Board R, Payne M, Rao A, Fusi A, Danson S, Eccles B, Carser J, Brown CO, Steven N, Bhattacharyya M, Brown E, Gonzalez M, Highley M, Pickering L, Kumar S, Waterston A, Burghel G, Demain L, Baker E, Wulff J, Qian W, Twelves S, Middleton M, Corrie P. A randomised phase 2 study of intermittent versus continuous dosing of dabrafenib plus trametinib in patients with BRAF V600 mutant advanced melanoma (INTERIM). Eur J Cancer 2024; 196:113455. [PMID: 38029480 DOI: 10.1016/j.ejca.2023.113455] [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: 09/05/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND BRAF+MEK inhibitors extend life expectancy of patients with BRAFV600 mutant advanced melanoma. Acquired resistance limits duration of benefit, but preclinical and case studies suggest intermittent dosing could overcome this limitation. INTERIM was a phase 2 trial evaluating an intermittent dosing regimen. METHODS Patients with BRAFV600 mutant advanced melanoma due to start dabrafenib+trametinib were randomised to receive either continuous (CONT), or intermittent (INT; dabrafenib d1-21, trametinib d1-14 every 28 days) dosing. A composite primary endpoint included progression-free survival (PFS) and quality of life (QoL). Secondary endpoints included response rate (ORR), overall survival (OS) and adverse events (AEs). Mutant BRAFV600E ctDNA was measured by droplet digital PCR (ddPCR), using mutant allele frequency of > 1 % as the detection threshold. RESULTS 79 patients (39 INT, 40 CONT) were recruited; median age 67 years, 65 % AJCC (7th ed) stage IV M1c, 29 % had brain metastases. With 19 months median follow-up, INT was inferior in all efficacy measures: median PFS 8.5 vs 10.7mo (HR 1.39, 95 %CI 0.79-2.45, p = 0.255); median OS 18.1mo vs not reached (HR 1.69, 95 %CI 0.87-3.28, p = 0.121), ORR 57 % vs 77 %. INT patients experienced fewer treatment-related AEs (76 % vs 88 %), but more grade > 3 AEs (53 % vs 42 %). QoL favoured CONT. Detection of BRAFV600E ctDNA prior to treatment correlated with worse OS (HR 2.55, 95 %CI 1.25-5.21, p = 0.01) in both arms. A change to undetected during treatment did not significantly predict better OS. CONCLUSION INTERIM findings are consistent with other recent clinical trials reporting that intermittent dosing does not improve efficacy of BRAF+MEK inhibitors.
Collapse
Affiliation(s)
- Alimu Dayimu
- Clinical Trials Unit, Department of Oncology, University of Cambridge, Cambridge, UK
| | | | - Rubeta N Matin
- Department of Dermatology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jenny Nobes
- Department of Oncology, Norfolk and Norwich University Hospital NHS Foundation Trust, Norfolk, UK
| | - Ruth Board
- Department of Oncology, Lancashire Teaching Hospitals NHS Trust, Preston, UK
| | - Miranda Payne
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - Ankit Rao
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Alberto Fusi
- Department of Medical Oncology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Sarah Danson
- Division of Clinical Medicine, The University of Sheffield, Sheffield, UK; Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - Bryony Eccles
- Department of Medical Oncology, University Hospitals Dorset NHS Foundation Trust, Poole, UK
| | - Judith Carser
- Department of Oncology, Belfast Health and Social Care Trust, Belfast, UK
| | | | - Neil Steven
- Department of Oncology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Ewan Brown
- Western General Hospital, Lothian NHS Board, Edinburgh, UK
| | - Michael Gonzalez
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Martin Highley
- Oncology Centre, Derriford Hospital, University Hospitals Plymouth NHS Trust, Plymouth, UK
| | - Lisa Pickering
- Skin and Renal Units, Royal Marsden NHS Foundation Trust, London, UK
| | - Satish Kumar
- Velindre Cancer Centre, Velindre University NHS Trust, Cardiff, UK
| | | | - George Burghel
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Leigh Demain
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Eleanor Baker
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jerome Wulff
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Wendi Qian
- Clinical Trials Unit, Department of Oncology, University of Cambridge, Cambridge, UK; Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sophie Twelves
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Mark Middleton
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK; Department of Oncology, University of Oxford, Oxford, UK
| | - Pippa Corrie
- Oncology Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| |
Collapse
|
3
|
Chahoud J, Anderson AR, Zhang J, Brown J, Gatenby RA. Evolutionary Dynamics and Intermittent Therapy for Metastatic Cancers. J Clin Oncol 2023; 41:4469-4471. [PMID: 37418680 PMCID: PMC10553063 DOI: 10.1200/jco.23.00647] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/09/2023] [Indexed: 07/09/2023] Open
Abstract
Mathematical models show that the induction period of intermittent cancer therapy drives cancer toward resistance
Collapse
Affiliation(s)
- Jad Chahoud
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| | | | - Jingsong Zhang
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Joel Brown
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Robert A. Gatenby
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
- Department of Radiology, Department of Integrated Mathematical Oncology H. Lee Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
4
|
Intermittent treatment of BRAF V600E melanoma cells delays resistance by adaptive resensitization to drug rechallenge. Proc Natl Acad Sci U S A 2022; 119:e2113535119. [PMID: 35290123 PMCID: PMC8944661 DOI: 10.1073/pnas.2113535119] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Preclinical studies of metastatic melanoma treated with targeted therapeutics have suggested that alternating periods of treatment and withdrawal might delay the onset of resistance. This has been attributed to drug addiction, where cells lose fitness upon drug removal due to the resulting hyperactivation of mitogen-activated protein (MAP) kinase signaling. This study presents evidence that the intermittent treatment response can also be explained by the resensitization of cells following drug removal and enhanced cell loss upon drug rechallenge. Resensitization is accompanied by adaptive transcriptomic switching and occurs despite the sustained expression of resistance genes throughout the intermittent treatment. Patients with melanoma receiving drugs targeting BRAFV600E and mitogen-activated protein (MAP) kinase kinases 1 and 2 (MEK1/2) invariably develop resistance and face continued progression. Based on preclinical studies, intermittent treatment involving alternating periods of drug withdrawal and rechallenge has been proposed as a method to delay the onset of resistance. The beneficial effect of intermittent treatment has been attributed to drug addiction, where drug withdrawal reduces the viability of resistant cells due to MAP kinase pathway hyperactivation. However, the mechanistic basis of the intermittent effect is incompletely understood. We show that intermittent treatment with the BRAFV600E inhibitor, LGX818/encorafenib, suppresses growth compared with continuous treatment in human melanoma cells engineered to express BRAFV600E, p61-BRAFV600E, or MEK2C125 oncogenes. Analysis of the BRAFV600E-overexpressing cells shows that, while drug addiction clearly occurs, it fails to account for the advantageous effect of intermittent treatment. Instead, growth suppression is best explained by resensitization during periods of drug removal, followed by cell death after drug readdition. Continuous treatment leads to transcriptional responses prominently associated with chemoresistance in melanoma. By contrast, cells treated intermittently reveal a subset of transcripts that reverse expression between successive cycles of drug removal and rechallenge and include mediators of cell invasiveness and the epithelial-to-mesenchymal transition. These transcripts change during periods of drug removal by adaptive switching, rather than selection pressure. Resensitization occurs against a background of sustained expression of melanoma resistance genes, producing a transcriptome distinct from that of the initial drug-naive cell state. We conclude that phenotypic plasticity leading to drug resensitization can underlie the beneficial effect of intermittent treatment.
