1
|
Zhang N, Guo H, Zhang M, Wang C, Tian R, Shi Y, Duan Z. Analyzing collaboration dynamics in new drug R&D: A case study of two lipid-lowering drugs. Front Pharmacol 2025; 16:1516882. [PMID: 40520161 PMCID: PMC12162338 DOI: 10.3389/fphar.2025.1516882] [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] [Received: 11/06/2024] [Accepted: 05/19/2025] [Indexed: 06/18/2025] Open
Abstract
Introduction The advancements in biotechnology have ushered in a new age for drug development, characterized by increased collaborative efforts. Academic institutions, pharmaceutical firms, hospitals, foundations, and various other entities across different sectors are now joining forces more frequently to accelerate new drug innovation. However, there remains a limited understanding of how scientific and technological advancements are influencing these research collaborations. Methods In this study, the development of two types of lipid-lowering drugs served as case studies. A detailed network analysis was performed at the levels of authors, institutions, and countries to quantify the evolutionary trends in research collaboration. Results In the clinical research segment of the academic chain, papers resulting from collaborations tend to receive a higher citation count compared to other areas. However, there were notably fewer collaborative connections between authors transitioning from basic to developmental research and beyond. Collaboration models involving universities with enterprises, hospitals, or both are becoming more prevalent in biologics R&D. These models demonstrate effects of similarity and proximity. Additionally, there has been an increase in the involvement of developing countries in the research and development of new biologic drugs on a national and regional scale. Conclusion New drug R&D research collaboration patterns evolve spontaneously with productivity updates. In the future, it is necessary to enhance the involvement of pharmaceutical companies in the basic research phase of new drug development, continuously strengthen the relationships across all segments of the academic chain, and thoroughly boost the efficiency of transforming new drug R&D into practical applications.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Health Administration, School of Management, Shanxi Medical University, Taiyuan, China
| | - Huaqing Guo
- Department of Humanistic Medicine, School of Humanities and Social Sciences, Shanxi Medical University, Taiyuan, China
| | - Mengchao Zhang
- Department of Health Administration, School of Management, Shanxi Medical University, Taiyuan, China
| | - Chen Wang
- Department of Humanistic Medicine, School of Humanities and Social Sciences, Shanxi Medical University, Taiyuan, China
| | - Ruonan Tian
- Department of Humanistic Medicine, School of Humanities and Social Sciences, Shanxi Medical University, Taiyuan, China
| | - Yan Shi
- Department of Humanistic Medicine, School of Humanities and Social Sciences, Shanxi Medical University, Taiyuan, China
| | - Zhiguang Duan
- Department of Health Administration, School of Management, Shanxi Medical University, Taiyuan, China
- Department of Humanistic Medicine, School of Humanities and Social Sciences, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Ingham J, Ruan JL, Coelho MA. Breaking barriers: we need a multidisciplinary approach to tackle cancer drug resistance. BJC REPORTS 2025; 3:11. [PMID: 40016372 PMCID: PMC11868516 DOI: 10.1038/s44276-025-00129-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 01/15/2025] [Accepted: 02/11/2025] [Indexed: 03/01/2025]
Abstract
Most cancer-related deaths result from drug-resistant disease(1,2). However, cancer drug resistance is not a primary focus in drug development. Effectively mitigating and treating drug-resistant cancer will require advancements in multiple fields, including early detection, drug discovery, and our fundamental understanding of cancer biology. Therefore, successfully tackling drug resistance requires an increasingly multidisciplinary approach. A recent workshop on cancer drug resistance, jointly organised by Cancer Research UK, the Rosetrees Trust, and the UKRI-funded Physics of Life Network, brought together experts in cell biology, physical sciences, computational biology, drug discovery, and clinicians to focus on these key challenges and devise interdisciplinary approaches to address them. In this perspective, we review the outcomes of the workshop and highlight unanswered research questions. We outline the emerging hallmarks of drug resistance and discuss lessons from the COVID-19 pandemic and antimicrobial resistance that could help accelerate information sharing and timely adoption of research discoveries into the clinic. We envisage that initiatives that drive greater interdisciplinarity will yield rich dividends in developing new ways to better detect, monitor, and treat drug resistance, thereby improving treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- James Ingham
- Department of Physics, University of Liverpool, Liverpool, UK
| | - Jia-Ling Ruan
- Department of Oncology, University of Oxford, Oxford, UK
| | - Matthew A Coelho
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, UK.
