1
|
Gentilini A, Neez E, Wong-Rieger D. Rare Disease Policy in High-Income Countries: An Overview of Achievements, Challenges, and Solutions. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2025; 28:680-685. [PMID: 39880194 DOI: 10.1016/j.jval.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 12/10/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025]
Abstract
OBJECTIVES To provide an overview of policy initiatives in high-income countries aimed at supporting the development and accessibility of treatments for rare diseases. METHODS We examine how legislative, research, and pricing policies in high-income countries address barriers that have historically hindered innovation and access to rare disease treatments. By analyzing examples from the European Union, United Kingdom, United States, Canada, Japan, and Australia, the article identifies ongoing initiatives, outlines current challenges, and explores proposed solutions to foster a sustainable, innovative, and accessible rare disease treatment ecosystem. RESULTS The review highlights policies such as legislative incentives in the European Union, United States, and Japan for orphan drug development, public-private partnerships to boost innovation, and patient registries to support research and clinical trials. Despite these efforts, major challenges persist, including high therapy costs, limited access to innovation for ultrarare diseases, and diagnostic delays, with significant disparities across regions. CONCLUSIONS Overcoming these challenges will require sustainable pricing and reimbursement frameworks, alongside stronger collaboration between stakeholders, particularly for ultrarare diseases. Advanced technologies, such as artificial intelligence, hold promise for improving diagnostic accuracy and data collection, supported by enhanced coding systems and registries to facilitate more robust research.
Collapse
Affiliation(s)
- Arianna Gentilini
- Department of Health Policy, London School of Economics and Political Science, London, England, UK; Department of Economics and Public Policy, Imperial College London, England, UK.
| | | | | |
Collapse
|
2
|
Nijim S, Korsunska A, Zinski J, Bolden SE, Zuccato M, Repasky M, Fajgenbaum DC. Rare Disease Drug Repurposing. JAMA Netw Open 2025; 8:e258330. [PMID: 40323602 PMCID: PMC12053540 DOI: 10.1001/jamanetworkopen.2025.8330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 03/01/2025] [Indexed: 05/08/2025] Open
Abstract
Importance Treatments are urgently needed for the more than 9500 rare diseases with no US Food and Drug Administration-approved therapies. Although repurposing can be less time- and cost-intensive compared with novel drug development, hurdles have impeded systematic repurposing. Rare disease nonprofit organizations (RDNPs) are well-positioned to overcome barriers and have spearheaded rare disease repurposing efforts for decades. However, no comprehensive data are available on the state of rare disease repurposing or features of successful efforts. Objective To characterize the state of rare disease drug repurposing, identify factors associated with successful outcomes, and share thematic insights using the interactive Repurposing of All Drugs, Mapping All Paths (ROADMAP) Project web tool. Design, Setting, and Participants The ROADMAP study was a qualitative study using a mixed-methods analysis of US-based RDNP leaders and their stakeholders, including a national survey and semistructured interviews of RDNP leaders, conducted from September 29, 2021, to January 6, 2022. Surveys and interviews revealed themes associated with RDNP strategies, timelines, and support mechanisms. Data were analyzed from January 22, 2024, to April 23, 2024. Main Outcomes and Measures The primary survey outcome was the repurposing project stage (abandoned, early, clinical, late, or successful). Qualitative outcomes included themes characterizing repurposing experiences. Two random forest models of drug- and disease- specific as well as organization-specific variables were used to evaluate factor importance toward inferring the project stage. Orthogonal significance testing was conducted using Spearman rank correlation, and P values in each model were corrected for multiple hypothesis testing using a Benjamini-Hochberg procedure. Results Representative organizations submitted survey responses, including 147 of 698 potential US-based RDNPs. The median RDNP age was 10 years (IQR, 5-20 years), and the median annual revenue was $355 390 (IQR, $90 028-$946 108). Among 34 leaders who were interviewed, representing 25 RDNPs, 23 were female (67.6%), and the RDNPs had a median age of 15 years (IQR, 6-19 years) and a median revenue of $670 719 (IQR, $193 587-$1 830 890). Among the surveyed RDNPs, 58 of 138 (42.0%) specifically identifying their involvement in repurposing supported repurposing projects, and 94 drugs were in various stages of repurposing, of which 23 met success criteria (5 with US Food and Drug Administration approval and 18 with off-label use with subjective benefit). Survey factors associated with successful outcomes included nonprofit-supported patient recruitment into trials (Gini importance, 3.90; ρ = 0.50; adjusted P < .001) and provision of nonfinancial research support (Gini importance, 0.69; ρ = 0.33; adjusted P = .02). Interview themes were synthesized into a 5-stage repurposing framework with roadblocks and recommendations that included (1) enabling drug repurposing, (2) identifying a drug therapy, (3) validating a drug therapy, (4) clinical use and testing, and (5) reaching an optimal end point for clinical practice. Conclusions and Relevance The findings of this qualitative study of RDNP repurposing suggest that several opportunities were associated with successful outcomes and can be considered to optimize systematic repurposing among RDNPs, external collaborators, and policymakers with the use of an interactive tool showcasing insights to facilitate data-driven drug repurposing.
Collapse
Affiliation(s)
- Sally Nijim
- Castleman Disease Collaborative Network, Paso Robles, California
- Center for Cytokine Storm Treatment and Laboratory, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Ania Korsunska
- Castleman Disease Collaborative Network, Paso Robles, California
| | - Joseph Zinski
- Center for Cytokine Storm Treatment and Laboratory, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Sarah E. Bolden
- School of Information Studies, Syracuse University, Syracuse, New York
| | - Mary Zuccato
- Castleman Disease Collaborative Network, Paso Robles, California
| | - Mileva Repasky
- Castleman Disease Collaborative Network, Paso Robles, California
| | - David C. Fajgenbaum
- Castleman Disease Collaborative Network, Paso Robles, California
- Center for Cytokine Storm Treatment and Laboratory, University of Pennsylvania Perelman School of Medicine, Philadelphia
| |
Collapse
|
3
|
Nishimura S, Ma C, Sidransky E, Ryan E. Obstacles to Early Diagnosis of Gaucher Disease. Ther Clin Risk Manag 2025; 21:93-101. [PMID: 39882275 PMCID: PMC11776414 DOI: 10.2147/tcrm.s388266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/11/2025] [Indexed: 01/31/2025] Open
Abstract
Gaucher disease (GD) is a rare lysosomal storage disorder resulting from a deficiency of the lysosomal enzyme glucocerebrosidase caused by biallelic variants in the GBA1 gene. Patients may present with a wide spectrum of disease manifestations, including hepatosplenomegaly, thrombocytopenia, bone manifestations, and in the case of GD types 2 and 3, neurodegeneration, cognitive delay, and/or oculomotor abnormalities. While there is no treatment for neuronopathic GD, non-neuronopathic manifestations can be efficiently managed with enzyme replacement therapy or substrate reduction therapy. However, many patients with GD experience a lengthy diagnostic odyssey, which can negatively affect their access to care and clinical outcomes. The cause of this diagnostic delay is multifaceted. Since genotype/phenotype correlations in GD are not always clear, it is difficult to predict the presence, severity, and onset of clinical manifestations. This heterogeneity, combined with the molecular complexity of the GBA1 locus, low disease prevalence, and limited knowledge of GD among providers serves as a barrier to early diagnosis of GD. In this review, we discuss such obstacles and challenges, considerations, and future steps toward improving the diagnostic journey for patients with GD.
Collapse
Affiliation(s)
- Samantha Nishimura
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charis Ma
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ellen Sidransky
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emory Ryan
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Scholte M, Grimm SE, Pauly B, Verbeeck F, Pasmooij AMG, Bouma B, van Duijn-Wiersma J, Guney E, Kesselheim AS, Schmidt HHHW, Joore MA. Revising EU pharmaceutical legislation: will it foster drug repurposing? Drug Discov Today 2025; 30:104286. [PMID: 39756647 DOI: 10.1016/j.drudis.2024.104286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
Repurposing off-patent drugs can be a potential source of low-cost treatments for patients with unmet medical needs. Here, we review the proposed new European Union (EU) pharmaceutical legislation in which two articles address drug repurposing. We find certain barriers hindering the adoption of these new incentives by academic and not-for-profit stakeholders, including lack of knowledge on regulatory aspects, pharmacovigilance, and restrictions in data protection. To further empower the intended stakeholders of the legislation, these initiatives can be strengthened by creating additional scientific, regulatory, and health technology assessment (HTA) support for not-for-profit repurposers, and by determining fair data protection periods and pricing policies. To support drug repurposing, Europe should work toward a comprehensive drug-repurposing strategy that fosters the repurposing of generic, shelved, and protected drugs.
Collapse
Affiliation(s)
- Mirre Scholte
- Department of Clinical Epidemiology & Medical Technology Assessment (KEMTA), Maastricht University Medical Centre, Maastricht, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands; Departments of IQ Health and Radiology, Radboudumc, Nijmegen, the Netherlands; REPO4EU consortium.
| | - Sabine E Grimm
- Department of Clinical Epidemiology & Medical Technology Assessment (KEMTA), Maastricht University Medical Centre, Maastricht, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands; REPO4EU consortium
| | - Bianca Pauly
- REPO4EU consortium; Regulatory Affairs Department, 3D-PharmXchange, Tilburg, the Netherlands
| | | | - Anna M G Pasmooij
- Dutch Medicines Evaluation Board, Utrecht, the Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Barend Bouma
- NLO - N.V. Nederlandsch Octrooibureau, the Netherlands
| | | | - Emre Guney
- REPO4EU consortium; Discovery and Data Science (DDS) Unit, STALICLA SL, Barcelona, Spain
| | - Aaron S Kesselheim
- REPO4EU consortium; Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Harald H H W Schmidt
- REPO4EU consortium; Department of Pharmacology and Personalised Medicine, Maastricht University, Maastricht, the Netherlands
| | - Manuela A Joore
- Department of Clinical Epidemiology & Medical Technology Assessment (KEMTA), Maastricht University Medical Centre, Maastricht, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands; REPO4EU consortium
| |
Collapse
|
5
|
Saha S, Chatterjee P, Nasipuri M, Basu S, Chakraborti T. Computational drug repurposing for viral infectious diseases: a case study on monkeypox. Brief Funct Genomics 2024; 23:570-578. [PMID: 38183212 DOI: 10.1093/bfgp/elad058] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024] Open
Abstract
The traditional method of drug reuse or repurposing has significantly contributed to the identification of new antiviral compounds and therapeutic targets, enabling rapid response to developing infectious illnesses. This article presents an overview of how modern computational methods are used in drug repurposing for the treatment of viral infectious diseases. These methods utilize data sets that include reviewed information on the host's response to pathogens and drugs, as well as various connections such as gene expression patterns and protein-protein interaction networks. We assess the potential benefits and limitations of these methods by examining monkeypox as a specific example, but the knowledge acquired can be applied to other comparable disease scenarios.
