1
|
Muhammad AM, Ismail A, Chong PP, Yap WH, Muhamad A, Alitheen NB, Kam A, Loo S, Lee KW. Skin-penetrating peptides (SKPs): Enhancing skin permeation for transdermal delivery of pharmaceuticals and cosmetic compounds. Int J Pharm 2025; 672:125339. [PMID: 39947363 DOI: 10.1016/j.ijpharm.2025.125339] [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: 11/19/2024] [Revised: 01/16/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Skin-penetrating peptides (SKPs) are emerging as a promising class of permeation enhancers that can facilitate macromolecule delivery across the skin. Although their pharmaceutical applications are under extensive study, SKPs are crucial for enhancing skin permeability, enabling larger molecules to penetrate the stratum corneum. This review explores the transformative role of SKPs in non-invasive transdermal drug delivery. Drawing from an extensive collection of literature, it provides insights into the current usage and application of SKPs as tools to enhance skin permeability and facilitate the delivery of larger molecules. Additionally, it highlights the opportunities, challenges, and future directions for SKP applications in transdermal drug delivery.
Collapse
Affiliation(s)
- Ameerah Montree Muhammad
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Alif Ismail
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Pei Pei Chong
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia; Digital Health and Medical Advancement Impact Lab, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Wei Hsum Yap
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia; Food Security and Nutrition Impact Lab, Taylor's University, Subang Jaya 47500 Selangor, Malaysia
| | - Azira Muhamad
- Malaysia Genome and Vaccine Institute, National Institutes of Biotechnology Malaysia Kajang Selangor Malaysia
| | - Noorjahan Banu Alitheen
- Faculty of Biotechnology & Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Antony Kam
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Shining Loo
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Khai Wooi Lee
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia; Digital Health and Medical Advancement Impact Lab, Taylor's University, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
2
|
Zhang J, Yang Z, Liu Y, Liu Y, Qu J, Pan X. Recent Advances in Smart Linkage Strategies for Developing Drug Conjugates for Targeted Delivery. Top Curr Chem (Cham) 2025; 383:13. [PMID: 40080285 DOI: 10.1007/s41061-025-00497-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: 06/11/2024] [Accepted: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Targeted drug delivery systems effectively solve the problem of off-target toxicity of chemotherapeutic drugs by combining chemotherapeutic drugs with antibodies or peptides, thereby promoting drug targeting to the tumor site and bringing further hope for cancer treatment. The development of stimulus-responsive smart linkage technologies has led to the emergence of drug conjugates. Linkage technologies play a crucial role in the design, synthesis, and in vivo circulation of drug conjugates, as they determine the release of cytotoxic drugs from the conjugates and their subsequent therapeutic efficacy. This article reviews some of the smart linkage strategies used in designing drug conjugates, with a focus on the tumor microenvironment and exogenous stimuli as conditions influencing controlled drug release. This review introduces linker classifications and cleavage mechanisms, discusses modular linkers that promote the efficient synthesis of conjugates, and discusses the differences between linkage strategies. Furthermore, this article focuses on the implementation of self-assembly in drug conjugates, which is currently of great interest. Related concepts are introduced and relevant examples of their applications are provided. Furthermore, a comprehensive discourse is presented on the challenges that may arise in the research and clinical implementation of diverse linkage strategies, along with the associated enhancement measures. Finally, the factors that should be considered when designing linkage strategies for drug conjugates are summarized, offering strategies and ideas for scientists involved in drug conjugate research. It is particularly noteworthy that appropriate linkage strategies allow for the intracellular release of drugs after internalization of the conjugates, thereby maximizing their tumor cell-killing effect.
