1
|
Fischer NG, Lin TY, Xiang Y, Sang T, Ye Z. Emerging supramolecular and living materials in oral medicine. Trends Biotechnol 2025:S0167-7799(25)00091-5. [PMID: 40199625 DOI: 10.1016/j.tibtech.2025.03.006] [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: 08/27/2024] [Revised: 01/12/2025] [Accepted: 03/09/2025] [Indexed: 04/10/2025]
Abstract
Conventional dental materials lack the ability to promote regeneration, necessitating innovative approaches for repairing dental, oral, and craniofacial (DOC) tissues. Supramolecular materials with reversible, tunable interactions, and engineered living materials (ELMs) that mimic natural tissue dynamics, present a promising pathway towards regenerative solutions in oral medicine. This review introduces the potential of these biomaterials, focusing on their applications in oral bioprinting, therapeutic delivery, and organ-on-a-chip (OOC) systems. We discuss the integration of these technologies into clinical applications, and offer insights into future developments that may redefine oral healthcare by enabling the regeneration of complex, dynamic tissue structures and improving therapeutic outcomes in oral diseases.
Collapse
Affiliation(s)
- Nicholas G Fischer
- Minnesota Dental Research Center for Biomaterials and Biomechanics (MDRCBB), University of Minnesota, Minneapolis, MN, USA.
| | - Tsung-Yi Lin
- Department of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Yuanhui Xiang
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Ting Sang
- School of Stomatology of Nanchang University and Key Laboratory of Oral Biomedicine, Nanchang, Jiangxi Province, China
| | - Zhou Ye
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, University of Hong Kong, Hong Kong.
| |
Collapse
|
2
|
Hervieu L, Groo AC, Bellien J, Guerrot D, Malzert-Fréon A. Glucuronidation of orally administered drugs and the value of nanocarriers in strategies for its overcome. Pharmacol Ther 2025; 266:108773. [PMID: 39647710 DOI: 10.1016/j.pharmthera.2024.108773] [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: 08/21/2024] [Revised: 10/25/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
The gastrointestinal tract (GIT) plays a pivotal role in the absorption of orally administered drugs, with the small intestine serving as the primary site due to its extensive surface area and specialized cell types, including enterocytes and M cells. After oral administration, drugs are generally transported via the portal vein to the liver, where they undergo first-pass metabolism. This process involves various enzymatic reactions, including glucuronidation, facilitated by uridine diphosphate-glucuronosyltransferase (UGT), a major phase 2 reaction in mammalian metabolism. UGTs conjugate glucuronic acid to a wide array of endogenous and exogenous substrates, enhancing their solubility and excretion, but significantly affecting the bioavailability and therapeutic efficacy of drugs. UGT enzymes are ubiquitously distributed across tissues, prominently in the liver, but also in the GIT, kidneys, brain, and other organs where they play crucial roles in xenobiotic metabolism. Species-specific differences in UGT expression and activity impact the selection of animal models for pharmacological studies. Various experimental models - ranging from computational simulations (in silico) to laboratory experiments (in vitro) and animal studies (in vivo) - are employed throughout drug discovery and development to evaluate drug metabolism, including UGT activity. Effective strategies to counter pre-systemic metabolism are critical for improving drug bioavailability. This review explores several approaches including prodrugs, co-administration of specific molecules or use of inhibiting excipients in formulations. Strategies incorporating these excipients in nanoformulations demonstrate notable increases in drug absorption and bioavailability. This review highlights the importance of targeted delivery systems and excipient selection in overcoming metabolic barriers, aiming to optimize drug efficacy and patient outcomes.
Collapse
Affiliation(s)
- Laura Hervieu
- Université de Caen Normandie, CERMN UR4258, Normandie Univ, 14000 Caen, France; Université de Rouen Normandie, INSERM UMR1096, Normandie Univ, 76000 Rouen, France
| | - Anne-Claire Groo
- Université de Caen Normandie, CERMN UR4258, Normandie Univ, 14000 Caen, France
| | - Jérémy Bellien
- Université de Rouen Normandie, INSERM UMR1096, Normandie Univ, 76000 Rouen, France; Pharmacology Department, Rouen University Hospital, 76000 Rouen, France
| | - Dominique Guerrot
- Université de Rouen Normandie, INSERM UMR1096, Normandie Univ, 76000 Rouen, France; Nephrology Department, Rouen University Hospital, 76000 Rouen, France
| | | |
Collapse
|
3
|
Zhang N, Song J, Han Y. Research Progress of Phospholipid Vesicles in Biological Field. Biomolecules 2024; 14:1628. [PMID: 39766335 PMCID: PMC11726895 DOI: 10.3390/biom14121628] [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: 11/08/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
Due to their high biocompatibility, biodegradability, and facile surface functionalization, phospholipid vesicles as carriers have garnered significant attention in the realm of disease diagnosis and treatment. On the one hand, phospholipid vesicles can function as probes for the detection of various diseases by encapsulating nanoparticles, thereby enabling the precise localization of pathological changes and the monitoring of disease progression. On the other hand, phospholipid vesicles possess the capability to selectively target and deliver therapeutic agents, including drug molecules, genes and immune modulators, to affected sites, thereby enhancing the sustained release of these agents and improving therapeutic efficacy. Recent advancements in nanotechnology have led to an increased focus on the application of phospholipid vesicles in drug delivery, biological detection, gene therapy, and cell mimics. This review aims to provide a concise overview of the structure, characteristics, and preparation techniques of phospholipid vesicles of varying sizes. Furthermore, we will summarize the latest research developments regarding their use as nanomedicines and gene carriers in disease treatment. Additionally, we will elucidate the potential of phospholipid vesicles in facilitating the internalization, controlled release, and targeted delivery of therapeutic substrates. Through this review, we aspire to enhance the understanding of the evolution of phospholipid vesicles within the biological field, outline prospective research, and address the forthcoming challenges associated with phospholipid vesicles in disease diagnosis and treatment.
Collapse
Affiliation(s)
- Na Zhang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China;
| | - Jie Song
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China;
| | - Yuchun Han
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, CAS Research/Education Center for Excellence in Molecular Sciences, Beijing National Laboratory for Molecular Sciences (BNLMS), Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
4
|
P. K, Bhattacharya A, Du L, Silswal A, Li M, Cao J, Zhou Q, Zheng W, Liu TM, Koner AL. Activity-Based Dicyanoisophorone Derivatives: Fluorogenic Toolbox Enables Direct Visualization and Monitoring of Esterase Activity in Tumor Models. Anal Chem 2024; 96:18278-18286. [PMID: 39483052 PMCID: PMC11561878 DOI: 10.1021/acs.analchem.4c04721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/03/2024]
Abstract
The visualization and spatiotemporal monitoring of endogenous esterase activity are crucial for clinical diagnostics and treatment of liver diseases. Our research adopts a novel substrate hydrolysis-enzymatic activity (SHEA) approach using dicyanoisophorone-based fluorogenic ester substrates DCIP-R (R = R1-R6) to evaluate esterase preferences on diverse substrate libraries. Esterase-mediated hydrolysis yielded fluorescent DCIP-OH with a nanomolar detection limit in vitro. These probes effectively monitor ester hydrolysis kinetics with a turnover number of 4.73 s-1 and catalytic efficiency (kcat/Km) of 106 M-1 s-1 (DCIP-R1). Comparative studies utilizing two-photon imaging have indicated that substrates containing alkyl groups (DCIP-R1) as recognition elements exhibit enhanced enzymatic cleavage compared to those containing phenyl substitution on alkyl chains (DCIP-R4). Time-dependent variations in endogenous esterase levels were tracked in healthy and liver tumor models, especially in diethylnitrosamine (DEN)-induced tumors and HepG2-transplanted liver tumors. Overall, fluorescence signal quantifications demonstrated the excellent proficiency of DCIP-R1 in detecting esterase activity both in vitro and in vivo, showing promising potential for biomedical applications.
Collapse
Affiliation(s)
- Kavyashree P.
- Bionanotechnology
Lab, Department of Chemistry, Indian Institute
of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya
Pradesh, India
| | - Atri Bhattacharya
- Bionanotechnology
Lab, Department of Chemistry, Indian Institute
of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya
Pradesh, India
- Department
of Chemistry, University of Texas at Austin, Austin, Texas 78712-1224, United States of
America
| | - Lidong Du
- Institute
of Translational Medicine, Faculty of Health Sciences & Ministry
of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau 999078, China
| | - Akshay Silswal
- Bionanotechnology
Lab, Department of Chemistry, Indian Institute
of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya
Pradesh, India
| | - Moxin Li
- Institute
of Translational Medicine, Faculty of Health Sciences & Ministry
of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau 999078, China
| | - Jiayue Cao
- Institute
of Translational Medicine, Faculty of Health Sciences & Ministry
of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau 999078, China
| | - Qingqing Zhou
- Institute
of Translational Medicine, Faculty of Health Sciences & Ministry
of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau 999078, China
| | - Weiming Zheng
- Translational
Medicine R&D Center, Zhuhai UM Science
and Technology Research Institute, Zhuhai 519000, China
| | - Tzu-Ming Liu
- Institute
of Translational Medicine, Faculty of Health Sciences & Ministry
of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau 999078, China
| | - Apurba Lal Koner
- Bionanotechnology
Lab, Department of Chemistry, Indian Institute
of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya
Pradesh, India
| |
Collapse
|
5
|
Reddiar SB, Xie Y, Abdallah M, Han S, Hu L, Feeney OM, Gracia G, Anshabo A, Lu Z, Farooq MA, Styles IK, Phillips ARJ, Windsor JA, Porter CJH, Cao E, Trevaskis NL. Intestinal Lymphatic Biology, Drug Delivery, and Therapeutics: Current Status and Future Directions. Pharmacol Rev 2024; 76:1326-1398. [PMID: 39179383 DOI: 10.1124/pharmrev.123.001159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Historically, the intestinal lymphatics were considered passive conduits for fluids, immune cells, dietary lipids, lipid soluble vitamins, and lipophilic drugs. Studies of intestinal lymphatic drug delivery in the late 20th century focused primarily on the drugs' physicochemical properties, especially high lipophilicity, that resulted in intestinal lymphatic transport. More recent discoveries have changed our traditional view by demonstrating that the lymphatics are active, plastic, and tissue-specific players in a range of biological and pathological processes, including within the intestine. These findings have, in turn, inspired exploration of lymph-specific therapies for a range of diseases, as well as the development of more sophisticated strategies to actively deliver drugs or vaccines to the intestinal lymph, including a range of nanotechnologies, lipid prodrugs, and lipid-conjugated materials that "hitchhike" onto lymphatic transport pathways. With the increasing development of novel therapeutics such as biologics, there has been interest in whether these therapeutics are absorbed and transported through intestinal lymph after oral administration. Here we review the current state of understanding of the anatomy and physiology of the gastrointestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. We summarize the current state-of-the-art approaches to deliver drugs and quantify their uptake into the intestinal lymphatic system. Finally, and excitingly, we discuss recent examples of significant pharmacokinetic and therapeutic benefits achieved via intestinal lymphatic drug delivery. We also propose approaches to advance the development and clinical application of intestinal lymphatic delivery strategies in the future. SIGNIFICANCE STATEMENT: This comprehensive review details the understanding of the anatomy and physiology of the intestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. It highlights current state-of-the-art approaches to deliver drugs to the intestinal lymphatics and the shift toward the use of these strategies to achieve pharmacokinetic and therapeutic benefits for patients.