| |
Collapse
|
3
|
Jeong JO, Kim M, Kim S, Lee KK, Choi H. Advanced Hydrogel Systems for Local Anesthetic Delivery: Toward Prolonged and Targeted Pain Relief. Gels 2025; 11:131. [PMID: 39996674 PMCID: PMC11854925 DOI: 10.3390/gels11020131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Local anesthetics (LAs) have been indispensable in clinical pain management, yet their limitations, such as short duration of action and systemic toxicity, necessitate improved delivery strategies. Hydrogels, with their biocompatibility, tunable properties, and ability to modulate drug release, have been extensively explored as platforms for enhancing LA efficacy and safety. This narrative review explores the historical development of LAs, their physicochemical properties, and clinical applications, providing a foundation for understanding the integration of hydrogels in anesthetic delivery. Advances in thermoresponsive, stimuli-responsive, and multifunctional hydrogels have demonstrated significant potential in prolonging analgesia and reducing systemic exposure in preclinical studies, while early clinical findings highlight the feasibility of thermoresponsive hydrogel formulations. Despite these advancements, challenges such as burst release, mechanical instability, and regulatory considerations remain critical barriers to clinical translation. Emerging innovations, including nanocomposite hydrogels, biofunctionalized matrices, and smart materials, offer potential solutions to these limitations. Future research should focus on optimizing hydrogel formulations, expanding clinical validation, and integrating advanced fabrication technologies such as 3D printing and artificial intelligence-driven design to enhance personalized pain management. By bridging materials science and anesthetic pharmacology, this review provides a comprehensive perspective on current trends and future directions in hydrogel-based LA delivery systems.
Collapse
Affiliation(s)
- Jin-Oh Jeong
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (J.-O.J.); (K.K.L.)
| | - Minjoo Kim
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Seonwook Kim
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
| | - Kyung Kwan Lee
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (J.-O.J.); (K.K.L.)
| | - Hoon Choi
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
4
|
Al-Tohamy A, Grove A. Targeting bacterial transcription factors for infection control: opportunities and challenges. Transcription 2025; 16:141-168. [PMID: 38126125 PMCID: PMC11970743 DOI: 10.1080/21541264.2023.2293523] [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: 08/31/2023] [Revised: 11/13/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
The rising threat of antibiotic resistance in pathogenic bacteria emphasizes the need for new therapeutic strategies. This review focuses on bacterial transcription factors (TFs), which play crucial roles in bacterial pathogenesis. We discuss the regulatory roles of these factors through examples, and we outline potential therapeutic strategies targeting bacterial TFs. Specifically, we discuss the use of small molecules to interfere with TF function and the development of transcription factor decoys, oligonucleotides that compete with promoters for TF binding. We also cover peptides that target the interaction between the bacterial TF and other factors, such as RNA polymerase, and the targeting of sigma factors. These strategies, while promising, come with challenges, from identifying targets to designing interventions, managing side effects, and accounting for changing bacterial resistance patterns. We also delve into how Artificial Intelligence contributes to these efforts and how it may be exploited in the future, and we touch on the roles of multidisciplinary collaboration and policy to advance this research domain.Abbreviations: AI, artificial intelligence; CNN, convolutional neural networks; DTI: drug-target interaction; HTH, helix-turn-helix; IHF, integration host factor; LTTRs, LysR-type transcriptional regulators; MarR, multiple antibiotic resistance regulator; MRSA, methicillin resistant Staphylococcus aureus; MSA: multiple sequence alignment; NAP, nucleoid-associated protein; PROTACs, proteolysis targeting chimeras; RNAP, RNA polymerase; TF, transcription factor; TFD, transcription factor decoying; TFTRs, TetR-family transcriptional regulators; wHTH, winged helix-turn-helix.
