The London School of Economics and Political Science Cancer Medicines

Total Page:16

File Type:pdf, Size:1020Kb

The London School of Economics and Political Science Cancer Medicines The London School of Economics and Political Science Cancer medicines: clinical impact, economics, and value Sebastian Salas-Vega A thesis submitted to the Department of Social Policy at the London School of Economics and Political Science for the degree of Doctor of Philosophy, London, April 2017 Declaration I certify that’s the thesis I have presented for examination fully MPhil/PhD degree at the London School of Economics and Political Science is solely my own work other than where I have clearly indicated that the work of others (in which case the extent of any work carried out jointly by me and any other persons clearly identified in it – see ‘Statement of Conjoint Work’). The copyright of this thesis rests with the author. Quotation from it is permitted, provided that full acknowledgement is made. This thesis may not be reproduced without the prior written consent of the author. I warrant that this authorization does not, to the best of my belief, infringe the rights of any third party. I declare that my thesis consists of 63,084 words (excluding bibliography and appendix). ii Statement of Conjoint Work The work that is presented in Chapter 2, and part of the work that is presented in Chapter 4, has been published.1 My co-authors on these publications include: Dr Elias Mossialos of the London School of Economics and Political Science (London, UK) and Dr Othon Iliopoulos of the Harvard Medical School and Massachusetts General Hospital (Boston, MA, US), who provided guidance on research methodologies and clinical input. Mr Rayden Llano of the London School of Economics and Political Science (London, UK) could not be named as co-author in one of these publications due to policies on publishing by federal employees. His contributions are nevertheless acknowledged: he assisted with data collection by serving as a second, independent reviewer of cancer drug appraisals in Chapter 2. For the same chapter, Dr Mossialos was asked to serve as a third reviewer if consensus with Mr Llano could not be reached. This consensus-based approach was used to minimize the introduction of investigator bias during the review of HTA appraisals. All major work related to Chapter 2, including analysis and interpretation of data, was conducted by me. Copyeditor Sarah Moncrieff assisted with designing Figure 5. For the published aspects of this thesis (Chapters 2, 4), I conceived and designed studies, acquired, analyzed, and interpreted data, and drafted manuscripts. Dr Mossialos offered feedback on the published aspects of this thesis, as well as all remaining chapters. Journal reviewers provided feedback, and suggested secondary analyses, that have been incorporated into this thesis. Additional publications are being 1 Salas-Vega S, Mossialos E. Cancer Drugs Provide Positive Value In Nine Countries, But The United States Lags In Health Gains Per Dollar Spent. Health Aff (Millwood). 2016;35(5):813–23. Salas-Vega S, Iliopoulos O, Mossialos E. Assessment of Overall Survival, Quality of Life, and Safety Benefits Associated With New Cancer Medicines. JAMA Oncol. 2016;116(14):3493–504. iii prepared from the work that is presented in the remaining chapters of this thesis. Other than where I have cited the relevant contributions of others, I confirm that I am responsible for the entirety of the work presented in this doctoral thesis. iv Abstract Background and Importance: There has been much debate recently over rapidly growing drug expenditures. Cancer medicines, in particular, have driven new brand spending over recent years, and US oncological expenditures have risen faster than for many other disease areas, in part because of rapidly growing drug prices, as well as increased rates of use. Objective: In the face of ongoing debates on how to reasonably control growth in pharmaceutical spending, while also providing patients with the best possible care, this thesis sets out to help address the question of whether growing pharmaceutical expenditures are providing value-for-money to patients and society. Novelty and Empirical Contributions: This thesis is based in part on a systematic review with narrative synthesis of English-language HTA appraisals of the comparative clinical risks and benefits of