1
|
Schuhmacher A, Gassmann O, Hinder M, Hartl D. Comparative analysis of FDA approvals by top 20 pharma companies (2014-2023). Drug Discov Today 2024; 29:104128. [PMID: 39097219 DOI: 10.1016/j.drudis.2024.104128] [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: 06/17/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
This article addresses the research and development (R&D) productivity challenge of the pharmaceutical industry, focusing on United States Food and Drug Administration (FDA)-related new drug approvals of the top 20 pharmaceutical companies (2014-2023). We evaluated the degree of innovation in new drugs to determine the innovativeness of these leading companies. A key finding of our analysis is the decline in the number of new drugs approved by the FDA for these leading companies over the investigated time period. This trend suggests that some of the leading companies are losing ground in R&D innovation, raising concerns about their ability to sustain competitive advantage, ensure long-term market success, and maintain viable business models.
Collapse
Affiliation(s)
- Alexander Schuhmacher
- Technische Hochschule Ingolstadt, THI Business School, Esplanade 10, D-85049 Ingolstadt, Germany; University of St. Gallen, Institute of Technology Management, Dufourstrasse 40a, CH-9000 St. Gallen, Switzerland.
| | - Oliver Gassmann
- University of St. Gallen, Institute of Technology Management, Dufourstrasse 40a, CH-9000 St. Gallen, Switzerland
| | - Markus Hinder
- Novartis, Development, Patient Safety, Forum 1, CH-4002 Basel, Switzerland; Fresenius University of Applied Sciences, Moritzstr. 17a, D-65185 Wiesbaden, Germany
| | - Dominik Hartl
- University of Tübingen, Hoppe-Seyler-Strasse 1, D-72076 Tübingen, Germany; Granite Bio, Aeschenvorstadt 36, CH-4051 Basel, Switzerland
| |
Collapse
|
2
|
Fuster-Martínez I, Calatayud S. The current landscape of antifibrotic therapy across different organs: A systematic approach. Pharmacol Res 2024; 205:107245. [PMID: 38821150 DOI: 10.1016/j.phrs.2024.107245] [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] [Received: 04/17/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Fibrosis is a common pathological process that can affect virtually all the organs, but there are hardly any effective therapeutic options. This has led to an intense search for antifibrotic therapies over the last decades, with a great number of clinical assays currently underway. We have systematically reviewed all current and recently finished clinical trials involved in the development of new antifibrotic drugs, and the preclinical studies analyzing the relevance of each of these pharmacological strategies in fibrotic processes affecting tissues beyond those being clinically studied. We analyze and discuss this information with the aim of determining the most promising options and the feasibility of extending their therapeutic value as antifibrotic agents to other fibrotic conditions.
Collapse
Affiliation(s)
- Isabel Fuster-Martínez
- Departamento de Farmacología, Universitat de València, Valencia 46010, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia 46020, Spain.
| | - Sara Calatayud
- Departamento de Farmacología, Universitat de València, Valencia 46010, Spain; CIBERehd (Centro de Investigación Biomédica en Red - Enfermedades Hepáticas y Digestivas), Spain.