Collapse
Affiliation(s)
- Ahmed Al-Tohamy
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
- Department of Cell Biology, Biotechnology Research Institute, National Research Centre, Cairo, Egypt
| | - Anne Grove
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
5
|
Mikhael EM, Al-Jumaili AA, Jamal MY, Abdulazeez ZD. Current status and perceived challenges of collaborative research in a leading pharmacy college in Iraq: a qualitative study. BMC MEDICAL EDUCATION 2025; 25:61. [PMID: 39806318 PMCID: PMC11730822 DOI: 10.1186/s12909-025-06653-6] [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: 06/19/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
BACKGROUND Interdisciplinary collaboration among academic pharmacists is crucial for enhancing scientific research, discovering new drugs and modifying existing ones, besides solving pharmaceutical problems. This study aimed to explore the perception and experience of academic pharmacists regarding research collaboration. METHODS A qualitative study through one-to-one face-to-face interviews with faculty members at the University of Baghdad/College of Pharmacy was conducted from May to July/2023. Purposive and convenience strategies were used to enroll study participants. Thematic-analysis approach was used to analyze the data. RESULTS Twenty-four faculty members were interviewed. Most participants were female with ≥ 10 years of academic experience. Five themes emerged from the obtained data. The first theme, entitled the collaborative research was conducted at three different levels: college, national, and to a lesser extent, international. The second theme was facilitators of collaborative research. This theme includes two subthemes the reasons behind research collaboration and the encouraging characteristics of the researcher. Seeking scientific and technical support were the main reported reasons behind domestic collaborations, while supervising postgraduate students was the main reason for international collaborations. The third theme was the barriers to collaborative research. The complicated-university laws, besides limited-resources & funds, were barriers to collaborative research. Academic workload was the main challenge for domestic collaborations, whereas poor professional-networking was the main challenge for international collaborations. Ethical challenge in collaborative research was the fourth study theme. The last theme was the recommendations to improve future research collaboration. In this regard, most participants recommended enhancing academics' research-skills, increasing research funding, and simplifying regulatory issues to improve collaborative research. CONCLUSION Research collaborations occur at the domestic and, to a limited extent, at the international level. Academic workload, shortage of resources, and complicated university regulations are the main challenges for research collaborations. Enhancing research-skills, increasing research funding, and simplifying university regulations can improve research collaborations.
Collapse
Affiliation(s)
- Ehab Mudher Mikhael
- Clinical Pharmacy Department, College of Pharmacy, University of Baghdad, Baghdad, Iraq.
| | - Ali Azeez Al-Jumaili
- Clinical Pharmacy Department, College of Pharmacy, University of Baghdad, Baghdad, Iraq
| | - Mohammed Yawuz Jamal
- Clinical Pharmacy Department, College of Pharmacy, University of Baghdad, Baghdad, Iraq
| | | |
Collapse
|
6
|
Wei J, Dong R, Ma Y, Wang J, Tian S, Tu X, Mu Z, Liu YQ. Single-cell sequencing reveals that specnuezhenide protects against osteoporosis via activation of METTL3 in LEPR + BMSCs. Eur J Pharmacol 2024; 981:176908. [PMID: 39154827 DOI: 10.1016/j.ejphar.2024.176908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Osteoporosis (OP) has garnered significant attention due to its substantial morbidity and mortality rates, imposing considerable health burdens on societies worldwide. However, the molecular mechanisms underlying osteoporosis pathogenesis remain largely elusive, and the available therapeutic interventions are limited. Therefore, there is an urgent need for innovative strategies in the treatment of osteoporosis. PURPOSE The primary objective of this study was to elucidate the molecular mechanisms underlying osteoporosis pathogenesis using single-cell RNA sequencing (scRNA-seq), thereby proposing novel therapeutic agents. METHODS The mice osteoporosis model was established through