Collapse
Affiliation(s)
- Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zeyu Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yu Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yuying Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jingkun Qu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
3
|
Zaleski MH, Chase LS, Hood ED, Wang Z, Nong J, Espy CL, Zamora ME, Wu J, Morrell LJ, Muzykantov VR, Myerson JW, Brenner JS. Conjugation Chemistry Markedly Impacts Toxicity and Biodistribution of Targeted Nanoparticles, Mediated by Complement Activation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2409945. [PMID: 39663706 PMCID: PMC11795710 DOI: 10.1002/adma.202409945] [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: 07/10/2024] [Revised: 11/21/2024] [Indexed: 12/13/2024]
Abstract
Conjugation chemistries are a major enabling technology for the development of drug delivery systems, from antibody-drug conjugates to antibody-targeted lipid nanoparticles inspired by the success of the COVID-19 vaccine. However, here it is shown that for antibody-targeted nanoparticles, the most popular conjugation chemistries directly participate in the activation of the complement cascade of plasma proteins. Their activation of complement leads to large changes in the biodistribution of nanoparticles (up to 140-fold increased uptake into phagocytes of the lungs) and multiple toxicities, including a 50% drop in platelet count. It is founded that the mechanism of complement activation varies dramatically between different conjugation chemistries. Dibenzocyclooctyne, a commonly used click-chemistry, caused aggregation of conjugated antibodies, but only on the surface of nanoparticles (not in bulk solution). By contrast, thiol-maleimide chemistry do not activate complement via its effects on antibodies, but rather because free maleimide bonded to albumin in plasma, and clustered albumin is then attacked by complement. Using these mechanistic insights, solutions are engineered that reduced the activation of complement for each class of conjugation chemistry. These results highlight that while conjugation chemistry is essential for the future of nanomedicine, it is not innocuous and must be designed with opsonins like complement in mind.
Collapse
Affiliation(s)
- Michael H. Zaleski
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Liam S. Chase
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Elizabeth D. Hood
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Zhicheng Wang
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Jia Nong
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Carolann L. Espy
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Marco E. Zamora
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Jichuan Wu
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Lianne J. Morrell
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Vladimir R. Muzykantov
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Jacob W. Myerson
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Jacob S. Brenner
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
- Department of MedicineUniversity of Pennsylvania3400 Civic Center BoulevardPhiladelphiaPA19104USA
| |
Collapse
|
4
|
Pande S. Factors affecting response variables with emphasis on drug release and loading for optimization of liposomes. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:334-344. [PMID: 38833335 DOI: 10.1080/21691401.2024.2360634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Drug delivery through Liposomes has shown tremendous potential in terms of the therapeutic application of nanoparticles. There are several drug-loaded liposomal formulations approved for clinical use that help mitigate harmful effects of life-threatening diseases. Developments in the field of liposomal formulations and drug delivery have made it possible for clinicians and researchers to find therapeutic solutions for complicated medical conditions. A key aspect in the development of drug-loaded liposomes is a careful review of optimization techniques to improve the overall formulation stability and efficacy. Optimization studies help in improving/modulating the various properties of drug-loaded liposomes and are vital for the development of this class of delivery systems. A comprehensive overview of the various process variables and factors involved in the optimization of drug-loaded liposomes is presented in this review. The influence of different independent variables on drug release and loading properties with the application of a statistical experimental design is also explained in this article.
Collapse
Affiliation(s)
- Shantanu Pande
- Drug Product Technical Services, Wave Life Sciences, MA, USA
| |
Collapse
|
5
|
Ma X, Tian Y, Yang R, Wang H, Allahou LW, Chang J, Williams G, Knowles JC, Poma A. Nanotechnology in healthcare, and its safety and environmental risks. J Nanobiotechnology 2024; 22:715. [PMID: 39548502 PMCID: PMC11566612 DOI: 10.1186/s12951-024-02901-x] [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: 07/22/2024] [Accepted: 10/03/2024] [Indexed: 11/18/2024] Open
Abstract
Nanotechnology holds immense promise in revolutionising healthcare, offering unprecedented opportunities in diagnostics, drug delivery, cancer therapy, and combating infectious diseases. This review explores the multifaceted landscape of nanotechnology in healthcare while addressing the critical aspects of safety and environmental risks associated with its widespread application. Beginning with an introduction to the integration of nanotechnology in healthcare, we first delved into its categorisation and various materials employed, setting the stage for a comprehensive understanding of its potential. We then proceeded to elucidate the diverse healthcare applications of nanotechnology, spanning medical diagnostics, tissue engineering, targeted drug delivery, gene delivery, cancer therapy, and the development of antimicrobial agents. The discussion extended to the current situation surrounding the clinical translation and commercialisation of these cutting-edge technologies, focusing on the nanotechnology-based healthcare products that have been approved globally to date. We also discussed the safety considerations of nanomaterials, both in terms of human health and environmental impact. We presented the in vivo health risks associated with nanomaterial exposure, in relation with transport mechanisms, oxidative stress, and physical interactions. Moreover, we highlighted the environmental risks, acknowledging the potential implications on ecosystems and biodiversity. Lastly, we strived to offer insights into the current regulatory landscape governing nanotechnology in healthcare across different regions globally. By synthesising these diverse perspectives, we underscore the imperative of balancing innovation with safety and environmental stewardship, while charting a path forward for the responsible integration of nanotechnology in healthcare.