| |
Collapse
|
3
|
Fernandes BS, Zhao Z. Improving drug development in precision psychiatry by ameliorating cognitive biases. Eur Neuropsychopharmacol 2023; 70:14-16. [PMID: 36796294 DOI: 10.1016/j.euroneuro.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023]
Affiliation(s)
- Brisa S Fernandes
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
4
|
Yang J, Zhang D, Cai Y, Yu K, Li M, Liu L, Chen X. Computational Prediction of Drug Phenotypic Effects Based on Substructure-Phenotype Associations. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:256-265. [PMID: 35239490 DOI: 10.1109/tcbb.2022.3155453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Identifying drug phenotypic effects, including therapeutic effects and adverse drug reactions (ADRs), is an inseparable part for evaluating the potentiality of new drug candidates (NDCs). However, current computational methods for predicting phenotypic effects of NDCs are mainly based on the overall structure of an NDC or a related target. These approaches often lead to inconsistencies between the structures and functions and limit the prediction space of NDCs. In this study, first, we constructed quantitative associations of substructure-domain, domain-ADR, and domain-ATC (Anatomical Therapeutic Chemical Classification System code) through L1LOG and L1SVM machine learning models. These associations represent relationships between phenotypes (ADRs and ATCs) and local structures of drugs and proteins. Then, based on these established associations, substructure-phenotype relationships were constructed which were utilized to quantify drug-phenotype relationships. Thus, this approach could achieve high-throughput and effective evaluations of the druggability of NDCs by referring to the established substructure-phenotype relationships and structural information of NDCs without additional prior knowledge. Using this computational pipeline, 83,205 drug-ATC relationships (including 1,479 drugs and 178 ATCs) and 306,421 drug-ADR relationships (including 1,752 drugs and 454 ADRs) were predicted in total. The prediction results were validated at four levels: five-fold cross validation, public databases, literature, and molecular docking. Furthermore, three case studies demonstrated the feasibility of our method. 79 ATCs and 269 ADRs were predicted to be related to Maraviroc, an approved drug, including the existing antiviral effect in clinical use. Additionally, we also found risk substructures of severe ADRs, for example, SUB215 (>= 1, saturated or only aromatic carbon ring size 7) can result in shock. And we analyzed the mechanism of action (MOA) of interested drugs based on the established drug-substructure-domain-protein associations. In a word, this approach through establishing drug-substructure-phenotype relationships can achieve quantitative prediction of phenotypes for a given NDC or drug without any prior knowledge except its structure information. Using that way, we can directly obtain the relationships between substructure and phenotype of a compound, which is more convenient to analyze the phenotypic mechanism of drugs and accelerate the process of rational drug design.
Collapse
|
5
|
Performance assessment and economic analysis of a human Liver-Chip for predictive toxicology. COMMUNICATIONS MEDICINE 2022; 2:154. [PMID: 36473994 PMCID: PMC9727064 DOI: 10.1038/s43856-022-00209-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 10/28/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Conventional preclinical models often miss drug toxicities, meaning the harm these drugs pose to humans is only realized in clinical trials or when they make it to market. This has caused the pharmaceutical industry to waste considerable time and resources developing drugs destined to fail. Organ-on-a-Chip technology has the potential improve success in drug development pipelines, as it can recapitulate organ-level pathophysiology and clinical responses; however, systematic and quantitative evaluations of Organ-Chips' predictive value have not yet been reported. METHODS 870 Liver-Chips were analyzed to determine their ability to predict drug-induced liver injury caused by small molecules identified as benchmarks by the Innovation and Quality consortium, who has published guidelines defining criteria for qualifying preclinical models. An economic analysis was also performed to measure the value Liver-Chips could offer if they were broadly adopted in supporting toxicity-related decisions as part of preclinical development workflows. RESULTS Here, we show that the Liver-Chip met the qualification guidelines across a blinded set of 27 known hepatotoxic and non-toxic drugs with a sensitivity of 87% and a specificity of 100%. We also show that this level of performance could generate over $3 billion annually for the pharmaceutical industry through increased small-molecule R&D productivity. CONCLUSIONS The results of this study show how incorporating predictive Organ-Chips into drug development workflows could substantially improve drug discovery and development, allowing manufacturers to bring safer, more effective medicines to market in less time and at lower costs.
Collapse
|
6
|
Hess AM. Real options or fallen angels: Examining the complexities of learning from terminated projects. CREATIVITY AND INNOVATION MANAGEMENT 2022. [DOI: 10.1111/caim.12527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Andrew M. Hess
- Ehrick Kilner Haight, Sr. Term Professor of Business Administration Washington and Lee University Lexington Virginia USA
| |
Collapse
|
7
|
Predictive validity in drug discovery: what it is, why it matters and how to improve it. Nat Rev Drug Discov 2022; 21:915-931. [PMID: 36195754 DOI: 10.1038/s41573-022-00552-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2022] [Indexed: 11/08/2022]
Abstract
Successful drug discovery is like finding oases of safety and efficacy in chemical and biological deserts. Screens in disease models, and other decision tools used in drug research and development (R&D), point towards oases when they score therapeutic candidates in a way that correlates with clinical utility in humans. Otherwise, they probably lead in the wrong direction. This line of thought can be quantified by using decision theory, in which 'predictive validity' is the correlation coefficient between the output of a decision tool and clinical utility across therapeutic candidates. Analyses based on this approach reveal that the detectability of good candidates is extremely sensitive to predictive validity, because the deserts are big and oases small. Both history and decision theory suggest that predictive validity is under-managed in drug R&D, not least because it is so hard to measure before projects succeed or fail later in the process. This article explains the influence of predictive validity on R&D productivity and discusses methods to evaluate and improve it, with the aim of supporting the application of more effective decision tools and catalysing investment in their creation.