Collapse
Affiliation(s)
- Sovan Saha
- Department of Computer Science and Engineering (Artificial Intelligence and Machine Learning), Techno Main Salt Lake, EM-4/1, Sector V, Bidhannagar, Kolkata, West Bengal 700091, India
| | - Piyali Chatterjee
- Department of Computer Science and Engineering, Netaji Subhash Engineering College, Garia, Kolkata-700152, India
| | - Mita Nasipuri
- Department of Computer Science and Engineering, Jadavpur University, Kolkata - 700032, India
| | - Subhadip Basu
- Department of Computer Science and Engineering, Jadavpur University, Kolkata - 700032, India
| | - Tapabrata Chakraborti
- Department of Medical Physics and Biomedical Engineering, University College London, UK
- Health Science Programme, The Alan Turing Institute, London, UK
- Linacre College, University of Oxford, UK
| |
Collapse
|
6
|
Shukla M, Malik S, Pandya A. Lab on chip for testing of repurposed drugs. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 205:71-90. [PMID: 38789187 DOI: 10.1016/bs.pmbts.2024.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
The lab-on-chip technique broadly comprises of microfluidics and aims to progress multidimensionally by changing the outlook of medicine and pharmaceuticals as it finds it roots in miniaturization. Moreover, microfluidics facilitates precise physiological simulation and possesses biological system-mimicking capabilities for drug development and repurposing. Thus, organs on chip could pave a revolutionary pathway in the field of drug development and repurposing by reducing animal testing and improving drug repurposing.
Collapse
Affiliation(s)
- Malvika Shukla
- Department of Biotechnology and Bioengineering, Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Saloni Malik
- Department of Biotechnology and Bioengineering, Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Alok Pandya
- Department of Biotechnology and Bioengineering, Institute of Advanced Research, Gandhinagar, Gujarat, India; Department of Nanoengineering, University of California San Diego, La Jolla, CA, United States.
| |
Collapse
|
7
|
Morris R, Ali R, Cheng F. Drug Repurposing Using FDA Adverse Event Reporting System (FAERS) Database. Curr Drug Targets 2024; 25:454-464. [PMID: 38566381 DOI: 10.2174/0113894501290296240327081624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
Drug repurposing is an emerging approach to reassigning existing pre-approved therapies for new indications. The FDA Adverse Event Reporting System (FAERS) is a large database of over 28 million adverse event reports submitted by medical providers, patients, and drug manufacturers and provides extensive drug safety signal data. In this review, four common drug repurposing strategies using FAERS are described, including inverse signal detection for a single disease, drug-drug interactions that mitigate a target ADE, identifying drug-ADE pairs with opposing gene perturbation signatures and identifying drug-drug pairs with congruent gene perturbation signatures. The purpose of this review is to provide an overview of these different approaches using existing successful applications in the literature. With the fast expansion of adverse drug event reports, FAERS-based drug repurposing represents a promising strategy for discovering new uses for existing therapies.
Collapse
Affiliation(s)
- Robert Morris
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL33612, USA
- Department of Biostatistics and Epidemiology, College of Public Health, University of South Florida, Tampa, FL33612, USA
| | - Rahinatu Ali
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL33612, USA
| | - Feng Cheng
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL33612, USA
- Department of Biostatistics and Epidemiology, College of Public Health, University of South Florida, Tampa, FL33612, USA
| |
Collapse
|
8
|
Lexchin J. Therapeutic Benefit From New Drugs From Pharmaceutical Companies. JAMA Intern Med 2024; 184:52-53. [PMID: 37983036 DOI: 10.1001/jamainternmed.2023.6256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Affiliation(s)
- Joel Lexchin
- School of Health Policy and Management, York University, Toronto, Ontario, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Alonso Ruiz A, Large K, Moon S, Vieira M. Pharmaceutical policy and innovation for rare diseases: A narrative review. F1000Res 2023; 12:211. [PMID: 38778810 PMCID: PMC11109548 DOI: 10.12688/f1000research.130809.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2023] [Indexed: 05/25/2024] Open
Abstract
This article aims to synthesize the existing literature on the implementation of public policies to incentivize the development of treatments for rare diseases, (diseases with very low prevalence and therefore with low commercial interest) otherwise known as orphan drugs. The implementation of these incentives in the United States (US), Japan, and in the European Union (EU) seems to be related to a substantial increase in treatments for these diseases, and has influenced the way the pharmaceutical research & development (R&D) system operates beyond this policy area. Despite the success of the Orphan Drug model, the academic literature also highlights the negative implications that these public policies have on affordability and access to orphan drugs, as well as on the prioritization of certain disease rare areas over others. The synthesis focuses mostly on the United States' Orphan Drug Act (ODA) as a model for subsequent policies in other regions and countries. It starts with a historical overview of the creation of the term "rare diseases", continues with a summary of the evidence available on the US ODA's positive and negative impacts, and provides a summary of the different proposals to reform these incentives in light of the negative outcomes described. Finally, it describes some key aspects of the Japanese and European policies, as well as some of the challenges captured in the literature related to their impact in Low- and Middle-Income Countries (LMICs).
Collapse
Affiliation(s)
- Adrián Alonso Ruiz
- Global Health Centre, Graduate Institute of International and Development Studies, Geneva, 1211, Switzerland
| | - Kaitlin Large
- Global Health Centre, Graduate Institute of International and Development Studies, Geneva, 1211, Switzerland
| | - Suerie Moon
- Global Health Centre, Graduate Institute of International and Development Studies, Geneva, 1211, Switzerland
| | - Marcela Vieira
- Global Health Centre, Graduate Institute of International and Development Studies, Geneva, 1211, Switzerland
| |
Collapse
|
10
|
Sánchez Fernández I, Gaínza-Lein M, Amengual-Gual M, Barcia Aguilar C, Romeu A, Torres A, Jonas R, Douglass LM. Evolution of antiseizure medication use and cost in the United States of America 2006-2021. Seizure 2023; 112:128-138. [PMID: 37832279 DOI: 10.1016/j.seizure.2023.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/15/2023] Open
Abstract
OBJECTIVE To describe the evolution in use and cost of antiseizure medications (ASM) in the United States of America (USA). METHODS Retrospective descriptive study using the IBM MarketScan Commercial Database (data of privately-insured patients) for the years 2006 to 2021. We identified patients with epilepsy who were on ASM. We adjusted cost for inflation with the Gross Domestic Product Implicit Price Deflator. RESULTS We evaluated 347,158 patients (46.9 % males; median (p25-p75) age: 33 (17-49) years; 28 % with pediatric-onset epilepsy and 72 % with adult-onset epilepsy) with a total of 1,385,382 person-years and 588,285,065 ASM prescription days. The most commonly prescribed (as percentage of prescription days) ASMs in 2006 were valproate (18 %) and lamotrigine (17 %) in pediatric-onset epilepsy and phenytoin (21 %) and carbamazepine (17 %) in adult-onset epilepsy, but starting in the 2010s, levetiracetam and lamotrigine became the most commonly prescribed ASMs in both pediatric-onset (in 2021, levetiracetam 25 %, lamotrigine 16 %) and adult-onset (in 2021, levetiracetam 27 %, lamotrigine 20 %) epilepsy. The proportion of generic ASM use increased 3.6-fold: from 23 % of prescription days in 2006 to 83 % of prescription days in 2021. The median (p25-p75) average wholesale price (AWP) per person-year increased by 102 % from $2,684 ($990-$5,509) in 2006 to $5,417 ($2,858-$12,310) in 2021. The increases were greater in absolute terms for brand-name ASMs by 419 %: $3,109 ($1,564-$5,068 in 2006 and $16,149 ($12,950-$23,377) in 2021 than for generic ASMs by 462 %: $699 ($457-$1,678) in 2006 and $3,931 ($2,618-$6,081) in 2021. The costs directly borne by the patient (copay, coinsurance, deductibles, and pharmacy processing fees) increased by 69 % for brand-name ASMs from $393 ($246-$570) in 2006 to $665 ($335-$1,308) in 2021, but decreased by 37 % for generic ASMs from $147 ($98-$213) in 2006 to $92 ($51-$141) in 2021. CONCLUSIONS The median cost of ASMs per person-year approximately doubled from 2006 to 2021. The increase in use of generic ASMs probably helped buffer the growing costs of ASMs. However, generic ASMs already represent 83 % of prescription days in 2021, with limited room to further contain costs by just increasing the proportion of generics.
Collapse
Affiliation(s)
- Iván Sánchez Fernández
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, The Boston University Chobanian and Avedisian School of Medicine, USA.
| | - Marina Gaínza-Lein
- Instituto de Pediatría, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile; Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marta Amengual-Gual
- Pediatric Neurology Unit, Department of Pediatrics, Hospital Universitari Son Llàtzer, Universitat de les Illes Balears, Palma, Spain
| | - Cristina Barcia Aguilar
- Department of Child Neurology, Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Amanda Romeu
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, The Boston University Chobanian and Avedisian School of Medicine, USA
| | - Alcy Torres
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, The Boston University Chobanian and Avedisian School of Medicine, USA
| | - Rinat Jonas
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, The Boston University Chobanian and Avedisian School of Medicine, USA
| | - Laurie M Douglass
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, The Boston University Chobanian and Avedisian School of Medicine, USA
| |
Collapse
|
11
|
Mandal N, Rath SL. Identification of inhibitors against SARS-CoV-2 variants of concern using virtual screening and metadynamics-based enhanced sampling. Chem Phys 2023; 573:111995. [PMID: 37342284 PMCID: PMC10265933 DOI: 10.1016/j.chemphys.2023.111995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/12/2023] [Accepted: 06/11/2023] [Indexed: 06/22/2023]
Abstract
Among the variants of SARS-CoV-2, some are more infectious than the Wild-type. Interestingly, these mutations enable the virus to evade the therapeutic efforts. Hence, there is a need for candidate drug molecules that can potently bind with all the variants. We have adopted a strategy combining virtual screening, molecular docking followed by rigorous sampling by metadynamics simulations to find candidate molecules. From our results we found four highly potent drug candidates that can bind to the Spike-RBD of all the variants of the virus. Additionally, we also found that certain signature residues on the RBM region commonly bind to each of these inhibitors. Thus, our study not only gives information on the chemical compounds, but also residues on the proteins which could be targeted for future drug and vaccine development studies.
