1
|
Chagas MDSDS, Moragas Tellis CJ, Silva AR, Brito MADSM, Teodoro AJ, de Barros Elias M, Ferrarini SR, Behrens MD, Gonçalves-de-Albuquerque CF. Luteolin: A novel approach to fight bacterial infection. Microb Pathog 2025; 204:107519. [PMID: 40164399 DOI: 10.1016/j.micpath.2025.107519] [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: 01/02/2025] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Diseases caused by bacteria significantly impact public health, causing both acute and chronic issues, sequelae, and death. The problems get even more significant, considering the antimicrobial resistance. Bacterial resistance occurs when antibacterial drugs fail to kill the microbes, leading to the persistence of infection and pathogen spread in the host. Thus, the search for new molecules with antibacterial activity dramatically impacts human health. Natural products have proven to be a prosperous source of these agents. Among them, the flavonoids deserve to be highlighted. They are secondary metabolites, primarily involved in plant signaling and protection. Thus, they play an essential role in plant adaptation to the environment. Herein, we will focus on luteolin because it is commonly found in edible plants and has diverse pharmacological properties such as anti-inflammatory, anticancer, antioxidant, and antimicrobial. We will further explore the luteolin antibacterial activity, mechanisms of action, structure-activity relationship, and toxicity of luteolin. Thus, we have included reports of luteolin with antibacterial activity recently published, as well as focused on nanotechnology as a pivotal and helpful approach for the clinical use of luteolin. This review aims to foster future research on luteolin as a therapeutic agent for treating bacterial infection.
Collapse
Affiliation(s)
- Maria do Socorro Dos Santos Chagas
- Programa de Pós-graduação em Ciências e Biotecnologia (PPBI), Instituto de Biologia, UFF, Brazil; Laboratório de Imunofarmacologia, Departamento de Ciências Fisiológicas, UNIRIO, Rio de Janeiro, RJ, Brazil; Laboratório de Imunofarmacologia, IOC, FIOCRUZ, Rio de Janeiro, RJ, Brazil; Laboratório de Produtos Naturais para Saúde Pública, Farmanguinhos, FIOCRUZ, RJ, Brazil
| | | | - Adriana R Silva
- Laboratório de Imunofarmacologia, IOC, FIOCRUZ, Rio de Janeiro, RJ, Brazil; Programa de Pós-Graduação em Neurociências, Instituto de Biologia, UFF, Niterói, Brazil
| | - Maria Alice Dos Santos Mascarenhas Brito
- Laboratório de Imunofarmacologia, Departamento de Ciências Fisiológicas, UNIRIO, Rio de Janeiro, RJ, Brazil; Laboratório de Imunofarmacologia, IOC, FIOCRUZ, Rio de Janeiro, RJ, Brazil; Programa de Pós-Graduação em Neurociências, Instituto de Biologia, UFF, Niterói, Brazil
| | - Anderson Junger Teodoro
- Laboratório de Biologia Celular e Nutrição (LABCEN) Universidade Federal Fluminense, UFF, Niteroi, Brazil
| | - Monique de Barros Elias
- Laboratório de Biologia Celular e Nutrição (LABCEN) Universidade Federal Fluminense, UFF, Niteroi, Brazil
| | - Stela Regina Ferrarini
- Laboratório de Nanotecnologia Farmacêutica, Universidade Federal do mato Grosso Campus Sinop - UFMT, Cuiabá, Brazil
| | - Maria Dutra Behrens
- Laboratório de Produtos Naturais para Saúde Pública, Farmanguinhos, FIOCRUZ, RJ, Brazil.
| | - Cassiano F Gonçalves-de-Albuquerque
- Programa de Pós-graduação em Ciências e Biotecnologia (PPBI), Instituto de Biologia, UFF, Brazil; Laboratório de Imunofarmacologia, Departamento de Ciências Fisiológicas, UNIRIO, Rio de Janeiro, RJ, Brazil; Programa de Pós-Graduação em Biologia Molecular e Celular (PPGBMC), UNIRIO, RJ, Brazil; Laboratório de Imunofarmacologia, IOC, FIOCRUZ, Rio de Janeiro, RJ, Brazil; Programa de Pós-Graduação em Neurociências, Instituto de Biologia, UFF, Niterói, Brazil.
| |
Collapse
|
2
|
Haque E, Georg GI. Medoxomil Prodrug Strategies. J Med Chem 2025; 68:9025-9036. [PMID: 40261681 DOI: 10.1021/acs.jmedchem.4c02967] [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: 04/24/2025]
Abstract
Drug discovery campaigns often face biopharmaceutical challenges, some of which can be solved by a prodrug approach. Prodrugs are enzymatically or chemically transformed in vivo to produce active drugs. Among these, medoxomil promoieties have been judiciously employed in multiple drug discovery campaigns, leading to three prodrugs gaining FDA approval: azilsartan medoxomil (6), olmesartan medoxomil (20), and ceftobiprole medocaril (29), and one approval in Japan: prulifloxacin (35). The promoiety can be easily appended to mask carboxylic acids, amines, zwitterionic compounds, and other polar groups, imparting lipophilicity to the parent compound. The promoiety has the added advantage of rapid and complete conversion to the parent drug by multiple enzymatic pathways across different tissues. The approach has been used for drugs spanning multiple classes to improve oral bioavailability, solubility, tissue localization, efflux, and side effect profiles. This Perspective analyzes the history and application of medoxomil prodrugs and discusses their potential for drug development.
Collapse
Affiliation(s)
- Ehfazul Haque
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, University of Minnesota, 717 Delaware Street, SE, Minneapolis, Minnesota 55414, United States
| | - Gunda I Georg
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, University of Minnesota, 717 Delaware Street, SE, Minneapolis, Minnesota 55414, United States
| |
Collapse
|
3
|
Geaneotes PJ, Floreancig PE. Strategy-Level Prodrug Synthesis. Chemistry 2025:e202501115. [PMID: 40317559 DOI: 10.1002/chem.202501115] [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: 03/19/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Organic synthesis uniquely provides opportunities to access molecules that serve defined purposes. Medicinal chemistry illustrates this attribute well with prodrug design, whereby a drug undergoes a late-stage conversion to a conditionally responsive active medicinal agent (AMA), being a notable example. Prodrugs are becoming increasingly important in medicinal chemistry but common approaches to introduce biologically responsive groups are limited in the chemoselectivity and scope of available functionalization reactions. This Concept article describes strategy-level prodrug synthesis, which is a powerful extension of classical prodrug formation that initiates sequences with the objective of introducing functionality early in a sequence to achieve greater scope, site-selectivity, and chemoselectivity for the incorporation of the biologically responsive group. Examples of functionalization using alkyne hydroamination, Curtius reaction, and alkene metathesis are highlighted along with the use of the prodrugs for biological applications.