Collapse
|
8
|
Alweis R, Salama A. Practical guidance to advisors of residents on the fellowship selection process. J Community Hosp Intern Med Perspect 2021; 11:421-424. [PMID: 34211641 PMCID: PMC8221151 DOI: 10.1080/20009666.2021.1938956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Inability to Match into a fellowship is usually not a reflection of some failure on the part of the resident, but rather a problem of supply and demand. Understanding how to advise residents to maximize their success in an environment with limited spots and limited fellowship faculty resources to perform holistic review remains one of the primary objectives of most residency mentors. Residents can alter the odds in their favor by engaging with local faculty and in national society mentorship programs, performing ‘enough’ research, building their ‘brand,’ and concentrating on high quality personal statements.
Collapse
Affiliation(s)
- Richard Alweis
- Graduate Medical Education, Rochester Regional Health, Rochester, NY, USA
| | - Amr Salama
- Sands-Constellation Heart Institute, Rochester General Hospital, Rochester, NY, USA
| |
Collapse
|
9
|
Schuhmacher A, Wilisch L, Kuss M, Kandelbauer A, Hinder M, Gassmann O. R&D efficiency of leading pharmaceutical companies - A 20-year analysis. Drug Discov Today 2021; 26:1784-1789. [PMID: 34022459 DOI: 10.1016/j.drudis.2021.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/28/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
Comparative analysis of the R&D efficiency of 14 leading pharmaceutical companies for the years 1999-2018 shows that there is a close positive correlation between R&D spending and the two investigated R&D output parameters, approved NMEs and the cumulative impact factor of their publications. In other words, higher R&D investments (input) were associated with higher R&D output. Second, our analyses indicate that there are 'economies of scale' (size) in pharmaceutical R&D.
Collapse
Affiliation(s)
- Alexander Schuhmacher
- Reutlingen University, Alteburgstrasse 150, DE-72762 Reutlingen, Germany; University of St Gallen, Dufourstrasse 40a, CH-9000 St Gallen, Switzerland.
| | - Lucas Wilisch
- University of St Gallen, Dufourstrasse 40a, CH-9000 St Gallen, Switzerland
| | - Michael Kuss
- PricewaterhouseCoopers AG, Birchstrasse 160, CH-8050 Zurich, Switzerland
| | | | - Markus Hinder
- Novartis Institute of BioMedical Research, Postfach, Forum 1, CH-4002 Basel, Switzerland
| | - Oliver Gassmann
- University of St Gallen, Dufourstrasse 40a, CH-9000 St Gallen, Switzerland
| |
Collapse
|
10
|
Wu SS, Fernando K, Allerton C, Jansen KU, Vincent MS, Dolsten M. Reviving an R&D pipeline: a step change in the Phase II success rate. Drug Discov Today 2020; 26:308-314. [PMID: 33129994 DOI: 10.1016/j.drudis.2020.10.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/30/2020] [Accepted: 10/20/2020] [Indexed: 01/18/2023]
Abstract
The pharmaceutical industry has faced declining research and development (R&D) productivity for decades. During the early 2010s, Pfizer saw its R&D productivity drop even more sharply than did its industry peers. As impactful medicines the company had developed and brought to patients in previous years lost exclusivity, Pfizer faced a steep patent cliff with a cumulative revenue impact of >US$28 billion through 2018. Since 2010, the company has embarked on a focused turnaround effort to improve R&D productivity. Although some efforts will need more time to prove themselves, there are early signs of a turnaround now, particularly in terms of Phase II success rates. Here, we share some learnings from a decade of experience as one of the largest R&D organizations in the industry.