Collapse
Affiliation(s)
- Nabanita Mandal
- Department of Biotechnology, National Institute of Technology Warangal, Telangana, India
| | - Soumya Lipsa Rath
- Department of Biotechnology, National Institute of Technology Warangal, Telangana, India
| |
Collapse
|
12
|
Moir J, Aggarwal S, Skondra D. Repurposing medications for treatment of age-related macular degeneration: Insights from novel approaches to data mining. Exp Biol Med (Maywood) 2023; 248:798-810. [PMID: 37452694 PMCID: PMC10468640 DOI: 10.1177/15353702231181188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
The economic and visual burdens associated with age-related macular degeneration (AMD) are expected to significantly increase in the coming years. As of now, interventions to delay or prevent AMD are limited. Hence, there is an urgent and unmet need to expand our therapeutic tools for AMD in a manner, that is, both efficient and cost-effective. In this review, we consider the idea of drug repurposing, in which existing medications with other indications can be re-imagined for treating AMD. We detail the results of several population-level studies that have shown associations between several candidates and decreased risk of AMD development or progression. Such candidates include the more extensively studied metformin and statins, in addition to recently identified candidates fluoxetine and l-DOPA (levodopa) that show promise. We then briefly explore results from an advanced bioinformatics study, which provides further evidence that existing medications are associated with AMD risk genes. Many of these candidates warrant further study in prospective, clinical trials, where their potential causal relationships with AMD can be thoroughly assessed.
Collapse
Affiliation(s)
- John Moir
- Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sarthak Aggarwal
- Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Dimitra Skondra
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
13
|
Ou SL, Luo J, Wei H, Qin XL, Jiang Q. Value assessment of PD-1/PD-L1 inhibitors in the treatment of oesophageal and gastrointestinal cancers. Front Pharmacol 2023; 14:1106961. [PMID: 37153768 PMCID: PMC10160363 DOI: 10.3389/fphar.2023.1106961] [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: 11/24/2022] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
Background: Evidence of efficacy and safety of programmed cell death 1 (PD-1) and programmed death ligand-1 (PD-L1) checkpoint inhibitors in oesophageal cancer (EC), gastric cancer (GC) and colorectal cancer (CRC) was inconsistent, obscuring their clinical application and decision-making. The aim of this study was to comprehensively evaluate the value of PD-1/PD-L1 inhibitors in EC, GC and CRC to select valuable PD-1/PD-L1 inhibitors, and to assess the association between the value and cost of PD-1/PD-L1 inhibitors. Methods: A comprehensive search of trials of PD-1/PD-L1 inhibitors in EC, GC and CRC was performed in Chinese and English medical databases with a cut-off date of 1 July 2022. Two authors independently applied the ASCO-VF and ESMO-MCBS to assess the value of PD-1/PD-L1 inhibitors. A receiver operating characteristic (ROC) curve was generated to establish the predictive value of the ASCO-VF score to meet the threshold of the ESMO-MCBS grade. Spearman's correlation was used to calculate the relationship between the cost and value of drugs. Results: Twenty-three randomized controlled trials were identified: ten (43.48%) in EC, five (21.74%) in CRC, and eight (34.78%) in GC or gastroesophageal junction cancer (GEJC). For advanced diseases, ASCO-VF scores ranged from -12.5 to 69, with a mean score of 26.5 (95% CI 18.4-34.6). Six (42.9%) therapeutic regimens met the ESMO-MCBS benefit threshold grade. The area under the ROC curve was 1.0 (p = 0.002). ASCO-VF scores and incremental monthly cost were negatively correlated (Spearman's ρ = -0.465, p = 0.034). ESMO-MCBS grades and incremental monthly cost were negatively correlated (Spearman's ρ = -0.211, p = 0.489). Conclusion: PD-1/PD-L1 inhibitors did not meet valuable threshold in GC/GEJC. Pembrolizumab met valuable threshold in advanced microsatellite instability-high CRC. The value of camrelizumab and toripalimab may be more worth paying in EC.
Collapse
Affiliation(s)
- Shun-Long Ou
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Luo
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hua Wei
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
| | - Xiao-Li Qin
- Department of Pharmacy, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Qian Jiang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Qian Jiang,
| |
Collapse
|
14
|
Krishnamurthy N, Grimshaw AA, Axson SA, Choe SH, Miller JE. Drug repurposing: a systematic review on root causes, barriers and facilitators. BMC Health Serv Res 2022; 22:970. [PMID: 35906687 PMCID: PMC9336118 DOI: 10.1186/s12913-022-08272-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Repurposing is a drug development strategy receiving heightened attention after the Food and Drug Administration granted emergency use authorization of several repurposed drugs to treat Covid-19. There remain knowledge gaps on the root causes, facilitators and barriers for repurposing. METHOD This systematic review used controlled vocabulary and free text terms to search ABI/Informa, Academic Search Premier, Business Source Complete, Cochrane Library, EconLit, Google Scholar, Ovid Embase, Ovid Medline, Pubmed, Scopus, and Web of Science Core Collection databases for the characteristics, reasons and example of companies deprioritizing development of promising drugs and barriers, facilitators and examples of successful re-purposing. RESULTS We identified 11,814 articles, screened 5,976 for relevance, found 437 eligible for full text review, 115 of which were included in full analysis. Most articles (66%, 76/115) discussed why promising drugs are abandoned, with lack of efficacy or superiority to other therapies (n = 59), strategic business reasons (n = 35), safety problems (n = 28), research design decisions (n = 12), the complex nature of a studied disease or drug (n = 7) and regulatory bodies requiring more information (n = 2) among top reasons. Key barriers to repurposing include inadequate resources (n = 42), trial data access and transparency around abandoned compounds (n = 20) and expertise (n = 11). Additional barriers include uncertainty about the value of repurposing (n = 13), liability risks (n = 5) and intellectual property (IP) challenges (n = 26). Facilitators include the ability to form multi-partner collaborations (n = 38), access to compound databases and database screening tools (n = 32), regulatory modifications (n = 5) and tax incentives (n = 2). CONCLUSION Promising drugs are commonly shelved due to insufficient efficacy or superiority to alternate therapies, poor market prospects, and industry consolidation. Inadequate resources and data access and challenges negotiating IP are key barriers to repurposing reaching its full potential as a core approach in drug development. Multi-partner collaborations and the availability and use of compound databases and tax incentives are key facilitators for repurposing. More research is needed on the current value of repurposing in drug development and how to better facilitate resources to support it, where valuable, especially financial, staffing for out-licensing shelved products, and legal expertise to negotiate IP agreements in multi-partner collaborations. TRIAL REGISTRATION The protocol was registered on Open Science Framework ( https://osf.io/f634k/ ) as it was not eligible for registration on PROSPERO as the review did not focus on a health-related outcome.
Collapse
Affiliation(s)
- Nithya Krishnamurthy
- Internal Medicine Department, Yale University School of Medicine, 367 Cedar Street, 4th Floor, New Haven, CT, 06520, USA
| | - Alyssa A Grimshaw
- Cushing/Whitney Medical Library, Yale University, 333 Cedar Street, Box 208014, New Haven, CT, 06520, USA
| | - Sydney A Axson
- Internal Medicine Department, Yale University School of Medicine, 367 Cedar Street, 4th Floor, New Haven, CT, 06520, USA
| | - Sung Hee Choe
- Milken Institute Center for Faster Cures, 730 15th Street NW, Washington, DC, 20005, USA
| | - Jennifer E Miller
- Internal Medicine Department, Yale University School of Medicine, 367 Cedar Street, 4th Floor, New Haven, CT, 06520, USA.
| |
Collapse
|
15
|
European patent protection for medical uses of known products and drug repurposing. Nat Biotechnol 2022; 40:465-471. [PMID: 35418638 DOI: 10.1038/s41587-022-01269-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
Affiliation(s)
- Robert Cook-Deegan
- School for the Future of Innovation in Society and Consortium for Science, Policy & Outcomes, College of Global Futures, Arizona State University, Washington, DC
| | - Aaron S Kesselheim
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ameet Sarpatwari
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
17
|
Shah S, Dooms MM, Amaral-Garcia S, Igoillo-Esteve M. Current Drug Repurposing Strategies for Rare Neurodegenerative Disorders. Front Pharmacol 2022; 12:768023. [PMID: 34992533 PMCID: PMC8724568 DOI: 10.3389/fphar.2021.768023] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Rare diseases are life-threatening or chronically debilitating low-prevalent disorders caused by pathogenic mutations or particular environmental insults. Due to their high complexity and low frequency, important gaps still exist in their prevention, diagnosis, and treatment. Since new drug discovery is a very costly and time-consuming process, leading pharmaceutical companies show relatively low interest in orphan drug research and development due to the high cost of investments compared to the low market return of the product. Drug repurposing–based approaches appear then as cost- and time-saving strategies for the development of therapeutic opportunities for rare diseases. In this article, we discuss the scientific, regulatory, and economic aspects of the development of repurposed drugs for the treatment of rare neurodegenerative disorders with a particular focus on Huntington’s disease, Friedreich’s ataxia, Wolfram syndrome, and amyotrophic lateral sclerosis. The role of academia, pharmaceutical companies, patient associations, and foundations in the identification of candidate compounds and their preclinical and clinical evaluation will also be discussed.
Collapse
Affiliation(s)
- Sweta Shah
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | |
Collapse
|
18
|
Simoens S, Huys I. How much do the public sector and the private sector contribute to biopharmaceutical R&D? Drug Discov Today 2021; 27:939-945. [PMID: 34863932 DOI: 10.1016/j.drudis.2021.11.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/11/2021] [Accepted: 11/25/2021] [Indexed: 11/03/2022]
Abstract
When examining the prices of new medicines, the question of how much the private and public sectors have contributed to their R&D is often raised. Contributions can be assessed in terms of the investment, authorship of publications, marketing authorizations and intellectual property rights associated with biopharmaceutical R&D. This review of the empirical evidence underlines the complementary and interwoven nature of the private and public sectors in supporting biopharmaceutical R&D. Both sectors invest in and contribute to biopharmaceutical R&D, with the public sector predominantly focusing on basic research and the private sector mainly targeting medicine discovery and development. Public-sector investment generates additional private-sector investment.