Collapse
Affiliation(s)
- Paul J Geaneotes
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA
| | - Paul E Floreancig
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA
| |
Collapse
|
4
|
Edupuganti VVSR, Matikonda SS, Lawer A, Fairhall JM, Lewin HM, Kueh JTB, Tyndall JDA, Gamble AB. Stimuli-Responsive Prodrug Linkers That Simultaneously Release Cargo and Neutralize In Situ Generated (Aza)Quinone Methides. Chemistry 2025:e202501278. [PMID: 40235087 DOI: 10.1002/chem.202501278] [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: 04/06/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/17/2025]
Abstract
Self-immolative linkers that use p-amino/hydroxy-benzyloxycarbonyl (PABC/PHBC) spacers are essential to the mechanism of many prodrugs. However, a highly reactive (aza)quinone methide is generated as a potential toxic byproduct. To remove the methide as it forms, we synthesized a series of novel tripartite prodrugs, comprising different triggers (nitro, amide, azide, boronate) and a PABC/PHBC-type self-immolative spacer with an integrated nucleophile (amine). Upon reductive, hydrolytic, or oxidative-trigger activation, the release of the cargo is facilitated via a 1,6-elimination that generates a reactive (aza)quinone methide. With the built-in nucleophile, the (aza)quinone methide is rapidly self-quenched to generate tetrahydroisoquinolines (THIQs). One of the selected THIQs does not exhibit an anti-proliferative effect on the A431 mammalian tumor cell line. The new prodrug strategy has broad scope, enabling the use of a trigger that matches the targeted stimulus, while allowing for a diverse range of drug/cargo attachment. This proof-of-concept study adds a new linker strategy that quenches the electrophilic (aza)quinone methide generated in many self-immolative linker systems and could find applications in prodrug and antibody-drug conjugate strategies, or as a linker for probes in chemical biology.
Collapse
Affiliation(s)
| | - Siddharth S Matikonda
- School of Pharmacy, University of Otago, 18 Frederick Street, Dunedin, 9054, New Zealand
| | - Aggie Lawer
- School of Pharmacy, University of Otago, 18 Frederick Street, Dunedin, 9054, New Zealand
| | - Jessica M Fairhall
- School of Pharmacy, University of Otago, 18 Frederick Street, Dunedin, 9054, New Zealand
| | - Harrison M Lewin
- School of Pharmacy, University of Otago, 18 Frederick Street, Dunedin, 9054, New Zealand
| | - Jui Thiang Brian Kueh
- Department of Chemistry, University of Otago, Union Place, Dunedin, 9016, New Zealand
| | - Joel D A Tyndall
- School of Pharmacy, University of Otago, 18 Frederick Street, Dunedin, 9054, New Zealand
| | - Allan B Gamble
- School of Pharmacy, University of Otago, 18 Frederick Street, Dunedin, 9054, New Zealand
| |
Collapse
|
5
|
Li H, Shen X, Chu Y, Yuan P, Shuai Q. Challenging and new opportunities for prodrug technology. Invest New Drugs 2025; 43:365-376. [PMID: 39966300 DOI: 10.1007/s10637-025-01515-w] [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: 12/14/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025]
Abstract
Research on prodrug technology has opened new avenues for site-directed chemotherapy rather than systemic chemotherapy. This distinctive strategy allows drug delivery to be activated by light-, irradiation-, or ultrasound (US)-tunable chemistries, which have been termed photopharmacology, radiopharmacology, and sonopharmacology, respectively. Prodrugs have emerged as a main strategy for improving pharmacokinetics, reducing side effects, and thus enhancing the therapeutic efficacy of drugs. This review summarizes stimuli-responsive drug release systems and the latest progress in exogenous stimuli-responsive prodrug activation, e.g., light, irradiation, and US, with a focus on the activation of small molecule prodrugs, antibody‒drug conjugates, and prodrug nanosystems. In addition, challenges encountered by Pt drugs and Pt(IV) prodrug nanotherapeutics are summarized and discussed. Moreover, this review presents the current state of precise treatment and discusses the opportunities and challenges for the clinical translation of these strategies.
Collapse
Affiliation(s)
- Helin Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
- Zhejiang Governor Triangle Biomedical Industrial Technology Research Park, Huzhou, 310014, Zhejiang, China
| | - Xuelian Shen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
- Zhejiang Governor Triangle Biomedical Industrial Technology Research Park, Huzhou, 310014, Zhejiang, China
| | - Yu Chu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
- Zhejiang Governor Triangle Biomedical Industrial Technology Research Park, Huzhou, 310014, Zhejiang, China
| | - Panhong Yuan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China.
| | - Qi Shuai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China.
- Zhejiang Governor Triangle Biomedical Industrial Technology Research Park, Huzhou, 310014, Zhejiang, China.
| |
Collapse
|
6
|
Miyaji K, Masaki Y, Seio K. Synthesis of Prodrug-Type Oligonucleotides Modified With a Galactosylated Self-Immolative Linker Cleavable by β-Galactosidase. Curr Protoc 2025; 5:e70128. [PMID: 40192303 DOI: 10.1002/cpz1.70128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
This protocol describes procedures for the preparation of a modified thymidine conjugated with galactose via a self-immolative linker at the O4-position, and the synthesis of β-galactosidase-responsive prodrug-type oligodeoxynucleotides (ONs) containing these modified thymidines. These prodrug-ONs are designed to be activated in response to β-galactosidase, enabling targeted activation in specific cells or tissues and potentially contributing to the reduction of adverse effects. © 2025 Wiley Periodicals LLC. Basic Protocol 1: Preparation of O4-modified thymidine phosphoramidite 7 Basic Protocol 2: Preparation of O4-modified thymidine phosphoramidite 15 Support Protocol 1: Preparation of triazolyl thymidine derivative 1 Support Protocol 2: Preparation of benzyl alcohol derivative 2 Basic Protocol 3: Preparation of β-galactosidase-responsive ODNs 1 to 5.