Collapse
|
5
|
Bédouelle E, Nguyen JM, Varey E, Khammari A, Dreno B. Should Targeted Therapy Be Continued in BRAF-Mutant Melanoma Patients after Complete Remission? Dermatology 2021; 238:517-526. [PMID: 34818219 DOI: 10.1159/000518718] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/03/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Targeted therapy is used to treat patients with a BRAF-mutated metastatic melanoma and is continued until disease progression or severe toxicity. No robust data on the management of patients achieving a complete remission (CR) are available. MAIN OBJECTIVE To determine the relapse rate in the first year after targeted therapy discontinuation in patients in CR. SECONDARY OBJECTIVES To determine the relapse rates throughout the follow-up and to identify prognostic factors for relapse at 1 year. METHODS A retrospective, monocentric observational study was conducted in patients with advanced melanoma included in the RIC-Mel database who discontinued targeted therapy after achieving a CR confirmed by CT scan and PET/CT scan. RESULTS Twenty-nine patients were included. Seventeen (58.6%) patients were treated with BRAF inhibitor (BRAFi) alone and 12 (41.4%) with a BRAFi combined with a MEK inhibitor (BRAFi + MEKi). The median treatment duration was 9.7 months. The relapse rates after discontinuation were 69% at 12 months (BRAFi: 70.6%; BRAFi + MEKi: 66.7%) and 76% at 36 months (BRAFi: 76.5%; BRAFi + MEKi: 75%). A non-significant trend towards a higher risk of relapse was found in women (p = 0.1; RR 3.36; 95% CI 0.77-17.07), in patients with an LDH level greater than the upper limits of normal (p = 0.58; RR 2.43; 95% CI 0.10-56.71), and when more than two metastatic sites were involved (p = 0.19; RR 4.6; 95% CI 0.46-46.51). After relapse, targeted therapy was resumed in 17 patients (7 with BRAFi; 10 with BRAFi + MEKi) with a response rate of 53%. CONCLUSIONS This real-life study provided long-term data in patients who discontinued targeted therapy after CR. Most patients experienced a relapse in the first year after targeted therapy discontinuation, of whom 50% were in the first 3 months. After targeted therapy resumption, 53% of relapsing patients achieved an objective response. Patients should be followed during the first year after treatment discontinuation. In addition, patients with less than 3 metastatic sites, a baseline LDH level with normal ranges, men, and patients responding rapidly to treatment would be more likely to maintain a CR after treatment discontinuation.
Collapse
Affiliation(s)
- Eve Bédouelle
- Department of Dermatology, CHU Nantes, CIC 1413, CRCINA Inserm U 1232, Nantes University, Nantes, France
| | | | - Emilie Varey
- Department of Dermatology, CHU Nantes, CIC 1413, CRCINA Inserm U 1232, Nantes University, Nantes, France
| | - Amir Khammari
- Department of Dermatology, CHU Nantes, CIC 1413, CRCINA Inserm U 1232, Nantes University, Nantes, France
| | - Brigitte Dreno
- Department of Dermatology, CHU Nantes, CIC 1413, CRCINA Inserm U 1232, Nantes University, Nantes, France
| |
Collapse
|
6
|
Biosse-Duplan G, Murphy S, Durrani A, Corrie P. Evolving treatment landscape: 18 years of managing melanoma in a single patient. BMJ Case Rep 2021; 14:14/11/e246070. [PMID: 34728513 PMCID: PMC8565563 DOI: 10.1136/bcr-2021-246070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A 57-year-old woman diagnosed with primary melanoma was managed with a wide variety of treatments over 18 years. Given her long history of disease, the array of therapies she has received range from those no longer recommended to those recently approved. This case highlights the extraordinary rate at which both the medical and surgical melanoma treatment landscape has evolved, alongside how professional consensus has changed over the past two decades. It also demonstrates the innovation and collaboration required between the patient and the multidisciplinary team, as well as how external factors such as national guidelines, eligibility for clinical trials and drug funding in the National Health Service (NHS) alter a management plan, presenting yet another set of challenges when managing cancer patients in the modern era.
Collapse
Affiliation(s)
| | - Suzanne Murphy
- Department of Plastic and Reconstructive Surgery, Addenbrooke's Hospital, Cambridge, UK
| | - Amer Durrani
- Department of Plastic and Reconstructive Surgery, Addenbrooke's Hospital, Cambridge, UK
| | - Pippa Corrie
- Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
7
|
Mitogen-activated protein kinase blockade in melanoma: intermittent versus continuous therapy, from preclinical to clinical data. Curr Opin Oncol 2021; 33:127-132. [PMID: 33315631 DOI: 10.1097/cco.0000000000000706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Although targeted therapy provides a high response rate and rapid disease control in advanced melanoma, most patients experience disease progression due to acquired resistance mechanisms leading to reactivation of mitogen-activated protein kinase pathway. The purpose of this article is to review the recently published data on the impact of an intermittent versus continuous dosing schedule of BRAF and MEK inhibition in advanced melanoma to determine the best approach in clinical practice. RECENT FINDINGS Some preclinical studies have highlighted the concept that drug-resistant cells may also display drug dependency, such that intermittent dosing of targeted therapy may prevent the emergence of lethal drug resistance. Moreover, clinical observations have suggested that repeated treatment after a break or an intervening therapy may provide clinical benefit. However, recent preclinical and clinical studies have also failed to demonstrate an advantage of intermittent dosing and showed a similar efficacy of the intermittent versus continuous regimens of BRAF and MEK inhibitors in mice models and phase 2 clinical trial. SUMMARY Owing to these discordant results, continuous dosing of BRAF and MEK inhibitors remains the optimal therapeutic approach until additional clinical data demonstrate the superiority of another combination or dosing regimen.