Collapse
Affiliation(s)
- Steven Simoens
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium.
| | - Isabelle Huys
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| |
Collapse
|
19
|
α-Lipoic Acid Exerts Its Antiviral Effect against Viral Hemorrhagic Septicemia Virus (VHSV) by Promoting Upregulation of Antiviral Genes and Suppressing VHSV-Induced Oxidative Stress. Virol Sin 2021; 36:1520-1531. [PMID: 34510367 PMCID: PMC8435143 DOI: 10.1007/s12250-021-00440-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/28/2021] [Indexed: 12/03/2022] Open
Abstract
Viral hemorrhagic septicemia virus (VHSV), belonging to the genus Novirhabdovirus, Rhabdoviridae family, is a causative agent of high mortality in fish and has caused significant losses to the aquaculture industry. Currently, no effective vaccines, Food and Drug Administration-approved inhibitors, or other therapeutic intervention options are available against VHSV. α-Lipoic Acid (LA), a potent antioxidant, has been proposed to have antiviral effects against different viruses. In this study, LA (CC50 = 472.6 μmol/L) was repurposed to exhibit antiviral activity against VHSV. In fathead minnow cells, LA significantly increased the cell viability post-VHSV infection (EC50 = 42.7 μmol/L), and exerted a dose-dependent inhibitory effect on VHSV induced-plaque, cytopathic effects, and VHSV glycoprotein expression. The time-of-addition assay suggested that the antiviral activity of LA occurred at viral replication stage. Survival assay revealed that LA could significantly upregulated the survival rate of VHSV-infected largemouth bass in both co-injection (38.095% vs. 1.887%, P < 0.01) and post-injection manner (38.813% vs. 8.696%, P < 0.01) compared with the control group. Additional comparative transcriptome and qRT-PCR analysis revealed LA treatment upregulated the expression of several antiviral genes, such as IRF7, Viperin, and ISG15. Moreover, LA treatment reduced VHSV-induced reactive oxygen species production in addition to Nrf2 and SOD1 expression. Taken together, these data demonstrated that LA suppressed VHSV replication by inducing antiviral genes expression and reducing VHSV-induced oxidative stress. These results suggest a new direction in the development of potential antiviral candidate drugs against VHSV infection.
Collapse
|
20
|
Hanisch M, Rake B. Repurposing without purpose? Early innovation responses to the COVID‐19 crisis: Evidence from clinical trials. R&D MANAGEMENT 2021; 51:393-409. [PMCID: PMC8014695 DOI: 10.1111/radm.12461] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 12/02/2020] [Accepted: 01/26/2021] [Indexed: 05/23/2023]
Abstract
The novel coronavirus has created one of the biggest social and economic challenges in recent decades. Since a critical issue in overcoming a large‐scale pandemic involves finding effective treatments for the disease, there is typically urgent pressure on the health‐care sector to develop innovations to combat the pandemic. Recently, scholars have argued that repurposing – that is, reusing an existing innovation in a different context – allows for such rapid innovation responses and can reduce costs, as the groundwork has already been laid. In this paper, we compare these benefits with the considerable disadvantages associated with innovation repurposing, including lowered barriers to entry, which can lead to declining average quality and duplicate work. Using data on 2,456 COVID‐19‐related clinical trials initiated between December 2019 and July 2020, we find that merely one‐third of the trials actually investigated drugs or vaccines, whereas the rest focused on diagnostics and crisis management issues. In the trials concerning drug testing, we find that drug repurposing is a predominant innovation strategy, but many trials tested the same (combination of) drugs. This indicates an inefficient use of resources and reductions in the average variety and novelty of clinical trials. Furthermore, the small percentage of biopharmaceutical firms involved in the search for COVID‐19 treatments raises the question of whether firms may have insufficient incentives to redirect innovation efforts to respond to the pandemic. Our paper contributes to crisis management research, the nascent debate on COVID‐19, and the emerging literature on innovation repurposing.
Collapse
Affiliation(s)
- Marvin Hanisch
- Department of Innovation Management & StrategyUniversity of GroningenNettelbosje 2Groningen9747 AEThe Netherlands
| | - Bastian Rake
- School of BusinessMaynooth UniversityMaynoothCo. KildareIreland
| |
Collapse
|
21
|
GPCR_LigandClassify.py; a rigorous machine learning classifier for GPCR targeting compounds. Sci Rep 2021; 11:9510. [PMID: 33947911 PMCID: PMC8097070 DOI: 10.1038/s41598-021-88939-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 04/12/2021] [Indexed: 02/02/2023] Open
Abstract
The current study describes the construction of various ligand-based machine learning models to be used for drug-repurposing against the family of G-Protein Coupled Receptors (GPCRs). In building these models, we collected > 500,000 data points, encompassing experimentally measured molecular association data of > 160,000 unique ligands against > 250 GPCRs. These data points were retrieved from the GPCR-Ligand Association (GLASS) database. We have used diverse molecular featurization methods to describe the input molecules. Multiple supervised ML algorithms were developed, tested and compared for their accuracy, F scores, as well as for their Matthews' correlation coefficient scores (MCC). Our data suggest that combined with molecular fingerprinting, ensemble decision trees and gradient boosted trees ML algorithms are on the accuracy border of the rather sophisticated deep neural nets (DNNs)-based algorithms. On a test dataset, these models displayed an excellent performance, reaching a ~ 90% classification accuracy. Additionally, we showcase a few examples where our models were able to identify interesting connections between known drugs from the Drug-Bank database and members of the GPCR family of receptors. Our findings are in excellent agreement with previously reported experimental observations in the literature. We hope the models presented in this paper synergize with the currently ongoing interest of applying machine learning modeling in the field of drug repurposing and computational drug discovery in general.
Collapse
|
22
|
Verbaanderd C, Rooman I, Huys I. Exploring new uses for existing drugs: innovative mechanisms to fund independent clinical research. Trials 2021; 22:322. [PMID: 33947441 PMCID: PMC8093905 DOI: 10.1186/s13063-021-05273-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 04/15/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Finding new therapeutic uses for existing medicines could lead to safe, affordable and timely new treatment options for patients with high medical needs. However, due to a lack of economic incentives, pharmaceutical developers are rarely interested to invest in research with approved medicines, especially when they are out of basic patent or regulatory protection. Consequently, potential new uses for these medicines are mainly studied in independent clinical trials initiated and led by researchers from academia, research institutes, or collaborative groups. Yet, additional financial support is needed to conduct expensive phase III clinical trials to confirm the results from exploratory research. METHODS In this study, scientific and grey literature was searched to identify and evaluate new mechanisms for funding clinical trials with repurposed medicines. Semi-structured interviews were conducted with 16 European stakeholders with expertise in clinical research, funding mechanisms and/or drug repurposing between November 2018 and February 2019 to consider the future perspectives of applying new funding mechanisms. RESULTS Traditional grant funding awarded by government and philanthropic organisations or companies is well known and widely implemented in all research fields. In contrast, only little research has focused on the application potential of newer mechanisms to fund independent clinical research, such as social impact bonds, crowdfunding or public-private partnerships. Interviewees stated that there is a substantial need for additional financial support in health research, especially in areas where there is limited commercial interest. However, the implementation of new funding mechanisms is facing several practical and financial challenges, such as a lack of expertise and guidelines, high transaction costs and difficulties to measure health outcomes. Furthermore, interviewees highlighted the need for increased collaboration and centralisation at a European and international level to make clinical research more efficient and reduce the need for additional funding. CONCLUSIONS New funding mechanisms to support clinical research may become more important in the future but the unresolved issues identified in the current study warrant further exploration.
Collapse
Affiliation(s)
- Ciska Verbaanderd
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
- Anticancer Fund, Strombeek-Bever, Belgium.
| | - Ilse Rooman
- Anticancer Fund, Strombeek-Bever, Belgium
- Oncology Research Centre, Vrije Universiteit Brussel, Brussels, Belgium
| | - Isabelle Huys
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
23
|
Stability of Antimicrobial Drug Molecules in Different Gravitational and Radiation Conditions in View of Applications during Outer Space Missions. Molecules 2021; 26:molecules26082221. [PMID: 33921448 PMCID: PMC8069917 DOI: 10.3390/molecules26082221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 11/16/2022] Open
Abstract
The evolution of different antimicrobial drugs in terrestrial, microgravity and hypergravity conditions is presented within this review, in connection with their implementation during human space exploration. Drug stability is of utmost importance for applications in outer space. Instabilities may be radiation-induced or micro-/hypergravity produced. The antimicrobial agents used in space may have diminished effects not only due to the microgravity-induced weakened immune response of astronauts, but also due to the gravity and radiation-altered pathogens. In this context, the paper provides schemes and procedures to find reliable ways of fighting multiple drug resistance acquired by microorganisms. It shows that the role of multipurpose medicines modified at the molecular scale by optical methods in long-term space missions should be considered in more detail. Solutions to maintain drug stability, even in extreme environmental conditions, are also discussed, such as those that would be encountered during long-duration space exploratory missions. While the microgravity conditions may not be avoided in space, the suggested approaches deal with the radiation-induced modifications in humans, bacteria and medicines onboard, which may be fought by novel pharmaceutical formulation strategies along with radioprotective packaging and storage.
Collapse
|
24
|
Korenstein D, Kaltenboeck A, Mamoor M, Chimonas S. Priceless Knowledge: Attitudes and Awareness Around Drug Pricing Among US Medical Students. MEDICAL SCIENCE EDUCATOR 2021; 31:489-494. [PMID: 34457906 PMCID: PMC8368429 DOI: 10.1007/s40670-020-01190-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/10/2020] [Indexed: 06/13/2023]
Abstract
UNLABELLED High US drug costs have garnered increasing attention, with multiple proposed reforms. While physicians are key stakeholders, medical education about drug pricing is not described, and medical students' understanding and attitudes are poorly understood. To assess students' awareness of drug pricing and its determinants, the authors conducted a cross-sectional, web-based survey of US medical students. Survey items included attitudes and knowledge around drug pricing and relevant education received (e.g., importance, quantity/quality of instruction). A composite knowledge score summed correct responses to 10 knowledge items. Descriptive statistics and t tests were used to evaluate associations. Among 815 viewers of the survey invitation, 361 visited the survey and 240 completed it (view rate 44%; participation rate 77%; completion rate 87%). Most participants were white (62%), in MD programs (82%), and female (53%). Nearly all (> 99%) said it was somewhat or very important to understand factors influencing drug pricing; over 90% were interested in learning more. Among year 3-4 students (n = 108), 59% reported receiving medical school instruction on pricing; few rated the quantity as adequate (7%) or the quality as excellent (3%) or good (8%). Among 10 knowledge questions, the median correct score was 6. Fewer than half (44%) knew that prices are uncorrelated with research/development costs. Knowledge was associated with year in school (p = 0.011) but not reported instructional quality or quantity. In sum, medical students report interest in drug pricing but inadequate instruction, and their knowledge is incomplete. Enhanced education is needed to equip future doctors to advocate effectively for patients around drug prices. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s40670-020-01190-x.