bilateral ovariectomy. Micro-computed tomography (μCT) and hematoxylin and eosin (H&E) staining were employed to assess the pathogenesis of osteoporosis. scRNA-seq was utilized to identify and analyze distinct molecular mechanisms and sub-clusters. Gradient dilution analysis was used to obtain specific sub-clusters, which were further validated by immunofluorescence staining and flow cytometry analysis. Molecular docking and cellular thermal shift assay (CETSA) were applied for screening potential agents in the TCMSPs database. Alkaline phosphatase (ALP) activity and alizarin red S (ARS) staining were performed to evaluate the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Osteogenic organoids analysis was employed to assess the proliferation and sphere-forming ability of BMSCs. Quantitative real-time PCR (qRT-PCR) and western blot analysis were conducted to investigate signaling pathways. Wound healing assay and tube formation analysis were employed to evaluate the angiogenesis of endothelial cells. RESULTS The scRNA-seq analysis revealed the crucial role of LEPR+ BMSCs in the pathogenesis of osteoporosis, which was confirmed by immunofluorescence staining of the epiphysis. Subsequently, the LEPR+ BMSCs were obtained by gradient dilution analysis and identified by immunofluorescence staining and flow cytometry. Accordingly, specnuezhenide (Spe) was screened and identified as a potential compound targeting METTL3 from the TCMSPs database. Spe promoted bone formation as evidenced by μ-CT, and H&E analysis. Additionally, Spe enhanced the osteogenic capacity of LEPR+ BMSCs through ALP and ARS assay. Notably, METTL3 pharmacological inhibitors S-Adenosylhomocysteine (SAH) attenuated the aforementioned osteo-protective effects of Spe. Particularly, Spe enhanced the LEPR+ BMSCs-dependent angiogenesis through the secretion of SLIT3, which was abolished by SAH in LEPR+ BMSCs. CONCLUSION Collectively, these findings suggest that Spe could enhance the osteogenic potential of LEPR+ BMSCs and promote LEPR+ BMSCs-dependent angiogenesis by activating METTL3 in LEPR+ BMSCs, indicating its potential as an ideal therapeutic agent for clinical treatment of osteoporosis.
Collapse
Affiliation(s)
- Jun Wei
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Renchao Dong
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Ma
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuo Tian
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinyi Tu
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenqiang Mu
- Chongqing Key Laboratory of High Active Traditional Chinese Medicine Delivery System & Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, China.
| | - Yan-Qiu Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
7
|
Chakraborty C, Bhattacharya M, Lee SS, Wen ZH, Lo YH. The changing scenario of drug discovery using AI to deep learning: Recent advancement, success stories, collaborations, and challenges. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102295. [PMID: 39257717 PMCID: PMC11386122 DOI: 10.1016/j.omtn.2024.102295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Due to the transformation of artificial intelligence (AI) tools and technologies, AI-driven drug discovery has come to the forefront. It reduces the time and expenditure. Due to these advantages, pharmaceutical industries are concentrating on AI-driven drug discovery. Several drug molecules have been discovered using AI-based techniques and tools, and several newly AI-discovered drug molecules have already entered clinical trials. In this review, we first present the data and their resources in the pharmaceutical sector for AI-driven drug discovery and illustrated some significant algorithms or techniques used for AI and ML which are used in this field. We gave an overview of the deep neural network (NN) models and compared them with artificial NNs. Then, we illustrate the recent advancement of the landscape of drug discovery using AI to deep learning, such as the identification of drug targets, prediction of their structure, estimation of drug-target interaction, estimation of drug-target binding affinity, design of de novo drug, prediction of drug toxicity, estimation of absorption, distribution, metabolism, excretion, toxicity; and estimation of drug-drug interaction. Moreover, we highlighted the success stories of AI-driven drug discovery and discussed several collaboration and the challenges in this area. The discussions in the article will enrich the pharmaceutical industry.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, Odisha 756020, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, Gangwon-Do 24252, Republic of Korea