Collapse
|
8
|
Clinical Implications of Acquired BRAF Inhibitors Resistance in Melanoma. Int J Mol Sci 2020; 21:ijms21249730. [PMID: 33419275 PMCID: PMC7766699 DOI: 10.3390/ijms21249730] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Understanding the role of mitogen-activated protein kinase (MAPK) pathway-activating mutations in the development and progression of melanoma and their possible use as therapeutic targets has substantially changed the management of this neoplasm, which, until a few years ago, was burdened by severe mortality. However, the presence of numerous intrinsic and extrinsic mechanisms of resistance to BRAF inhibitors compromises the treatment responses’ effectiveness and durability. The strategy of overcoming these resistances by combination therapy has proved successful, with the additional benefit of reducing side effects derived from paradoxical activation of the MAPK pathway. Furthermore, the use of other highly specific inhibitors, intermittent dosing schedules and the association of combination therapy with immune checkpoint inhibitors are promising new therapeutic strategies. However, numerous issues related to dose, tolerability and administration sequence still need to be clarified, as is to be expected from currently ongoing trials. In this review, we describe the clinical results of using BRAF inhibitors in advanced melanoma, with a keen interest in strategies aimed at overcoming resistance.
Collapse
|
9
|
Guo C, Chénard-Poirier M, Roda D, de Miguel M, Harris SJ, Candilejo IM, Sriskandarajah P, Xu W, Scaranti M, Constantinidou A, King J, Parmar M, Turner AJ, Carreira S, Riisnaes R, Finneran L, Hall E, Ishikawa Y, Nakai K, Tunariu N, Basu B, Kaiser M, Lopez JS, Minchom A, de Bono JS, Banerji U. Intermittent schedules of the oral RAF-MEK inhibitor CH5126766/VS-6766 in patients with RAS/RAF-mutant solid tumours and multiple myeloma: a single-centre, open-label, phase 1 dose-escalation and basket dose-expansion study. Lancet Oncol 2020; 21:1478-1488. [PMID: 33128873 DOI: 10.1016/s1470-2045(20)30464-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 01/12/2023]
Abstract
BACKGROUND CH5126766 (also known as VS-6766, and previously named RO5126766), a novel MEK-pan-RAF inhibitor, has shown antitumour activity across various solid tumours; however, its initial development was limited by toxicity. We aimed to investigate the safety and toxicity profile of intermittent dosing schedules of CH5126766, and the antitumour activity of this drug in patients with solid tumours and multiple myeloma harbouring RAS-RAF-MEK pathway mutations. METHODS We did a single-centre, open-label, phase 1 dose-escalation and basket dose-expansion study at the Royal Marsden National Health Service Foundation Trust (London, UK). Patients were eligible for the study if they were aged 18 years or older, had cancers that were refractory to conventional treatment or for which no conventional therapy existed, and if they had a WHO performance status score of 0 or 1. For the dose-escalation phase, eligible patients had histologically or cytologically confirmed advanced or metastatic solid tumours. For the basket dose-expansion phase, eligible patients had advanced or metastatic solid tumours or multiple myeloma harbouring RAS-RAF-MEK pathway mutations. During the dose-escalation phase, we evaluated three intermittent oral schedules (28-day cycles) in patients with solid tumours: (1) 4·0 mg or 3·2 mg CH5126766 three times per week; (2) 4·0 mg CH5126766 twice per week; and (3) toxicity-guided dose interruption schedule, in which treatment at the recommended phase 2 dose (4·0 mg CH5126766 twice per week) was de-escalated to 3 weeks on followed by 1 week off if patients had prespecified toxic effects (grade 2 or worse diarrhoea, rash, or creatinine phosphokinase elevation). In the basket dose-expansion phase, we evaluated antitumour activity at the recommended phase 2 dose, determined from the dose-escalation phase, in biomarker-selected patients. The primary endpoints were the recommended phase 2 dose at which no more than one out of six patients had a treatment-related dose-limiting toxicity, and the safety and toxicity profile of each dosing schedule. The key secondary endpoint was investigator-assessed response rate in the dose-expansion phase. Patients who received at least one dose of the study drug were evaluable for safety and patients who received one cycle of the study drug and underwent baseline disease assessment were evaluable for response. This trial is registered with ClinicalTrials.gov, NCT02407509. FINDINGS Between June 5, 2013, and Jan 10, 2019, 58 eligible patients were enrolled to the study: 29 patients with solid tumours were included in the dose-escalation cohort and 29 patients with solid tumours or multiple myeloma were included in the basket dose-expansion cohort (12 non-small-cell lung cancer, five gynaecological malignancy, four colorectal cancer, one melanoma, and seven multiple myeloma). Median follow-up at the time of data cutoff was 2·3 months (IQR 1·6-3·5). Dose-limiting toxicities included grade 3 bilateral retinal pigment epithelial detachment in one patient who received 4·0 mg CH5126766 three times per week, and grade 3 rash (in two patients) and grade 3 creatinine phosphokinase elevation (in one patient) in those who received 3·2 mg CH5126766 three times per week. 4·0 mg CH5126766 twice per week (on Monday and Thursday or Tuesday and Friday) was established as the recommended phase 2 dose. The most common grade 3-4 treatment-related adverse events were rash (11 [19%] patients), creatinine phosphokinase elevation (six [11%]), hypoalbuminaemia (six [11%]), and fatigue (four [7%]). Five (9%) patients had serious treatment-related adverse events. There were no treatment-related deaths. Eight (14%) of 57 patients died during the trial due to disease progression. Seven (27% [95% CI 11·6-47·8]) of 26 response-evaluable patients in the basket expansion achieved objective responses. INTERPRETATION To our knowledge, this is the first study to show that highly intermittent schedules of a RAF-MEK inhibitor has antitumour activity across various cancers with RAF-RAS-MEK pathway mutations, and that this inhibitor is tolerable. CH5126766 used as a monotherapy and in combination regimens warrants further evaluation. FUNDING Chugai Pharmaceutical.