Collapse
Affiliation(s)
- Sanjeevini Babu Reddiar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Yining Xie
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Mohammad Abdallah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Luojuan Hu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Orlagh M Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Gracia Gracia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Abel Anshabo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Zijun Lu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Anthony R J Phillips
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - John A Windsor
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Enyuan Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| |
Collapse
|
6
|
Han S, Zou J, Xiao F, Xian J, Liu Z, Li M, Luo W, Feng C, Kong N. Nanobiotechnology boosts ferroptosis: opportunities and challenges. J Nanobiotechnology 2024; 22:606. [PMID: 39379969 PMCID: PMC11460037 DOI: 10.1186/s12951-024-02842-5] [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: 06/14/2024] [Accepted: 09/07/2024] [Indexed: 10/10/2024] Open
Abstract
Ferroptosis, distinct from apoptosis, necrosis, and autophagy, is a unique type of cell death driven by iron-dependent phospholipid peroxidation. Since ferroptosis was defined in 2012, it has received widespread attention from researchers worldwide. From a biochemical perspective, the regulation of ferroptosis is strongly associated with cellular metabolism, primarily including iron metabolism, lipid metabolism, and redox metabolism. The distinctive regulatory mechanism of ferroptosis holds great potential for overcoming drug resistance-a major challenge in treating cancer. The considerable role of nanobiotechnology in disease treatment has been widely reported, but further and more systematic discussion on how nanobiotechnology enhances the therapeutic efficacy on ferroptosis-associated diseases still needs to be improved. Moreover, while the exciting therapeutic potential of ferroptosis in cancer has been relatively well summarized, its applications in other diseases, such as neurodegenerative diseases, cardiovascular and cerebrovascular diseases, and kidney disease, remain underreported. Consequently, it is necessary to fill these gaps to further complete the applications of nanobiotechnology in ferroptosis. In this review, we provide an extensive introduction to the background of ferroptosis and elaborate its regulatory network. Subsequently, we discuss the various advantages of combining nanobiotechnology with ferroptosis to enhance therapeutic efficacy and reduce the side effects of ferroptosis-associated diseases. Finally, we analyze and discuss the feasibility of nanobiotechnology and ferroptosis in improving clinical treatment outcomes based on clinical needs, as well as the current limitations and future directions of nanobiotechnology in the applications of ferroptosis, which will not only provide significant guidance for the clinical applications of ferroptosis and nanobiotechnology but also accelerate their clinical translations.
Collapse
Affiliation(s)
- Shiqi Han
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Jianhua Zou
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Fan Xiao
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Jing Xian
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Ziwei Liu
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Meng Li
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Wei Luo
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Chan Feng
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China.
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
7
|
Martula E, Morak-Młodawska B, Jeleń M, Okechukwu PN. Analysis of Lipophilicity and Pharmacokinetic Parameters of Dipyridothiazine Dimers with Anticancer Potency. Pharmaceutics 2024; 16:1235. [PMID: 39339271 PMCID: PMC11435374 DOI: 10.3390/pharmaceutics16091235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Lipophilicity is an essential parameter of a compound that determines the solubility and pharmacokinetic properties that determine the transport of the drug to the molecular target. Dimers of dipyridothiazines are diazaphenothiazine derivatives exhibiting diverse anticancer potential in vitro, which is related to their affinity for histone deacetylase. In this study, the lipophilicity of 16 isomeric dipyridothiazine dimers was investigated theoretically and experimentally by reversed-phase thin-layer chromatography (RP-TLC) in an acetone-TRIS buffer (pH = 7.4). The relative lipophilicity parameter RM0 and specific hydrophobic surface area b were significantly intercorrelated, showing congeneric classes of dimers. The parameter RM0 was transformed into parameter logPTLC by use of the calibration curve. Molecular descriptors, ADMET parameters and probable molecular targets were determined in silico for analysis of the pharmacokinetic profile of the tested compounds showing anticancer activity. The analyzed compounds were tested in the context of Lipinski's rule of five, Ghose's rule and Veber's rule, confirming their bioavailability.
Collapse
Affiliation(s)
- Emilia Martula
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Małgorzata Jeleń
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Patrick Nwabueze Okechukwu
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
8
|
Anjali PB, Jawahar N, Praharsh Kumar MR, Jubie S, Selvamuthukumar S. Exploring the Anticonvulsant Properties of a Celecoxib-Phospholipid Conjugate: Synthesis, Activation, and Evaluation of Cytotoxicity. Drug Res (Stuttg) 2024; 74:296-301. [PMID: 38968953 DOI: 10.1055/a-2331-7114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
BACKGROUND Epilepsy poses a significant global health challenge, particularly in regions with limited financial resources hindering access to treatment. Recent research highlights neuroinflammation, particularly involving cyclooxygenase-2 (COX-2) pathways, as a promising avenue for epilepsy management. METHODS This study aimed to develop a Cyclooxygenase-2 inhibitor with potential anticonvulsant properties. A promising drug candidate was identified and chemically linked with phospholipids through docking analyses. The activation of this prodrug was assessed using phospholipase A2 (PLA2)-mediated hydrolysis studies. The conjugate's confirmation and cytotoxicity were evaluated using Fourier Transform Infrared Spectroscopy (FT-IR), Differential Scanning Calorimetry (DSC), and Sulphoramide B (SRB) assays. RESULTS Docking studies revealed that the Celecoxib-Phospholipid conjugate exhibited a superior affinity for PLA2 compared to other drug-phospholipid conjugates. FT-IR spectroscopy confirmed the successful synthesis of the conjugate, while DSC analysis confirmed its purity and formation. PLA2-mediated hydrolysis experiments demonstrated selective activation of the prodrug depending on PLA2 concentration. SRB experiments indicated dose-dependent cytotoxic effects of Celecoxib, phospholipid non-toxicity, and efficient celecoxib-phospholipid conjugation. CONCLUSION This study successfully developed a Celecoxib-phospholipid conjugate with potential anticonvulsant properties. The prodrug's specific activation and cytotoxicity profile makes it a promising therapeutic candidate. Further investigation into underlying mechanisms and in vivo studies is necessary to assess its translational potential fully.
Collapse
Affiliation(s)
- Puthusserikkunnu B Anjali
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Natarajan Jawahar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Mandadhi R Praharsh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Selvaraj Jubie
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | | |
Collapse
|
9
|
Ashique S, Bhowmick M, Pal R, Khatoon H, Kumar P, Sharma H, Garg A, Kumar S, Das U. Multi drug resistance in Colorectal Cancer- approaches to overcome, advancements and future success. ADVANCES IN CANCER BIOLOGY - METASTASIS 2024; 10:100114. [DOI: 10.1016/j.adcanc.2024.100114] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
|
10
|
Klimoszek D, Jeleń M, Dołowy M, Morak-Młodawska B. Study of the Lipophilicity and ADMET Parameters of New Anticancer Diquinothiazines with Pharmacophore Substituents. Pharmaceuticals (Basel) 2024; 17:725. [PMID: 38931392 PMCID: PMC11206290 DOI: 10.3390/ph17060725] [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: 05/16/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Lipophilicity is one of the principal parameters that describe the pharmacokinetic behavior of a drug, including its absorption, distribution, metabolism, elimination, and toxicity. In this study, the lipophilicity and other physicochemical, pharmacokinetic, and toxicity properties that affect the bioavailability of newly synthesized dialkylaminoalkyldiquinothiazine hybrids as potential drug candidates are presented. The lipophilicity, as RM0, was determined experimentally by the RP-TLC method using RP18 plates and acetone-TRIS buffer (pH 7.4) as the mobile phase. The chromatographic parameters of lipophilicity were compared to computationally calculated partition coefficients obtained by various types of programs such as iLOGP, XLOGP3, WLOGP, MLOGP, SILCOS-IT, LogP, logP, and milogP. In addition, the selected ADMET parameters were determined in silico using the SwissADME and pkCSM platforms and correlated with the experimental lipophilicity descriptors. The results of the lipophilicity study confirm that the applied algorithms can be useful for the rapid prediction of logP values during the first stage of study of the examined drug candidates. Of all the algorithms used, the biggest similarity to the chromatographic value (RM0) for certain compounds was seen with iLogP. It was found that both the SwissADME and pkCSM web tools are good sources of a wide range of ADMET parameters that describe the pharmacokinetic profiles of the studied compounds and can be fast and low-cost tools in the evaluation of examined drug candidates during the early stages of the development process.
Collapse
Affiliation(s)
- Daria Klimoszek
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska Street 4, 41-200 Sosnowiec, Poland; (D.K.); (M.D.)
| | - Małgorzata Jeleń
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska Street 4, 41-200 Sosnowiec, Poland;
| | - Małgorzata Dołowy
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska Street 4, 41-200 Sosnowiec, Poland; (D.K.); (M.D.)
| | - Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska Street 4, 41-200 Sosnowiec, Poland;
| |
Collapse
|
11
|
Dang Q, Li B, Jin B, Ye Z, Lou X, Wang T, Wang Y, Pan X, Hu Q, Li Z, Ji S, Zhou C, Yu X, Qin Y, Xu X. Cancer immunometabolism: advent, challenges, and perspective. Mol Cancer 2024; 23:72. [PMID: 38581001 PMCID: PMC10996263 DOI: 10.1186/s12943-024-01981-5] [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: 12/26/2023] [Accepted: 03/06/2024] [Indexed: 04/07/2024] Open
Abstract
For decades, great strides have been made in the field of immunometabolism. A plethora of evidence ranging from basic mechanisms to clinical transformation has gradually embarked on immunometabolism to the center stage of innate and adaptive immunomodulation. Given this, we focus on changes in immunometabolism, a converging series of biochemical events that alters immune cell function, propose the immune roles played by diversified metabolic derivatives and enzymes, emphasize the key metabolism-related checkpoints in distinct immune cell types, and discuss the ongoing and upcoming realities of clinical treatment. It is expected that future research will reduce the current limitations of immunotherapy and provide a positive hand in immune responses to exert a broader therapeutic role.