Collapse
Affiliation(s)
- Kento Miyaji
- School of Life Science and Technology, Institute of Science Tokyo, Kanagawa, Japan
| | - Yoshiaki Masaki
- School of Life Science and Technology, Institute of Science Tokyo, Kanagawa, Japan
- Nucleotide and Peptide Drug Discovery Center, Institute of Science Tokyo, Tokyo, Japan
| | - Kohji Seio
- School of Life Science and Technology, Institute of Science Tokyo, Kanagawa, Japan
| |
Collapse
|
7
|
de Souza MM, Gini ALR, Moura JA, Scarim CB, Chin CM, dos Santos JL. Prodrug Approach as a Strategy to Enhance Drug Permeability. Pharmaceuticals (Basel) 2025; 18:297. [PMID: 40143076 PMCID: PMC11946379 DOI: 10.3390/ph18030297] [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: 01/06/2025] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 03/28/2025] Open
Abstract
Absorption and permeability are critical physicochemical parameters that must be balanced to achieve optimal drug uptake. These key factors are closely linked to the maximum absorbable dose required to provide appropriate plasma levels of drugs. Among the various strategies employed to enhance drug solubility and permeability, prodrug design stands out as a highly effective and versatile approach for improving physicochemical properties and enabling the optimization of biopharmaceutical and pharmacokinetic parameters while mitigating adverse effects. Prodrugs are compounds with reduced or no activity that, through bio-reversible chemical or enzymatic processes, release an active parental drug. The application of this technology has led to significant advancements in drug optimization during the design phase, and it offers broad potential for further development. Notably, approximately 13% of the drugs approved by the U.S. Food and Drug Administration (FDA) between 2012 and 2022 were prodrugs. In this review article, we will explore the application of prodrug strategies to enhance permeability, describing examples of market drugs. We also describe the use of the prodrug approach to optimize PROteolysis TArgeting Chimeras (PROTACs) permeability by using conjugation technologies. We will highlight some new technologies in prodrugs to enrich permeability properties, contributing to developing new effective and safe prodrugs.
Collapse
Affiliation(s)
- Mateus Mello de Souza
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Ana Luísa Rodriguez Gini
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Jhonnathan Alves Moura
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-900, SP, Brazil;
| | - Cauê Benito Scarim
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Chung Man Chin
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
- Union of the Colleges of the Great Lakes (UNILAGO), School of Medicine, Advanced Research Center in Medicine (CEPAM), Sao Jose do Rio Preto 15030-070, SP, Brazil
| | - Jean Leandro dos Santos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-900, SP, Brazil;
| |
Collapse
|
8
|
von Glasenapp V, C Almeida A, Chang D, Gasic I, Winssinger N, Gotta M. Spatio-temporal control of mitosis using light via a Plk1 inhibitor caged for activity and cellular permeability. Nat Commun 2025; 16:1599. [PMID: 39971898 PMCID: PMC11840123 DOI: 10.1038/s41467-025-56746-5] [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: 03/26/2024] [Accepted: 01/28/2025] [Indexed: 02/21/2025] Open
Abstract
The ability to control the activity of kinases spatially and temporally is essential to elucidate the role of signalling pathways in development and physiology. Progress in this direction has been hampered by the lack of tools to manipulate kinase activity in a highly controlled manner in vivo. Here we report a strategy to modify BI2536, the well characterized inhibitor of the conserved and essential mitotic kinase Polo-like kinase 1 (Plk1). We introduce the same coumarin photolabile protecting group (PPG) at two positions of the inhibitor. At one position, the coumarin prevents the interaction with Plk1, at the second it masks an added carboxylic acid, important for cellular retention. Exposure to light results in removal of both PPGs, leading to the activation of the inhibitor and its trapping inside cells. We demonstrate the efficacy of the caged inhibitor in three-dimensional spheroid cultures: by uncaging it with a single light pulse, we can inhibit Plk1 and arrest cell division, a highly dynamic process, with spatio-temporal control. Our design can be applied to other small molecules, providing a solution to control their activity in living cells with unprecedented precision.
Collapse
Affiliation(s)
- Victoria von Glasenapp
- Department of Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Ana C Almeida
- Department of Molecular and Cellular Biology, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Dalu Chang
- NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
- Department of Organic Chemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Ivana Gasic
- Department of Molecular and Cellular Biology, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Nicolas Winssinger
- NCCR Chemical Biology, University of Geneva, Geneva, Switzerland.
- Department of Organic Chemistry, Faculty of Science, University of Geneva, Geneva, Switzerland.
| | - Monica Gotta
- Department of Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- NCCR Chemical Biology, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
9
|
Faisal AF, Mustafa YF. The Multifaceted Chemistry of Chili Peppers: A Biodiversity Treasure for Nutrition and Biomedicine. Chem Biodivers 2025:e202402690. [PMID: 39898594 DOI: 10.1002/cbdv.202402690] [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: 10/21/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/04/2025]
Abstract
Due to its biodiversity, traditional medicine has been recognized worldwide for centuries and continues to affect the development of complementary and alternative therapies. A wide variety of spices, herbs, and trees are known for their curative effects. Chili pepper (Ch-p), a spice-utilizing fruit, is rich in natural medicinally bioactive compounds, such as flavonoids, capsaicinoids, and many other phytochemicals and phytonutrients. Operating in synergy and consortium, these compounds demonstrate their functionality, in comparison to lonely treatment, as active agents in handling many disorders. These may include abnormal coagulation, oxidative stress, obesity, diabetes, inflammation, cancer, and microbe-inducing diseases. Recently, capsaicinoids, particularly capsaicin, have been shown to manage the symptoms of significant viral diseases, including COVID-19. Capsaicin also has the potential to be an effective anesthetic agent and enables Ch-p to be expandedly employed as a topical preparation in relieving pain as well. The phytochemicals of Ch-p are not only beneficial and inexpensive phyto-alternatives in disease management, but they can also be used as scaffolds for the production of novel medicines. The study also substantiates the role of the TRPV1 receptor in the mitigation of chronic diseases in conjunction with capsaicin. Nevertheless, the consumption of Ch-p is the subject of limited medicinal research, necessitating the confirmation of the results from animal studies. The nutritional and biomedical prospection of Ch-p-derived products has been addressed in an accessible format in this artifact, with the potential to precisely enhance and enrich our pharmaceutical industries in the pursuit of human well-being.