Collapse
Affiliation(s)
- Christina Guo
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Maxime Chénard-Poirier
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Desamparados Roda
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Maria de Miguel
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Samuel J Harris
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Irene Moreno Candilejo
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Priya Sriskandarajah
- Division of Cancer Therapeutics, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Wen Xu
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Mariana Scaranti
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Anastasia Constantinidou
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Jenny King
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Mona Parmar
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Alison J Turner
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | | | - Ruth Riisnaes
- Cancer Biomarkers, The Institute of Cancer Research, London, UK
| | - Laura Finneran
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Emma Hall
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Yuji Ishikawa
- Translational Research Division, Chugai Pharmaceutical, Tokyo, Japan
| | - Kiyohiko Nakai
- Translational Research Division, Chugai Pharmaceutical, Tokyo, Japan
| | - Nina Tunariu
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Bristi Basu
- Department of Oncology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Martin Kaiser
- Division of Molecular Pathology and Myeloma Molecular Therapy Group, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Juanita Suzanne Lopez
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Anna Minchom
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Johann S de Bono
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Udai Banerji
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK.
| |
Collapse
|
10
|
Ravindran Menon D, Hammerlindl H, Torrano J, Schaider H, Fujita M. Epigenetics and metabolism at the crossroads of stress-induced plasticity, stemness and therapeutic resistance in cancer. Theranostics 2020; 10:6261-6277. [PMID: 32483452 PMCID: PMC7255038 DOI: 10.7150/thno.42523] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the recent advances in the treatment of cancers, acquired drug resistance remains a major challenge in cancer management. While earlier studies suggest Darwinian factors driving acquired drug resistance, recent studies point to a more dynamic process involving phenotypic plasticity and tumor heterogeneity in the evolution of acquired drug resistance. Chronic stress after drug treatment induces intrinsic cellular reprogramming and cancer stemness through a slow-cycling persister state, which subsequently drives cancer progression. Both epigenetic and metabolic mechanisms play an important role in this dynamic process. In this review, we discuss how epigenetic and metabolic reprogramming leads to stress-induced phenotypic plasticity and acquired drug resistance, and how the two reprogramming mechanisms crosstalk with each other.
Collapse
|
11
|
BRAF inhibitor treatment in classic hairy cell leukemia: a long-term follow-up study of patients treated outside clinical trials. Leukemia 2019; 34:1454-1457. [PMID: 31740808 DOI: 10.1038/s41375-019-0646-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 10/17/2019] [Accepted: 11/03/2019] [Indexed: 12/21/2022]
|
12
|
Garnier L, Ducray F, Verlut C, Mihai MI, Cattin F, Petit A, Curtit E. Prolonged Response Induced by Single Agent Vemurafenib in a BRAF V600E Spinal Ganglioglioma: A Case Report and Review of the Literature. Front Oncol 2019; 9:177. [PMID: 30984614 PMCID: PMC6448025 DOI: 10.3389/fonc.2019.00177] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 03/04/2019] [Indexed: 12/11/2022] Open
Abstract
Spinal ganglioglioma is a rare low-grade, slow-growing tumor of the central nervous system affecting mostly children and young adults. After surgery, some patients show tumor recurrence and/or malignant transformation. Gangliogliomas harbor molecular deficiencies such as mutations in the B-rapidly accelerated fibrosarcoma (BRAF) gene, resulting in activation of a downstream signaling pathway and cancer development. Vemurafenib is a BRAF inhibitor used to treat patients with BRAF V600E-mutated cancer. Although a few studies have reported the clinical responses in gangliogliomas, the sequence and duration of treatment have not been established. We describe a case of an adult with a progressive BRAF V600E mutant spinal cord ganglioglioma 9 years after surgery who was treated with vemurafenib. This treatment resulted in a partial response within 2 months, which was sustained for more than a year. The patient then decided to stop treatment because of side effects. Despite this decision, the tumor showed no sign of progression 21 months after treatment discontinuation. This is the first reported case of a response to vemurafenib in an adult with progressive spinal cord BRAF V600E-mutated ganglioglioma which was sustained after treatment discontinuation.
Collapse
Affiliation(s)
- Louis Garnier
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - François Ducray
- Department of Neuro-Oncology, Hospices Civils de Lyon, Lyon, France
| | - Clotilde Verlut
- Department of Neurology, University Hospital of Besançon, Besançon, France
| | | | - Françoise Cattin
- Department of Radiology, University Hospital of Besançon, Besançon, France
| | - Antoine Petit
- Department of Neurosurgery, University Hospital of Besançon, Besançon, France
| | - Elsa Curtit
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| |
Collapse
|
13
|
Djebbari F, Stoner N, Lavender VT. A systematic review of non-standard dosing of oral anticancer therapies. BMC Cancer 2018; 18:1154. [PMID: 30466406 PMCID: PMC6249819 DOI: 10.1186/s12885-018-5066-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 11/07/2018] [Indexed: 12/03/2022] Open
Abstract
Background The use of oral systemic anticancer therapies (SACT) has increased and led to improved cancer survival outcomes, particularly with the introduction of small molecule targeted agents and immunomodulators. Oral targeted SACT are, however, associated with toxicities, which might result in reduced quality of life and non-adherence. To reduce treatment-related toxicity, the practice of non-standard dosing is increasing; however guidance to govern this practice is limited. A systematic review was conducted to identify evidence of, and outcomes from, non-standard dosing of oral SACT in oncology and malignant haematology. Methods A comprehensive search of 78 oral SACT was conducted in the following databases: MEDLINE®, EMBASE®, Cochrane Library©, and Cumulative Index to Nursing and Allied Health Literature (CINAHL©). Studies were selected based on predefined inclusion/exclusion criteria, and were critically appraised. Extracted data were tabulated to summarise key findings. Due to diversity of study designs and heterogeneity of reported outcomes, studies were categorised and evidence was synthesised in three main themes: dose interruption; dose reduction; and other dosing strategies. Results Thirty-four studies were eligible for inclusion: four clinical trials, fifteen cohort studies and fifteen case reports. Evidence for non-standard dosing was reported for eleven oral SACT. Dose interruptions were the most commonly reported strategy (14 studies); nine studies reported dose reductions; and eleven reported other dosing strategies. Eight retrospective cohort studies reported dose interruption of sunitinib in renal cell carcinoma and showed either similar or improved responses and survival outcomes, and fewer or equivalent high grade toxicities, compared to the standard schedule. Four cohort studies retrospectively evaluated dose reductions of imatinib, gefitinib or erlotinib, for chronic myeloid leukaemia and non-small cell lung cancer, respectively. Other dosing strategies included alternate-day dosing. The quality of the evidence was limited by the small sample size in many studies, retrospective study designs, and lack of reported toxicity and/or QoL outcomes. Conclusions This review identified limited evidence to support current non-standard dosing strategies, but some of findings, e.g. dose interruption of sunitinib, warrant further investigation in large-scale prospective clinical trials. Electronic supplementary material The online version of this article (10.1186/s12885-018-5066-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Faouzi Djebbari
- Oxford Cancer and Haematology Centre & NIHR Oxford Biomedical Research Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Old Road, Headington, Oxford, OX3 7LE, UK.