Collapse
Affiliation(s)
- Shuang S Wu
- Pfizer, Inc, 1 Portland Street, Cambridge, MA 02139, USA
| | - Kathy Fernando
- Pfizer Inc, 235 East 42nd Street, New York City, NY 10017, USA
| | | | | | | | - Mikael Dolsten
- Pfizer Inc, 235 East 42nd Street, New York City, NY 10017, USA.
| |
Collapse
|
11
|
Burt T, Young G, Lee W, Kusuhara H, Langer O, Rowland M, Sugiyama Y. Phase 0/microdosing approaches: time for mainstream application in drug development? Nat Rev Drug Discov 2020; 19:801-818. [PMID: 32901140 DOI: 10.1038/s41573-020-0080-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
Phase 0 approaches - which include microdosing - evaluate subtherapeutic exposures of new drugs in first-in-human studies known as exploratory clinical trials. Recent progress extends phase 0 benefits beyond assessment of pharmacokinetics to include understanding of mechanism of action and pharmacodynamics. Phase 0 approaches have the potential to improve preclinical candidate selection and enable safer, cheaper, quicker and more informed developmental decisions. Here, we discuss phase 0 methods and applications, highlight their advantages over traditional strategies and address concerns related to extrapolation and developmental timelines. Although challenges remain, we propose that phase 0 approaches be at least considered for application in most drug development scenarios.
Collapse
Affiliation(s)
- Tal Burt
- Burt Consultancy LLC. talburtmd.com, New York, NY, USA. .,Phase-0/Microdosing Network. Phase-0Microdosing.org, New York, NY, USA.
| | - Graeme Young
- GlaxoSmithKline Research and Development Ltd, Ware, UK
| | - Wooin Lee
- Seoul National University, Seoul, Republic of Korea
| | | | - Oliver Langer
- Medical University of Vienna, Vienna, Austria.,AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | | | | |
Collapse
|
12
|
Resource interdependence and project termination: An analysis in the biopharmaceutical industry. INTERNATIONAL JOURNAL OF PROJECT MANAGEMENT 2020. [DOI: 10.1016/j.ijproman.2020.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
13
|
Kalaria DR, Parker K, Reynolds GK, Laru J. An industrial approach towards solid dosage development for first-in-human studies: Application of predictive science and lean principles. Drug Discov Today 2020; 25:505-518. [DOI: 10.1016/j.drudis.2019.12.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/16/2019] [Accepted: 12/30/2019] [Indexed: 01/24/2023]
|
14
|
Feijoo F, Palopoli M, Bernstein J, Siddiqui S, Albright TE. Key indicators of phase transition for clinical trials through machine learning. Drug Discov Today 2020; 25:414-421. [DOI: 10.1016/j.drudis.2019.12.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 12/22/2019] [Accepted: 12/30/2019] [Indexed: 02/08/2023]
|
15
|
McMeekin P, Lendrem DW, Lendrem BC, Pratt AG, Peck R, Isaacs JD, Jones D. Schrödinger's pipeline and the outsourcing of pharmaceutical innovation. Drug Discov Today 2019; 25:480-484. [PMID: 31835019 DOI: 10.1016/j.drudis.2019.11.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/19/2019] [Accepted: 11/30/2019] [Indexed: 12/22/2022]
Abstract
In the wake of the Global Financial Crisis (2007-2008) cheaper, softer money flooded the worldwide markets. Faced with historically low capital costs, the pharmaceutical industry chose to pay down debt through share buybacks rather than invest in research and development (R&D). Instead, the industry explored new R&D models for open innovation, such as open-sourcing, crowd-sourcing, public-private partnerships, innovation centres, Science Parks, and the wholesale outsourcing of pharmaceutical R&D. However, economic Greater Fool Theory suggests that outsourcing R&D was never likely to increase innovation. Ten years on, the period of cheaper and softer money is coming to an end. So how are things looking? And what happens next?