new cancer medicines, as well as on the novel use of (b) (4) methods to generate comparative evidence on their use and cost. Adapting established methods, these data are then used to examine existing questions over whether growing expenditures are worth the cost to patients and society. This thesis makes five major contributions to the literature on value-based spending on cancer medicines: 1) approximately one in three newly licensed cancer medicines provide no known overall survival benefit, while one in five provide no known overall survival, quality of life, or safety benefit; 2) novel use of methodologies to model treatment course and duration reveals that cancer drug use and costs vary greatly between individual medicines, and across Australia, France, the UK, and the US; 3) the monetized value of survival gains attributable to cancer drug innovation, net of growth in cancer drug v spending, varies across individual medicines, and, at a country-level, remains unambiguously positive in Australia, France, and the UK, but negative in the US; 4) spending on new cancer medicines is often only weakly associated with their clinical benefits; and 5) the strength of this association nevertheless varies across countries, with the UK demonstrating the strongest evidence of value-based spending on new cancer medicines. Clinical and Policy Implications: Findings from this thesis provide a resource for value- based clinical decision-making by patients and physicians. Moreover, growing expenditures on cancer medicines may only weakly be associated with meaningful clinical benefits, though the extent to which this is true differs across countries. These findings highlight the important role that health policy can have in encouraging value- based cancer drug spending. In particular, it is argued that managed access schemes promoting access and evidence development, as well as the use of value-based spending policies, can help expedite access to new treatments, incentivize the development of clinically meaningful medicines, and rationalize growing cancer drug expenditures. Future Research Directions: The comparative clinical risks and benefits from new cancer medicines using real-world data, and how they compare with trial-based results; how evidence on the comparative impact from new treatments is measured, weighted, and rewarded in decision-making by regulators and payers; and how it is effectively linked through policy and regulation to cancer drug spending. vi Acknowledgments I would like to express my deepest gratitude to all who have offered their guidance and expertise throughout my PhD. In particular, I would like to sincerely thank my doctoral supervisors, Prof Elias Mossialos (1°) and Prof Alistair McGuire (2°), for their mentorship, patience, and, when most needed, encouragement. I am grateful to Mr Rayden Llano of the London School of Economics (London, UK) for assisting with the review of HTA appraisals, and to Dr Irini Moustaki, Dr Zlatko Nikoloski, Dr Anwen Zhang, and Dr Jon Cylus of the London School of Economics (London, UK) for offering feedback on regression and statistical methods. I would also like to thank Dr Othon Iliopoulos of the Harvard Medical School and Massachusetts General Hospital (Boston, MA, USA) and experts from the Food and Drug Administration (Silver Spring, MD, USA) for offering clinical and regulatory feedback on some of the research findings that are presented in this thesis. I must acknowledge the organizations offering the publicly available data that was used in this work: the Australian Bureau of Statistics, European Medicines Agency, Haute Autorité de Santé, Institute for Health Metrics and Evaluation, National Institute for Health and Care Excellence, NHS Digital, Organisation for Economic Co-operation and Development, Pharmaceutical Benefits Advisory Committee, Therapeutic Goods Administration, US Centers for Disease Control and Prevention, US Food and Drug Administration, World Bank, and World Health Organization. Their investments in the public disclosure of information are a testament of their commitment to research and public health. vii I would also like to thank the many individuals who offered insights that contributed to this research: Dr Caroline Rudisill (LSE), Dr Walter Holland (LSE), Aris Angelis (LSE), Sahan Jayawardana (LSE), Felix Cornehl (LSE), Dr Victoria Simpkin (LSE), Emmy Shearer (Stanford