| | - Nicola Stoner
- Oxford Cancer and Haematology Centre & Oxford Cancer Research Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Old Road, Headington, Oxford, OX3 7LE, UK
| | - Verna Teresa Lavender
- Faculty of Health and Life Sciences, Oxford Brookes University, Marston Road, Oxford, OX3 0FL, UK
| |
Collapse
|
14
|
Chen P, Chen F, Zhou B. Systematic review and meta-analysis of prevalence of dermatological toxicities associated with vemurafenib treatment in patients with melanoma. Clin Exp Dermatol 2018; 44:243-251. [PMID: 30280426 DOI: 10.1111/ced.13751] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2017] [Indexed: 12/12/2022]
Affiliation(s)
- P. Chen
- Department of Pharmacy; Renmin Hospital of Wuhan University; Wuhan China
| | - F. Chen
- Department of Pharmacy; Dongfeng Hospital; Hubei University of Medicine; Shiyan China
| | - B. Zhou
- Department of Pharmacy; Renmin Hospital of Wuhan University; Wuhan China
- School of Pharmaceutical Sciences; Wuhan University; Wuhan China
| |
Collapse
|
15
|
Gowda R, Dinavahi SS, Iyer S, Banerjee S, Neves RI, Pameijer CR, Robertson. GP. Nanoliposomal delivery of cytosolic phospholipase A 2 inhibitor arachidonyl trimethyl ketone for melanoma treatment. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2018; 14:863-873. [PMID: 29317343 PMCID: PMC5899023 DOI: 10.1016/j.nano.2017.12.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/15/2017] [Accepted: 12/27/2017] [Indexed: 12/19/2022]
Abstract
Drug resistance and toxicity are major limitations of cancer treatment and frequently occurs during melanoma therapy. Nanotechnology can decrease drug resistance by improving drug delivery, with limited toxicity. This study details the development of nanoparticles containing arachidonyl trifluoromethyl ketone (ATK), a cytosolic phospholipase A2 inhibitor, which can inhibit multiple key pathways responsible for the development of recurrent resistant disease. Free ATK is toxic, limiting its efficacy as a therapeutic agent. Hence, a novel nanoliposomal delivery system called NanoATK was developed, which loads 61.7% of the compound and was stable at 4oC for 12 weeks. The formulation decreased toxicity-enabling administration of higher doses, which was more effective at inhibiting melanoma cell growth compared to free-ATK. Mechanistically, NanoATK decreased cellular proliferation and triggered apoptosis to inhibit melanoma xenograft tumor growth without affecting animal weight. Functionally, it inhibited the cPLA2, AKT, and STAT3 pathways. Our results suggest the successful preclinical development of a unique nanoliposomal formulation containing ATK for the treatment of melanoma.
Collapse
Affiliation(s)
- Raghavendra Gowda
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033,The Penn State Melanoma and Skin Cancer Center The Pennsylvania State University College of Medicine, Hershey, PA 17033,Penn State Melanoma Therapeutics Program The Pennsylvania State University College of Medicine, Hershey, PA 17033,Foreman Foundation for Melanoma Research The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Saketh S. Dinavahi
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Soumya Iyer
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Shubhadeep Banerjee
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Rogerio I. Neves
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033,Department of Dermatology and The Pennsylvania State University College of Medicine, Hershey, PA 17033 The Pennsylvania State University College of Medicine, Hershey, PA 17033,Department of Surgery The Pennsylvania State University College of Medicine, Hershey, PA 17033,The Penn State Melanoma and Skin Cancer Center The Pennsylvania State University College of Medicine, Hershey, PA 17033,Penn State Melanoma Therapeutics Program The Pennsylvania State University College of Medicine, Hershey, PA 17033,Foreman Foundation for Melanoma Research The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Colette R. Pameijer
- Department of Surgery The Pennsylvania State University College of Medicine, Hershey, PA 17033,The Penn State Melanoma and Skin Cancer Center The Pennsylvania State University College of Medicine, Hershey, PA 17033,Penn State Melanoma Therapeutics Program The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Gavin P. Robertson.
- Department of Pharmacology The Pennsylvania State University College of Medicine, Hershey, PA 17033,Department of Pathology The Pennsylvania State University College of Medicine, Hershey, PA 17033,Department of Dermatology and The Pennsylvania State University College of Medicine, Hershey, PA 17033 The Pennsylvania State University College of Medicine, Hershey, PA 17033,Department of Surgery The Pennsylvania State University College of Medicine, Hershey, PA 17033,The Penn State Melanoma and Skin Cancer Center The Pennsylvania State University College of Medicine, Hershey, PA 17033,Penn State Melanoma Therapeutics Program The Pennsylvania State University College of Medicine, Hershey, PA 17033,Foreman Foundation for Melanoma Research The Pennsylvania State University College of Medicine, Hershey, PA 17033
| |
Collapse
|
16
|
Rechallenge with BRAF-directed treatment in metastatic melanoma: A multi-institutional retrospective study. Eur J Cancer 2018; 91:116-124. [DOI: 10.1016/j.ejca.2017.12.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 12/02/2017] [Indexed: 12/11/2022]
|
17
|
Kong X, Kuilman T, Shahrabi A, Boshuizen J, Kemper K, Song JY, Niessen HWM, Rozeman EA, Geukes Foppen MH, Blank CU, Peeper DS. Cancer drug addiction is relayed by an ERK2-dependent phenotype switch. Nature 2017; 550:270-274. [PMID: 28976960 PMCID: PMC5640985 DOI: 10.1038/nature24037] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 08/25/2017] [Indexed: 12/29/2022]
Abstract
Drug addiction denotes the dependency of tumors on the same therapeutic
drugs to which they have acquired resistance. Observations from cultured
cells1–3, animal models4 and
patients5–7 raise the possibility that cancer drug addiction can
instigate a potential cancer vulnerability, which may be used therapeutically.
However, for this trait to become of clinical interest, it is imperative to
first define the underlying mechanism. Therefore, we performed an unbiased
CRISPR-Cas9 knockout screen to functionally mine the genome of melanoma cells
that are both resistant and addicted to BRAF inhibition for “addiction
genes”. Here, we describe a signaling pathway comprising ERK2, JUNB and
FRA1, disruption of which allows tumor cells to reverse addiction and survive
upon treatment discontinuation. This occurred both in culture and mice, and was
irrespective of the acquired drug resistance mechanism. In melanoma and lung
cancer cells, death induced by drug withdrawal was preceded by a specific
ERK2-dependent phenotype switch, alongside transcriptional reprogramming
reminiscent of EMT. In melanoma, this caused shutdown of the lineage survival
oncoprotein MITF, restoration of which reversed both phenotype switching and
drug addiction-associated lethality. In melanoma patients who had progressed on
BRAF inhibition, treatment cessation was followed by increased expression of the
phenotype switch-associated receptor tyrosine kinase AXL. Drug discontinuation
synergized with the melanoma chemotherapeutic dacarbazine by further suppressing
MITF and its prosurvival target BCL2 while inducing DNA damage. Our results
uncover a pathway driving cancer drug addiction, which may guide alternating
therapeutic strategies for enhanced clinical responses of drug-resistant
cancers.