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Yi-Hao Lo
- Department of Family Medicine, Zuoying Armed Forces General Hospital, Kaohsiung 813204, Taiwan
- Shu-Zen Junior College of Medicine and Management, Kaohsiung 821004, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| |
Collapse
|
8
|
Liu X, Chen H, Liu Y, Zou J, Tian J, Tsomo T, Li M, Yu W. Social network analysis of a decade-long collaborative innovation network between hospitals and the biomedical industry in China. Sci Rep 2024; 14:11374. [PMID: 38762652 PMCID: PMC11102486 DOI: 10.1038/s41598-024-62082-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024] Open
Abstract
Collaborative innovation between hospitals and biomedical enterprises is crucial for ensuring breakthroughs in their development. This study explores the structural characteristics and examines the main roles of associated key actors of collaborative innovation between hospitals and biomedical enterprises in China. Using the jointly owned patent data within the country's healthcare industry, a decade-long collaborative innovation network between hospitals and biomedical enterprises in China was established and analyzed through social network analysis. The results revealed that the overall levels of collaborative innovation network density, collaborative frequency, and network connectivity were significantly low, especially in less-developed regions. In terms of actors with higher degree centrality, hospitals accounted for the majority, whereas a biomedical enterprise in Shenzhen had the highest degree centrality. Organizations in underdeveloped and northwest regions and small players were more likely to implement collaborative innovation. In conclusion, a collaborative innovation network between hospitals and biomedical enterprises in China demonstrated high dispersion and poor development levels. Stimulating organizations' initiatives for collaborative innovation may enhance quality and quantity of such innovation. Policy support and economic investments, strategic collaborative help, and resource and partnership optimization, especially for small players and in less-developed and northwest regions, should be encouraged to enhance collaborative innovation between hospitals and the biomedical industry in China and other similar countries or regions.
Collapse
Affiliation(s)
- Xiang Liu
- Affiliated Xihu Hospital, Hangzhou Medical College, Hangzhou, 310000, China
- Department of Respiratory Disease, The 903Rd Hospital of PLA, Hangzhou, 310000, China
| | - Hong Chen
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yue Liu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Zou
- Department of Pharmacy, The 903Rd Hospital of PLA, Hangzhou, 310000, China
| | - Jiahe Tian
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tenzin Tsomo
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Meina Li
- Department of Military Medical Service, Faculty of Military Health Service, Naval Medical University, Shanghai, 200433, China.
| | - Wenya Yu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
9
|
Sullivan JA, Gold ER. Exploring regulatory flexibility to create novel incentives to optimize drug discovery. Front Med (Lausanne) 2024; 11:1379966. [PMID: 38808140 PMCID: PMC11131371 DOI: 10.3389/fmed.2024.1379966] [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: 01/31/2024] [Accepted: 04/17/2024] [Indexed: 05/30/2024] Open
Abstract
Efforts by governments, firms, and patients to deliver pioneering drugs for critical health needs face a challenge of diminishing efficiency in developing those medicines. While multi-sectoral collaborations involving firms, researchers, patients, and policymakers are widely recognized as crucial for countering this decline, existing incentives to engage in drug development predominantly target drug manufacturers and thereby do little to stimulate collaborative innovation. In this mini review, we consider the unexplored potential within pharmaceutical regulations to create novel incentives to encourage a diverse set of actors from the public and private spheres to engage in the kind of collaborative knowledge exchange requisite for fostering enhanced innovation in early drug development.