Collapse
Affiliation(s)
- Xiaohan Ma
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, Royal Free Hospital, University College London, Rowland Hill Street, London, NW3 2PF, UK.
| | - Yaxin Tian
- United InnoMed (Shanghai) Limited, F/2, E-1, No.299, Kangwei Rd, Pudong District, Shanghai, China
| | - Ren Yang
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, Royal Free Hospital, University College London, Rowland Hill Street, London, NW3 2PF, UK
| | - Haowei Wang
- Centre for Precision Healthcare, UCL Division of Medicine, University College London, London, WC1E 6JF, UK
| | - Latifa W Allahou
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, Royal Free Hospital, University College London, Rowland Hill Street, London, NW3 2PF, UK
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Jinke Chang
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Division of Surgery & Interventional Science, University College London, London, NW3 2PF, UK
| | - Gareth Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Jonathan C Knowles
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, Royal Free Hospital, University College London, Rowland Hill Street, London, NW3 2PF, UK
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Med-Icine, Dankook University, Cheonan, 31116, South Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, South Korea
| | - Alessandro Poma
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, Royal Free Hospital, University College London, Rowland Hill Street, London, NW3 2PF, UK.
| |
Collapse
|
6
|
Verma VS, Pandey A, Jha AK, Badwaik HKR, Alexander A, Ajazuddin. Polyethylene Glycol-Based Polymer-Drug Conjugates: Novel Design and Synthesis Strategies for Enhanced Therapeutic Efficacy and Targeted Drug Delivery. Appl Biochem Biotechnol 2024; 196:7325-7361. [PMID: 38519751 DOI: 10.1007/s12010-024-04895-6] [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] [Accepted: 03/04/2024] [Indexed: 03/25/2024]
Abstract
Due to their potential to enhance therapeutic results and enable targeted drug administration, polymer-drug conjugates that use polyethylene glycol (PEG) as both the polymer and the linker for drug conjugation have attracted much research. This study seeks to investigate recent developments in the design and synthesis of PEG-based polymer-drug conjugates, emphasizing fresh ideas that fill in existing knowledge gaps and satisfy the increasing need for more potent drug delivery methods. Through an extensive review of the existing literature, this study identifies key challenges and proposes innovative strategies for future investigations. The paper presents a comprehensive framework for designing and synthesizing PEG-based polymer-drug conjugates, including rational molecular design, linker selection, conjugation methods, and characterization techniques. To further emphasize the importance and adaptability of PEG-based polymer-drug conjugates, prospective applications are highlighted, including cancer treatment, infectious disorders, and chronic ailments.
Collapse
Affiliation(s)
- Vinay Sagar Verma
- Faculty of Pharmaceutical Sciences, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai, Durg, Chhattisgarh, 490023, India
| | - Aakansha Pandey
- Faculty of Pharmaceutical Sciences, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India
| | - Arvind Kumar Jha
- Shri Shankaracharya Professional University, Junwani, Bhilai, 490020, Chhattisgarh, India
| | - Hemant Kumar Ramchandra Badwaik
- Shri Shankaracharya College of Pharmaceutical Sciences, Junwani, Bhilai, 490020, Chhattisgarh, India.