University), Ilias Kyriopoulos (LSE), Dr Jia Hu (University of Toronto), Dr José- Luis Fernández (LSE), Dr Bo Hu (LSE), Dr Rebecca Roylance (University College London), Dr David Dodwell (Public Health England), Dr Mauricio Avendano (King’s College London), Dr Isabelle Soerjomataram (WHO), Alex Carter (Imperial College London), Dr Wei Yang (University of Kent), (b) (4) Dr Alexandre Barna (OECD), (b) (4) Thomas Wilkinson (NICE), Dr Josep Figueras (WHO), Kai Schulze (University of Cambridge), Dr Lawrence Brown (Columbia University), Dr Guillaume Dedet (WHO), and Adria Haimann (Health Navigator). I would also like to acknowledge librarians Ellen Wilkinson and Alex Taylor (LSE) for their support. I am thankful to Dr Panos Kanavos and Dr Joachim Marti for their helpful feedback during my doctoral defense. Their comments have helped me strengthen the work that is presented here. I would like to thank QuintilesIMS and Imperial
Recommended publications
  • Potential High-Impact Interventions Report Priority Area 02: Cancer
    AHRQ Healthcare Horizon Scanning System – Potential High-Impact Interventions Report Priority Area 02: Cancer Prepared for: Agency for Healthcare Research and Quality U.S. Department of Health and Human Services 540 Gaither Road Rockville, MD 20850 www.ahrq.gov Contract No. HHSA290201000006C Prepared by: ECRI Institute 5200 Butler Pike Plymouth Meeting, PA 19462 December 2012 Statement of Funding and Purpose This report incorporates data collected during implementation of the Agency for Healthcare Research and Quality (AHRQ) Healthcare Horizon Scanning System by ECRI Institute under contract to AHRQ, Rockville, MD (Contract No. HHSA290201000006C). The findings and conclusions in this document are those of the authors, who are responsible for its content, and do not necessarily represent the views of AHRQ. No statement in this report should be construed as an official position of AHRQ or of the U.S. Department of Health and Human Services. This report’s content should not be construed as either endorsements or rejections of specific interventions. As topics are entered into the System, individual topic profiles are developed for technologies and programs that appear to be close to diffusion into practice in the United States. Those reports are sent to various experts with clinical, health systems, health administration, and/or research backgrounds for comment and opinions about potential for impact. The comments and opinions received are then considered and synthesized by ECRI Institute to identify interventions that experts deemed, through the comment process, to have potential for high impact. Please see the methods section for more details about this process. This report is produced twice annually and topics included may change depending on expert comments received on interventions issued for comment during the preceding 6 months.
    [Show full text]
  • Investigator Initiated Study IRB-29839 an Open-Label Pilot Study To
    Investigator Initiated Study IRB-29839 An open-label pilot study to evaluate the efficacy and safety of a combination treatment of Sonidegib and BKM120 for the treatment of advanced basal cell carcinomas Version 05 September 2016 NCT02303041 DATE: 12Dec2018 1 Coordinating Center Stanford Cancer Center 875 Blake Wilbur Drive Stanford, CA 94305 And 450 Broadway, MC 5334 Redwood City, CA 94603 Protocol Director and Principal Investigator Anne Lynn S Chang, MD, Director of Dermatological Clinical Trials 450 Broadway St, MC 5334 Redwood City, CA 94603 [email protected] Co-Investigator Anthony Oro, MD PhD 450 Broadway St, MC 5334 Redwood City, CA 94603 [email protected] Biostatistician Shufeng Li, MS 450 Broadway St, MC 5334 Redwood City, CA 94603 [email protected] Study Coordinator Ann Moffat 450 Broadway St, MC 5334 Redwood City, CA 94603 [email protected] 2 Table of Contents 1 Background ................................................................. 7 1.1 Disease Background ..................................................... 7 1.2 Hedgehog Pathway and mechanism of action ............................... 7 1.3 PI3K Pathway and mechanism of action ................................... 9 1.4 Sonidegib Compound Information ............ Error! Bookmark not defined. 1.4.1 Preclinical Studies for Sonidegib ....................................................................11 1.4.2 Muscular system...............................................................................................13 1.4.3 Skeletal system ................................................................................................13