Collapse
Affiliation(s)
- Peter McMeekin
- School of Health, Community and Education Studies, Northumbria University, Newcastle upon Tyne, UK; Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK.
| | - Dennis W Lendrem
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; NIHR Biomedical Research Centre at Newcastle University & Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - B Clare Lendrem
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; NIHR Newcastle In Vitro Diagnostics Cooperative, Newcastle University, Newcastle upon Tyne, UK
| | - Arthur G Pratt
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; NIHR Biomedical Research Centre at Newcastle University & Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Richard Peck
- Pharma Research and Exploratory Development, Roche Innovation Center, Basel, Switzerland
| | - John D Isaacs
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; NIHR Biomedical Research Centre at Newcastle University & Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - David Jones
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; NIHR Biomedical Research Centre at Newcastle University & Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| |
Collapse
|
16
|
Lee DW, Lee SH, Choi N, Sung JH. Construction of pancreas–muscle–liver microphysiological system (MPS) for reproducing glucose metabolism. Biotechnol Bioeng 2019; 116:3433-3445. [DOI: 10.1002/bit.27151] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/28/2019] [Accepted: 08/13/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Dong Wook Lee
- Department of Chemical EngineeringHongik UniversitySeoul Republic of Korea
| | - Seung Hwan Lee
- Department of Bionano EngineeringHanyang UniversityAnsan Republic of Korea
- Nanosensor Research InstituteHanyang UniversityAnsan Republic of Korea
- Department of BionanotechnologyHanyang UniversityAnsan Republic of Korea
| | - Nakwon Choi
- Center for BioMicrosystems, Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical EngineeringHongik UniversitySeoul Republic of Korea
| |
Collapse
|
17
|
Nance E. Careers in nanomedicine and drug delivery. Adv Drug Deliv Rev 2019; 144:180-189. [PMID: 31260712 DOI: 10.1016/j.addr.2019.06.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/19/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
Abstract
Nanomedicine continues to be a rapidly growing and increasingly interdisciplinary field. The career opportunities available in nanomedicine are also numerous, yet not always obvious to the early-career scientist determining their individual track for maximal impact. This perspective provides a brief overview of the field of nanomedicine, then delves into the many career trajectories one could take in this field. The article concludes with thoughts on how to provide diverse training to increase supply for the variety of career paths, and the role that mentors can play in young scientists' development and exploration of these career paths.
Collapse
Affiliation(s)
- Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, United States of America; Department of Radiology, University of Washington, Seattle, WA, United States of America; Center on Human Development & Disability, University of Washington, Seattle, WA, United States of America; eScience Institute, Seattle, WA, United States of America; Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, United States of America.
| |
Collapse
|
18
|
Morgan P, Brown DG, Lennard S, Anderton MJ, Barrett JC, Eriksson U, Fidock M, Hamrén B, Johnson A, March RE, Matcham J, Mettetal J, Nicholls DJ, Platz S, Rees S, Snowden MA, Pangalos MN. Impact of a five-dimensional framework on R&D productivity at AstraZeneca. Nat Rev Drug Discov 2018; 17:167-181. [DOI: 10.1038/nrd.2017.244] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
19
|
Ringel MS, Choy MK. Do large mergers increase or decrease the productivity of pharmaceutical R&D? Drug Discov Today 2017. [DOI: 10.1016/j.drudis.2017.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
20
|
Visser SA, Bueters TJ. Assessment of translational risk in drug research: Role of biomarker classification and mechanism-based PKPD concepts. Eur J Pharm Sci 2017; 109S:S72-S77. [DOI: 10.1016/j.ejps.2017.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 08/12/2017] [Indexed: 01/10/2023]
|
21
|
Drug discovery effectiveness from the standpoint of therapeutic mechanisms and indications. Nat Rev Drug Discov 2017; 17:19-33. [PMID: 29075002 DOI: 10.1038/nrd.2017.194] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The productivity of the pharmaceutical industry has been widely discussed in recent years, particularly with regard to concerns that substantial expenditures on research and development have failed to translate into approved drugs. Various analyses of this productivity challenge have focused on aspects such as attrition rates at particular clinical phases or the physicochemical properties of drug candidates, but relatively little attention has been paid to how the industry has performed from the standpoint of the choice of therapeutic mechanisms and their intended indications. This article examines what the pharmaceutical industry has achieved in this respect by analysing comprehensive industry-wide data on the mechanism-indication pairs that have been investigated during the past 20 years. Our findings indicate several points and trends that we hope will be useful in understanding and improving the productivity of the industry, including areas in which the industry has had substantial success or failure and the relative extent of novelty in completed and ongoing projects.