Collapse
Affiliation(s)
- Deborah Korenstein
- Center for Health Policy & Outcomes, Memorial Sloan Kettering Cancer Center, 485 Lexington Avenue, New York, NY 10017 USA
| | - Anna Kaltenboeck
- Center for Health Policy & Outcomes, Memorial Sloan Kettering Cancer Center, 485 Lexington Avenue, New York, NY 10017 USA
| | - Maha Mamoor
- Center for Health Policy & Outcomes, Memorial Sloan Kettering Cancer Center, 485 Lexington Avenue, New York, NY 10017 USA
| | - Susan Chimonas
- Center for Health Policy & Outcomes, Memorial Sloan Kettering Cancer Center, 485 Lexington Avenue, New York, NY 10017 USA
| |
Collapse
|
25
|
Darrow JJ, Light DW. Beyond The High Prices Of Prescription Drugs: A Framework To Assess Costs, Resource Allocation, And Public Funding. Health Aff (Millwood) 2021; 40:281-288. [PMID: 33523733 DOI: 10.1377/hlthaff.2020.00328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
During the past century, an accumulation of laws, organizations, and policy mechanisms has led to increasing transfers of public funds to private drug manufacturers, straining budgets and enabling industry revenues beyond what markets could ordinarily sustain. Tax benefits and fee waivers subsidize industry research, while public institutions and charities help fund the creation of new products and pay for their use once they are approved. New exclusivities increase prices by delaying competition, and payment programs such as Medicare Part D help guarantee that prices will be paid no matter how high they rise. Members of the public thus pay for pharmaceuticals in more ways than is commonly recognized. This article provides a more comprehensive framework for legislators and scholars to use in assessing the total societal costs of drugs. Greater transparency is needed to clarify individual drug costs, facilitate appropriate resource allocation, and ensure that the amount of public funding is justified by the value of the drugs. Congress should direct the Government Accountability Office to study public contributions underlying the highest-cost drugs and should require periodic reporting by drug manufacturers of the direct and indirect public funding they receive.
Collapse
Affiliation(s)
- Jonathan J Darrow
- Jonathan J. Darrow is an assistant professor in the Program on Regulation, Therapeutics, and Law at Harvard Medical School and an associate scientist at Brigham and Women's Hospital, both in Boston, Massachusetts
| | - Donald W Light
- Donald W. Light is a professor of psychiatry and medical sociology at the Rowan University School of Osteopathic Medicine, in Stratford, New Jersey
| |
Collapse
|
26
|
Barenie RE, Avorn J, Tessema FA, Kesselheim AS. Public funding for transformative drugs: the case of sofosbuvir. Drug Discov Today 2021; 26:273-281. [PMID: 33011345 PMCID: PMC7528745 DOI: 10.1016/j.drudis.2020.09.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/03/2020] [Accepted: 09/23/2020] [Indexed: 11/21/2022]
Abstract
The approval of sofosbuvir (Sovaldi) in 2013 transformed chronic hepatitis C virus (HCV) care, but its high cost was criticized in part because of reports of substantial public involvement in its development. We developed a methodology to assess the public's contribution through the National Institutes of Health (NIH) in developing sofosbuvir. Using key terms from the timeline of sofosbuvir, we identified articles in PubMed; linked them to federal funding using the NIH RePORTER; reviewed the title, organization, and investigator of each resulting award for relatedness; and converted related awards to 2018 US dollars. Of 6043 unique awards, we identified 29 that were directly (US$7.7 million) and 110 that were indirectly (US$53.2 million) related awards made to major academic institutions and companies engaged in the development of the drug. These findings indicate that public funding had a key role in developing sofosbuvir, with an estimated US$60.9 million provided in NIH funding.
Collapse
Affiliation(s)
- Rachel E Barenie
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 1620 Tremont St, Suite 3030, Boston, MA 02120, USA.
| | - Jerry Avorn
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 1620 Tremont St, Suite 3030, Boston, MA 02120, USA
| | - Frazer A Tessema
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 1620 Tremont St, Suite 3030, Boston, MA 02120, USA
| | - Aaron S Kesselheim
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 1620 Tremont St, Suite 3030, Boston, MA 02120, USA
| |
Collapse
|
27
|
Abstract
In the debate over prescription drug pricing, some pharmaceutical industry critics claim that U.S. taxpayers pay twice for costly therapies, because publicly supported research is a major contributor to drug discovery and American taxpayers are inadequately rewarded for their research investment due to high drug prices. In fact, the empirical evidence supporting these claims is weak, and the pay twice argument distracts from important efforts to ensure that impactful new drugs continue to be developed and made widely available to patients who need them.
Collapse
Affiliation(s)
- Rena M Conti
- Questrom School of Business, Boston University, Boston, MA, 02215, USA.,Institute for Health System Innovation and Policy, Boston University, Boston, MA, 02215, USA
| | - Frank S David
- Pharmagellan LLC, Milton, MA, 02186, USA.,Harvard-MIT Center for Regulatory Science, Boston, MA, 02115, USA
| |
Collapse
|
28
|
Marshall LJ, Triunfol M, Seidle T. Patient-Derived Xenograft vs. Organoids: A Preliminary Analysis of Cancer Research Output, Funding and Human Health Impact in 2014-2019. Animals (Basel) 2020; 10:ani10101923. [PMID: 33092060 PMCID: PMC7593921 DOI: 10.3390/ani10101923] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer remains a major threat to mortality and morbidity globally, despite intense research and generous funding. Patient-derived xenograft (PDX) models-where tumor biopsies are injected into an animal-were developed to improve the predictive capacity of preclinical animal models. However, recent observations have called into question the clinical relevance, and therefore the translational accuracy, of these. Patient-derived organoids (PDO) use patient tumor samples to create in vitro models that maintain aspects of tumor structure and heterogeneity. We undertook a preliminary analysis of the number of breast, colorectal, and lung cancer research studies using PDX or PDO published worldwide between 2014-2019. We looked for evidence of impacts of this research on human health. The number of publications that focused on PDO is gradually increasing over time, but is still very low compared to publications using PDX models. Support for new research projects using PDO is gradually increasing, a promising indicator of a shift towards more human-relevant approaches to understanding human disease. Overall, increases in total funding for these three major cancer types does not appear to be translating to any consequential increase in outputs, defined for this purpose as publications associated with clinical trials. With increasing public discomfort in research using animals and demands for 'alternative' methods, it is timely to consider how to implement non-animal methods more effectively.
Collapse
Affiliation(s)
- Lindsay J. Marshall
- Humane Society International and the Humane Society of the United States, Washington, DC 20037, USA
- Correspondence:
| | - Marcia Triunfol
- Humane Society International, Washington, DC, 20037, USA; (M.T.); (T.S.)
| | - Troy Seidle
- Humane Society International, Washington, DC, 20037, USA; (M.T.); (T.S.)
| |
Collapse
|
29
|
Sarpatwari A, Avorn J, Kesselheim AS. Accounting for US public funding in drug development: how can we better balance access, affordability, and innovation? BMJ 2020; 371:m3841. [PMID: 33032982 DOI: 10.1136/bmj.m3841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Ameet Sarpatwari
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA 02120, USA
| | - Jerry Avorn
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA 02120, USA
| | - Aaron S Kesselheim
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA 02120, USA
| |
Collapse
|
30
|
Anwar A, Khan NA, Siddiqui R. Repurposing of Drugs Is a Viable Approach to Develop Therapeutic Strategies against Central Nervous System Related Pathogenic Amoebae. ACS Chem Neurosci 2020; 11:2378-2384. [PMID: 32073257 DOI: 10.1021/acschemneuro.9b00613] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Brain-eating amoebae including Acanthamoeba spp., Naegleria fowleri, and Balamuthia mandrillaris cause rare infections of the central nervous system that almost always result in death. The high mortality rate, lack of interest for drug development from pharmaceutical industries, and no available effective drugs present an alarming challenge. The current drugs employed in the management and therapy of these devastating diseases are amphotericin B, miltefosine, chlorhexidine, pentamidine, and voriconazole which are generally used in combination. However, clinical evidence shows that these drugs have limited efficacy and high host cell cytotoxicity. Repurposing of drugs is a practical approach to utilize commercially available, U.S. Food and Drug Administration approved drugs for one disease against rare diseases caused by brain-eating amoebae. In this Perspective, we highlight some of the success stories of drugs repositioned against neglected parasitic diseases and identify future potential for effective and sustainable drug development against brain-eating amoebae infections.
Collapse
Affiliation(s)
- Ayaz Anwar
- Department of Biological Sciences, School of Science and Technology, Sunway University, Subang Jaya 47500, Selangor, Malaysia
| | - Naveed Ahmed Khan
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Ruqaiyyah Siddiqui
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah 26666, United Arab Emirates
| |
Collapse
|
31
|
Franzen N, Retèl VP, Schats W, van Harten WH. Evidence Underlying Policy Proposals for Sustainable Anticancer Drug Prices. JAMA Oncol 2020; 6:909-916. [DOI: 10.1001/jamaoncol.2019.6846] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Nora Franzen
- Department of Health Technology and Services Research, University of Twente, Enschede, the Netherlands
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Valesca P. Retèl
- Department of Health Technology and Services Research, University of Twente, Enschede, the Netherlands
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Winnie Schats
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Wim H. van Harten
- Department of Health Technology and Services Research, University of Twente, Enschede, the Netherlands
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Rijnstate Hospital, Arnhem, the Netherlands
| |
Collapse
|
32
|
Hsiue EHC, Moore TJ, Alexander GC. Estimated costs of pivotal trials for U.S. Food and Drug Administration-approved cancer drugs, 2015-2017. Clin Trials 2020; 17:119-125. [PMID: 32114790 DOI: 10.1177/1740774520907609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Pivotal clinical trials provide critical evidence to regulators regarding a product's suitability for marketing approval. The objectives of this study are (1) to characterize select features of trials for oncology products approved by the U.S. Food and Drug Administration between 2015 and 2017; and (2) to quantify the costs of these trials and how such costs varied based on trial characteristics. METHODS We identified novel oncology therapeutic drugs, and their respective pivotal trials, approved between 2015 and 2017 using annual summary reports from the Food and Drug Administration. Cost estimates for each pivotal trial were calculated using IQVIA's CostPro, a clinical trial cost estimating tool based on executed contracts between pharmaceutical manufacturers and contract research organizations. Measures of drug and trial characteristics included trial design, end point, patient enrollment, and regulatory pathway. We also performed sensitivity analyses that varied assumptions regarding how efficiently each trial was conducted. RESULTS A total of 39 pivotal clinical trials provided the basis for Food and Drug Administration approval of 30 new oncology drugs from 2015 to 2017. Among these trials, primary end points were objective response rate in 20 (51.3%), progression-free survival in 13 (33.3%), and overall survival in 6 (15.4%). Twenty trials (51.3%) were single-arm studies. The median estimated cost of oncology pivotal trials was $31.7 million (interquartile range = $17.0-$60.4 million). Trials with objective response rate as primary end point had a median estimate of $17.7 million (interquartile range = $11.9-$27.1 million), compared with trials examining progression-free survival ($42.3 million, interquartile range = $34.6-$101.2 million) or overall survival ($79.4 million, interquartile range = $56.9-$97.7 million) (p < 0.001). Estimated costs for single-arm trials ($17.7 million, interquartile range = $11.9-$23.7 million) were less than for trials with a placebo-controlled ($56.7 million, interquartile range = $40.9-$103.9 million) or active control arm ($67.6 million, IQR = $35.5-$93.5 million) (p < 0.001). CONCLUSIONS Relative to the estimated costs of drug development, the costs of these oncology pivotal trials were modest, with trials that produced more valuable scientific information costing more than their counterparts.