    [Show full text]
  • Regimen Reference Order – GAST – FOLFIRI
    ADULT Updated: January 8, 2018 Regimen Reference Order – GAST – FOLFIRI ARIA: GAST – [FOLFIRI] Planned Course: Every 14 days until disease progression or unacceptable toxicity Indication for Use: Colorectal Cancer Metastatic CVAD: Required (Ambulatory Pump) Proceed with treatment if: ANC equal to or greater than 1.5 x 109/L AND Platelets equal to or greater than 100 x 109/L Contact Physician if parameters not met SEQUENCE OF MEDICATION ADMINISTRATION Pre-treatment Requirements Drug Dose CCMB Administration Guideline Not Applicable Treatment Regimen – GAST - FOLFIRI Establish primary solution 500 mL of: normal saline Drug Dose CCMB Administration Guideline ondansetron 16 mg Orally 30 minutes pre-chemotherapy dexamethasone 12 mg Orally 30 minutes pre-chemotherapy atropine 0.6 mg IV Push over 2 – 3 minutes pre-irinotecan May be repeated once if diarrhea occurs during irinotecan infusion irinotecan 180 mg/m2 IV in 500 mL D5W over 90 minutes irinotecan can be infused at the same time as leucovorin through a Y site leucovorin 400 mg/m2 IV in 500 mL D5W over 90 minutes fluorouracil 400 mg/m2 IV Push over 5 minutes fluorouracil 2400 mg/m2 IV in D5W continuously over 46 hours by ambulatory infusion device All doses will be automatically rounded that fall within the DSG Approved Dose Bands. See GAST DSG – Dose Banding document for more information Flush after each medication: 50 mL over 6 minutes (500 mL/hr) In the event of an infusion-related hypersensitivity reaction, refer to the ‘Hypersensitivity Reaction Standing Order’ Please refer to CCMB
    [Show full text]
  • And High-Dose Cyclophosphamide
    141-146 6/6/06 13:01 Page 141 ONCOLOGY REPORTS 16: 141-146, 2006 141 Different mechanisms for anti-tumor effects of low- and high-dose cyclophosphamide YASUHIDE MOTOYOSHI1,2, KAZUHISA KAMINODA1,3, OHKI SAITOH1, KEISUKE HAMASAKI2, KAZUHIKO NAKAO3, NOBUKO ISHII3, YUJI NAGAYAMA1 and KATSUMI EGUCHI2 Departments of 1Medical Gene Technology, Atomic Bomb Disease Institute; 2Division of Immunology, Endocrinology and Metabolism, Department of Medical and Dental Sciences, and 3Division of Clinical Pharmaceutics and Health Research Center, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8501, Japan Received February 3, 2006; Accepted April 7, 2006 Abstract. It is known that, besides its direct cytotoxic effect appear to be highly crucial in a clinical setting of combined as an alkylating chemotherapeutic agent, cyclophosphamide chemotherapy and immunotherapy for cancer treatment. also has immuno-modulatory effects, such as depletion of CD4+CD25+ regulatory T cells. However, its optimal concen- Introduction tration has not yet been fully elucidated. Therefore, we first compared the effects of different doses of cyclophosphamide Chemotherapy and immunotherapy are generally regarded on T cell subsets including CD4+CD25+ T cells in mice. as unrelated or even mutually exclusive in cancer treatment, Cyclophosphamide (20 mg/kg) decreased the numbers of because chemotherapy kills not only target cancer cells but splenocytes, CD4+ and CD8+ T cells by ~50%, while a decline also immune cells, inducing systemic immune suppression in CD4+CD25+ T cell number was more profound, leading to and dampening the therapeutic efficacy of immunotherapy. the remarkably lower ratios of CD4+CD25+ T cells to CD4+ In this regard, however, cyclophosphamide appears an T cells.
    [Show full text]
  • Assessment of Overall Survival, Quality of Life, And
    Supplementary Online Content Salas-Vega S, Iliopoulos O, Mossialos E. Assesment of overall survival, quality of life, and safety benefits associated with new cancer medicines [published online December 29, 2016]. JAMA Oncol. doi:10.1001/jamaoncol.2016.4166 eTable 1. Therapeutic profile of all cancer medicines approved by the FDA between 2003- 2013 eTable 2. Regulatory evidence in support of classification of drug clinical benefits eFigure 1. Number of cancer drugs that were evaluated by all three HTA agencies, sorted by magnitude of clinical benefits eTable 3. Interagency agreement–Krippendorff’s alpha coefficients eMethods This supplementary material has been provided by the authors to give readers additional information about their work. Online-Only Supplement © 2016 American Medical Association. All rights reserved. Downloaded From: https://jamanetwork.com/ on 09/25/2021 Clinical value of cancer medicines Contents eExhibits ......................................................................................................................................................... 