Collapse
|
22
|
Yu PY, Gardner HL, Roberts R, Cam H, Hariharan S, Ren L, LeBlanc AK, Xiao H, Lin J, Guttridge DC, Mo X, Bennett CE, Coss CC, Ling Y, Phelps MA, Houghton P, London CA. Target specificity, in vivo pharmacokinetics, and efficacy of the putative STAT3 inhibitor LY5 in osteosarcoma, Ewing's sarcoma, and rhabdomyosarcoma. PLoS One 2017; 12:e0181885. [PMID: 28750090 PMCID: PMC5531494 DOI: 10.1371/journal.pone.0181885] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/07/2017] [Indexed: 12/21/2022] Open
Abstract
Background STAT3 is a transcription factor involved in cytokine and receptor kinase signal transduction that is aberrantly activated in a variety of sarcomas, promoting metastasis and chemotherapy resistance. The purpose of this work was to develop and test a novel putative STAT3 inhibitor, LY5. Methods and findings An in silico fragment-based drug design strategy was used to create LY5, a small molecule inhibitor that blocks the STAT3 SH2 domain phosphotyrosine binding site, inhibiting homodimerization. LY5 was evaluated in vitro demonstrating good biologic activity against rhabdomyosarcoma, osteosarcoma and Ewing’s sarcoma cell lines at high nanomolar/low micromolar concentrations, as well as specific inhibition of STAT3 phosphorylation without effects on other STAT3 family members. LY5 exhibited excellent oral bioavailability in both mice and healthy dogs, and drug absorption was enhanced in the fasted state with tolerable dosing in mice at 40 mg/kg BID. However, RNAi-mediated knockdown of STAT3 did not phenocopy the biologic effects of LY5 in sarcoma cell lines. Moreover, concentrations needed to inhibit ex vivo metastasis growth using the PuMA assay were significantly higher than those needed to inhibit STAT3 phosphorylation in vitro. Lastly, LY5 treatment did not inhibit the growth of sarcoma xenografts or prevent pulmonary metastasis in mice. Conclusions LY5 is a novel small molecule inhibitor that effectively inhibits STAT3 phosphorylation and cell proliferation at nanomolar concentrations. LY5 demonstrates good oral bioavailability in mice and dogs. However LY5 did not decrease tumor growth in xenograft mouse models and STAT3 knockdown did not induce concordant biologic effects. These data suggest that the anti-cancer effects of LY5 identified in vitro were not mediated through STAT3 inhibition.
Collapse
Affiliation(s)
- Peter Y. Yu
- Medical Student Research Program, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Heather L. Gardner
- Department of Veterinary Biosciences and Clinical Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Ryan Roberts
- Center for Childhood Cancer, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Hakan Cam
- Center for Childhood Cancer, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Seethalakshmi Hariharan
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Ling Ren
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amy K. LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hui Xiao
- Center for Childhood Cancer, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Jiayuh Lin
- Center for Childhood Cancer, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Denis C. Guttridge
- Arthur G. James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Chad E. Bennett
- Medicinal Chemistry Shared Resource, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Christopher C. Coss
- Pharmacoanalytic Shared Resource, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Yonghua Ling
- Pharmacoanalytic Shared Resource, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Mitch A. Phelps
- Pharmacoanalytic Shared Resource, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Peter Houghton
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Cheryl A. London
- Department of Veterinary Biosciences and Clinical Sciences, The Ohio State University, Columbus, Ohio, United States of America
- Cummings School of Veterinary Medicine, Tufts University, Grafton, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
23
|
van Hoogdalem E. Take Care of the Fast-in-Human Study. Clin Transl Sci 2017; 10:122-123. [PMID: 27981812 PMCID: PMC5421727 DOI: 10.1111/cts.12437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 12/12/2016] [Indexed: 11/29/2022] Open
|
24
|