Collapse
Affiliation(s)
- Qin Dang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Borui Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bing Jin
- School of Clinical Medicine, Zhengzhou University, Zhengzhou, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xin Lou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Ting Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xuan Pan
- Department of Hepatobiliary Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Qiangsheng Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chenjie Zhou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Zhang M, Miao Y, Zhao C, Liu T, Wang X, Wang Z, Zhong W, He Z, Tian C, Sun J. Fine-tuning the activation behaviors of ternary modular cabazitaxel prodrugs for efficient and on-target oral anti-cancer therapy. Asian J Pharm Sci 2024; 19:100908. [PMID: 38623486 PMCID: PMC11017284 DOI: 10.1016/j.ajps.2024.100908] [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: 12/06/2023] [Revised: 01/27/2024] [Accepted: 02/25/2024] [Indexed: 04/17/2024] Open
Abstract
The disulfide bond plays a crucial role in the design of anti-tumor prodrugs due to its exceptional tumor-specific redox responsiveness. However, premature breaking of disulfide bonds is triggered by small amounts of reducing substances (e.g., ascorbic acid, glutathione, uric acid and tea polyphenols) in the systemic circulation. This may lead to toxicity, particularly in oral prodrugs that require more frequent and high-dose treatments. Fine-tuning the activation kinetics of these prodrugs is a promising prospect for more efficient on-target cancer therapies. In this study, disulfide, steric disulfide, and ester bonds were used to bridge cabazitaxel (CTX) to an intestinal lymph vessel-directed triglyceride (TG) module. Then, synthetic prodrugs were efficiently incorporated into self-nanoemulsifying drug delivery system (corn oil and Maisine CC were used as the oil phase and Cremophor EL as the surfactant). All three prodrugs had excellent gastric stability and intestinal permeability. The oral bioavailability of the disulfide bond-based prodrugs (CTX-(C)S-(C)S-TG and CTX-S-S-TG) was 11.5- and 19.1-fold higher than that of the CTX solution, respectively, demonstrating good oral delivery efficiency. However, the excessive reduction sensitivity of the disulfide bond resulted in lower plasma stability and safety of CTX-S-S-TG than that of CTX-(C)S-(C)S-TG. Moreover, introducing steric hindrance into disulfide bonds could also modulate drug release and cytotoxicity, significantly improving the anti-tumor activity even compared to that of intravenous CTX solution at half dosage while minimizing off-target adverse effects. Our findings provide insights into the design and fine-tuning of different disulfide bond-based linkers, which may help identify oral prodrugs with more potent therapeutic efficacy and safety for cancer therapy.
Collapse
Affiliation(s)
- Mingyang Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yifan Miao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Can Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tong Liu
- Liaoning Provincial Institute of Drug Inspection and Testing, Shenyang 110036, China
| | - Xiyan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zixuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wenxin Zhong
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Chutong Tian
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, Hangzhou 310058, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| |
Collapse
|
13
|
Wang K, Liao PY, Chang WC, Yang CR, Su YT, Wu PC, Wu YC, Hung YC, Akhtar N, Lai HC, Ma WL. Linoleate-pazopanib conjugation as active pharmacological ingredient to abolish hepatocellular carcinoma growth. Front Pharmacol 2024; 14:1281067. [PMID: 38293667 PMCID: PMC10824963 DOI: 10.3389/fphar.2023.1281067] [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: 08/21/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Small molecule compounds targeting multiple kinases involved in neoangiogenesis have shown survival benefits in patients with unresectable hepatocellular carcinoma (HCC). Nonetheless, despite the beneficial effects of multikinase inhibitors (MKIs), a lack of boosting adjuvant limits their objective response rate. Lipid conjugates have been used to improve delivery efficacy or pharmaceutical benefits for decades. However, the feasibility of utilizing lipid-drug conjugates (LDCs) in HCC regimens remains untested. In this study, oral feeding of linoleate-fluorescein isothiocyanate conjugates showed that the compound was well distributed in a spontaneous HCC mouse model. Therefore, a rationale design was developed for chemically synthesizing a linoleate-pazopanib conjugate (LAPC). The LAPC showed a significantly improved cytotoxicity compared to the parental drug pazopanib. Pazopanib's angiogenic suppressing signals were not observed in LAPC-treated HCC cells, potentially suggesting an altered mechanism of action (MOA). In an efficacy trial comparing placebo, oral pazopanib, and LAPC treatments in the hepatitis B virus transgene-related spontaneous HCC mouse model (HBVtg-HCC), the LAPC treatment demonstrated superior tumor ablating capacity in comparison to both placebo and pazopanib treatments, without any discernible systemic toxicity. The LAPC exposure is associated with an apoptosis marker (Terminal deoxynucleotidyl transferase dUTP nick end labeling [TUNEL]) and an enhanced ferroptosis (glutathione peroxidase 4 [GPX4]) potential in HBVtg-HCC tumors. Therefore, the LAPC showed excellent HCC ablative efficacy with altered MOA. The molecular mechanisms of the LAPC and LDCs for HCC therapeutics are of great academic interest. Further comprehensive preclinical trials (e.g., chemical-manufacture-control, toxicity, distribution, and pharmacokinetics/pharmacodynamics) are expected.
Collapse
Affiliation(s)
- Ke Wang
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Chinese Medicine Research and Development Center, and Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Pei-Yin Liao
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Chun Chang
- Department of Medical Research, Chinese Medicine Research and Development Center, and Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Cian-Ru Yang
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
| | - Yu-Ting Su
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
| | - Ping-Ching Wu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Institute of Oral Medicine and Department of Stomatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, Taiwan Innovation Center of Medical Devices and Technology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Yang-Chang Wu
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yao-Ching Hung
- Department of Medical Research, Chinese Medicine Research and Development Center, and Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
- Department of Obstetrics and Gynecology, Asia University Hospital, Taichung, Taiwan
| | - Najim Akhtar
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
| | - Hsueh-Chou Lai
- Department of Medical Research, Chinese Medicine Research and Development Center, and Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Lung Ma
- Graduate Institute of Biomedical Sciences, and Ph.D. Program for Health Science and Industry, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Chinese Medicine Research and Development Center, and Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
14
|
Chen A, Cai P, Peng Y, Guo M, Su Y, Cai T. The role of alkyl chain length in the melt and solution crystallization of paliperidone aliphatic prodrugs. IUCRJ 2024; 11:23-33. [PMID: 37962472 PMCID: PMC10833388 DOI: 10.1107/s2052252523009582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023]
Abstract
Fatty acid-derivative prodrugs have been utilized extensively to improve the physicochemical, biopharmaceutical and pharmacokinetic properties of active pharmaceutical ingredients. However, to our knowledge, the crystallization behavior of prodrugs modified with different fatty acids has not been explored. In the present work, a series of paliperidone aliphatic prodrugs with alkyl chain lengths ranging from C4 to C16 was investigated with respect to crystal structure, crystal morphology and crystallization kinetics. The paliperidone derivatives exhibited isostructural crystal packing, despite the different alkyl chain lengths, and crystallized with the dominant (100) face in both melt and solution. The rate of crystallization for paliperidone derivatives in the melt increases with alkyl chain length owing to greater molecular mobility. In contrast, the longer chains prolong the nucleation induction time and reduce the crystal growth kinetics in solution. The results show a correlation between difficulty of nucleation in solution and the interfacial energy. This work provides insight into the crystallization behavior of paliperidone aliphatic prodrugs and reveals that the role of alkyl chain length in the crystallization behavior has a strong dependence on the crystallization method.
Collapse
Affiliation(s)
- An Chen
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Peishan Cai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yayun Peng
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Minshan Guo
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yuan Su
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Ting Cai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| |
Collapse
|
15
|
Zhang M, Su Y, Li J, Chang C, Gu L, Yang Y. Fabrication of phosphatidylcholine-EGCG nanoparticles with sustained release in simulated gastrointestinal digestion and their transcellular permeability in a Caco-2 monolayer model. Food Chem 2023; 437:137580. [PMID: 39491254 DOI: 10.1016/j.foodchem.2023.137580] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/24/2023] [Accepted: 09/21/2023] [Indexed: 11/05/2024]
Abstract
In this study, we prepared phosphatidylcholine (PC)-EGCG complex nanoparticles (P-E NPs) by solvent reflux method. The physicochemical properties, in vitro digestion, uptake in Caco-2 cells, and bidirectional permeability of P-E NPs were systematically investigated. The constructed P-E1.5:1 NPs had an average particle size of 118 nm, a ζ-potential of -37.8 mV, and a polymerization dispersion index (PDI) of 0.16. The encapsulation efficiency (EE) of EGCG was 85.0% and the loading capacity (LC) was 24.4%. UV spectra, FTIR, XRD and intermolecular force results indicated that hydrophobic, electrostatic and hydrogen bonding interactions contributed to formate P-E1.5:1 NPs. P-E1.5:1 NPs exhibited first-order kinetics sustained release properties in simulated gastrointestinal digestion. Furthermore, P-E1.5:1 NPs were able to enhance absorptive transport and inhibit efflux transport mediated by MRP2 and P-gp compared to EGCG. These results indicated that P-E1.5:1 NPs may be a potential strategy to ameliorate EGCG bioavailability.
Collapse
Affiliation(s)
- Mengya Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yujie Su
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Junhua Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Cuihua Chang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Luping Gu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yanjun Yang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
16
|
Yang W, Lipert M, Nofsinger R. Current screening, design, and delivery approaches to address low permeability of chemically synthesized modalities in drug discovery and early clinical development. Drug Discov Today 2023; 28:103685. [PMID: 37356613 DOI: 10.1016/j.drudis.2023.103685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/09/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
A drug's permeability across biological membranes is a key property associated with the successful development of an orally absorbed drug candidate. Although a variety of methods are available for predicting and assessing permeability, some are more preferred than others at specific stages of drug discovery and development across the pharmaceutical industry. Permeability measurements may be interpreted differently depending on the chosen method. Herein, we present a refreshed perspective on the screening approaches and philosophy in permeability evaluation, from early drug discovery to early clinical development. Additionally, we review and discuss chemical design and drug delivery technologies that can be leveraged to overcome permeability challenges, which are increasingly being used with emerging modalities.
Collapse
Affiliation(s)
- Wenzhan Yang
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, USA.
| | - Maya Lipert
- Molecular Profiling and Drug Delivery, Small Molecule CMC Development, AbbVie, Inc., North Chicago, IL, USA
| | | |
Collapse
|
17
|
Li Y, Zhan H, Wu J, Yu J, Cao G, Wu X, Guo B, Liu X, Fan Y, Hu J, Li X, Wu H, Wang Y, Chen Y, Xu X, Yu P, Zhang J. Population Pharmacokinetics and Exposure-Safety of Lipophilic Conjugates Prodrug DP-VPA in Healthy Chinese Subjects for Dose Regime Exploring. Eur J Pharm Biopharm 2023:S0939-6411(23)00111-X. [PMID: 37142130 DOI: 10.1016/j.ejpb.2023.04.023] [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: 02/01/2023] [Revised: 04/07/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
Phospholipid-valproic acid (DP-VPA)is a prodrug for treating epilepsy. The present study explored the pharmacokinetics (PK) and exposure safety of DP-VPA to provide a basis for future studies exploring the safe dosage and therapeutic strategies for epilepsy. The study included a randomized placebo-controlled dose-escalation tolerance evaluation trial and a randomized triple crossover food-effect trial in healthy Chinese volunteers. A population pharmacokinetic (PopPK) model was established to analyze the PK of DP-VPA and active metabolite VPA. The exposure safety was assessed with the adverse drug reaction (ADR) in CNS. The PopPK of DP-VPA and metabolite VPA fitted a two-compartment model coupling one-compartment with Michaelis-Menten metabolite kinetics and first-order elimination. The absorption processes after single oral administration of DP-VPA tablet demonstrated nonlinear characteristics, including 0-order kinetic phase and time-dependent phase fitting Weibull distribution. The final model indicated that the DP-VPA PK was significantly affected by dosage and food. The exposure-safety relationship demonstrated a generalized linear regression; mild/moderate ADRs occurred in some subjects with 600 mg and all subjects with 1500 mg of DP-VPA, and no severe ADRs were reported up to 2400 mg. In conclusion, the study established a PopPK model describing the processing of DP-VPA and VPA in healthy Chinese subjects. DP-VPA showed good tolerance after a single dose of 600-2400 mg with nonlinear PK and was affected by dosage and food. Based on the association between neurological ADRs and higher exposure to DP-VPA by exposure-safety analysis, 900-1200 mg was recommended for subsequent study of safety and clinical effectiveness.