3 eTable 1. Therapeutic profile of all cancer medicines approved by the FDA between 2003- 2013 (Summary of eTable 2) ................................................................................................................... 3 eTable 2. Regulatory evidence in support of classification of drug clinical benefits ....................... 6 eFigure 1. Number of cancer drugs that were evaluated by all three HTA agencies, sorted by magnitude of clinical benefits
    [Show full text]
  • Intratumoral Immunotherapy for Early Stage Solid Tumors
    Author Manuscript Published OnlineFirst on February 18, 2020; DOI: 10.1158/1078-0432.CCR-19-3642 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Intratumoral immunotherapy for early-stage solid tumors 2 3 Wan Xing Hong1,2*, Sarah Haebe2,3*, Andrew S. Lee4,5, C. Benedikt Westphalen3,6,7, 4 Jeffrey A. Norton1,2, Wen Jiang8, Ronald Levy2# 5 6 1. Department of Surgery, Stanford University School of Medicine 7 2. Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University 8 3. Department of Medicine III, University Hospital, LMU Munich, Germany 9 4. Department of Pathology, Stanford University School of Medicine 10 5. Shenzhen Bay Labs, Cancer Research Institute, Shenzhen, China 11 6. Comprehensive Cancer Center Munich, Munich, Germany 12 7. Deutsches Konsortium für Translationale Krebsforschung (DKTK), DKTK Partner Site Munich, 13 Germany 14 8. Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, 15 Texas, USA 16 * These authors contributed equally to this work 17 # Correspondence: [email protected]. 18 CCSR 1105, Stanford, California 94305-5151 19 (650) 725-6452 (office) 20 21 Running title: Intratumoral immunotherapy for early-stage solid tumors 22 23 24 Conflict of Interest 25 Dr. Ronald Levy serves on the advisory boards for Five Prime, Corvus, Quadriga, BeiGene, GigaGen, 26 Teneobio, Sutro, Checkmate, Nurix, Dragonfly, Abpro, Apexigen, Spotlight, 47 Inc, XCella, 27 Immunocore, Walking Fish. 28 Dr. Benedikt Westphalen is on the advisory boards for Celgene, Shire/Baxalta Rafael Pharmaceuticals, 29 Redhill and Roche. He receives research support from Roche.
    [Show full text]
  • AN ANTICANCER DRUG COCKTAIL of Three Kinase Inhibitors Improved Response to a DENDRITIC CELL–BASED CANCER VACCINE
    Author Manuscript Published OnlineFirst on July 2, 2019; DOI: 10.1158/2326-6066.CIR-18-0684 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 AN ANTICANCER DRUG COCKTAIL OF Three Kinase Inhibitors Improved Response to a DENDRITIC CELL–BASED CANCER VACCINE Jitao Guo1, Elena Muse1, Allison J. Christians1, Steven J. Swanson2, and Eduardo Davila1, 3, 4 1Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States; 2Immunocellular Therapeutics Ltd., Westlake Village, California, United States; 3 Human Immunology and Immunotherapy Initiative, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States; and 4University of Colorado Comprehensive Cancer Center, Aurora, Colorado, United States. Running Title: REPURPOSING ANTICANCER DRUGS FOR DC CANCER VACCINE Keywords: Dendritic cell; cancer vaccine; MK2206; NU7441; Trametinib Financial supports: This work was supported by R01CA207913, a research award from ImmunoCellular Therapeutics, VA Merit Award BX002142, University of Maryland School of Medicine Marlene and Stewart Greenebaum Comprehensive Cancer Center P30CA134274 and the University of Colorado Denver Comprehensive Cancer Center P30CA046934. Corresponding author: Dr. Eduardo Davila Mail stop 8117,12801 E. 17th Avenue, Aurora, CO 80045 Email: [email protected] Phone: (303) 724-0817; Fax:(303) 724-3889 Conflict of Interest Disclosures: These studies were partly funded by a grant from ImmunoCellular Therapeutics (IMUC). S.S. currently serves as SVP Research IMUC but the remaining authors declare no competing financial interests. Counts: Abstract, 242 words; Manuscript, 4523 words; Figures, 5; References,50. Downloaded from cancerimmunolres.aacrjournals.org on October 2, 2021. © 2019 American Association for Cancer Research.