Yildirim O, Gottwald M, Schüler P, Michel MC. Opportunities and Challenges for Drug Development: Public-Private Partnerships, Adaptive Designs and Big Data. Front Pharmacol 2016; 7:461. [PMID: 27999543 PMCID: PMC5138214 DOI: 10.3389/fphar.2016.00461] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/16/2016] [Indexed: 01/18/2023] Open
Abstract
Drug development faces the double challenge of increasing costs and increasing pressure on pricing. To avoid that lack of perceived commercial perspective will leave existing medical needs unmet, pharmaceutical companies and many other stakeholders are discussing ways to improve the efficiency of drug Research and Development. Based on an international symposium organized by the Medical School of the University of Duisburg-Essen (Germany) and held in January 2016, we discuss the opportunities and challenges of three specific areas, i.e., public–private partnerships, adaptive designs and big data. Public–private partnerships come in many different forms with regard to scope, duration and type and number of participants. They range from project-specific collaborations to strategic alliances to large multi-party consortia. Each of them offers specific opportunities and faces distinct challenges. Among types of collaboration, investigator-initiated studies are becoming increasingly popular but have legal, ethical, and financial implications. Adaptive trial designs are also increasingly discussed. However, adaptive should not be used as euphemism for the repurposing of a failed trial; rather it requires carefully planning and specification before a trial starts. Adaptive licensing can be a counter-part of adaptive trial design. The use of Big Data is another opportunity to leverage existing information into knowledge useable for drug discovery and development. Respecting limitations of informed consent and privacy is a key challenge in the use of Big Data. Speakers and participants at the symposium were convinced that appropriate use of the above new options may indeed help to increase the efficiency of future drug development.
Collapse
Affiliation(s)
- Oktay Yildirim
- Institute of Pharmacology, University Duisburg-Essen Essen, Germany
| | | | - Peter Schüler
- Department of Drug Development Services CNS, ICON Clinical Research Langen, Germany
| | - Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University Mainz, Germany
| |
Collapse
|
25
|
Yuh Liou S, Nakade S. Translational science in drug development: challenges and possible solutions. Nihon Yakurigaku Zasshi 2016; 148:290-295. [PMID: 27904006 DOI: 10.1254/fpj.148.290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
26
|
Vasaikar S, Bhatia P, Bhatia PG, Chu Yaiw K. Complementary Approaches to Existing Target Based Drug Discovery for Identifying Novel Drug Targets. Biomedicines 2016; 4:E27. [PMID: 28536394 PMCID: PMC5344266 DOI: 10.3390/biomedicines4040027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 02/07/2023] Open
Abstract
In the past decade, it was observed that the relationship between the emerging New Molecular Entities and the quantum of R&D investment has not been favorable. There might be numerous reasons but few studies stress the introduction of target based drug discovery approach as one of the factors. Although a number of drugs have been developed with an emphasis on a single protein target, yet identification of valid target is complex. The approach focuses on an in vitro single target, which overlooks the complexity of cell and makes process of validation drug targets uncertain. Thus, it is imperative to search for alternatives rather than looking at success stories of target-based drug discovery. It would be beneficial if the drugs were developed to target multiple components. New approaches like reverse engineering and translational research need to take into account both system and target-based approach. This review evaluates the strengths and limitations of known drug discovery approaches and proposes alternative approaches for increasing efficiency against treatment.
Collapse
Affiliation(s)
- Suhas Vasaikar
- Integrative Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Pooja Bhatia
- School of Biological Sciences, Indian Institute of Technology, Delhi 110016, India.
| | - Partap G Bhatia
- Department of Pharmaceutics and Pharmaceutical Microbiology, Usmanu Danfodiyo University, Sokoto 840231, Nigeria.
| | - Koon Chu Yaiw
- Experimental Cardiovascular Research Unit, Department of Medicine-Solna, Center for Molecular Medicine, Karolinska Institute, Stockholm 17177, Sweden.