Collapse
Affiliation(s)
- Ayman Faris Faisal
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
10
|
Zhu J, Jia X, Ren S, Zhang Z, Li H, Wang J, Song B, Wu W, Peng C. Inhibition of Polo-Like Kinase 1 Dampens the Replication of Vaccinia Virus in Mammalian Cells. J Med Virol 2025; 97:e70240. [PMID: 39953955 DOI: 10.1002/jmv.70240] [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: 10/20/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Since the eradication of smallpox, zoonotic poxviruses, such as the mpox virus (MPXV), continue to pose a threat to public health. Identifying drugs that reduce poxvirus infection and replication, as well as understanding their molecular mechanisms, is essential for epidemic control. Polo-like kinase 1 (PLK1) has been shown to facilitate vaccinia virus (VACV) infection and replication. This study confirms the effects of the PLK1 inhibitors HMN-214 and ON-01910 on VACV replication in A549 cells. Both viral titers and DNA loads were significantly reduced in treated cells after infection. Additionally, ON-01910 demonstrated broad-spectrum antiviral activity against the lumpy skin disease virus (LSDV) and the infectious bovine rhinotracheitis virus (IBRV) in vitro. PLK1 knockdown in A549 cells also led to a reduction in VACV protein expression, viral titers, and DNA levels. Further analysis showed that VACV infection leads to the accumulation of PLK1 near viral factories. However, despite its strong in vitro effects, ON-01910 did not significantly reduce VACV replication in mice. These findings highlight the critical role of PLK1 in VACV replication and its potential as a target for antiviral therapy against orthopoxviruses.
Collapse
Affiliation(s)
- Junda Zhu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xuejiao Jia
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shuning Ren
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zihui Zhang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hua Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jing Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Baifen Song
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wenxue Wu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chen Peng
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
11
|
Kurian R, Wang H. Prodrugs in Oncology: Bioactivation and Impact on Therapeutic Efficacy and Toxicity. Int J Mol Sci 2025; 26:988. [PMID: 39940757 PMCID: PMC11816641 DOI: 10.3390/ijms26030988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
A prodrug is a molecule that lacks pharmacological activity, but upon enzymatic bioactivation, it can generate a therapeutically active molecule. The primary reason behind the design of a prodrug is to help circumvent challenges associated with the physicochemical properties of a drug molecule, such as solubility, absorption, distribution, and instability. Chemotherapy has been at the forefront of cancer treatment for over 70 years due to its ability to target rapidly proliferating tumor cells. However, a major concern with conventional chemotherapy is the lack of selectivity and its associated side toxicity, which can severely impact patients' quality of life. In oncology, prodrugs have been explored to enhance the bioavailability, improve efficacy, and minimize systemic toxicity of chemotherapeutic agents. Prodrugs activated by enzymes unique to a tumor microenvironment can significantly increase targeted delivery of chemotherapeutic drugs. This review aims to highlight commonly used chemotherapeutic prodrugs, including both alkylating and non-alkylating agents, and discuss their clinical relevance, mechanisms of bioactivation, and toxicity concerns.
Collapse
Affiliation(s)
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA;
| |
Collapse
|
12
|
Jacob S, Kather FS, Boddu SHS, Attimarad M, Nair AB. Nanosuspension Innovations: Expanding Horizons in Drug Delivery Techniques. Pharmaceutics 2025; 17:136. [PMID: 39861782 PMCID: PMC11768797 DOI: 10.3390/pharmaceutics17010136] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/08/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Nanosuspensions (NS), with their submicron particle sizes and unique physicochemical properties, provide a versatile solution for enhancing the administration of medications that are not highly soluble in water or lipids. This review highlights recent advancements, future prospects, and challenges in NS-based drug delivery, particularly for oral, ocular, transdermal, pulmonary, and parenteral routes. The conversion of oral NS into powders, pellets, granules, tablets, and capsules, and their incorporation into film dosage forms to address stability concerns is thoroughly reviewed. This article summarizes key stabilizers, polymers, surfactants, and excipients used in NS formulations, along with ongoing clinical trials and recent patents. Furthermore, a comprehensive analysis of various methods for NS preparation is provided. This article also explores various in vitro and in vivo characterization techniques, as well as scale-down technologies and bottom-up methods for NS preparation. Selected examples of commercial NS drug products are discussed. Rapid advances in the field of NS could resolve issues related to permeability-limited absorption and hepatic first-pass metabolism, offering promise for medications based on proteins and peptides. The evolution of novel stabilizers is essential to overcome the current limitations in NS formulations, enhancing their stability, bioavailability, targeting ability, and safety profile, which ultimately accelerates their clinical application and commercialization.
Collapse
Affiliation(s)
- Shery Jacob
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates;
| | - Fathima Sheik Kather
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates;
| | - Sai H. S. Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman P.O. Box 346, United Arab Emirates;
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Mahesh Attimarad
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (M.A.); (A.B.N.)
| | - Anroop B. Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (M.A.); (A.B.N.)
| |
Collapse
|
13
|
Ge Y, Yang M, Yu X, Zhou Y, Zhang Y, Mou M, Chen Z, Sun X, Ni F, Fu T, Liu S, Han L, Zhu F. MolBiC: the cell-based landscape illustrating molecular bioactivities. Nucleic Acids Res 2025; 53:D1683-D1691. [PMID: 39373530 PMCID: PMC11701603 DOI: 10.1093/nar/gkae868] [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: 08/15/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024] Open
Abstract
The measurement of cell-based molecular bioactivity (CMB) is critical for almost every step of drug development. With the booming application of AI in biomedicine, it is essential to have the CMB data to promote the learning of cell-based patterns for guiding modern drug discovery, but no database providing such information has been constructed yet. In this study, we introduce MolBiC, a knowledge base designed to describe valuable data on molecular bioactivity measured within a cellular context. MolBiC features 550 093 experimentally validated CMBs, encompassing 321 086 molecules and 2666 targets across 988 cell lines. Our MolBiC database is unique in describing the valuable data of CMB, which meets the critical demands for CMB-based big data promoting the learning of cell-based molecular/pharmaceutical pattern in drug discovery and development. MolBiC is now freely accessible without any login requirement at: https://idrblab.org/MolBiC/.