Collapse
Affiliation(s)
- Emily Han-Chung Hsiue
- Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Thomas J Moore
- Institute for Safe Medication Practices, Alexandria, VA, USA
- Department of Epidemiology, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - G Caleb Alexander
- Center for Drug Safety and Effectiveness, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Division of General Internal Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| |
Collapse
|
33
|
Verbaanderd C, Rooman I, Meheus L, Huys I. On-Label or Off-Label? Overcoming Regulatory and Financial Barriers to Bring Repurposed Medicines to Cancer Patients. Front Pharmacol 2020; 10:1664. [PMID: 32076405 PMCID: PMC7006723 DOI: 10.3389/fphar.2019.01664] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
Repurposing of medicines has gained a lot of interest from the research community in recent years as it could offer safe, timely, and affordable new treatment options for cancer patients with high unmet needs. Increasingly, questions arise on how new uses will be translated into clinical practice, especially in case of marketed medicinal products that are out of basic patent or regulatory protection. The aim of this study was to portray the regulatory framework relevant for making repurposed medicines available to cancer patients in Europe and propose specific policy recommendations to address the current regulatory and financial barriers. We outlined two routes relevant to the clinical adoption of a repurposed medicine. First, a new indication can be approved, and thus brought on-label, via the marketing authorization procedures established in European and national legislation. Such procedures initiate a detailed and independent assessment of the quality and the benefit-risk balance of a medicinal product in a specific indication, benefiting both prescribers and patients as it reassures them that the scientific evidence is robust. However, the process of marketing authorization for new therapeutic indications entails a high administrative burden and significant costs while the return-on-investment for the pharmaceutical industry is expected to be low or absent for medicines that are out of basic patent and regulatory protection. Moreover, most of the repurposing research is conducted by independent or academic researchers who do not have the expertise or resources to get involved in regulatory procedures. A second option is to prescribe a medicine off-label for the new indication, which is managed at the national level in Europe. While off-label use could provide timely access to treatments for patients with urgent medical needs, it also entails important safety, liability and financial risks for patients, physicians, and society at large. In view of that, we recommend finding solutions to facilitate bringing new uses on-label, for example by developing a collaborative framework between not-for-profit and academic organizations, pharmaceutical industry, health technology assessment bodies, payers, and regulators.
Collapse
Affiliation(s)
- Ciska Verbaanderd
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.,Anticancer Fund, Strombeek-Bever, Belgium
| | - Ilse Rooman
- Anticancer Fund, Strombeek-Bever, Belgium.,Oncology Research Centre, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Isabelle Huys
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
34
|
Trotta F, Mayer F, Barone-Adesi F, Esposito I, Punreddy R, Da Cas R, Traversa G, Perrone F, Martini N, Gyawali B, Addis A. Anticancer drug prices and clinical outcomes: a cross-sectional study in Italy. BMJ Open 2019; 9:e033728. [PMID: 31826897 PMCID: PMC6924817 DOI: 10.1136/bmjopen-2019-033728] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/10/2019] [Accepted: 11/11/2019] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE To investigate whether the prices of new anticancer drugs correlated with their relative benefit despite negotiation. DESIGN Retrospective cross-sectional study correlating new anticancer drugs prices with clinical outcomes. SETTING We did a retrospective cross-sectional study including all new anticancer drugs approved by the European Medicines Agency (EMA) (2010-2016) and reimbursed in Italy. MAIN OUTCOMES AND MEASURES Information on clinical outcomes-in terms of median overall survival (OS), median progression-free survival (PFS) and objective response rate (ORR)-was extracted from pivotal trials as reported in the European Public Assessment Reports available on the EMA website. Cost of a full course treatment was estimated on negotiated official and discounted prices. Regression coefficients β, their levels of significance p and the coefficients of determination R2 were estimated adjusting by tumour type. RESULTS Overall, 30 new anticancer drugs (with 35 indications) were available for analysis. Where data on OS were available, we observed no correlation between the improvement in median OS (in weeks) and negotiated price (R2=0.067, n=16 drugs for 17 indications). When the clinical outcomes were expressed as improvements in the median PFS or ORR, 25 drugs (29 indications) were available for the analysis, and again, there was no correlation with prices (R2=0.004 and 0.006, respectively). CONCLUSIONS AND RELEVANCE Our results suggest that the prices of anticancer drugs in Italy do not reflect their therapeutic benefit. Drug price negotiations, which is mandatory by law in Italy, do not seem to ensure that prices correlate with clinical benefits provided by the cancer drugs. These results call for further efforts to establish the standard determinants of drug prices available at the time of negotiation. These findings need to be confirmed in other countries where price negotiations are in place. Moreover, further investigations may verify whether outcome data obtained after drug marketing would improve the correlation between prices and therapeutic benefit.
Collapse
Affiliation(s)
- Francesco Trotta
- Department of Epidemiology of the Regional Health Service Lazio, Rome, Italy
| | - Flavia Mayer
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Italy
| | | | | | | | - Roberto Da Cas
- National Centre for Epidemiology, Italian National Institute of Health, Rome, Italy
| | - Giuseppe Traversa
- National Centre for Epidemiology, Italian National Institute of Health, Rome, Italy
| | - Francesco Perrone
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | | | | | - Antonio Addis
- Department of Epidemiology of the Regional Health Service Lazio, Rome, Italy
| |
Collapse
|
35
|
Long G. Federal government-interest patent disclosures for recent top-selling drugs. J Med Econ 2019; 22:1261-1267. [PMID: 31190582 DOI: 10.1080/13696998.2019.1631832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Objective: To provide updated evidence on government-interest patent disclosures in US patents for top-selling small-molecule drugs.Methods: IQVIA National Sales Perspectives data identified 300 top-selling drugs, defined by peak 2013-2017 US sales. For the 197 approved through New Drug Applications (NDAs), data were collected from a recently-released dataset of all patents listed in 1985-2016 Annual Editions of the FDA Orange Book. Data on patent assignees and Government Interest Statements (if any) were collected from the US Patent and Trademark Office online database. The percentage of drugs with at least one government-interest patent disclosure was calculated, as was the percentage of patents with a disclosure, by type (drug product, drug substance, or method of use). Government-interest patent disclosures were defined as those for which: the patent application contained a Government Interest Statement; and/or any of the patent assignees was a US government agency.Results: Few patents for the top-selling drugs had a government-interest patent disclosure, 2.6% on average. By patent type, figures ranged between 1.6% (for patents with drug product claims) and 3.6% (for patents with drug substance claims). Accounting for multiple patents per drug, 8.6% of top-selling drugs analyzed had at least one patent with a Government Interest Statement; 1.5% had at least one with a US government agency assignee; and 10.2% met either criterion (none met both).Limitations: Analyses were limited to top-selling NDA-approved drugs (generally excluding biologics) and Orange Book-listed patents. Patents with government-interest patent disclosures could also have relied on non-government funding. Patents were not characterized by relative economic investments, importance in the discovery and development process, or contribution to clinical value.Conclusion: Results were generally comparable to a prior analysis that found that 9.0% of new drugs approved between 1988 and 2005 had either a Government Interest Statement disclosure or a government agency first-listed patent assignee.
Collapse
|
36
|
Choudhury MC, Saberwal G. The work, goals, challenges, achievements, and recommendations of orphan medicinal product organizations in India: an interview-based study. Orphanet J Rare Dis 2019; 14:241. [PMID: 31684990 PMCID: PMC6829914 DOI: 10.1186/s13023-019-1224-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 10/09/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Orphan medicinal products (OMPs) are intended for the diagnosis, prevention, management or treatment of rare diseases (RDs). Each RD affects only a small fraction of the population, and therefore, historically, industry hesitated to undertake relevant research and development (R&D). In response, the governments of many countries came up with orphan drug policies and RD policies which were hugely successful in incentivizing companies to do so. In India, in the absence of any such policy until recently, there are very few organizations involved in RD R&D. OBJECTIVES We wished to understand (i) the OMP Organizations' (OMPOs') areas of work and the nature of their work, (ii) their goals, (iii) the challenges they faced and how they were overcoming them, (iv) their achievements, and (v) their recommendations to the government to help their R&D, their success as commercial entities (where applicable), and patients' access to their products or services. RESULTS Ten of the 14 OMPOs are companies, whereas four are not-for-profit organizations. Almost all of the OMPOs are heavily into R&D. Six have already made their products or services available to patients. Four plan to out-license their products after the pre-clinical phase or phase 1 trials, eight plan to cater to patients directly and two of the OMPOs have been established only recently and thus do not yet have any product or service to offer patients. Nine OMPOs import about 90% of the components in the production process, which comprises either capital or recurrent expenditure. For most, locally manufactured alternatives are not available or are of inadequate quality. Most of the OMPOs have had productive collaborations with local or foreign academics or hospitals for R&D, animal efficacy studies, clinical trials or providing services to patients. The main challenges for the OMPOs are the lack of adequate funding, supportive government policies, and a conducive ecosystem. CONCLUSIONS These OMPOs are pioneers in their respective fields in India, and despite the challenges, have achieved new levels of innovation. With suitable government policies, they could scale up and provide relevant products and services to the large number of RD patients in the country whose medical needs are largely unmet.