    [Show full text]
  • Signal Transduction Guide
    Signal Transduction Product Guide | 2007 NEW! Selective T-type Ca2+ channel blockers, NNC 55-0396 and Mibefradil ZM 447439 – Novel Aurora Kinase Inhibitor NEW! Antibodies for Cancer Research EGFR-Kinase Selective Inhibitors – BIBX 1382 and BIBU 1361 DRIVING RESEARCH FURTHER Calcium Signaling Agents ...................................2 G Protein Reagents ...........................................12 Cell Cycle and Apoptosis Reagents .....................3 Ion Channel Modulators ...................................13 Cyclic Nucleotide Related Tools ...........................7 Lipid Signaling Agents ......................................17 Cytokine Signaling Agents ..................................9 Nitric Oxide Tools .............................................19 Enzyme Inhibitors/Substrates/Activators ..............9 Protein Kinase Reagents....................................22 Glycobiology Agents .........................................12 Protein Phosphatase Reagents ..........................33 Neurochemicals | Signal Transduction Agents | Peptides | Biochemicals Signal Transduction Product Guide Calcium Signaling Agents ......................................................................................................................2 Calcium Binding Protein Modulators ...................................................................................................2 Calcium ATPase Modulators .................................................................................................................2 Calcium Sensitive Protease
    [Show full text]
  • In the United States Court of Appeals for the Federal Circuit
    Case: 18-1959 Document: 16 Page: 1 Filed: 08/20/2018 No. 18-1959 In the United States Court of Appeals for the Federal Circuit GENENTECH, INC., APPELLANT v. HOSPIRA, INC., APPELLEE ON APPEAL FROM THE UNITED STATES PATENT AND TRADEMARK OFFICE PATENT TRIAL AND APPEAL BOARD IN NO. IPR2016-01771 BRIEF OF APPELLANT GENENTECH, INC. PAUL B. GAFFNEY ADAM L. PERLMAN THOMAS S. FLETCHER WILLIAMS & CONNOLLY LLP 725 Twelfth Street, N.W. Washington, DC 20005 (202) 434-5000 Case: 18-1959 Document: 16 Page: 2 Filed: 08/20/2018 CERTIFICATE OF INTEREST Pursuant to Federal Circuit Rule 47.4, undersigned counsel for appellant certifies the following: 1. The full name of the party represented by me is Genentech, Inc. 2. The name of the real party in interest represented by me is the same. 3. Genentech, Inc. is a wholly-owned subsidiary of Roche Holdings Inc. Roche Holdings Inc.’s ultimate parent, Roche Holdings Ltd, is a publicly held Swiss corporation traded on the Swiss Stock Exchange. Upon information and belief, more than 10% of Roche Holdings Ltd’s voting shares are held either directly or indirectly by Novartis AG, a publicly held Swiss corporation. 4. The following attorneys appeared for Genentech, Inc. in proceedings below or are expected to appear in this Court and are not already listed on the docket for the current case: Teagan J. Gregory and Christopher A. Suarez of Williams & Connolly LLP, 725 Twelfth Street, N.W., Washington, D.C. 20005. 5. The title and number of any case known to counsel to be pending in this or any other court or agency that will directly affect or be directly affected by this court’s decision in this pending appeal are Genentech, Inc.
    [Show full text]
  • Identification of Candidate Repurposable Drugs to Combat COVID-19 Using a Signature-Based Approach
    www.nature.com/scientificreports OPEN Identifcation of candidate repurposable drugs to combat COVID‑19 using a signature‑based approach Sinead M. O’Donovan1,10, Ali Imami1,10, Hunter Eby1, Nicholas D. Henkel1, Justin Fortune Creeden1, Sophie Asah1, Xiaolu Zhang1, Xiaojun Wu1, Rawan Alnafsah1, R. Travis Taylor2, James Reigle3,4, Alexander Thorman6, Behrouz Shamsaei4, Jarek Meller4,5,6,7,8 & Robert E. McCullumsmith1,9* The COVID‑19 pandemic caused by the novel SARS‑CoV‑2 is more contagious than other coronaviruses and has higher rates of mortality than infuenza. Identifcation of efective therapeutics is a crucial tool to treat those infected with SARS‑CoV‑2 and limit the spread of this novel disease globally. We deployed a bioinformatics workfow to identify candidate drugs for the treatment of COVID‑19. Using an “omics” repository, the Library of Integrated Network‑Based Cellular Signatures (LINCS), we simultaneously probed transcriptomic signatures of putative COVID‑19 drugs and publicly available SARS‑CoV‑2 infected cell lines to identify novel therapeutics. We identifed a shortlist of 20 candidate drugs: 8 are already under trial for the treatment of COVID‑19, the remaining 12 have antiviral properties and 6 have antiviral efcacy against coronaviruses specifcally, in vitro. All candidate drugs are either FDA approved or are under investigation. Our candidate drug fndings are discordant with (i.e., reverse) SARS‑CoV‑2 transcriptome signatures generated in vitro, and a subset are also identifed in transcriptome signatures generated from COVID‑19 patient samples, like the MEK inhibitor selumetinib. Overall, our fndings provide additional support for drugs that are already being explored as therapeutic agents for the treatment of COVID‑19 and identify promising novel targets that are worthy of further investigation.