Collapse
Affiliation(s)
- Yichao Ge
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai 200040, China
- Greater Bay Area Institute of Precision Medicine, School of Life Sciences, Guangzhou, Guangzhou 511458, China
| | - Mengjie Yang
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Xinyuan Yu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yintao Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Zhen Chen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Xiuna Sun
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Feng Ni
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Tingting Fu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Shuiping Liu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Lianyi Han
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai 200040, China
- Greater Bay Area Institute of Precision Medicine, School of Life Sciences, Guangzhou, Guangzhou 511458, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
14
|
Imai T, Isasaka M, Oyama Y, Takagi Y, Ohura K, Kotani S, Nakada Y. Functional Analysis of Modified Caco-2 Cells Carrying CES2A1 as a Model for Intestinal Absorption of Prodrugs. Biol Pharm Bull 2025; 48:162-171. [PMID: 40010717 DOI: 10.1248/bpb.b24-00645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Carboxylesterase (CES) plays an important role in the metabolism of ester-containing drugs such as prodrugs and is highly expressed in the human intestine and liver. The ideal prodrug is barely hydrolyzed by human intestinal CES (CES2A1) but is extensively converted to an active drug by human hepatic CES (CES1A). It is, therefore, important to evaluate CES2A1-mediated hydrolysis during intestinal absorption. Unfortunately, Caco-2 cells, the most common enterocyte model for drug permeability, are not suitable for permeability studies of prodrugs due to their high and extremely low expression of CES1A and CES2A1, respectively. Previously, we have prepared CES2/Caco-2CES1KD cells with reduced human CES1A and highly expressed CES2A1. In the present study, the metabolic and transport properties of CES2/Caco-2CES1KD cells were characterized. The expression of transporters and metabolizing enzymes other than CESs was similar in CES2/Caco-2CES1KD and Caco-2 cells. However, the expression of CES2A1 in CES2/Caco-2CES1KD was about 7-10 fold higher than that of CES1A in Caco-2 cells and comparable to levels found in the human intestine. Hydrolysis during transport across cell monolayers was analyzed using ethyl and butyl esters of p-aminobenzoic acid (PABA). Ethyl PABA, a better substrate for CES1A than CES2A1, was similarly hydrolyzed in Caco-2 and CES2/Caco-2CES1KD cell monolayers due to the high expression of CES2A1 in CES2/Caco-2CES1KD cells. Butyl PABA, a good substrate for CES2A1, was substantially hydrolyzed in CES2/Caco-2CES1KD cell monolayers, in contrast to negligible hydrolysis in Caco-2 cell monolayers. N-Acetylation of PABA derived from PABA esters showed similar activity in Caco-2 and CES2/Caco-2CES1KD cell monolayers.
Collapse
Affiliation(s)
- Teruko Imai
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
- Daiichi University of Pharmacy, 22-1 Tamagawa-machi, Minami-ku, Fukuoka 815-8511, Japan
| | - Masanari Isasaka
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yusuke Oyama
- School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yusuke Takagi
- School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Kayoko Ohura
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
- Pharmacy Talent Development Department, Sogo Medical Co., Ltd., PRIO Fukuoka Building, 2‒9‒23 Daimyo, Chuo-ku, Fukuoka 810‒0041, Japan
| | - Shunsuke Kotani
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yuichiro Nakada
- Faculty of Pharmacy, Osaka Ohtani University, 3-11-1 Nishikiori-kita, Tondabayashi, Osaka 584-0066, Japan
| |
Collapse
|
15
|
Xu W, Jia A, Lei Z, Wang J, Jiang H, Wang S, Wang Q. Stimuli-responsive prodrugs with self-immolative linker for improved cancer therapy. Eur J Med Chem 2024; 279:116928. [PMID: 39362023 DOI: 10.1016/j.ejmech.2024.116928] [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: 08/26/2024] [Revised: 09/19/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
Self-immolative prodrugs have gained significant attention as an innovative approach for targeted cancer therapy. These prodrugs are engineered to release the active anticancer agents in response to specific triggers within the tumor microenvironment, thereby improving therapeutic precision while reducing systemic toxicity. This review focuses on the molecular architecture and design principles of self-immolative prodrugs, emphasizing the role of stimuli-responsive linkers and activation mechanisms that enable controlled drug release. Recent advancements in this field include the development of prodrugs that incorporate targeting moieties for enhanced site-specificity. Moreover, the review discusses the incorporation of targeting moieties to achieve site-specific drug delivery, thereby improving the selectivity of treatment. By summarizing key research from the past five years, this review highlights the potential of self-immolative prodrugs to revolutionize cancer treatment strategies and pave the way for their integration into clinical practice.