Collapse
Affiliation(s)
- Jacqueline A. Sullivan
- Department of Philosophy, Rotman Institute of Philosophy and Western Institute for Neuroscience, Western University, London, ON, Canada
| | - E. Richard Gold
- Faculty of Law and Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
10
|
Marques L, Costa B, Pereira M, Silva A, Santos J, Saldanha L, Silva I, Magalhães P, Schmidt S, Vale N. Advancing Precision Medicine: A Review of Innovative In Silico Approaches for Drug Development, Clinical Pharmacology and Personalized Healthcare. Pharmaceutics 2024; 16:332. [PMID: 38543226 PMCID: PMC10975777 DOI: 10.3390/pharmaceutics16030332] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 11/12/2024] Open
Abstract
The landscape of medical treatments is undergoing a transformative shift. Precision medicine has ushered in a revolutionary era in healthcare by individualizing diagnostics and treatments according to each patient's uniquely evolving health status. This groundbreaking method of tailoring disease prevention and treatment considers individual variations in genes, environments, and lifestyles. The goal of precision medicine is to target the "five rights": the right patient, the right drug, the right time, the right dose, and the right route. In this pursuit, in silico techniques have emerged as an anchor, driving precision medicine forward and making this a realistic and promising avenue for personalized therapies. With the advancements in high-throughput DNA sequencing technologies, genomic data, including genetic variants and their interactions with each other and the environment, can be incorporated into clinical decision-making. Pharmacometrics, gathering pharmacokinetic (PK) and pharmacodynamic (PD) data, and mathematical models further contribute to drug optimization, drug behavior prediction, and drug-drug interaction identification. Digital health, wearables, and computational tools offer continuous monitoring and real-time data collection, enabling treatment adjustments. Furthermore, the incorporation of extensive datasets in computational tools, such as electronic health records (EHRs) and omics data, is also another pathway to acquire meaningful information in this field. Although they are fairly new, machine learning (ML) algorithms and artificial intelligence (AI) techniques are also resources researchers use to analyze big data and develop predictive models. This review explores the interplay of these multiple in silico approaches in advancing precision medicine and fostering individual healthcare. Despite intrinsic challenges, such as ethical considerations, data protection, and the need for more comprehensive research, this marks a new era of patient-centered healthcare. Innovative in silico techniques hold the potential to reshape the future of medicine for generations to come.
Collapse
Affiliation(s)
- Lara Marques
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (L.M.); (B.C.); (M.P.); (A.S.); (J.S.); (L.S.); (I.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Bárbara Costa
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (L.M.); (B.C.); (M.P.); (A.S.); (J.S.); (L.S.); (I.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Mariana Pereira
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (L.M.); (B.C.); (M.P.); (A.S.); (J.S.); (L.S.); (I.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- ICBAS—School of Medicine and Biomedical Sciences, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Abigail Silva
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (L.M.); (B.C.); (M.P.); (A.S.); (J.S.); (L.S.); (I.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Joana Santos
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (L.M.); (B.C.); (M.P.); (A.S.); (J.S.); (L.S.); (I.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Leonor Saldanha
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (L.M.); (B.C.); (M.P.); (A.S.); (J.S.); (L.S.); (I.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Isabel Silva
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (L.M.); (B.C.); (M.P.); (A.S.); (J.S.); (L.S.); (I.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Paulo Magalhães
- Coimbra Institute for Biomedical Imaging and Translational Research, Edifício do ICNAS, Polo 3 Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, 6550 Sanger Road, Office 465, Orlando, FL 328227-7400, USA;
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (L.M.); (B.C.); (M.P.); (A.S.); (J.S.); (L.S.); (I.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
11
|
Cheng F, Wang F, Tang J, Zhou Y, Fu Z, Zhang P, Haines JL, Leverenz JB, Gan L, Hu J, Rosen-Zvi M, Pieper AA, Cummings J. Artificial intelligence and open science in discovery of disease-modifying medicines for Alzheimer's disease. Cell Rep Med 2024; 5:101379. [PMID: 38382465 PMCID: PMC10897520 DOI: 10.1016/j.xcrm.2023.101379] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 08/15/2023] [Accepted: 12/19/2023] [Indexed: 02/23/2024]
Abstract
The high failure rate of clinical trials in Alzheimer's disease (AD) and AD-related dementia (ADRD) is due to a lack of understanding of the pathophysiology of disease, and this deficit may be addressed by applying artificial intelligence (AI) to "big data" to rapidly and effectively expand therapeutic development efforts. Recent accelerations in computing power and availability of big data, including electronic health records and multi-omics profiles, have converged to provide opportunities for scientific discovery and treatment development. Here, we review the potential utility of applying AI approaches to big data for discovery of disease-modifying medicines for AD/ADRD. We illustrate how AI tools can be applied to the AD/ADRD drug development pipeline through collaborative efforts among neurologists, gerontologists, geneticists, pharmacologists, medicinal chemists, and computational scientists. AI and open data science expedite drug discovery and development of disease-modifying therapeutics for AD/ADRD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA.