| |
Collapse
|
27
|
Dickinson PA, Kesisoglou F, Flanagan T, Martinez MN, Mistry HB, Crison JR, Polli JE, Cruañes MT, Serajuddin AT, Müllertz A, Cook JA, Selen A. Optimizing Clinical Drug Product Performance: Applying Biopharmaceutics Risk Assessment Roadmap (BioRAM) and the BioRAM Scoring Grid. J Pharm Sci 2016; 105:3243-3255. [DOI: 10.1016/j.xphs.2016.07.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/20/2016] [Accepted: 07/26/2016] [Indexed: 01/22/2023]
|
28
|
Applying the best of oncology drug development paradigms to the non-malignant space. Drug Discov Today 2016; 21:1869-1872. [PMID: 27393488 DOI: 10.1016/j.drudis.2016.06.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/28/2016] [Accepted: 06/30/2016] [Indexed: 01/25/2023]
Abstract
With 80-90% of drugs entering the clinic not meeting regulatory approval (a high cost of failure), there is a major need for innovation in the clinical development space. Features of the new era of practice-changing innovation in oncology have included novel clinical trial designs incorporating multiple new molecular entities and/or multiple patient types, patient selection strategies (which allow detection of early signs of efficacy), and use of surrogate endpoints to achieve speedy regulatory approval. Disease areas beyond oncology could benefit from the application of specific aspects of these approaches. Here, we describe several such potential adaptations of the approaches, with scenarios and prerequisites, which could help reduce the costs of, and accelerate, clinical drug development with confidence.
Collapse
|
29
|
Tollman P, Panier V, Dosik D, Biondi P, Cuss F. Organizational effectiveness: a key to R&D productivity. Nat Rev Drug Discov 2016; 15:441-2. [DOI: 10.1038/nrd.2016.91] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
30
|
Leeson PD. Molecular inflation, attrition and the rule of five. Adv Drug Deliv Rev 2016; 101:22-33. [PMID: 26836397 DOI: 10.1016/j.addr.2016.01.018] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/12/2016] [Accepted: 01/18/2016] [Indexed: 12/18/2022]
Abstract
Physicochemical properties underlie all aspects of drug action and are critical for solubility, permeability and successful formulation. Specific physicochemical properties shown to be relevant to oral drugs are size, lipophilicity, ionisation, hydrogen bonding, polarity, aromaticity and shape. The rule of 5 (Ro5) and subsequent studies have raised awareness of the importance of compound quality amongst bioactive molecules. Lipophilicity, probably the most important physical property of oral drugs, has on average changed little over time in oral drugs, until increases in drugs published after 1990. In contrast other molecular properties such as average size have increased significantly. Factors influencing property inflation include the targets pursued, where antivirals frequently violate the Ro5, risk/benefit considerations, and variable drug discovery practices. The compounds published in patents from the pharmaceutical industry are on average larger, more lipophilic and less complex than marketed oral drugs. The variation between individual companies' patented compounds is due to different practices and not to the targets pursued. Overall, there is demonstrable physical property attrition in moving from patents to candidate drugs to marketed drugs. The pharmaceutical industry's recent poor productivity has been due, in part, to progression of molecules that are unable to unambiguously test clinical efficacy, and attrition can therefore be improved by ensuring candidate drug quality is 'fit for purpose.' The combined ligand efficiency (LE) and lipophilic ligand efficiency (LLE) values of many marketed drugs are optimised relative to other molecules acting at the same target. Application of LLE in optimisation can help identify improved leads, even with challenging targets that seem to require lipophilic ligands. Because of their targets, some projects may need to pursue 'beyond Ro5' physicochemical space; such projects will require non-standard lead generation and optimisation and should not dominate in a well-balanced portfolio. Compound quality is controllable by lead selection and optimisation and should not be a cause of clinical failure.
Collapse
Affiliation(s)
- Paul D Leeson
- Paul Leeson Consulting Ltd, The Malt House, Main Street, Congerstone, Nuneaton, Warks CV13 6LZ, UK.
| |
Collapse
|
31
|
Meanwell NA. Improving Drug Design: An Update on Recent Applications of Efficiency Metrics, Strategies for Replacing Problematic Elements, and Compounds in Nontraditional Drug Space. Chem Res Toxicol 2016; 29:564-616. [DOI: 10.1021/acs.chemrestox.6b00043] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Nicholas A. Meanwell
- Department of Discovery Chemistry, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut 06492, United States
| |
Collapse
|