Collapse
Affiliation(s)
- Wenting Xu
- Department of Pediatric Intensive Care Medicine, Hainan Women and Children's Medical Center, Haikou, China
| | - Ang Jia
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Zhixian Lei
- Department of Pediatric Intensive Care Medicine, Hainan Women and Children's Medical Center, Haikou, China
| | - Jianing Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Hongfei Jiang
- School of Pharmacy, Qingdao University, Qingdao, 266071, China.
| | - Shuai Wang
- Department of Radiotherapy, School of Medical Imaging, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, Shandong, China.
| | - Qi Wang
- Department of Pediatric Intensive Care Medicine, Hainan Women and Children's Medical Center, Haikou, China.
| |
Collapse
|
16
|
Baró EL, Catti F, Estarellas C, Ghashghaei O, Lavilla R. Drugs from drugs: New chemical insights into a mature concept. Drug Discov Today 2024; 29:104212. [PMID: 39442750 DOI: 10.1016/j.drudis.2024.104212] [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: 07/30/2024] [Revised: 10/01/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Developing new drugs from marketed ones is a well-established and successful approach in drug discovery. We offer a unified view of this field, focusing on the new chemical aspects of the involved approaches: (a) chemical transformation of the original drugs (late-stage modifications, molecular editing), (b) prodrug strategies, and (c) repurposing as a tool to develop new hits/leads. Special focus is placed on the molecular structure of the drugs and their synthetic feasibility. The combination of experimental advances and new computational approaches, including artificial intelligence methods, paves the way for the evolution of the drugs from drugs concept.
Collapse
Affiliation(s)
- Eloy Lozano Baró
- Laboratory of Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona and Institute of Biomedicine UB (IBUB), Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Federica Catti
- Faculty of Science and Mathematics, Arkansas State University Campus Querétaro, Carretera Estatal 100, km 17.5. C.P. 76270, Municipio de Colón, Estado de Querétaro, Mexico
| | - Carolina Estarellas
- Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Institut de Química Teòrica i Computacional, University of Barcelona, Barcelona, Spain
| | - Ouldouz Ghashghaei
- Laboratory of Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona and Institute of Biomedicine UB (IBUB), Av. Joan XXIII, 27-31, 08028 Barcelona, Spain.
| | - Rodolfo Lavilla
- Laboratory of Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona and Institute of Biomedicine UB (IBUB), Av. Joan XXIII, 27-31, 08028 Barcelona, Spain.
| |
Collapse
|
17
|
Urbaniak T, Milasheuski Y, Musiał W. Zero-Order Kinetics Release of Lamivudine from Layer-by-Layer Coated Macromolecular Prodrug Particles. Int J Mol Sci 2024; 25:12921. [PMID: 39684632 DOI: 10.3390/ijms252312921] [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: 10/30/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
To reduce the risk of side effects and enhance therapeutic efficiency, drug delivery systems that offer precise control over active ingredient release while minimizing burst effects are considered advantageous. In this study, a novel approach for the controlled release of lamivudine (LV) was explored through the fabrication of polyelectrolyte-coated microparticles. LV was covalently attached to poly(ε-caprolactone) via ring-opening polymerization, resulting in a macromolecular prodrug (LV-PCL) with a hydrolytic release mechanism. The LV-PCL particles were subsequently coated using the layer-by-layer (LbL) technique, with polyelectrolyte multilayers assembled to potentially modify the carrier's properties. The LbL assembly process was comprehensively analyzed, including assessments of shell thickness, changes in ζ-potential, and thermodynamic properties, to provide insights into the multilayer structure and interactions. The sustained LV release over 7 weeks was observed, following zero-order kinetics (R2 > 0.99), indicating a controlled and predictable release mechanism. Carriers coated with polyethylene imine/heparin and chitosan/heparin tetralayers exhibited a distinct increase in the release rate after 6 weeks and 10 weeks, respectively, suggesting that this coating can facilitate the autocatalytic degradation of the polyester microparticles. These findings indicate the potential of this system for long-term, localized drug delivery applications, requiring sustained release with minimal burst effects.
Collapse
Affiliation(s)
- Tomasz Urbaniak
- Department of Physical Chemistry and Biophysics, Pharmaceutical Faculty, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland
| | - Yauheni Milasheuski
- Department of Physical Chemistry and Biophysics, Pharmaceutical Faculty, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland
| | - Witold Musiał
- Department of Physical Chemistry and Biophysics, Pharmaceutical Faculty, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland
| |
Collapse
|
18
|
Mashweu AR, Azov VA. Nanotechnology in Drug Delivery: Anatomy and Molecular Insight into the Self-Assembly of Peptide-Based Hydrogels. Molecules 2024; 29:5654. [PMID: 39683812 PMCID: PMC11643151 DOI: 10.3390/molecules29235654] [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: 10/24/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
The bioavailability, release, and stability of pharmaceuticals under physicochemical conditions is the major cause of drug candidates failing during their clinical trials. Therefore, extensive efforts have been invested in the development of novel drug delivery systems that are able to transport drugs to a desired site and improve bioavailability. Hydrogels, and peptide hydrogels in particular, have been extensively investigated due to their excellent biocompatibility and biodegradability properties. However, peptide hydrogels often have weak mechanical strength, which limits their therapeutic efficacy. Therefore, a number of methods for improving their rheological properties have been established. This review will cover the broad area of drug delivery, focusing on the recent developments in this research field. We will discuss the variety of different types of nanocarrier drug delivery systems and then, more specifically, the significance and perspectives of peptide-based hydrogels. In particular, the interplay of intermolecular forces that govern the self-assembly of peptide hydrogels, progress made in understanding the distinct morphologies of hydrogels, and applications of non-canonical amino acids in hydrogel design will be discussed in more detail.
Collapse
Affiliation(s)
- Adelaide R. Mashweu
- Department of Chemistry, University of the Free State, P.O. Box 339, Bloemfontein 9300, South Africa
| | - Vladimir A. Azov
- Department of Chemistry, University of the Free State, P.O. Box 339, Bloemfontein 9300, South Africa
| |
Collapse
|
19
|
A M Subbaiah M, Ramar T, Reddy M, Sivaprasad Lvj S, Yeshwante S, Sridhar S, Desai S, Chiney M, Dierks EA, Mathur A, Moslin R, Weinstein DS. Prodrug Strategy to Address Impaired Oral Absorption of a Weakly Basic TYK2 Inhibitor Caused by a Gastric Acid-Reducing Agent. J Med Chem 2024; 67:20664-20681. [PMID: 39560351 DOI: 10.1021/acs.jmedchem.4c02219] [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: 11/20/2024]
Abstract
The pH-dependent solubility of the weakly basic TYK2 inhibitor 1 posed a risk to its advancement, given that drugs with such profiles have exhibited drug-drug interaction (DDI) with stomach acid-reducing agents in humans. In a rat model of pH dependence, preadministration of famotidine caused a 2.4-fold lower exposure of 1 when compared to control rats, implying that pH-dependent oral absorption can reduce the active drug's exposure and translate to subtherapeutic treatment. As part of risk mitigation, a prodrug strategy was explored by synthesizing solubility-enhancing prodrugs, resulting in the identification of lead prodrug 3c with acceptable stability and solubility profiles. In rats, the prodrug eliminated the significant difference in AUC and Cmax between pentagastrin and famotidine arms, thereby effectively mitigating the impaired drug absorption at the elevated pH relevant for absorption and DDI with famotidine. The prodrug also facilitated dose-proportional systemic exposure of 1 following dose escalation in rats and monkeys.