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Jian Tang
- Mila-Quebec Institute for Learning Algorithms and CIFAR AI Research Chair, HEC Montreal, Montréal, QC H3T 2A7, Canada
| | - Yadi Zhou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Zhimin Fu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Pengyue Zhang
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN 46037, USA
| | - Jonathan L Haines
- Cleveland Institute for Computational Biology, and Department of Population & Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jianying Hu
- IBM Research, Yorktown Heights, New York, NY 10598, USA
| | - Michal Rosen-Zvi
- AI for Accelerated Healthcare and Life Sciences Discovery, IBM Research Labs, Haifa 3498825, Israel; Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9190500, Israel
| | - Andrew A Pieper
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland OH 44106, USA; Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA; Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, UNLV, Las Vegas, NV 89154, USA
| |
Collapse
|
12
|
Li W, Kim M, Zhang K, Chen H, Jiang X, Harmanci A. COLLAGENE enables privacy-aware federated and collaborative genomic data analysis. Genome Biol 2023; 24:204. [PMID: 37697426 PMCID: PMC10496350 DOI: 10.1186/s13059-023-03039-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 08/16/2023] [Indexed: 09/13/2023] Open
Abstract
Growing regulatory requirements set barriers around genetic data sharing and collaborations. Moreover, existing privacy-aware paradigms are challenging to deploy in collaborative settings. We present COLLAGENE, a tool base for building secure collaborative genomic data analysis methods. COLLAGENE protects data using shared-key homomorphic encryption and combines encryption with multiparty strategies for efficient privacy-aware collaborative method development. COLLAGENE provides ready-to-run tools for encryption/decryption, matrix processing, and network transfers, which can be immediately integrated into existing pipelines. We demonstrate the usage of COLLAGENE by building a practical federated GWAS protocol for binary phenotypes and a secure meta-analysis protocol. COLLAGENE is available at https://zenodo.org/record/8125935 .
Collapse
Affiliation(s)
- Wentao Li
- Center for Secure Artificial Intelligence For hEalthcare (SAFE), D. Bradley McWilliams School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Miran Kim
- Department of Mathematics, Department of Computer Science, Hanyang University, Seoul, 04763, Republic of Korea
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763, Republic of Korea
- Bio-BigData Center, Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, 04763, Republic of Korea
| | - Kai Zhang
- Center for Secure Artificial Intelligence For hEalthcare (SAFE), D. Bradley McWilliams School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Center for Precision Health, D. Bradley McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Xiaoqian Jiang
- Center for Secure Artificial Intelligence For hEalthcare (SAFE), D. Bradley McWilliams School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Arif Harmanci
- Center for Secure Artificial Intelligence For hEalthcare (SAFE), D. Bradley McWilliams School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, 77030, USA.