Collapse
Affiliation(s)
- Yi Li
- National Clinical Research Center for Geriatric Diseases, Huashan Hospital, Fudan University, Shanghai, China; Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; China Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Shanghai, China
| | - Huizhong Zhan
- Office of Drug Clinical Trail Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Jufang Wu
- Phase I Clinical Research Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jicheng Yu
- Phase I Clinical Research Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Guoying Cao
- Phase I Clinical Research Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaojie Wu
- Phase I Clinical Research Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Beining Guo
- National Clinical Research Center for Geriatric Diseases, Huashan Hospital, Fudan University, Shanghai, China; Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; China Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Shanghai, China
| | - Xiaofen Liu
- National Clinical Research Center for Geriatric Diseases, Huashan Hospital, Fudan University, Shanghai, China; Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; China Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Shanghai, China
| | - Yaxin Fan
- National Clinical Research Center for Geriatric Diseases, Huashan Hospital, Fudan University, Shanghai, China; Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; China Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Shanghai, China
| | - Jiali Hu
- National Clinical Research Center for Geriatric Diseases, Huashan Hospital, Fudan University, Shanghai, China; Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; China Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Shanghai, China
| | - Xin Li
- National Clinical Research Center for Geriatric Diseases, Huashan Hospital, Fudan University, Shanghai, China; Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; China Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Shanghai, China
| | - Hailan Wu
- National Clinical Research Center for Geriatric Diseases, Huashan Hospital, Fudan University, Shanghai, China; Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; China Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Shanghai, China
| | - Yu Wang
- National Clinical Research Center for Geriatric Diseases, Huashan Hospital, Fudan University, Shanghai, China; Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; China Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Shanghai, China
| | - Yuancheng Chen
- Phase I Clinical Research Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoyong Xu
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Peimin Yu
- Institute of Neurology, Huashan Hospital, Fudan University, WHO Collaborating Centre for Research and Training in Neurosciences, Shanghai, China.
| | - Jing Zhang
- Phase I Clinical Research Center, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Geriatric Diseases, Huashan Hospital, Fudan University, Shanghai, China; Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; China Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Shanghai, China.
| |
Collapse
|
18
|
Cheng Y, Zhong C, Yan S, Chen C, Gao X. Structure modification: a successful tool for prodrug design. Future Med Chem 2023; 15:379-393. [PMID: 36946236 DOI: 10.4155/fmc-2022-0309] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
Prodrug strategy is critical for innovative drug development. Structural modification is the most straightforward and effective method to develop prodrugs. Improving drug defects and optimizing the physical and chemical properties of a drug, such as lipophilicity and water solubility, changing the way of administration can be achieved through specific structural modification. Designing prodrugs by linking microenvironment-responsive groups to the prototype drugs is of great help in enhancing drug targeting. In the meantime, making connections between prodrugs and suitable drug delivery systems could realize drug loading increases, greater stability, bioavailability and drug release control. In this paper, lipidic, water-soluble, pH-responsive, redox-sensitive and enzyme-activatable prodrugs are reviewed on the basis of structural modification.
Collapse
Affiliation(s)
- Yuexuan Cheng
- College of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | - Chunhong Zhong
- College of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | - Shujing Yan
- College of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | - Chunli Chen
- College of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
- Engineering Research Center of Xinjiang & Central Asian Medicine Resources, Ministry of Education, Urumqi, Xinjiang, 830011, China
| | - Xiaoli Gao
- College of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
- Engineering Research Center of Xinjiang & Central Asian Medicine Resources, Ministry of Education, Urumqi, Xinjiang, 830011, China
| |
Collapse
|
19
|
smProdrugs: A repository of small molecule prodrugs. Eur J Med Chem 2023; 249:115153. [PMID: 36724634 DOI: 10.1016/j.ejmech.2023.115153] [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: 10/20/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
In modern drug discovery and development, the prodrug approach has become a crucial strategy for enhancing the pharmacokinetic profiles of drugs. A prodrug is a chemical compound, which gets metabolized into a pharmacologically active form (drug) inside the body after its administration. In the current work, we report 'smProdrugs' (http://cheminfolab.in/databases/prodrug/), which is one of the first exclusive databases on small molecule prodrugs. It stores the structures, physicochemical properties and experimental ADMET data manually curated from literature. SmProdrugs lists 626 small molecule prodrugs and their active compounds with the above mentioned experimental data from 1808 research articles and 61 patents have been stored. The information page of each record gives the structures and properties of the prodrug and the active drug side by side which makes it easy for the user to instantly compare them. The structural modifications in the prodrug/active drugs are highlighted in a different colour for easy comparison. Experimental data has been curated from the downloaded PubMed and patent articles and were catalogued in a tabular form with more than 25 fields under sub-sections i) name and structures of the prodrugs and their active compounds, ii) mode of activation of the prodrug and enzyme/biocatalyst involved in the conversion, iii) indications/disease, iv) pharmacological target, v) experimental pharmacokinetic properties such as solubility, absorption, volume of distribution, half-life, clearance etc. and vi) information on the purpose/gain from the prodrug strategies. Considering the ever expanding utility of the prodrug approach smProdrugs will be of great use to the scientific community working on rational design of small molecule prodrugs.
Collapse
|
20
|
Zhang Y, Wang Y, Li X, Nie D, Liu C, Gan Y. Ligand-modified nanocarriers for oral drug delivery: Challenges, rational design, and applications. J Control Release 2022; 352:813-832. [PMID: 36368493 DOI: 10.1016/j.jconrel.2022.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/15/2022]
Abstract
Ligand-modified nanocarriers (LMNCs) specific to their targets have attracted increasing interest for enhanced oral drug delivery in recent decades. Although the design of LMNCs for enhanced endocytosis and improved exposure of the loaded drugs through the oral route has received abundant attention, it remains unclear how the design influences their transcellular process, especially the key factors affecting their functions. This review discusses the extracellular and cellular barriers to orally administered LMNCs in the gastrointestinal (GI) tract and new discoveries regarding the GI protein corona and the sequential transport barriers that impede the preplanned movements of LMNCs after oral administration. Furthermore, innovative progress in considering key factors (including target selection, ligand properties, and other important factors) in the rational design of LMNCs for oral drug delivery is presented. In particular, some factors that endow LMNCs with efficient transcytosis rather than only endocytosis are highlighted. Finally, the prospects of orally administered LMNCs in disease therapy for the enhanced oral/local bioavailability of active pharmaceutical ingredients, as well as emerging delivery routes, such as lymphatic drug delivery and systemic location-specific drug release based on oral transcellular LMNCs, are discussed.
Collapse
Affiliation(s)
- Yaqi Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaying Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Di Nie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chang Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yong Gan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, Beijing 100050, China.
| |
Collapse
|
21
|
Gao F, Huang H, Sheng C, He S. Efficient synthesis of artificial pharmaceutical solid-phase modules for constructing aptamer-drug conjugates. Bioorg Chem 2022; 126:105919. [DOI: 10.1016/j.bioorg.2022.105919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/22/2022] [Accepted: 05/28/2022] [Indexed: 02/07/2023]
|
22
|
A lymphatic-absorbed multi-targeted kinase inhibitor for myelofibrosis therapy. Nat Commun 2022; 13:4730. [PMID: 35977945 PMCID: PMC9386018 DOI: 10.1038/s41467-022-32486-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 07/19/2022] [Indexed: 11/09/2022] Open
Abstract
Activation of compensatory signaling nodes in cancer often requires combination therapies that are frequently plagued by dose-limiting toxicities. Intestinal lymphatic drug absorption is seldom explored, although reduced toxicity and sustained drug levels would be anticipated to improve systemic bioavailability. A potent orally bioavailable multi-functional kinase inhibitor (LP-182) is described with intrinsic lymphatic partitioning for the combined targeting of phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signaling pathways without observable toxicity. We demonstrate selectivity and therapeutic efficacy through reduction of downstream kinase activation, amelioration of disease phenotypes, and improved survival in animal models of myelofibrosis. Our further characterization of synthetic and physiochemical properties for small molecule lymphatic uptake will support continued advancements in lymphatropic therapy for altering disease trajectories of a myriad of human disease indications. Combination therapies simultaneously inhibiting different therapeutic targets in cancer is challenged by individual pharmacokinetic profiles. Here, the authors generate an orally provided multi-targeted kinase inhibitor that is lymphatic absorbed and increases survival in a murine model of myelofibrosis.
Collapse
|
23
|
Long chain triglyceride-lipid formulation promotes the oral absorption of the lipidic prodrugs through coincident intestinal behaviors. Eur J Pharm Biopharm 2022; 176:122-132. [DOI: 10.1016/j.ejpb.2022.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/24/2022] [Accepted: 05/21/2022] [Indexed: 11/22/2022]
|
24
|
Karthika C, Sureshkumar R, Zehravi M, Akter R, Ali F, Ramproshad S, Mondal B, Kundu MK, Dey A, Rahman MH, Antonescu A, Cavalu S. Multidrug Resistance in Cancer Cells: Focus on a Possible Strategy Plan to Address Colon Carcinoma Cells. Life (Basel) 2022; 12:811. [PMID: 35743842 PMCID: PMC9224881 DOI: 10.3390/life12060811] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 05/26/2022] [Indexed: 12/20/2022] Open
Abstract
Even though various treatment methods are available for cancer, the death curve is not reducing. The diagnosis of cancer at the fourth stage and drug resistance are the leading reasons for treatment failure and lower survival rates. In this review article, we summarize the possible pitfalls during cancer treatment in general, which mainly include multidrug resistance, and propose a hypothesis for colorectal cancer specifically. We also evaluate multidrug resistance in cancer in general and colorectal cancer in particular and hypothesize a concept based on combination therapy with 5-fluorouracil, curcumin, and lipids for the possible management of colorectal cancer. In addition, a hypothetical approach, combining a synthetic agent and a natural chemotherapeutic agent, to treating colorectal cancer is also discussed. This hypothesis could improve the management of colorectal cancer.