Collapse
Affiliation(s)
- Mohua Chakraborty Choudhury
- Institute of Bioinformatics and Applied Biotechnology, Biotech Park, Electronics City Phase 1, Bengaluru, Karnataka, 560100, India
| | - Gayatri Saberwal
- Institute of Bioinformatics and Applied Biotechnology, Biotech Park, Electronics City Phase 1, Bengaluru, Karnataka, 560100, India.
| |
Collapse
|
37
|
Nayak RK, Avorn J, Kesselheim AS. Public sector financial support for late stage discovery of new drugs in the United States: cohort study. BMJ 2019; 367:l5766. [PMID: 31645328 PMCID: PMC6812612 DOI: 10.1136/bmj.l5766] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2019] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To determine the extent to which late stage development of new drugs relies on support from public funding. DESIGN Cohort study. SETTING All new drugs containing one or more new molecular entities approved by the US Food and Drug Administration (FDA) between January 2008 and December 2017 via the new drug application pathway. MAIN OUTCOME MEASURES Patents or drug development histories documenting late stage research contributions by a public sector research institution or a spin-off company, as well as each drug's regulatory approval pathway and first-in-class designation. RESULTS Over the 10 year study period, the FDA approved 248 drugs containing one or more new molecular entities. Of these drugs, 48 (19%) had origins in publicly supported research and development and 14 (6%) originated in companies spun off from a publicly supported research program. Drugs in these groups were more likely to receive expedited FDA approval (68% v 47%, P=0.005) or be designated first in class (45% v 26%, P=0.007), indicating therapeutic importance. CONCLUSIONS A review of the patents associated with new drugs approved over the past decade indicates that publicly supported research had a major role in the late stage development of at least one in four new drugs, either through direct funding of late stage research or through spin-off companies created from public sector research institutions. These findings could have implications for policy makers in determining fair prices and revenue flows for these products.
Collapse
Affiliation(s)
- Rahul K Nayak
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 1620 Tremont Street, Boston, MA 02120, USA
| | - Jerry Avorn
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 1620 Tremont Street, Boston, MA 02120, USA
| | - Aaron S Kesselheim
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 1620 Tremont Street, Boston, MA 02120, USA
| |
Collapse
|
38
|
The use or generation of biomedical data and existing medicines to discover and establish new treatments for patients with rare diseases - recommendations of the IRDiRC Data Mining and Repurposing Task Force. Orphanet J Rare Dis 2019; 14:225. [PMID: 31615551 PMCID: PMC6794821 DOI: 10.1186/s13023-019-1193-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022] Open
Abstract
The number of available therapies for rare diseases remains low, as fewer than 6% of rare diseases have an approved treatment option. The International Rare Diseases Research Consortium (IRDiRC) set up the multi-stakeholder Data Mining and Repurposing (DMR) Task Force to examine the potential of applying biomedical data mining strategies to identify new opportunities to use existing pharmaceutical compounds in new ways and to accelerate the pace of drug development for rare disease patients. In reviewing past successes of data mining for drug repurposing, and planning for future biomedical research capacity, the DMR Task Force identified four strategic infrastructure investment areas to focus on in order to accelerate rare disease research productivity and drug development: (1) improving the capture and sharing of self-reported patient data, (2) better integration of existing research data, (3) increasing experimental testing capacity, and (4) sharing of rare disease research and development expertise. Additionally, the DMR Task Force also recommended a number of strategies to increase data mining and repurposing opportunities for rare diseases research as well as the development of individualized and precision medicine strategies.
Collapse
|
39
|
Microgravity protein crystallization for drug development: a bold example of public sector entrepreneurship. JOURNAL OF TECHNOLOGY TRANSFER 2019. [DOI: 10.1007/s10961-019-09743-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
40
|
Karas L, Lu CY, Agrawal PB, Asgari MM. The impact of the Orphan Drug Act on Food and Drug Administration-approved therapies for rare skin diseases and skin-related cancers. J Am Acad Dermatol 2019; 81:867-877. [PMID: 31103566 DOI: 10.1016/j.jaad.2019.05.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/07/2019] [Accepted: 05/10/2019] [Indexed: 12/18/2022]
Abstract
The Orphan Drug Act of 1983 (ODA) put in place a set of financial and marketing incentives to stimulate the development of drugs to treat rare diseases, and since its passage, more than 600 orphan drug and biologic products have been brought to market in the United States. Rapid growth in orphan drug approvals in conjunction with high orphan drug prices have triggered concern that drug makers are exploiting certain aspects of the ODA for financial gain and that some pharmaceutical drugs are receiving orphan status where it is not warranted. The landscape of approved therapies for rare skin diseases has not been well described. In this article, we provide a descriptive analysis of the United States Food and Drug Administration-approved orphan drugs for the treatment of rare dermatologic conditions and skin-related cancers since the enactment of the ODA. We discuss policy issues that emerge from the analysis and suggest areas for future research. Next, we elucidate ODA loopholes using dermatologic drugs as examples and propose potential reforms. Finally, we consider future directions for orphan drug development in the field of dermatology.
Collapse
Affiliation(s)
- Laura Karas
- Department of Population Medicine, Harvard Medical School and the Harvard Pilgrim Health Care Institute, Boston, Massachusetts.
| | - Christine Y Lu
- Department of Population Medicine, Harvard Medical School and the Harvard Pilgrim Health Care Institute, Boston, Massachusetts
| | - Pankaj B Agrawal
- Divisions of Genetics & Genomics and Newborn Medicine, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School Boston, Massachusetts
| | - Maryam M Asgari
- Department of Population Medicine, Harvard Medical School and the Harvard Pilgrim Health Care Institute, Boston, Massachusetts; Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
41
|
Gleeson D, Townsend B, Lopert R, Lexchin J, Moir H. Financial costs associated with monopolies on biologic medicines in Australia. AUST HEALTH REV 2019; 43:36-42. [DOI: 10.1071/ah17031] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 09/28/2017] [Indexed: 11/23/2022]
Abstract
Objectives
The aim of the study was to estimate the potential savings to the Pharmaceutical Benefits Scheme (PBS) and the Repatriation Pharmaceutical Benefits Scheme (RPBS) in 2015–16 if biosimilar versions of selected biologic medicines (biologics) had been available and listed on the PBS.
Methods
The research involved retrospective analysis of Australian Medicare expenditure data and PBS price data from 2015–16 for biologics, for which biosimilar competition may be available in future, listed on the PBS.
Results
Australian Government expenditure on biologics on the PBS and RPBS was estimated at A$2.29 billion dollars in 2015–16. If biosimilar versions of these medicines had been listed on the PBS in 2015–16, at least A$367million dollars would have been saved in PBS and RPBS subsidies. Modelling based on price decreases following listing of biosimilars on the PBS suggests that annual PBS outlays on biologics could be reduced by as much as 24% through the timely introduction of biosimilars.
Conclusions
Biologic medicines represent a large proportion of government expenditure on pharmaceuticals. Reducing the length of monopoly protections on these medicines could generate savings of hundreds of millions of dollars per year.
What is known about the topic?
Biologics take up an increasing share of pharmaceutical expenditure, but no previous published studies have examined Australian Government expenditure on biologics or the potential savings from reducing the duration of monopoly protection.
What does this paper add?
This paper provides new evidence about Australian Government expenditure on biologics and potential savings for selected medicines that are still subject to monopoly protection and thus are not yet subject to biosimilar competition. In 2015–16 Australian Government expenditure on biologics through the PBS and RPBS was estimated at A$2.29 billion dollars. If biosimilar versions of these medicines had been listed on the PBS at that time, at least A$367million dollars would have been saved.
What are the implications for practitioners?
Reducing the duration of monopoly protection on biologic medicines could save hundreds of millions of dollars annually that could be redirected to other areas of the healthcare system.
Collapse
|
42
|
Fitipaldi H, McCarthy MI, Florez JC, Franks PW. A Global Overview of Precision Medicine in Type 2 Diabetes. Diabetes 2018; 67:1911-1922. [PMID: 30237159 PMCID: PMC6152339 DOI: 10.2337/dbi17-0045] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/07/2018] [Indexed: 01/01/2023]
Abstract
The detailed characterization of human biology and behaviors is now possible at scale owing to innovations in biomarkers, bioimaging, and wearable technologies; "big data" from electronic medical records, health insurance databases, and other platforms becoming increasingly accessible; and rapidly evolving computational power and bioinformatics methods. Collectively, these advances are creating unprecedented opportunities to better understand diabetes and many other complex traits. Identifying hidden structures within these complex data sets and linking these structures to outcome data may yield unique insights into the risk factors and natural history of diabetes, which in turn may help optimize the prevention and management of the disease. This emerging area is broadly termed "precision medicine." In this Perspective, we give an overview of the evidence and barriers to the development and implementation of precision medicine in type 2 diabetes. We also discuss recently presented paradigms through which complex data might enhance our understanding of diabetes and ultimately our ability to tackle the disease more effectively than ever before.