- Center for Precision Health, D. Bradley McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
13
|
Ozieranski P, Saito H, Rickard E, Mulinari S, Ozaki A. International comparison of pharmaceutical industry payment disclosures in the UK and Japan: implications for self-regulation, public regulation, and transparency. Global Health 2023; 19:14. [PMID: 36869318 PMCID: PMC9985252 DOI: 10.1186/s12992-022-00902-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 12/20/2022] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Self-regulation of payment disclosure by pharmaceutical industry trade groups is a major global approach to increasing transparency of financial relationships between drug companies and healthcare professionals and organisations. Nevertheless, little is known about the relative strengths and weaknesses of self-regulation across countries, especially beyond Europe. To address this gap in research and stimulate international policy learning, we compare the UK and Japan, the likely strongest cases of self-regulation of payment disclosure in Europe and Asia, across three dimensions of transparency: disclosure rules, practices, and data. RESULTS The UK and Japanese self-regulation of payment disclosure had shared as well unique strengths and weaknesses. The UK and Japanese pharmaceutical industry trade groups declared transparency as the primary goal of payment disclosure, without, however, explaining the link between the two. The rules of payment disclosure in each country provided more insight into some payments but not others. Both trade groups did not reveal the recipients of certain payments by default, and the UK trade group also made the disclosure of some payments conditional on recipient consent. Drug company disclosure practices were more transparent in the UK, allowing for greater availability and accessibility of payment data and insight into underreporting or misreporting of payments by companies. Nevertheless, the share of payments made to named recipients was three times higher in Japan than in the UK, indicating higher transparency of disclosure data. CONCLUSIONS The UK and Japan performed differently across the three dimensions of transparency, suggesting that any comprehensive analysis of self-regulation of payment disclosure must triangulate analysis of disclosure rules, practices, and data. We found limited evidence to support key claims regarding the strengths of self-regulation, while often finding it inferior to public regulation of payment disclosure. We suggest how the self-regulation of payment disclosure in each country can be enhanced and, in the long run, replaced by public regulation to strengthen the industry's accountability to the public.
Collapse
Affiliation(s)
- Piotr Ozieranski
- Department of Social and Policy Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| | - Hiroaki Saito
- Department of Gastroenterology, Sendai Kousei Hospital, Sendai, Miyagi, Japan
| | - Emily Rickard
- Department of Social and Policy Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Shai Mulinari
- Department of Sociology, Lund University, Lund, Sweden
| | - Akihiko Ozaki
- Department of Breast Surgery, Jyoban Hospital of Tokiwa Foundation, Iwaki, Fukushima, Japan
| |
Collapse
|
14
|
Vinayak, Raghuvanshi A, kshitij A. Signatures of capacity development through research collaborations in artificial intelligence and machine learning. J Informetr 2023. [DOI: 10.1016/j.joi.2022.101358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
15
|
Multimodal single-cell omics analysis identifies epithelium-immune cell interactions and immune vulnerability associated with sex differences in COVID-19. Signal Transduct Target Ther 2021; 6:292. [PMID: 34330889 PMCID: PMC8322111 DOI: 10.1038/s41392-021-00709-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/25/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
Sex differences in the susceptibility of SARS-CoV-2 infection and severity have been controversial, and the underlying mechanisms of COVID-19 in a sex-specific manner remain understudied. Here we inspected sex differences in SARS-CoV-2 infection, hospitalization, admission to the intensive care unit (ICU), sera inflammatory biomarker profiling, and single-cell RNA-sequencing (scRNA-seq) profiles across nasal, bronchoalveolar lavage fluid (BALF), and peripheral blood mononuclear cells (PBMCs) from COVID-19 patients with varying degrees of disease severities. Our propensity score-matching observations revealed that male individuals have a 29% elevated likelihood of SARS-CoV-2 positivity, with a hazard ratio (HR) 1.32 (95% confidence interval [CI] 1.18–1.48) for hospitalization and HR 1.51 (95% CI 1.24–1.84) for admission to ICU. Sera from male patients at hospital admission had elevated neutrophil–lymphocyte ratio and elevated expression of inflammatory markers (C-reactive protein and procalcitonin). We found that SARS-CoV-2 entry factors, including ACE2, TMPRSS2, FURIN, and NRP1, have elevated expression in nasal squamous cells from male individuals with moderate and severe COVID-19. We observed male-biased transcriptional activation in SARS-CoV-2-infected macrophages from BALF and sputum samples, which offers potential molecular mechanism for sex-biased susceptibility to viral infection. Cell–cell interaction network analysis reveals potential epithelium–immune cell interactions and immune vulnerability underlying male-elevated disease severity and mortality in COVID-19. Mechanistically, monocyte-elevated expression of Toll-like receptor 7 (TLR7) and Bruton tyrosine kinase (BTK) is associated with severe outcomes in males with COVID-19. In summary, these findings provide basis to decipher immune responses underlying sex differences and designing sex-specific targeted interventions and patient care for COVID-19.
Collapse
|