Collapse
|
20
|
Song G, Yang Z, Huang Y, Bai H, Lv F, Wang S. Chemically engineered exogenous organic reactions in living cells for in situ fluorescence imaging and biomedical applications. J Mater Chem B 2024; 12:11852-11866. [PMID: 39485083 DOI: 10.1039/d4tb01925c] [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: 11/03/2024]
Abstract
The unique microenvironment within living cells, characterized by high glutathione levels, reactive oxygen species concentrations, and active enzymes, facilitates the execution of chemical reactions. Recent advances in organic chemistry and chemical biology have leveraged living cells as reactors for chemical synthesis. This review summarizes recent reports on key intracellular in situ synthesis processes, including the synthesis of near-infrared fluorescent dyes, intracellular oxidative cross-linking, bioorthogonal reactions, and intracellular polymerization reactions. These methods have been applied to fluorescence imaging, tumor treatment, and the enhancement of biological functions. Finally, we discuss the challenges and opportunities in the field of in situ intracellular synthesis. We aim to guide the design of chemical molecules for in situ synthesis, improving the efficiency and control of artificial reactions in living cells, and ultimately achieving cell factory-like exogenous biological synthesis, biological function enhancement, and biomedical applications.
Collapse
Affiliation(s)
- Gang Song
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiwen Yang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiming Huang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Haotian Bai
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Fengting Lv
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
21
|
Abo Qoura L, Morozova E, Ramaa СS, Pokrovsky VS. Smart nanocarriers for enzyme-activated prodrug therapy. J Drug Target 2024; 32:1029-1051. [PMID: 39045650 DOI: 10.1080/1061186x.2024.2383688] [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: 03/18/2024] [Revised: 06/26/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
Exogenous enzyme-activated prodrug therapy (EPT) is a potential cancer treatment strategy that delivers non-human enzymes into or on the surface of the cell and subsequently converts a non-toxic prodrug into an active cytotoxic substance at a specific location and time. The development of several pharmacological pairs based on EPT has been the focus of anticancer research for more than three decades. Numerous of these pharmacological pairs have progressed to clinical trials, and a few have achieved application in specific cancer therapies. The current review highlights the potential of enzyme-activated prodrug therapy as a promising anticancer treatment. Different microbial, plant, or viral enzymes and their corresponding prodrugs that advanced to clinical trials have been listed. Additionally, we discuss new trends in the field of enzyme-activated prodrug nanocarriers, including nanobubbles combined with ultrasound (NB/US), mesoscopic-sized polyion complex vesicles (PICsomes), nanoparticles, and extracellular vesicles (EVs), with special emphasis on smart stimuli-triggered drug release, hybrid nanocarriers, and the main application of nanotechnology in improving prodrugs.
Collapse
Affiliation(s)
- Louay Abo Qoura
- Research Institute of Molecular and Cellular Medicine, People's Friendship University of Russia (RUDN University), Moscow, Russia
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Elena Morozova
- Engelhardt Institute of Molecular Biology of the, Russian Academy of Sciences, Moscow, Russia
| | - С S Ramaa
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth's College of Pharmacy, Mumbai, India
| | - Vadim S Pokrovsky
- Research Institute of Molecular and Cellular Medicine, People's Friendship University of Russia (RUDN University), Moscow, Russia
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
22
|
Miyaji K, Masaki Y, Seio K. Inhibitory Effects on RNA Binding and RNase H Induction Activity of Prodrug-Type Oligodeoxynucleotides Modified with a Galactosylated Self-Immolative Linker Cleavable by β-Galactosidase. Bioconjug Chem 2024. [PMID: 39376088 DOI: 10.1021/acs.bioconjchem.4c00376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Prodrug-type oligonucleotides (prodrug-ONs) are a class of oligonucleotide designed for activation under specific intracellular conditions or external stimuli. Prodrug-ONs can be activated in the target tissues or cells, thereby reducing the risk of adverse effects. In this study, we synthesized prodrug-type oligodeoxynucleotides activated by β-galactosidase, an enzyme that is overexpressed in cancer and senescent cells. These oligodeoxynucleotides (ODNs) contain a modified thymidine conjugated with galactose via a self-immolative linker at the O4-position. UV-melting analysis revealed that the modifications decreased the melting temperature (Tm) compared with that of the unmodified ODN when hybridized with complementary RNA. Furthermore, cleavage of the glycosidic bond by β-galactosidase resulted in the spontaneous removal of the linker from the nucleobase moiety, generating unmodified ODNs. Additionally, the introduction of multiple modified thymidines into ODNs completely inhibited the RNase H-mediated cleavage of complementary RNA. These findings suggest the possibility of developing prodrug-ONs, which are specifically activated in cancer cells or senescent cells with high β-galactosidase expression.