Collapse
Affiliation(s)
- Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, India;
| | - Raman Sureshkumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, India;
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy Girls Section, Prince Sattam Bin Abdul Aziz University, Alkharj 11942, Saudi Arabia;
| | - Rokeya Akter
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, 24, Wonju 26426, Korea;
| | - Faraat Ali
- Department of Licensing and Enforcement, Laboratory Services, Botswana Medicines Regulatory Authority (BoMRA), Gaborone 999106, Botswana;
| | - Sarker Ramproshad
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (S.R.); (B.M.)
| | - Banani Mondal
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (S.R.); (B.M.)
| | | | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, India;
| | - Md. Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, 24, Wonju 26426, Korea;
| | - Angela Antonescu
- Faculty of Medicine and Pharmacy, University of Oradea, Pta 1 Decembrie 10, 410087 Oradea, Romania;
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Pta 1 Decembrie 10, 410087 Oradea, Romania;
| |
Collapse
|
25
|
Quach T, Hu L, Han S, Lim SF, Senyschyn D, Yadav P, Trevaskis NL, Simpson JS, Porter CJH. Triglyceride-Mimetic Prodrugs of Buprenorphine Enhance Oral Bioavailability via Promotion of Lymphatic Transport. Front Pharmacol 2022; 13:879660. [PMID: 35496278 PMCID: PMC9039622 DOI: 10.3389/fphar.2022.879660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/15/2022] [Indexed: 11/24/2022] Open
Abstract
Buprenorphine (BUP) is a potent opioid analgesic that is widely used for severe pain management and opioid replacement therapy. The oral bioavailability of BUP, however, is significantly limited by first-pass metabolism. Previous studies have shown that triglyceride (TG) mimetic prodrugs of the steroid hormone testosterone circumvent first-pass metabolism by directing drug transport through the intestinal lymphatics, bypassing the liver. The current study expanded this prodrug strategy to BUP. Here different self-immolative (SI) linkers were evaluated to conjugate BUP to the 2 position of the TG backbone via the phenol group on BUP. The SI linkers were designed to promote drug release in plasma. Lipolysis of the prodrug in the intestinal tract was examined via incubation with simulated intestinal fluid (SIF), and potential for parent drug liberation in the systemic circulation was evaluated via incubation in rat plasma. Lymphatic transport and bioavailability studies were subsequently conducted in mesenteric lymph duct or carotid artery-cannulated rats, respectively. TG prodrug derivatives were efficiently transported into the lymphatics (up to 45% of the dose in anaesthetised rats, vs. less than 0.1% for BUP). Incorporation of the SI linkers facilitated BUP release from the prodrugs in the plasma and in concert with high lymphatic transport led to a marked enhancement in oral bioavailability (up to 22-fold) compared to BUP alone. These data suggest the potential to develop an orally bioavailable BUP product which may have advantages with respect to patient preference when compared to current sublingual, transdermal patch or parenteral formulations.
Collapse
Affiliation(s)
- Tim Quach
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Luojuan Hu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- *Correspondence: Sifei Han, ; Christopher J. H. Porter,
| | - Shea F. Lim
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Danielle Senyschyn
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Preeti Yadav
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Natalie L. Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Jamie S. Simpson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Christopher J. H. Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- *Correspondence: Sifei Han, ; Christopher J. H. Porter,
| |
Collapse
|
26
|
Markovic M, Ben-Shabat S, Nagendra Manda J, Abramov-Harpaz K, Regev C, Miller Y, Aponick A, Zimmermann EM, Dahan A. PLA 2-Triggered Activation of Cyclosporine-Phospholipid Prodrug as a Drug Targeting Approach in Inflammatory Bowel Disease Therapy. Pharmaceutics 2022; 14:pharmaceutics14030675. [PMID: 35336048 PMCID: PMC8950246 DOI: 10.3390/pharmaceutics14030675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Oral medication with activity specifically at the inflamed sites throughout the gastrointestinal tract and limited systemic exposure would be a major advance in our therapeutic approach to inflammatory bowel disease (IBD). For this purpose, we have designed a prodrug by linking active drug moiety to phospholipid (PL), the substrate of phospholipase A2 (PLA2). PLA2 expression and activity is significantly elevated in the inflamed intestinal tissues of IBD patients. Since PLA2 enzyme specifically hydrolyses the sn-2 bond within PLs, in our PL-based prodrug approach, the sn-2 positioned FA is replaced with cyclosporine, so that PLA2 may be exploited as the prodrug-activating enzyme, releasing the free drug from the PL-complex. Owing to the enzyme overexpression, this may effectively target free cyclosporine to the sites of inflammation. Four PL-cyclosporine prodrugs were synthesized, differing by their linker length between the PL and the drug moiety. To study the prodrug activation, a novel enzymatically enriched model was developed, the colonic brush border membrane vesicles (cBBMVs); in this model, tissue vesicles were produced from colitis-induced (vs. healthy) rat colons. PLA2 overexpression (3.4-fold) was demonstrated in diseased vs. healthy cBBMVs. Indeed, while healthy cBBMVs induced only marginal activation, substantial prodrug activation was evident by colitis-derived cBBMVs. Together with the PLA2 overexpression, these data validate our drug targeting strategy. In the diseased cBBMVs, quick and complete activation of the entire dose was obtained for the 12-carbon linker prodrug, while slow and marginal activation was obtained for the 6/8-carbon linkers. The potential to target the actual sites of inflammation and treat any localizations throughout the GIT, together with the extended therapeutic index, makes this orally delivered prodrug approach an exciting new therapeutic strategy for IBD treatment.
Collapse
Affiliation(s)
- Milica Markovic
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | - Shimon Ben-Shabat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | | | - Karina Abramov-Harpaz
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.); (Y.M.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Clil Regev
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.); (Y.M.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Yifat Miller
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.); (Y.M.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Aaron Aponick
- Department of Chemistry, University of Florida, Gainesville, FL 32603, USA; (J.N.M.); (A.A.)
| | - Ellen M. Zimmermann
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL 32610, USA;
| | - Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
- Correspondence:
| |
Collapse
|
27
|
Markovic M, Abramov-Harpaz K, Regev C, Ben-Shabat S, Aponick A, Zimmermann EM, Miller Y, Dahan A. Prodrug-Based Targeting Approach for Inflammatory Bowel Diseases Therapy: Mechanistic Study of Phospholipid-Linker-Cyclosporine PLA 2-Mediated Activation. Int J Mol Sci 2022; 23:ijms23052673. [PMID: 35269813 PMCID: PMC8910962 DOI: 10.3390/ijms23052673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/14/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023] Open
Abstract
Therapeutics with activity specifically at the inflamed sites throughout the gastrointestinal tract (GIT) would be a major advance in our therapeutic approach to inflammatory bowel disease (IBD). We aimed to develop the prodrug approach that can allow such site-specific drug delivery. Currently, using cyclosporine as a drug of choice in IBD is limited to the most severe cases due to substantial systemic toxicities and narrow therapeutic index of this drug. Previously, we synthesized a series of a phospholipid-linker-cyclosporine (PLC) prodrugs designed to exploit the overexpression of phospholipase A2 (PLA2) in the inflamed intestinal tissues, as the prodrug-activating enzyme. Nevertheless, the extent and rate of prodrug activation differed significantly. In this study we applied in-vitro and modern in-silico tools based on molecular dynamics (MD) simulation, to gain insight into the dynamics and mechanisms of the PLC prodrug activation. We aimed to elucidate the reason for the significant activation change between different linker lengths in our prodrug design. Our work reveals that the PLC conjugate with the 12-carbon linker length yields the optimal prodrug activation by PLA2 in comparison to shorter linker length (6-carbons). This optimized length efficiently allows cyclosporine to be released from the prodrug to the active pocket of PLA2. This newly developed mechanistic approach, presented in this study, can be applied for future prodrug optimization to accomplish optimal prodrug activation and drug targeting in various conditions that include overexpression of PLA2.
Collapse
Affiliation(s)
- Milica Markovic
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | - Karina Abramov-Harpaz
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Clil Regev
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Shimon Ben-Shabat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | - Aaron Aponick
- Department of Chemistry, University of Florida, Gainesville, FL 32603, USA;
| | - Ellen M. Zimmermann
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL 32610, USA;
| | - Yifat Miller
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Correspondence: (Y.M.); (A.D.)
| | - Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
- Correspondence: (Y.M.); (A.D.)
| |
Collapse
|
28
|
Prodrug Therapies for Infectious and Neurodegenerative Diseases. Pharmaceutics 2022; 14:pharmaceutics14030518. [PMID: 35335894 PMCID: PMC8953076 DOI: 10.3390/pharmaceutics14030518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Prodrugs are bioreversible drug derivatives which are metabolized into a pharmacologically active drug following chemical or enzymatic modification. This approach is designed to overcome several obstacles that are faced by the parent drug in physiological conditions that include rapid drug metabolism, poor solubility, permeability, and suboptimal pharmacokinetic and pharmacodynamic profiles. These suboptimal physicochemical features can lead to rapid drug elimination, systemic toxicities, and limited drug-targeting to disease-affected tissue. Improving upon these properties can be accomplished by a prodrug design that includes the careful choosing of the promoiety, the linker, the prodrug synthesis, and targeting decorations. We now provide an overview of recent developments and applications of prodrugs for treating neurodegenerative, inflammatory, and infectious diseases. Disease interplay reflects that microbial infections and consequent inflammation affects neurodegenerative diseases and vice versa, independent of aging. Given the high prevalence, personal, social, and economic burden of both infectious and neurodegenerative disorders, therapeutic improvements are immediately needed. Prodrugs are an important, and might be said a critical tool, in providing an avenue for effective drug therapy.
Collapse
|
29
|
De Oliveira TC, Tavares ME, Soares-Sobrinho JL, Chaves LL. The role of nanocarriers for transdermal application targeted to lymphatic drug delivery: Opportunities and challenges. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
30
|
Huang L, Yang J, Wang T, Gao J, Xu D. Engineering of small-molecule lipidic prodrugs as novel nanomedicines for enhanced drug delivery. J Nanobiotechnology 2022; 20:49. [PMID: 35073914 PMCID: PMC8785568 DOI: 10.1186/s12951-022-01257-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/10/2022] [Indexed: 12/31/2022] Open
Abstract
AbstractA widely established prodrug strategy can effectively optimize the unappealing properties of therapeutic agents in cancer treatment. Among them, lipidic prodrugs extremely uplift the physicochemical properties, site-specificity, and antitumor activities of therapeutic agents while reducing systemic toxicity. Although great perspectives have been summarized in the progress of prodrug-based nanoplatforms, no attention has been paid to emphasizing the rational design of small-molecule lipidic prodrugs (SLPs). With the aim of outlining the prospect of the SLPs approach, the review will first provide an overview of conjugation strategies that are amenable to SLPs fabrication. Then, the rational design of SLPs in response to the physiological barriers of chemotherapeutic agents is highlighted. Finally, their biomedical applications are also emphasized with special functions, followed by a brief introduction of the promising opportunities and potential challenges of SLPs-based drug delivery systems (DDSs) in clinical application.