Collapse
Affiliation(s)
- Xiangjun Kong
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Thomas Kuilman
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Aida Shahrabi
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Julia Boshuizen
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Kristel Kemper
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Ji-Ying Song
- Division of Experimental Animal Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Hans W M Niessen
- Department of Pathology and Cardiac Surgery, VU University Medical Center, ACS, 1007 MB Amsterdam, The Netherlands
| | - Elisa A Rozeman
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Marnix H Geukes Foppen
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Christian U Blank
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Daniel S Peeper
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
18
|
BRAF inhibitor discontinuation and rechallenge in advanced melanoma patients with a complete initial treatment response. Melanoma Res 2017; 27:281-287. [DOI: 10.1097/cmr.0000000000000350] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
19
|
Marconcini R, Galli L, Antonuzzo A, Bursi S, Roncella C, Fontanini G, Sensi E, Falcone A. Metastatic BRAF K601E-mutated melanoma reaches complete response to MEK inhibitor trametinib administered for over 36 months. Exp Hematol Oncol 2017; 6:6. [PMID: 28344857 PMCID: PMC5361706 DOI: 10.1186/s40164-017-0067-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 03/09/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The BRAF K601E mutation occurs in 5% of patients with melanoma, and is the third most common type of BRAF mutation. However, treatment with BRAF and mitogen-activated extracellular signal-regulated kinase (MEK) inhibitors is only approved in patients with BRAF V600-positive melanoma, and patients with K601E-mutated melanoma do not have access to such drugs. CASE PRESENTATION A female patient was diagnosed with high tumor burden metastatic melanoma harboring the BRAF K601E mutation. After chemotherapy failure, she underwent compassionate treatment with trametinib. Trametinib showed good activity and efficacy, with 48% shrinkage of a metastatic lymphadenopathy after 4 months' treatment. However, the patient reported treatment-related skin toxicity that required dosage reduction and a personalized intermittent trametinib dosing schedule. After over 36 months from the first trametinib administration, and resection of a metastatic lymphadenopathy, the patient experienced complete response. CONCLUSIONS This case report shows that trametinib could be a valid therapeutic option in patients with metastatic melanoma harboring the rare BRAF K601E mutation.
Collapse
Affiliation(s)
- Riccardo Marconcini
- Department of Oncology, Azienda Ospedaliero-Universitaria Pisana and University of Pisa, Istituto Toscano Tumori, Santa Chiara Hospital, Via Roma 67, 56100 Pisa, Italy
| | - Luca Galli
- Department of Oncology, Azienda Ospedaliero-Universitaria Pisana and University of Pisa, Istituto Toscano Tumori, Santa Chiara Hospital, Via Roma 67, 56100 Pisa, Italy
| | - Andrea Antonuzzo
- Department of Oncology, Azienda Ospedaliero-Universitaria Pisana and University of Pisa, Istituto Toscano Tumori, Santa Chiara Hospital, Via Roma 67, 56100 Pisa, Italy
| | - Simona Bursi
- Department of Oncology, Ospedale Civile-Istituto Toscano Tumori, Livorno, Italy
| | - Claudia Roncella
- Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| | - Gabriella Fontanini
- Units of Pathological Anatomy, Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Elisa Sensi
- Units of Pathological Anatomy, Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Alfredo Falcone
- Department of Oncology, Azienda Ospedaliero-Universitaria Pisana and University of Pisa, Istituto Toscano Tumori, Santa Chiara Hospital, Via Roma 67, 56100 Pisa, Italy
| |
Collapse
|
20
|
Spain L, Goode E, McGovern Y, Joshi K, Larkin J. The combination of vemurafenib and cobimetinib in advanced melanoma. Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2016.1241707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
21
|
Abstract
INTRODUCTION Cobimetinib combined with vemurafenib is a new approved MEK inhibitor for first line treatment of metastatic melanoma patients with BRAF V600 mutations. It improves tumor response rates and progression free survival compared to vemurafenib alone, while decreasing toxicities due to the paradoxical activation of the MAPK signaling pathway. AREAS COVERED This review covers the pharmacology, efficacy, and toxicity data derived from clinical and preclinical studies on cobimetinib. It also reports ongoing trials evaluating cobimetinib to better understand future developments for this drug. EXPERT OPINION The combination of cobimetinib and vemurafenib seems to be more toxic than the combination therapy dabrafenib and trametinib even if these four drugs have never been compared in a randomized trial. The future of this combination depends on its capacity to be combined simultaneously or sequentially with immune based therapies to improve the durability of responses.
Collapse
Affiliation(s)
- Amélie Boespflug
- a Dermatology Unit , Hospices Civils de Lyon , Lyon , France.,b INSERM U1052 , Cancer Research Center of Lyon , Lyon , France.,c CNRS UMR 5286 , Cancer Research Center of Lyon , Lyon , France.,d Université Lyon1 , Department of Medecine , Lyon , France
| | - Luc Thomas
- a Dermatology Unit , Hospices Civils de Lyon , Lyon , France.,b INSERM U1052 , Cancer Research Center of Lyon , Lyon , France.,c CNRS UMR 5286 , Cancer Research Center of Lyon , Lyon , France.,d Université Lyon1 , Department of Medecine , Lyon , France
| |
Collapse
|
22
|
Gillis NK, McLeod HL. The pharmacogenomics of drug resistance to protein kinase inhibitors. Drug Resist Updat 2016; 28:28-42. [PMID: 27620953 PMCID: PMC5022787 DOI: 10.1016/j.drup.2016.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/17/2016] [Accepted: 06/29/2016] [Indexed: 01/05/2023]
Abstract
Dysregulation of growth factor cell signaling is a major driver of most human cancers. This has led to development of numerous drugs targeting protein kinases, with demonstrated efficacy in the treatment of a wide spectrum of cancers. Despite their high initial response rates and survival benefits, the majority of patients eventually develop resistance to these targeted therapies. This review article discusses examples of established mechanisms of drug resistance to anticancer therapies, including drug target mutations or gene amplifications, emergence of alternate signaling pathways, and pharmacokinetic variation. This reveals a role for pharmacogenomic analysis to identify and monitor for resistance, with possible therapeutic strategies to combat chemoresistance.