Collapse
Affiliation(s)
- Hugo Fitipaldi
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, U.K
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
| | - Jose C Florez
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Paul W Franks
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, U.K
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
43
|
Sinha MS, Kesselheim AS. The Tax Cuts and Jobs Act of 2017 and the Pharmaceutical Industry. THE JOURNAL OF LAW, MEDICINE & ETHICS : A JOURNAL OF THE AMERICAN SOCIETY OF LAW, MEDICINE & ETHICS 2018; 46:806-808. [PMID: 30336090 DOI: 10.1177/1073110518804246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Affiliation(s)
- Michael S Sinha
- Michael S. Sinha, M.D., J.D., M.P.H., is with the Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmaco-economics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA. Aaron S. Kesselheim, M.D., J.D., M.P.H., is an Associate Professor of Medicine at Harvard Medical School and Director of the Program on Regulation, Therapeutics, and Law (PORTAL) in the Division of Pharmacoepidemiology and Pharmacoeconomics at the Brigham and Women's Hospital and a faculty member at the Harvard Center for Bioethics in Boston. He is also a faculty member at the Harvard Center for Bioethics
| | - Aaron S Kesselheim
- Michael S. Sinha, M.D., J.D., M.P.H., is with the Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmaco-economics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA. Aaron S. Kesselheim, M.D., J.D., M.P.H., is an Associate Professor of Medicine at Harvard Medical School and Director of the Program on Regulation, Therapeutics, and Law (PORTAL) in the Division of Pharmacoepidemiology and Pharmacoeconomics at the Brigham and Women's Hospital and a faculty member at the Harvard Center for Bioethics in Boston. He is also a faculty member at the Harvard Center for Bioethics
| |
Collapse
|
44
|
Fonseca R, Peneva D, Clancy Z, Abouzaid S, Jena AB. The Importance of Economic Trade-offs in Cancer Drug Pricing. Mayo Clin Proc 2018; 93:976-979. [PMID: 29945732 DOI: 10.1016/j.mayocp.2018.05.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/30/2018] [Accepted: 05/15/2018] [Indexed: 10/28/2022]
Affiliation(s)
- Rafael Fonseca
- Division of Hematology and Oncology, Mayo Clinic, Phoenix, AZ.
| | - Desi Peneva
- Health Policy, Innovation, and Value Practice, Precision Health Economics, Los Angeles, CA
| | - Zoe Clancy
- US Health Economics & Outcomes Research, Hematology/Oncology, Celgene Corporation, Summit, NJ
| | - Safiya Abouzaid
- Multiple Myeloma Marketing, Patient & Caregiver, Celgene Corporation, Summit, NJ
| | - Anupam B Jena
- Department of Health Care Policy, Harvard Medical School, Boston, MA
| |
Collapse
|
45
|
Shameer K, Glicksberg BS, Hodos R, Johnson KW, Badgeley MA, Readhead B, Tomlinson MS, O’Connor T, Miotto R, Kidd BA, Chen R, Ma’ayan A, Dudley JT. Systematic analyses of drugs and disease indications in RepurposeDB reveal pharmacological, biological and epidemiological factors influencing drug repositioning. Brief Bioinform 2018; 19:656-678. [PMID: 28200013 PMCID: PMC6192146 DOI: 10.1093/bib/bbw136] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/29/2016] [Indexed: 12/22/2022] Open
Abstract
Increase in global population and growing disease burden due to the emergence of infectious diseases (Zika virus), multidrug-resistant pathogens, drug-resistant cancers (cisplatin-resistant ovarian cancer) and chronic diseases (arterial hypertension) necessitate effective therapies to improve health outcomes. However, the rapid increase in drug development cost demands innovative and sustainable drug discovery approaches. Drug repositioning, the discovery of new or improved therapies by reevaluation of approved or investigational compounds, solves a significant gap in the public health setting and improves the productivity of drug development. As the number of drug repurposing investigations increases, a new opportunity has emerged to understand factors driving drug repositioning through systematic analyses of drugs, drug targets and associated disease indications. However, such analyses have so far been hampered by the lack of a centralized knowledgebase, benchmarking data sets and reporting standards. To address these knowledge and clinical needs, here, we present RepurposeDB, a collection of repurposed drugs, drug targets and diseases, which was assembled, indexed and annotated from public data. RepurposeDB combines information on 253 drugs [small molecules (74.30%) and protein drugs (25.29%)] and 1125 diseases. Using RepurposeDB data, we identified pharmacological (chemical descriptors, physicochemical features and absorption, distribution, metabolism, excretion and toxicity properties), biological (protein domains, functional process, molecular mechanisms and pathway cross talks) and epidemiological (shared genetic architectures, disease comorbidities and clinical phenotype similarities) factors mediating drug repositioning. Collectively, RepurposeDB is developed as the reference database for drug repositioning investigations. The pharmacological, biological and epidemiological principles of drug repositioning identified from the meta-analyses could augment therapeutic development.
Collapse
Affiliation(s)
- Khader Shameer
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Benjamin S Glicksberg
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
| | - Rachel Hodos
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
- New York University, New York, NY, USA
| | - Kipp W Johnson
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
| | - Marcus A Badgeley
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
| | - Ben Readhead
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Max S Tomlinson
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | | | - Riccardo Miotto
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Brian A Kidd
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Rong Chen
- Clinical Genome Informatics, Icahn Institute of Genetics and Multiscale
Biology, Mount Sinai Health System, New York, NY
| | - Avi Ma’ayan
- Mount Sinai Center for Bioinformatics, Mount Sinai Health System, New York,
NY
| | - Joel T Dudley
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
- Department of Genetics and Genomic Sciences, Mount Sinai Health System, New
York, NY, USA
- Department of Population Health Science and Policy, Mount Sinai Health System,
New York, NY, USA
- Director of Biomedical Informatics, Icahn School of Medicine at Mount Sinai,
Mount Sinai Health System, New York, NY
| |
Collapse
|
46
|
Tran G, Zafar SY. Financial toxicity and implications for cancer care in the era of molecular and immune therapies. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:166. [PMID: 29911114 DOI: 10.21037/atm.2018.03.28] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Molecular and immune therapies have revolutionized cancer treatment and improved patient outcomes and survival. However, the pricing of these drugs has become an issue as the cost of cancer care continues to rise significantly. Cost sharing policies have increased out-of-pocket expenses for patients, leading to poorer financial well-being, quality of life, psychosocial health, and treatment adherence. In this review, we briefly examine some factors affecting the pricing of these new targeted therapies; the effects of financial toxicity on patients; and highlight potential health policy and patient-provider level interventions to address these issues.
Collapse
Affiliation(s)
- George Tran
- Duke University School of Medicine, Durham, NC, USA
| | - S Yousuf Zafar
- Duke University School of Medicine, Durham, NC, USA.,Duke Cancer Institute, Duke-Margolis Center for Health Policy, Durham, NC, USA
| |
Collapse
|
47
|
Affiliation(s)
- Mohammed AlQuraishi
- Mohammed AlQuraishi is a Fellow in Therapeutic Science at Harvard Medical School, Boston, MA 02115, USA.Peter K. Sorger is Professor of Systems Biology, Head of the Harvard Program in Therapeutic Sciences, and Director of the Laboratory of Systems Biology at Harvard Medical School, Boston, MA 02115, USA.
| | - Peter K Sorger
- Mohammed AlQuraishi is a Fellow in Therapeutic Science at Harvard Medical School, Boston, MA 02115, USA.Peter K. Sorger is Professor of Systems Biology, Head of the Harvard Program in Therapeutic Sciences, and Director of the Laboratory of Systems Biology at Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
48
|
Mimouni M, Krauthammer M, Abualhasan H, Badarni H, Imtanis K, Allon G, Berkovitz L, Blumenthal EZ, Mimouni FB, Amarilyo G. Publication outcome of abstracts submitted to the American Academy of Ophthalmology meeting. J Med Libr Assoc 2018; 106:57-64. [PMID: 29339934 PMCID: PMC5764594 DOI: 10.5195/jmla.2018.314] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/01/2017] [Indexed: 11/24/2022] Open
Abstract
Objective Abstracts submitted to meetings are subject to less rigorous peer review than full-text manuscripts. This study aimed to explore the publication outcome of abstracts presented at the American Academy of Ophthalmology (AAO) annual meeting. Methods Abstracts presented at the 2008 AAO meeting were analyzed. Each presented abstract was sought via PubMed to identify if it had been published as a full-text manuscript. The publication outcome, journal impact factor (IF), and time to publication were recorded. Results A total of 690 abstracts were reviewed, of which 39.1% were subsequently published. They were published in journals with a median IF of 2.9 (range 0–7.2) and a median publication time of 426 days (range 0–2,133 days). A quarter were published in the journal Ophthalmology, with a shorter time to publication (median 282 vs. 534 days, p=0.003). Oral presentations were more likely to be published than poster presentations (57.8% vs. 35.9%, p<0.001) and in journals with higher IFs (3.2 vs. 2.8, p=0.02). Abstracts describing rare diseases had higher publication rates (49.4% vs. 38.0%, p=0.04) and were published in higher IF journals (3.7 vs. 2.9, p=0.03), within a shorter period of time (358 vs. 428 days, p=0.03). In multivariate analysis, affiliation with an institute located in the United States (p=0.002), abstracts describing rare diseases (p=0.03), and funded studies (p=0.03) were associated with publication in higher IF journals. Conclusions Almost 40% of abstracts were published. Factors that correlated with publication in journals with higher IF were a focus on rare diseases, affiliation with a US institute, and funding.
Collapse
|
49
|
Murtazalieva KA, Druzhilovskiy DS, Goel RK, Sastry GN, Poroikov VV. How good are publicly available web services that predict bioactivity profiles for drug repurposing? SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2017; 28:843-862. [PMID: 29183230 DOI: 10.1080/1062936x.2017.1399448] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 10/29/2017] [Indexed: 06/07/2023]
Abstract
Drug repurposing provides a non-laborious and less expensive way for finding new human medicines. Computational assessment of bioactivity profiles shed light on the hidden pharmacological potential of the launched drugs. Currently, several freely available computational tools are available via the Internet, which predict multitarget profiles of drug-like compounds. They are based on chemical similarity assessment (ChemProt, SuperPred, SEA, SwissTargetPrediction and TargetHunter) or machine learning methods (ChemProt and PASS). To compare their performance, this study has created two evaluation sets, consisting of (1) 50 well-known repositioned drugs and (2) 12 drugs recently patented for new indications. In the first set, sensitivity values varied from 0.64 (TarPred) to 1.00 (PASS Online) for the initial indications and from 0.64 (TarPred) to 0.98 (PASS Online) for the repurposed indications. In the second set, sensitivity values varied from 0.08 (SuperPred) to 1.00 (PASS Online) for the initial indications and from 0.00 (SuperPred) to 1.00 (PASS Online) for the repurposed indications. Thus, this analysis demonstrated that the performance of machine learning methods surpassed those of chemical similarity assessments, particularly in the case of novel repurposed indications.
Collapse
Affiliation(s)
- K A Murtazalieva
- a Institute of Biomedical Chemistry , Moscow , Russia
- b Pirogov Russian National Research Medical University , Moscow , Russia
| | | | - R K Goel
- c Punjabi University , Patiala , Punjab , India
| | - G N Sastry
- d CSIR-Indian Institute of Chemical Technology , Hyderabad , India
| | - V V Poroikov
- a Institute of Biomedical Chemistry , Moscow , Russia
| |
Collapse
|
50
|
Hwang TJ, Kesselheim AS. Challenges in the Development of Novel Cardiovascular Therapies. Clin Pharmacol Ther 2017. [DOI: 10.1002/cpt.703] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- TJ Hwang
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine; Brigham and Women's Hospital and Harvard Medical School; Boston Massachusetts USA
| | - AS Kesselheim
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine; Brigham and Women's Hospital and Harvard Medical School; Boston Massachusetts USA
| |
Collapse
|