Graphical Abstract
Collapse
|
31
|
Ryu S, Jin M, Lee HK, Wang MH, Baek JS, Cho CW. Effects of lipid nanoparticles on physicochemical properties, cellular uptake, and lymphatic uptake of 6-methoxflavone. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-021-00557-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
32
|
Elz AS, Trevaskis NL, Porter CJH, Bowen JM, Prestidge CA. Smart design approaches for orally administered lipophilic prodrugs to promote lymphatic transport. J Control Release 2021; 341:676-701. [PMID: 34896450 DOI: 10.1016/j.jconrel.2021.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/22/2022]
Abstract
Challenges to effective delivery of drugs following oral administration has attracted growing interest over recent decades. Small molecule drugs (<1000 Da) are generally absorbed across the gastrointestinal tract into the portal blood and further transported to the systemic circulation via the liver. This can result in a significant reduction to the oral bioavailability of drugs that are metabolically labile and ultimately lead to ineffective exposure and treatment. Targeting drug delivery to the intestinal lymphatics is attracting increased attention as an alternative route of drug transportation providing multiple benefits. These include bypassing hepatic first-pass metabolism and selectively targeting disease reservoirs residing within the lymphatic system. The particular physicochemical requirements for drugs to be able to access the lymphatics after oral delivery include high lipophilicity (logP>5) and high long-chain triglyceride solubility (> 50 mg/g), properties required to enable drug association with the lipoprotein transport pathway. The majority of small molecule drugs, however, are not this lipophilic and therefore not substantially transported via the intestinal lymph. This has contributed to a growing body of investigation into prodrug approaches to deliver drugs to the lymphatic system by chemical manipulation. Optimised lipophilic prodrugs have the potential to increase lymphatic transport thereby improving oral pharmacokinetics via a reduction in first pass metabolism and may also target of disease-specific reservoirs within the lymphatics. This may provide advantages for current pharmacotherapy approaches for a wide array of pathological conditions, e.g. immune disease, cancer and metabolic disease, and also presents a promising approach for advanced vaccination strategies. In this review, specific emphasis is placed on medicinal chemistry strategies that have been successfully employed to design lipophilic prodrugs to deliberately enable lymphatic transport. Recent progress and opportunities in medicinal chemistry and drug delivery that enable new platforms for efficacious and safe delivery of drugs are critically evaluated.
Collapse
Affiliation(s)
- Aurelia S Elz
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| | - Natalie L Trevaskis
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia.
| | - Christopher J H Porter
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia.
| | - Joanne M Bowen
- School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Clive A Prestidge
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
33
|
Zhang Z, Lu Y, Qi J, Wu W. An update on oral drug delivery via intestinal lymphatic transport. Acta Pharm Sin B 2021; 11:2449-2468. [PMID: 34522594 PMCID: PMC8424224 DOI: 10.1016/j.apsb.2020.12.022] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/14/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022] Open
Abstract
Orally administered drug entities have to survive the harsh gastrointestinal environment, penetrate the enteric epithelia and circumvent hepatic metabolism before reaching the systemic circulation. Whereas the gastrointestinal stability can be well maintained by taking proper measures, hepatic metabolism presents as a formidable barrier to drugs suffering from first-pass metabolism. The pharmaceutical academia and industries are seeking alternative pathways for drug transport to circumvent problems associated with the portal pathway. Intestinal lymphatic transport is emerging as a promising pathway to this end. In this review, we intend to provide an updated overview on the rationale, strategies, factors and applications involved in intestinal lymphatic transport. There are mainly two pathways for peroral lymphatic transport-the chylomicron and the microfold cell pathways. The underlying mechanisms are being unraveled gradually and nowadays witness increasing research input and applications.
Collapse
Key Words
- ACQ, aggregation-caused quenching
- ASRT, apical sodium-dependent bile acid transporter
- AUC, area under curve
- BCS, biopharmaceutics classification system
- CM, chylomicron
- Chylomicron
- DC, dendritic cell
- DDT, dichlorodiphenyltrichloroethane
- DTX, docetaxel
- Drug absorption
- Drug carriers
- Drug delivery
- FA, fatty acid
- FAE, follicle-associated epithelia
- FRET, Föster resonance energy transfer
- GIT, gastrointestinal tract
- HBsAg, hepatitis B surface antigen
- HIV, human immunodeficiency virus
- LDL, low-density lipoprotein
- LDV, Leu-Asp-Val
- LDVp, LDV peptidomimetic
- Lymphatic transport
- M cell, microfold cells
- MG, monoglyceride
- MPA, mycophenolic acid
- MPS, mononuclear phagocyte system
- Microfold cell
- Nanoparticles
- OA, oleate
- Oral
- PCL, polycaprolactone
- PEG-PLA, polyethylene glycol-poly(lactic acid)
- PEI, polyethyleneimine
- PLGA, poly(lactic-co-glycolic acid)
- PVA, poly(vinyl alcohol)
- RGD, Arg-Gly-Asp
- RGDp, RGD peptidomimetic
- SEDDS, self-emulsifying drug delivery system
- SLN, solid lipid nanoparticles
- SNEDDS, self-nanoemulsifying drug delivery system
- TEM, transmission electron microscopy
- TG, triglyceride
- TPGS, D-α-tocopherol polyethylene glycol 1000 succinate
- TU, testosterone undecanoate
- WGA, wheat germ agglutinin
- YCW, yeast cell wall
Collapse
Affiliation(s)
- Zichen Zhang
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi Lu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jianping Qi
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| |
Collapse
|
34
|
Chen KJ, Plaunt AJ, Leifer FG, Kang JY, Cipolla D. Recent advances in prodrug-based nanoparticle therapeutics. Eur J Pharm Biopharm 2021; 165:219-243. [PMID: 33979661 DOI: 10.1016/j.ejpb.2021.04.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 03/10/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022]
Abstract
Extensive research into prodrug modification of active pharmaceutical ingredients and nanoparticle drug delivery systems has led to unprecedented levels of control over the pharmacological properties of drugs and resulted in the approval of many prodrug or nanoparticle-based therapies. In recent years, the combination of these two strategies into prodrug-based nanoparticle drug delivery systems (PNDDS) has been explored as a way to further advance nanomedicine and identify novel therapies for difficult-to-treat indications. Many of the PNDDS currently in the clinical development pipeline are expected to enter the market in the coming years, making the rapidly evolving field of PNDDS highly relevant to pharmaceutical scientists. This review paper is intended to introduce PNDDS to the novice reader while also updating those working in the field with a comprehensive summary of recent efforts. To that end, first, an overview of FDA-approved prodrugs is provided to familiarize the reader with their advantages over traditional small molecule drugs and to describe the chemistries that can be used to create them. Because this article is part of a themed issue on nanoparticles, only a brief introduction to nanoparticle-based drug delivery systems is provided summarizing their successful application and unfulfilled opportunities. Finally, the review's centerpiece is a detailed discussion of rationally designed PNDDS formulations in development that successfully leverage the strengths of prodrug and nanoparticle approaches to yield highly effective therapeutic options for the treatment of many diseases.
Collapse
|
35
|
Zhao C, Cai L, Nie M, Shang L, Wang Y, Zhao Y. Cheerios Effect Inspired Microbubbles as Suspended and Adhered Oral Delivery Systems. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2004184. [PMID: 33854900 PMCID: PMC8025035 DOI: 10.1002/advs.202004184] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/18/2021] [Indexed: 05/09/2023]
Abstract
Oral drug administration has an important role in medical treatment. Attempts to develop drug microcarriers with desired features for extended duration and improved absorption is highly sought. Herein, inspired by the physical phenomenon of the Cheerios effect, a novel microfluidic electrospray microbubble carrier is presented that can suspend and actively adhere to the stomach for durable oral delivery. Compared with conventional fabrication methods, the present strategy shows stability and controllability of the product. Benefiting from their uniform hollow structure, the resultant microbubbles present the same behavior of the Cheerios and can float in the gastric juice, adhere and remain to the stomach wall, which thus enhance the duration and absorption of the loaded drugs. Based on these, it is demonstrated as a proof of concept that the dexamethasone-loaded hollow microbubbles can be applied to oral administration and remain suspended and adhered to the stomach of murine for more than 1 d, showing good therapeutic effect in treating lupus erythematosus. Thus, it is believed that the microbubbles floating system will find important values in long-term oral administration.
Collapse
Affiliation(s)
- Cheng Zhao
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210002China
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Lijun Cai
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Min Nie
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210002China
| | - Luoran Shang
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210002China
- Zhongshan‐Xuhui HospitalThe Shanghai Key Laboratory of Medical Epigeneticsthe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and Technology, and Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Yongan Wang
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210002China
- State Key Laboratory of Toxicology and Medical CountermeasuresInstitute of Pharmacology and ToxicologyAcademy of Military Medical SciencesBeijing100850China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210002China
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| |
Collapse
|
36
|
Gao F, Zhou J, Sun Y, Yang C, Zhang S, Wang R, Tan W. Programmable Repurposing of Existing Drugs as Pharmaceutical Elements for the Construction of Aptamer-Drug Conjugates. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9457-9463. [PMID: 33356116 DOI: 10.1021/acsami.0c18846] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Converting marketed drug molecules into phosphoramidites may present a potential strategy to facilitate the development of aptamer-drug conjugates (ApDCs) by a DNA synthesizer in a programmable way; however, quite limited methods were reported. Herein, we demonstrated a general approach by repurposing camptothecin (CPT) species. Commonly used inactive ingredients in pharmaceuticals are investigated and selected as a bonding moiety, from which 2-mercaptoethoxy ethanol and thioglycerol were efficiently incorporated with CPT to give the precursors. Cell viability and molecular docking results of the precursors supported that incorporation of the bonding moiety would not interrupt the inhibitory activity. Therefore, corresponding phosphoramidites were prepared as pharmaceutical elements, and a series of ApDCs were constructed automatically by solid-phase synthesis. Biological studies revealed that CPT elements could be specifically delivered to HCT116 cells by an aptamer and released inside cells. This kind of programmable repurposing may take advantage of established safety data and efficacy of existing drugs resulting in a faster development of ApDCs.
Collapse
Affiliation(s)
- Fei Gao
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Institute of Translation Medicine, Shanghai University, Shanghai 200444, China
| | - Jinming Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua 321004, China
| | - Yang Sun
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cai Yang
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shiyan Zhang
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruowen Wang
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- The Cancer Hospital of the University of Chinese Academy of Sciences, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
37
|
Thanki K, Date T, Jain S. Enabling Oral Amphotericin B Delivery by Merging the Benefits of Prodrug Approach and Nanocarrier-Mediated Drug Delivery. ACS Biomater Sci Eng 2021. [PMID: 33587853 DOI: 10.1021/acsbiomaterials.0c01505] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Amphotericin B (AmB) is gold standard therapy for leishmaniasis and fungal infections. Considering the global disease burden, nearly 90% of cases occur in economically vulnerable countries, making the cost of AmB therapy a critical healthcare challenge in controlling disease burden. All currently marketed AmB products are administered through an intravenous (i.v.) route and involve high treatment costs. Designing an orally effective AmB formulation can substantially reduce the cost of therapy and improve patient compliance. However, it is a challenging task because of the distinctive physicochemical properties of AmB. Previously, we developed a lipid-based prodrug of AmB, AmB-oleyl conjugate (AmB-OA), which showcased remarkable stability in the gastrointestinal (GI) environment and improved intestinal permeation. Hereby, we have developed self-nanoemulsifiying drug delivery system (SNEDDS) of AmB-OA to further enhance the oral bioavailability of AmB and potentiate its therapeutic benefits. SNEDDS was developed by screening a wide range of oils, surfactants, and cosurfactants, and formulation composition was optimized using extreme vertices design. AmB-OA SNEDDS possessed the ability of quick self-nanoemulsification on dilution (droplet size ∼56 nm) along with remarkable stability in the GI environment. Accelerated stability (40 °C/75% relative humidity) studies and freeze-thaw cycling studies proved that the formulation was stable at tropical conditions as well as temperature cycling stress. Drug transport analysis in Caco-2 cells revealed a remarkable increase in drug transport for AmB-OA SNEDDS compared to AmB along with minimal cellular toxicities. AmB-OA SNEDDS showcased ∼8.9-fold higher AUCTot than AmB in in vivo pharmacokinetic study, proving the effectiveness of formulation to enhance oral bioavailability. In vivo toxicity analysis highlighted the ameliorated toxicity risk associated with SNEDDS formulation. Therefore, the AmB-OA SNEDDS formulation may provide a cost-friendly and effective strategy to resolve the oral AmB drug delivery challenge.