Collapse
Affiliation(s)
- Nancy K Gillis
- Eshelman School of Pharmacy, Center for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC, United States; H. Lee Moffitt Cancer Center and Research Institute, DeBartolo Family Personalized Medicine Institute, Tampa, FL, United States
| | - Howard L McLeod
- H. Lee Moffitt Cancer Center and Research Institute, DeBartolo Family Personalized Medicine Institute, Tampa, FL, United States; Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
23
|
Dooley AJ, Gupta A, Middleton MR. Ongoing Response in BRAF V600E-Mutant Melanoma After Cessation of Intermittent Vemurafenib Therapy: A Case Report. Target Oncol 2016; 11:557-63. [PMID: 26857260 PMCID: PMC5309307 DOI: 10.1007/s11523-015-0410-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The selective BRAF inhibitors vemurafenib and dabrafenib yield high response rates and improved overall survival in patients with BRAF V600E-mutant metastatic melanoma. Treatment traditionally continues until disease progression or the development of unacceptable toxicity. Acquired drug resistance and toxicity are key challenges with the use of these drugs. Resistance to vemurafenib usually develops within 6-8 months. Management of drug toxicity typically involves stopping vemurafenib until resolution, before restarting at a lower dose, or permanently ceasing vemurafenib therapy. We have recently considered whether intermittent dosing could be used as an alternative to dose reduction/termination in the management of vemurafenib toxicity. One patient treated with intermittent vemurafenib was an 89-year-old woman with metastatic melanoma, who initially showed a good response to continuous dosing. Recurrent toxicity meant that the continuous vemurafenib dosage was repeatedly ceased before restarting at a lower dose. Ten months after vemurafenib was first begun, an intermittent dosing regimen was introduced in an attempt to control toxicity. This continued for 2 months, before cessation due to continued unacceptable toxicity. A further 24 months later, the patient remains fit and well in complete clinical remission, with no recurrence of her previous melanoma and no new primary malignancies. To the best of our knowledge, a continued response after the cessation of selective BRAF inhibitors has never before been described in melanoma. Induction of an immune response and/or epigenetic changes could explain continued disease response after cessation of vemurafenib therapy. Care should be taken when extrapolating the findings from the continued response after vemurafenib cessation to other tumour types. We recommend the collection and analysis of data to investigate the clinical responses seen after cessation of vemurafenib due to intolerable toxicities, which could help further explain vemurafenib's mechanism of action.
Collapse
Affiliation(s)
- Andrew J Dooley
- University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Avinash Gupta
- Department of Oncology, NIHR Biomedical Research Centre, Oxford Cancer and Haematology Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
| | - Mark R Middleton
- Department of Oncology, NIHR Biomedical Research Centre, Oxford Cancer and Haematology Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK.
| |
Collapse
|
24
|
Tate SC, Burke TF, Hartman D, Kulanthaivel P, Beckmann RP, Cronier DM. Optimising the combination dosing strategy of abemaciclib and vemurafenib in BRAF-mutated melanoma xenograft tumours. Br J Cancer 2016; 114:669-79. [PMID: 26978007 PMCID: PMC4800303 DOI: 10.1038/bjc.2016.40] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/15/2016] [Accepted: 01/28/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Resistance to BRAF inhibition is a major cause of treatment failure for BRAF-mutated metastatic melanoma patients. Abemaciclib, a cyclin-dependent kinase 4 and 6 inhibitor, overcomes this resistance in xenograft tumours and offers a promising drug combination. The present work aims to characterise the quantitative pharmacology of the abemaciclib/vemurafenib combination using a semimechanistic pharmacokinetic/pharmacodynamic modelling approach and to identify an optimum dosing regimen for potential clinical evaluation. METHODS A PK/biomarker model was developed to connect abemaciclib/vemurafenib concentrations to changes in MAPK and cell cycle pathway biomarkers in A375 BRAF-mutated melanoma xenografts. Resultant tumour growth inhibition was described by relating (i) MAPK pathway inhibition to apoptosis, (ii) mitotic cell density to tumour growth and, under resistant conditions, (iii) retinoblastoma protein inhibition to cell survival. RESULTS The model successfully described vemurafenib/abemaciclib-mediated changes in MAPK pathway and cell cycle biomarkers. Initial tumour shrinkage by vemurafenib, acquisition of resistance and subsequent abemaciclib-mediated efficacy were successfully captured and externally validated. Model simulations illustrate the benefit of intermittent vemurafenib therapy over continuous treatment, and indicate that continuous abemaciclib in combination with intermittent vemurafenib offers the potential for considerable tumour regression. CONCLUSIONS The quantitative pharmacology of the abemaciclib/vemurafenib combination was successfully characterised and an optimised, clinically-relevant dosing strategy was identified.
Collapse
Affiliation(s)
- Sonya C Tate
- Global PK/PD, Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK
| | - Teresa F Burke
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Daisy Hartman
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | | | - Richard P Beckmann
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Damien M Cronier
- Global PK/PD, Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK
| |
Collapse
|
25
|
Vemurafenib-associated neutrophilic panniculitis in a patient with metastatic amelanotic melanoma presenting as cancer of unknown primary origin. DERMATOL SIN 2016. [DOI: 10.1016/j.dsi.2015.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
26
|
Affiliation(s)
- Mark Minden
- Princess Margaret Cancer Centre, Toronto, Canada
| |
Collapse
|
27
|
BRAF inhibition in hairy cell leukemia with low-dose vemurafenib. Blood 2016; 127:2847-55. [PMID: 26941398 DOI: 10.1182/blood-2015-11-680074] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/13/2016] [Indexed: 11/20/2022] Open
Abstract
The activating mutation of the BRAF serine/threonine protein kinase (BRAF V600E) is the key driver mutation in hairy cell leukemia (HCL), suggesting opportunities for therapeutic targeting. We analyzed the course of 21 HCL patients treated with vemurafenib outside of trials with individual dosing regimens (240-1920 mg/d; median treatment duration, 90 days). Vemurafenib treatment improved blood counts in all patients, with platelets, neutrophils, and hemoglobin recovering within 28, 43, and 55 days (median), respectively. Complete remission was achieved in 40% (6/15 of evaluable patients) and median event-free survival was 17 months. Response rate and kinetics of response were independent of vemurafenib dosing. Retreatment with vemurafenib led to similar response patterns (n = 6). Pharmacodynamic analysis of BRAF V600E downstream targets showed that vemurafenib (480 mg/d) completely abrogated extracellular signal-regulated kinase phosphorylation of hairy cells in vivo. Typical side effects also occurred at low dosing regimens. We observed the development of acute myeloid lymphoma (AML) subtype M6 in 1 patient, and the course suggested disease acceleration triggered by vemurafenib. The phosphatidylinositol 3-kinase hotspot mutation (E545K) was identified in the AML clone, providing a potential novel mechanism for paradoxical BRAF activation. These data provide proof of dependence of HCL on active BRAF signaling. We provide evidence that antitumor and side effects are observed with 480 mg vemurafenib, suggesting that dosing regimens in BRAF-driven cancers could warrant reassessment in trials with implications for cost of cancer care.