    [Show full text]
  • Combined BRAF and MEK Inhibition with Vemurafenib and Cobimetinib for Patients with Advanced Melanoma
    Review Melanoma Combined BRAF and MEK Inhibition with Vemurafenib and Cobimetinib for Patients with Advanced Melanoma Antonio M Grimaldi, Ester Simeone, Lucia Festino, Vito Vanella and Paolo A Ascierto Melanoma, Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”, Napoli, Italy cquired resistance is the most common cause of BRAF inhibitor monotherapy treatment failure, with the majority of patients experiencing disease progression with a median progression-free survival of 6-8 months. As such, there has been considerable A focus on combined therapy with dual BRAF and MEK inhibition as a means to improve outcomes compared with monotherapy. In the COMBI-d and COMBI-v trials, combined dabrafenib and trametinib was associated with significant improvements in outcomes compared with dabrafenib or vemurafenib monotherapy, in patients with BRAF-mutant metastatic melanoma. The combination of vemurafenib and cobimetinib has also been investigated. In the phase III CoBRIM study in patients with unresectable stage III-IV BRAF-mutant melanoma, treatment with vemurafenib and cobimetinib resulted in significantly longer progression-free survival and overall survival (OS) compared with vemurafenib alone. One-year OS was 74.5% in the vemurafenib and cobimetinib group and 63.8% in the vemurafenib group, while 2-year OS rates were 48.3% and 38.0%, respectively. The combination was also well tolerated, with a lower incidence of cutaneous squamous-cell carcinoma and keratoacanthoma compared with monotherapy. Dual inhibition of both MEK and BRAF appears to provide a more potent and durable anti-tumour effect than BRAF monotherapy, helping to prevent acquired resistance as well as decreasing adverse events related to BRAF inhibitor-induced activation of the MAPK-pathway.
    [Show full text]
  • Dangerous Liaisons: Flirtations Between Oncogenic BRAF and GRP78 in Drug-Resistant Melanomas
    Dangerous liaisons: flirtations between oncogenic BRAF and GRP78 in drug-resistant melanomas Shirish Shenolikar J Clin Invest. 2014;124(3):973-976. https://doi.org/10.1172/JCI74609. Commentary BRAF mutations in aggressive melanomas result in kinase activation. BRAF inhibitors reduce BRAFV600E tumors, but rapid resistance follows. In this issue of the JCI, Ma and colleagues report that vemurafenib activates ER stress and autophagy in BRAFV600E melanoma cells, through sequestration of the ER chaperone GRP78 by the mutant BRAF and subsequent PERK activation. In preclinical studies, treating vemurafenib-resistant melanoma with a combination of vemurafenib and an autophagy inhibitor reduced tumor load. Further work is needed to establish clinical relevance of this resistance mechanism and demonstrate efficacy of autophagy and kinase inhibitor combinations in melanoma treatment. Find the latest version: https://jci.me/74609/pdf commentaries F2-2012–279017) and NEUROKINE net- from the brain maintains CNS immune tolerance. 17. Benner EJ, et al. Protective astrogenesis from the J Clin Invest. 2014;124(3):1228–1241. SVZ niche after injury is controlled by Notch mod- work (EU Framework 7 ITN project). 8. Sawamoto K, et al. New neurons follow the flow ulator Thbs4. Nature. 2013;497(7449):369–373. of cerebrospinal fluid in the adult brain. Science. 18. Sabelstrom H, et al. Resident neural stem cells Address correspondence to: Gianvito Mar- 2006;311(5761):629–632. restrict tissue damage and neuronal loss after spinal tino, Neuroimmunology Unit, Institute 9. Butti E, et al. Subventricular zone neural progeni- cord injury in mice. Science. 2013;342(6158):637–640. tors protect striatal neurons from glutamatergic 19.
    [Show full text]