Collapse
Affiliation(s)
- Kaushik Thanki
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), Sector 67, Punjab 160062, India
| | - Tushar Date
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), Sector 67, Punjab 160062, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), Sector 67, Punjab 160062, India
| |
Collapse
|
38
|
Opportunities and challenges of fatty acid conjugated therapeutics. Chem Phys Lipids 2021; 236:105053. [PMID: 33484709 DOI: 10.1016/j.chemphyslip.2021.105053] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/20/2020] [Accepted: 01/16/2021] [Indexed: 01/03/2023]
Abstract
Instability, poor cellular uptake and unfavorable pharmacokinetics and biodistribution of many therapeutic molecules require modification in their physicochemical properties. The conjugation of these APIs with fatty acids has demonstrated an enhancement in their lipophilicity and stability. The improvement in the formulations that resulted from the conjugation of a drug with a fatty acid includes increased half-life, enhanced cellular uptake and retention, targeted tumor delivery, reduced chemoresistance in cancer, and improved blood-brain-barrier (BBB) penetration. In this review, various therapeutic molecules, including small molecules, peptides and oligonucleotides, that have been conjugated with fatty acid have been thoroughly discussed along with various conjugation strategies. The application of nano-system based delivery is gaining a lot of attention due to its ability to provide controlled drug release, targeting and reducing the extent of side effects. This review also covers various nano-carriers that have been utilized for the delivery of fatty acid drug conjugates. The enhanced lipophilicity of the drug-fatty acid conjugate has shown to enhance the affinity of the drug towards these carriers, thereby increasing the entrapment efficiency and formulation performance.
Collapse
|
39
|
Abstract
Ischemic stroke, which is caused by a sudden clot in the blood vessels, may cause severe brain tissue damage and has become a leading cause of death globally. Currently, thrombolysis is the gold standard primary treatment of ischemic stroke in clinics. However, the short therapeutic window of opportunity limits thrombolysis utility. Secondary cerebral damage caused by stroke is also an urgent problem. In this review, we discuss the present methods of treating ischemic stroke in clinics and their limitations. Various new drug delivery strategies targeting ischemic stroke lesions have also been summarized, including pharmaceutical methods, diagnostic approaches and other routes. These strategies could change the pharmacokinetic behavior, improve targeted delivery or minimize side effects. A better understanding of the novel approaches utilized to facilitate drug delivery in ischemic stroke would improve outcomes.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, P. R. China
| | - Rong Yan
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, P. R. China
| | - Jingjing Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, P. R. China
| |
Collapse
|
40
|
Patel R, Barker J, ElShaer A. Pharmaceutical Excipients and Drug Metabolism: A Mini-Review. Int J Mol Sci 2020; 21:E8224. [PMID: 33153099 PMCID: PMC7662502 DOI: 10.3390/ijms21218224] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Conclusions from previously reported articles have revealed that many commonly used pharmaceutical excipients, known to be pharmacologically inert, show effects on drug transporters and/or metabolic enzymes. Thus, the pharmacokinetics (absorption, distribution, metabolism and elimination) of active pharmaceutical ingredients are possibly altered because of their transport and metabolism modulation from the incorporated excipients. The aim of this review is to present studies on the interaction of various commonly-used excipients on pre-systemic metabolism by CYP450 enzymes. Excipients such as surfactants, polymers, fatty acids and solvents are discussed. Based on all the reported outcomes, the most potent inhibitors were found to be surfactants and the least effective were organic solvents. However, there are many factors that can influence the inhibition of CYP450, for instance type of excipient, concentration of excipient, type of CYP450 isoenzyme, incubation condition, etc. Such evidence will be very useful in dosage form design, so that the right formulation can be designed to maximize drug bioavailability, especially for poorly bioavailable drugs.
Collapse
Affiliation(s)
| | | | - Amr ElShaer
- Drug Discovery, Delivery and Patient Care (DDDPC), School of Life Sciences, Pharmacy and Chemistry, Kingston University, Kingston upon Thames, Surrey KT1 2EE, UK; (R.P.); (J.B.)
| |
Collapse
|
41
|
Markovic M, Ben-Shabat S, Dahan A. Prodrugs for Improved Drug Delivery: Lessons Learned from Recently Developed and Marketed Products. Pharmaceutics 2020; 12:pharmaceutics12111031. [PMID: 33137942 PMCID: PMC7692606 DOI: 10.3390/pharmaceutics12111031] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/18/2020] [Accepted: 10/23/2020] [Indexed: 12/18/2022] Open
Abstract
Prodrugs are bioreversible, inactive drug derivatives, which have the ability to convert into a parent drug in the body. In the past, prodrugs were used as a last option; however, nowadays, prodrugs are considered already in the early stages of drug development. Optimal prodrug needs to have effective absorption, distribution, metabolism, and elimination (ADME) features to be chemically stable, to be selective towards the particular site in the body, and to have appropriate safety. Traditional prodrug approach aims to improve physicochemical/biopharmaceutical drug properties; modern prodrugs also include cellular and molecular parameters to accomplish desired drug effect and site-specificity. Here, we present recently investigated prodrugs, their pharmaceutical and clinical advantages, and challenges facing the overall prodrug development. Given examples illustrate that prodrugs can accomplish appropriate solubility, increase permeability, provide site-specific targeting (i.e., to organs, tissues, enzymes, or transporters), overcome rapid drug metabolism, decrease toxicity, or provide better patient compliance, all with the aim to provide optimal drug therapy and outcome. Overall, the prodrug approach is a powerful tool to decrease the time/costs of developing new drug entities and improve overall drug therapy.
Collapse
Affiliation(s)
| | | | - Arik Dahan
- Correspondence: ; Tel.: +972-8-6479483; Fax: +972-8-6479303
| |
Collapse
|
42
|
Wiemer AJ. Metabolic Efficacy of Phosphate Prodrugs and the Remdesivir Paradigm. ACS Pharmacol Transl Sci 2020; 3:613-626. [PMID: 32821882 PMCID: PMC7409933 DOI: 10.1021/acsptsci.0c00076] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Indexed: 02/08/2023]
Abstract
![]()
Drugs that contain phosphates (and
phosphonates or phosphinates)
have intrinsic absorption issues and are therefore often delivered
in prodrug forms to promote their uptake. Effective prodrug forms
distribute their payload to the site of the intended target and release
it efficiently with minimal byproduct toxicity. The ability to balance
unwanted payload release during transit with desired release at the
site of action is critical to prodrug efficacy. Despite decades of
research on prodrug forms, choosing the ideal prodrug form remains
a challenge which is often solved empirically. The recent emergency
use authorization of the antiviral remdesivir for COVID-19 exemplifies
a new approach for delivery of phosphate prodrugs by parenteral dosing,
which minimizes payload release during transit and maximizes tissue
payload distribution. This review focuses on the role of metabolic
activation in efficacy during oral and parenteral dosing of phosphate,
phosphonate, and phosphinate prodrugs. Through examining prior structure–activity
studies on prodrug forms and the choices that led to development of
remdesivir and other clinical drugs and drug candidates, a better
understanding of their ability to distribute to the planned site of
action, such as the liver, plasma, PBMCs, or peripheral tissues, can
be gained. The structure–activity relationships described here
will facilitate the rational design of future prodrugs.
Collapse
Affiliation(s)
- Andrew J Wiemer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269, United States.,Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|
43
|
Lu B, Xiao F, Wang Z, Wang B, Pan Z, Zhao W, Zhu Z, Zhang J. Redox-Sensitive Hyaluronic Acid Polymer Prodrug Nanoparticles for Enhancing Intracellular Drug Self-Delivery and Targeted Cancer Therapy. ACS Biomater Sci Eng 2020; 6:4106-4115. [DOI: 10.1021/acsbiomaterials.0c00762] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Beibei Lu
- State Key Laboratory of Advanced Welding and Joining, Harbin Institute of Technology, Shenzhen 518055, China
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen 518055, China
| | - Fan Xiao
- School of Materials Science and Engineering, Harbin Institute of Technology, Nangang District, Harbin 150001, China
| | - Zhenyuan Wang
- State Key Laboratory of Advanced Welding and Joining, Harbin Institute of Technology, Shenzhen 518055, China
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen 518055, China
| | - Binshen Wang
- State Key Laboratory of Advanced Welding and Joining, Harbin Institute of Technology, Shenzhen 518055, China
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen 518055, China
| | - Zuchen Pan
- State Key Laboratory of Advanced Welding and Joining, Harbin Institute of Technology, Shenzhen 518055, China
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen 518055, China
| | - Weiwei Zhao
- State Key Laboratory of Advanced Welding and Joining, Harbin Institute of Technology, Shenzhen 518055, China
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen 518055, China
| | - Zhenye Zhu
- State Key Laboratory of Advanced Welding and Joining, Harbin Institute of Technology, Shenzhen 518055, China
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen 518055, China
| | - Jiaheng Zhang
- State Key Laboratory of Advanced Welding and Joining, Harbin Institute of Technology, Shenzhen 518055, China
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen 518055, China
| |
Collapse
|
44
|
Markovic M, Ben-Shabat S, Dahan A. Computational Simulations to Guide Enzyme-Mediated Prodrug Activation. Int J Mol Sci 2020; 21:ijms21103621. [PMID: 32443905 PMCID: PMC7279318 DOI: 10.3390/ijms21103621] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
Prodrugs are designed to improve pharmaceutical/biopharmaceutical characteristics, pharmacokinetic/pharmacodynamic properties, site-specificity, and more. A crucial step in successful prodrug is its activation, which releases the active parent drug, exerting a therapeutic effect. Prodrug activation can be based on oxidation/reduction processes, or through enzyme-mediated hydrolysis, from oxidoreductases (i.e., Cytochrome P450) to hydrolytic enzymes (i.e., carboxylesterase). This study provides an overview of the novel in silico methods for the optimization of enzyme-mediated prodrug activation. Computational methods simulating enzyme-substrate binding can be simpler like molecular docking, or more complex, such as quantum mechanics (QM), molecular mechanics (MM), and free energy perturbation (FEP) methods such as molecular dynamics (MD). Examples for MD simulations used for elucidating the mechanism of prodrug (losartan, paclitaxel derivatives) metabolism via CYP450 enzyme are presented, as well as an MD simulation for optimizing linker length in phospholipid-based prodrugs. Molecular docking investigating quinazolinone prodrugs as substrates for alkaline phosphatase is also presented, as well as QM and MD simulations used for optimal fit of different prodrugs within the human carboxylesterase 1 catalytical site. Overall, high quality computational simulations may show good agreement with experimental results, and should be used early in the prodrug development process.