Collapse
|
28
|
Ebert PJR, Cheung J, Yang Y, McNamara E, Hong R, Moskalenko M, Gould SE, Maecker H, Irving BA, Kim JM, Belvin M, Mellman I. MAP Kinase Inhibition Promotes T Cell and Anti-tumor Activity in Combination with PD-L1 Checkpoint Blockade. Immunity 2016; 44:609-621. [PMID: 26944201 DOI: 10.1016/j.immuni.2016.01.024] [Citation(s) in RCA: 528] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/26/2016] [Accepted: 01/29/2016] [Indexed: 12/11/2022]
Abstract
Targeted inhibition of mitogen-activated protein kinase (MAPK) kinase (MEK) can induce regression of tumors bearing activating mutations in the Ras pathway but rarely leads to tumor eradication. Although combining MEK inhibition with T-cell-directed immunotherapy might lead to more durable efficacy, T cell responses are themselves at least partially dependent on MEK activity. We show here that MEK inhibition did profoundly block naive CD8(+) T cell priming in tumor-bearing mice, but actually increased the number of effector-phenotype antigen-specific CD8(+) T cells within the tumor. MEK inhibition protected tumor-infiltrating CD8(+) T cells from death driven by chronic TCR stimulation while sparing cytotoxic activity. Combining MEK inhibition with anti-programmed death-ligand 1 (PD-L1) resulted in synergistic and durable tumor regression even where either agent alone was only modestly effective. Thus, despite the central importance of the MAP kinase pathway in some aspects of T cell function, MEK-targeted agents can be compatible with T-cell-dependent immunotherapy.
Collapse
Affiliation(s)
| | - Jeanne Cheung
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yagai Yang
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Erin McNamara
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Rebecca Hong
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | - Bryan A Irving
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jeong M Kim
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Marcia Belvin
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ira Mellman
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
29
|
Jain T, Bryce A. Intermittent BRAF Inhibition Can Achieve Prolonged Disease Control in BRAF Mutant Melanoma. Cureus 2015; 7:e410. [PMID: 26824010 PMCID: PMC4725676 DOI: 10.7759/cureus.410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BRAF V600E is the most common somatic mutation seen in patients with metastatic melanoma. BRAF inhibitors (BRAFi), along with MEK inhibitors (MEKi), have been shown to improve overall survival in these patients with a median time to resistance of 6-10 months. We describe a patient with an ongoing response of 48 months on intermittent BRAFi therapy. She was started on vemurafenib at initial diagnosis, which was discontinued after a total of 39 weeks of therapy, and achieved a complete response due to cumulative toxicity. Upon evidence of progression on serial imaging following 81 weeks of disease-free status, BRAFi was resumed with dabrafenib, along with trametinib. Complete response was seen with seven weeks of treatment. Therapy was discontinued again, due to side effects, with an intention to pursue intermittent therapy. Serial imaging, so far, has shown no progression or recurrence of disease after over a year (66 weeks and ongoing) since discontinuation of therapy. This case underscores the clinical feasibility of intermittent BRAFi therapy in patients while still achieving a prolonged response. Disease control of 48 months, to date, has been achieved using therapy only “as needed” and keeping toxicities to the minimum.
Collapse
Affiliation(s)
- Tania Jain
- Internal Medicine, Mayo Clinic, Scottsdale, AZ
| | - Alan Bryce
- Hematology Oncology, Mayo Clinic, Scottsdale, AZ
| |
Collapse
|
30
|
Frontline nilotinib in patients with chronic myeloid leukemia in chronic phase: results from the European ENEST1st study. Leukemia 2015; 30:57-64. [PMID: 26437782 PMCID: PMC4705425 DOI: 10.1038/leu.2015.270] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/14/2015] [Accepted: 09/18/2015] [Indexed: 12/20/2022]
Abstract
The Evaluating Nilotinib Efficacy and Safety in Clinical Trials as First-Line Treatment (ENEST1st) study included 1089 patients with newly diagnosed chronic myeloid leukemia in chronic phase. The rate of deep molecular response (MR4 (BCR-ABL1⩽0.01% on the International Scale or undetectable BCR-ABL1 with ⩾10 000 ABL1 transcripts)) at 18 months was evaluated as the primary end point, with molecular responses monitored by the European Treatment and Outcome Study network of standardized laboratories. This analysis was conducted after all patients had completed 24 months of study treatment (80.9% of patients) or discontinued early. In patients with typical BCR-ABL1 transcripts and ⩽3 months of prior imatinib therapy, 38.4% (404/1052) achieved MR4 at 18 months. Six patients (0.6%) developed accelerated or blastic phase, and 13 (1.2%) died. The safety profile of nilotinib was consistent with that of previous studies, although the frequencies of some nilotinib-associated adverse events were lower (for example, rash, 21.4%). Ischemic cardiovascular events occurred in 6.0% of patients. Routine monitoring of lipid and glucose levels was not mandated in the protocol. These results support the use of frontline nilotinib, particularly when achievement of a deep molecular response (a prerequisite for attempting treatment-free remission in clinical trials) is a treatment goal.
Collapse
|
31
|
BRAF inhibitors: the current and the future. Curr Opin Pharmacol 2015; 23:68-73. [PMID: 26072431 DOI: 10.1016/j.coph.2015.05.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 05/22/2015] [Accepted: 05/26/2015] [Indexed: 01/07/2023]
Abstract
The introduction of BRAF inhibitors (BRAFi), vemurafenib and dabrafenib, revolutionized BRAFV600-mutated metastatic melanoma treatment with improved response rate and overall survival compared to standard chemotherapy. However, the mechanism related cutaneous toxicity remains a concern. In addition, intrinsic and acquired resistance remain the key challenges in BRAFi therapy. Extensive efforts to elucidate the mechanisms have led to an improved understanding of disease biology and resulted in exploration of multiple new therapeutic options. While the future looks bright with multiple new therapeutic strategies in exploration and possible new generations of BRAFi, there are questions remaining to be answered to enable more efficient use of BRAFi in cancer therapy.
Collapse
|