Collapse
Affiliation(s)
- Kento Miyaji
- Department of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Yoshiaki Masaki
- Department of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
- Nucleotide and Peptide Drug Discovery Center, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Kohji Seio
- Department of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| |
Collapse
|
23
|
Sankar J, Chauhan A, Singh R, Mahajan D. Isoniazid-historical development, metabolism associated toxicity and a perspective on its pharmacological improvement. Front Pharmacol 2024; 15:1441147. [PMID: 39364056 PMCID: PMC11447295 DOI: 10.3389/fphar.2024.1441147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/30/2024] [Indexed: 10/05/2024] Open
Abstract
Despite the extraordinary anti-tubercular activity of isoniazid (INH), the drug-induced hepatotoxicity and peripheral neuropathy pose a significant challenge to its wider clinical use. The primary cause of INH-induced hepatotoxicity is in vivo metabolism involving biotransformation on its terminal -NH2 group owing to its high nucleophilic nature. The human N-acetyltransferase-2 enzyme (NAT-2) exploits the reactivity of INH's terminal -NH2 functional group and inactivates it by transferring the acetyl group, which subsequently converts to toxic metabolites. This -NH2 group also tends to react with vital endogenous molecules such as pyridoxine, leading to their deficiency, a major cause of peripheral neuropathy. The elevation of liver functional markers is observed in 10%-20% of subjects on INH treatment. INH-induced risk of fatal hepatitis is about 0.05%-1%. The incidence of peripheral neuropathy is 2%-6.5%. In this review, we discuss the genesis and historical development of INH, and different reported mechanisms of action of INH. This is followed by a brief review of various clinical trials in chronological order, highlighting treatment-associated adverse events and their occurrence rates, including details such as geographical location, number of subjects, dosing concentration, and regimen used in these clinical studies. Further, we elaborated on various known metabolic transformations highlighting the involvement of the terminal -NH2 group of INH and corresponding host enzymes, the structure of different metabolites/conjugates, and their association with hepatotoxicity or neuritis. Post this deliberation, we propose a hydrolysable chemical derivatives-based approach as a way forward to restrict this metabolism.
Collapse
Affiliation(s)
- Jishnu Sankar
- Centre for Drug Discovery, BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Anjali Chauhan
- Centre for Drug Discovery, BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Centre for Tuberculosis Research, BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Ramandeep Singh
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, New Delhi, India
| | - Dinesh Mahajan
- Centre for Drug Discovery, BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Centre for Tuberculosis Research, BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
24
|
Keydel T, Link A. Synthetic Approaches, Properties, and Applications of Acylals in Preparative and Medicinal Chemistry. Molecules 2024; 29:4451. [PMID: 39339447 PMCID: PMC11434492 DOI: 10.3390/molecules29184451] [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: 08/09/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Diesters of geminal diols (R-CH(O-CO-R')2, RR'C(OCOR″)2, etc. with R = H, aryl or alkyl) are termed acylals according to IUPAC recommendations (Rule P-65.6.3.6 Acylals) if the acids involved are carboxylic acids. Similar condensation products can be obtained from various other acidic structures as well, but these related "non-classical acylals", as one might call them, differ in various aspects from classical acylals and will not be discussed in this article. Carboxylic acid diesters of geminal diols play a prominent role in organic chemistry, not only in their application as protective groups for aldehydes and ketones but also as precursors in the total synthesis of natural compounds and in a variety of organic reactions. What is more, acylals are useful as a key structural motif in clinically validated prodrug approaches. In this review, we summarise the syntheses and chemical properties of such classical acylals and show what potentially under-explored possibilities exist in the field of drug design, especially prodrugs, and classify this functional group in medicinal chemistry.
Collapse
Affiliation(s)
| | - Andreas Link
- Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany;
| |
Collapse
|
25
|
Maria C, de Matos AM, Rauter AP. Antibacterial Prodrugs to Overcome Bacterial Antimicrobial Resistance. Pharmaceuticals (Basel) 2024; 17:718. [PMID: 38931385 PMCID: PMC11206681 DOI: 10.3390/ph17060718] [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: 05/10/2024] [Revised: 05/25/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Antimicrobial resistance (AMR) is an increasingly concerning phenomenon that requires urgent attention because it poses a threat to human and animal health. Bacteria undergo continuous evolution, acquiring novel resistance mechanisms in addition to their intrinsic ones. Multidrug-resistant and extensively drug-resistant bacterial strains are rapidly emerging, and it is expected that bacterial AMR will claim the lives of 10 million people annually by 2050. Consequently, the urgent need for the development of new therapeutic agents with new modes of action is evident. The antibacterial prodrug approach, a strategy that includes drug repurposing and derivatization, integration of nanotechnology, and exploration of natural products, is highlighted in this review. Thus, this publication aims at compiling the most pertinent research in the field, spanning from 2021 to 2023, offering the reader a comprehensive insight into the AMR phenomenon and new strategies to overcome it.
Collapse
Affiliation(s)
| | | | - Amélia P. Rauter
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal; (C.M.); (A.M.d.M.)
| |
Collapse
|
26
|
Yao SY, Ying AK, Jiang ZT, Cheng YQ, Geng WC, Hu XY, Cai K, Guo DS. Single Molecular Nanomedicines Based on Macrocyclic Carrier-Drug Conjugates for Concentration-Independent Encapsulation and Precise Activation of Drugs. J Am Chem Soc 2024; 146:14203-14212. [PMID: 38733560 DOI: 10.1021/jacs.4c03238] [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: 05/13/2024]
Abstract
Nanomedicines often rely on noncovalent self-assembly and encapsulation for drug loading and delivery. However, challenges such as reproducibility issues due to the multicomponent nature, off-target activation caused by premature drug release, and complex pharmacokinetics arising from assembly dissociation have hindered their clinical translation. In this study, we introduce an innovative design concept termed single molecular nanomedicine (SMNM) based on macrocyclic carrier-drug conjugates. Through the covalent linkage of two chemotherapy drugs to a hypoxia-cleavable macrocyclic carrier, azocalix[4]arene, we obtained two self-included complexes to serve as SMNMs. The intramolecular inclusion feature of the SMNMs has not only demonstrated comprehensive shielding and protection for the drugs but also effectively prevented off-target drug leakage, thereby significantly reducing their side effects and enhancing their antitumor therapeutic efficacy. Additionally, the attributes of being a single component and molecularly dispersed confer advantages such as ease of preparation and good reproducibility for SMNMs, which is desirable for clinical applications.
Collapse
Affiliation(s)
- Shun-Yu Yao
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - An-Kang Ying
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Ze-Tao Jiang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Yuan-Qiu Cheng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Wen-Chao Geng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Xin-Yue Hu
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Kang Cai
- College of Chemistry, Nankai University, Tianjin 300071, China
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, College of Chemistry and Environmental Sciences, Kashi University, Kashi 844000, China
| |
Collapse
|