Collapse
|
45
|
Li Q, Li X, Zhao C. Strategies to Obtain Encapsulation and Controlled Release of Small Hydrophilic Molecules. Front Bioeng Biotechnol 2020; 8:437. [PMID: 32478055 PMCID: PMC7237580 DOI: 10.3389/fbioe.2020.00437] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/16/2020] [Indexed: 12/03/2022] Open
Abstract
The therapeutic effect of small hydrophilic molecules is limited by the rapid clearance from the systemic circulation or a local site of administration. The unsuitable pharmacokinetics and biodistribution can be improved by encapsulating them in drug delivery systems. However, the high-water solubility, very hydrophilic nature, and low molecular weight make it difficult to encapsulate small hydrophilic molecules in many drug delivery systems. In this mini-review, we highlight three strategies to efficiently encapsulate small hydrophilic molecules and achieve controlled release: physical encapsulation in micro/nanocapsules, physical adsorption via electronic interactions, and covalent conjugation. The principles, advantages, and disadvantages of each strategy are discussed. This review paper could be a guide for scientists, engineers, and medical doctors who want to improve the therapeutic efficacy of small hydrophilic drugs.
Collapse
Affiliation(s)
| | | | - Chao Zhao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, United States
| |
Collapse
|
46
|
Zhang F, Wu Q, Liu H. NIR light-triggered nanomaterials-based prodrug activation towards cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1643. [PMID: 32394638 DOI: 10.1002/wnan.1643] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 01/10/2023]
Abstract
Nanomaterials-based prodrug activation systems have been widely explored in cancer therapy, aiming at overcoming limited dosage formulation, systemic toxicity, and insufficient pharmacokinetic performance of parent drugs. For better delivery control, various stimuli systems, especially nanomaterials-based ones, have come to the forefront. Among them, near-infrared (NIR) light takes advantage of on-demand/site-specific regulation and non-invasiveness. In this review, we will address the developments of nanomaterials-based prodrug over the last decade, the activation mechanisms, and bioapplications under NIR light triggering. The advantages and limitations of NIR-triggered prodrug activation strategies and the perspectives of the next-generation prodrug nanomedicine will also be summarized. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Fengrong Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, China
| | - Qingyuan Wu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, China
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
47
|
Markovic M, Ben-Shabat S, Aponick A, Zimmermann EM, Dahan A. Lipids and Lipid-Processing Pathways in Drug Delivery and Therapeutics. Int J Mol Sci 2020; 21:ijms21093248. [PMID: 32375338 PMCID: PMC7247327 DOI: 10.3390/ijms21093248] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/28/2020] [Accepted: 05/02/2020] [Indexed: 12/19/2022] Open
Abstract
The aim of this work is to analyze relevant endogenous lipid processing pathways, in the context of the impact that lipids have on drug absorption, their therapeutic use, and utilization in drug delivery. Lipids may serve as biomarkers of some diseases, but they can also provide endogenous therapeutic effects for certain pathological conditions. Current uses and possible clinical benefits of various lipids (fatty acids, steroids, triglycerides, and phospholipids) in cancer, infectious, inflammatory, and neurodegenerative diseases are presented. Lipids can also be conjugated to a drug molecule, accomplishing numerous potential benefits, one being the improved treatment effect, due to joined influence of the lipid carrier and the drug moiety. In addition, such conjugates have increased lipophilicity relative to the parent drug. This leads to improved drug pharmacokinetics and bioavailability, the ability to join endogenous lipid pathways and achieve drug targeting to the lymphatics, inflamed tissues in certain autoimmune diseases, or enable overcoming different barriers in the body. Altogether, novel mechanisms of the lipid role in diseases are constantly discovered, and new ways to exploit these mechanisms for the optimal drug design that would advance different drug delivery/therapy aspects are continuously emerging.
Collapse
Affiliation(s)
- Milica Markovic
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | - Shimon Ben-Shabat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | - Aaron Aponick
- Department of Chemistry, University of Florida, Gainesville, FL 32603, USA;
| | - Ellen M. Zimmermann
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL 32610, USA;
| | - Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
- Correspondence:
| |
Collapse
|
48
|
Anti-echinococcal activity of menthol and a novel prodrug, menthol-pentanol, against Echinococcus multilocularis. Acta Trop 2020; 205:105411. [PMID: 32101761 DOI: 10.1016/j.actatropica.2020.105411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/14/2020] [Accepted: 02/19/2020] [Indexed: 11/20/2022]
Abstract
Alveolar echinococcosis is one of the most dangerous parasitic zoonoses. This disease, widely distributed in the northern hemisphere, is caused by the metacestode stage of the tapeworm Echinococcus multilocularis. All surgical and non-surgical patients should perform chemotherapy with benzimidazoles, mainly with albendazole. However, the efficacy of albendazole is variable due to its deficient pharmacokinetic properties. Therefore, the need to find new therapeutic alternatives for the treatment of alveolar echinococcosis is evident. Menthol is a natural compound of low toxicity, used in industries such as cosmetics and gastronomy and generally recognized as safe by the Food and Drug Administration. In addition, menthol has important pharmacological effects and is effective against a wide variety of organisms. The development of prodrugs allows improving the pharmacokinetic properties of the parental drug. To improve lipophilicity and therefore the bioavailability of menthol, a novel prodrug called menthol-pentanol was developed by masking the functional polar group of menthol by linking n-pentanol by a carbonate bond. The aim of the current work was to evaluate the in vitro and in vivo efficacy of menthol and menthol-pentanol against E. multilocularis. Menthol-pentanol had a greater protoscolicidal effect than menthol. In addition, the prodrug demonstrated a similar clinical efficacy to albendazole. The increase in lipophilicity of the prodrug with respect to menthol was reflected in an increase in its antiparasitic activity against E. multilocularis. Thus, menthol-pentanol appears as a promising candidate for further evaluation as a potential alternative for the treatment of alveolar echinococcosis.
Collapse
|
49
|
Takalani F, Kumar P, Kondiah PPD, Choonara YE, Pillay V. Lipid-drug conjugates and associated carrier strategies for enhanced antiretroviral drug delivery. Pharm Dev Technol 2019; 25:267-280. [PMID: 31744408 DOI: 10.1080/10837450.2019.1694037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mortality rate of patients infected with HIV-1 has been significantly reduced by using HAART. However, the virus to date has not been eradicated. Transmission of HIV-1 infection through sexual intercourse remains an ongoing challenge, with increased risk of infection occurring in women. Interestingly, ARV drugs can be chemically linked with lipids to produce lipid-drug conjugates (LDCs). This alters pharmacokinetic properties of ARV drugs and thereby resulting in improved effectiveness. Although LDCs can be administered without a delivery carrier, they are usually incorporated into suitable delivery systems such as lipid nanoparticles, polymeric nanoparticles, micelles, liposomes, emulsions, and carbon nanotubes. Given that LDCs have the potential to improve oral bioavailability, lipophilicity, toxicity, and drug targeting, it is of our great interest to review strategies of lipid-drug conjugation together with their delivery systems for enhanced antiretroviral efficacy.
Collapse
Affiliation(s)
- Funanani Takalani
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Pierre P D Kondiah
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Viness Pillay
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
50
|
Gu Y, Ma J, Fu Z, Xu Y, Gao B, Yao J, Xu W, Chu K, Chen J. Development Of Novel Liposome-Encapsulated Combretastatin A4 Acylated Derivatives: Prodrug Approach For Improving Antitumor Efficacy. Int J Nanomedicine 2019; 14:8805-8818. [PMID: 31806973 PMCID: PMC6844228 DOI: 10.2147/ijn.s210938] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 10/21/2019] [Indexed: 12/26/2022] Open
Abstract
Purpose The objective of the present study was to develop a liposomal drug delivery system based on combretastatin A4 (CA4) prodrugs modified with varying alkyl chains and investigate the in vitro drug conversion from prodrug and in vivo antitumor effect. Methods The prodrug of CA4 was synthesized with stearyl chloride (18-carbon chain), palmitoyl chloride (16-carbon chain), myristoyl chloride (14-carbon chain), decanoyl chloride (10-carbon chain), and hexanoyl chloride (6-carbon chain) at the 3′-position of the CA4. Subsequently, it was encapsulated with liposomes through the thin-film evaporation method. Furthermore, the characteristics of prodrug-liposome were evaluated using in vitro drug release, conversion, and cytotoxicity assays, as well as in vivo pharmacokinetic, antitumor, and biodistribution studies. Results The liposome system with loaded CA4 derivatives was successfully developed with nano-size and electronegative particles. The rate of in vitro drug release and conversion was reduced as the fatty acid carbon chain lengthened. On the contrary, in vivo antitumor effects were improved with the enlargement of the fatty acid carbon chain. The results of the in vivo pharmacokinetic and tissue distribution studies indicated that the reduced rate of CA4 release with a long carbon chain could prolong the circulation time and increase the drug concentration in the tumor tissue. Conclusion These results suggested that the release or hydrolysis of the parent drug from the prodrug was closely related with the in vitro and in vivo properties. The slow drug release of CA4 modified with longer acyl chain could prolong the circulation time and increase the concentration of the drug in the tumor tissue. These effects play a critical role in increasing the antitumor efficacy.
Collapse
Affiliation(s)
- Yongwei Gu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350108, People's Republic of China.,Shanghai Wei Er Biopharmaceutical Technology Co., Ltd., Shanghai 201707, People's Republic of China
| | - Juanjuan Ma
- Shanghai Wei Er Biopharmaceutical Technology Co., Ltd., Shanghai 201707, People's Republic of China
| | - Zhiqin Fu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350108, People's Republic of China.,Shanghai Wei Er Biopharmaceutical Technology Co., Ltd., Shanghai 201707, People's Republic of China
| | - Youfa Xu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350108, People's Republic of China.,Shanghai Wei Er Biopharmaceutical Technology Co., Ltd., Shanghai 201707, People's Republic of China
| | - Baoan Gao
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350108, People's Republic of China.,Shanghai Wei Er Biopharmaceutical Technology Co., Ltd., Shanghai 201707, People's Republic of China
| | - Jianzhong Yao
- School of Pharmacy, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Wei Xu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350108, People's Republic of China
| | - Kedan Chu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350108, People's Republic of China
| | - Jianming Chen
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350108, People's Republic of China.,Shanghai Wei Er Biopharmaceutical Technology Co., Ltd., Shanghai 201707, People's Republic of China
| |
Collapse
|