- Shri Shankaracharya Institute of Pharmaceutical Sciences and Research, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India.
| | - Amit Alexander
- Department of Pharmaceuticals, National Institute of Pharmaceutical Education and Research, Ministry of Chemical and Fertilizers, Guwahati, 781101, Assam, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai, Durg, Chhattisgarh, 490023, India.
| |
Collapse
|
7
|
Chen Y, Liu F, Pal S, Hu Q. Proteolysis-targeting drug delivery system (ProDDS): integrating targeted protein degradation concepts into formulation design. Chem Soc Rev 2024; 53:9582-9608. [PMID: 39171633 DOI: 10.1039/d4cs00411f] [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: 08/23/2024]
Abstract
Targeted protein degradation (TPD) has emerged as a revolutionary paradigm in drug discovery and development, offering a promising avenue to tackle challenging therapeutic targets. Unlike traditional drug discovery approaches that focus on inhibiting protein function, TPD aims to eliminate proteins of interest (POIs) using modular chimeric structures. This is achieved through the utilization of proteolysis-targeting chimeras (PROTACs), which redirect POIs to E3 ubiquitin ligases, rendering them for degradation by the cellular ubiquitin-proteasome system (UPS). Additionally, other TPD technologies such as lysosome-targeting chimeras (LYTACs) and autophagy-based protein degraders facilitate the transportation of proteins to endo-lysosomal or autophagy-lysosomal pathways for degradation, respectively. Despite significant growth in preclinical TPD research, many chimeras fail to progress beyond this stage in the drug development. Various factors contribute to the limited success of TPD agents, including a significant hurdle of inadequate delivery to the target site. Integrating TPD into delivery platforms could surmount the challenges of in vivo applications of TPD strategies by reshaping their pharmacokinetics and pharmacodynamic profiles. These proteolysis-targeting drug delivery systems (ProDDSs) exhibit superior delivery performance, enhanced targetability, and reduced off-tissue side effects. In this review, we will survey the latest progress in TPD-inspired drug delivery systems, highlight the importance of introducing delivery ideas or technologies to the development of protein degraders, outline design principles of protein degrader-inspired delivery systems, discuss the current challenges, and provide an outlook on future opportunities in this field.
Collapse
Affiliation(s)
- Yu Chen
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Fengyuan Liu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Samira Pal
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
8
|
Yang Y, Chu Y, Li C, Fan L, Lu H, Zhan C, Zhang Z. Brain-targeted drug delivery by in vivo functionalized liposome with stable D-peptide ligand. J Control Release 2024; 373:240-251. [PMID: 38977135 DOI: 10.1016/j.jconrel.2024.07.014] [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: 04/09/2024] [Revised: 06/17/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
Brain-targeted drug delivery poses a great challenge due to the blood-brain barrier (BBB). In a previous study, we have developed a peptide-modified stealth liposome (SP-sLip) to enhance BBB penetration via the adsorption of apolipoproteins in plasma. SP is an 11-amino acid peptide derived from 25 to 35 of the Amyloid β peptide (Aβ1-42), which is a nature ligand of apolipoproteins. Although freshly prepared SP-sLip exhibited efficient brain targeting performance, it occured self-aggregation and instability in storage. In this study, we developed a D-peptide ligand according to the reverse sequence of SP with D-amino acids, known as DSP, to improve the stability in storage. Notably, DSP exhibited a reduced tendency for self-aggregation and improved stability in comparison to the SP peptide. Furthermore, compared to SP-sLip, DSP-modified sLip (DSP-sLip) demonstrated enhanced stability (>2 weeks), prolonged blood circulation (AUC increased 44.4%), reduced liver and spleen accumulation (reduced by 2.23 times and 1.86 times) with comparable brain-targeting efficiency. Similar to SP-sLip, DSP-sLip selectively adsorbed apolipoprotein A1, E, and J in the blood to form functionalized protein corona, thus crossing BBB via apolipoprotein receptor-mediated transcytosis. These findings underscored the importance of ligand stability in the in vitro and in vivo performance of brain-targeted liposomes, therefore paving the way for the design and optimization of efficient and stable nanocarriers.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201399, China; Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Yuxiu Chu
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Cheng Li
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Lianfeng Fan
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Huiping Lu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201399, China.
| | - Changyou Zhan
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201399, China; Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China.
| | - Zui Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China.
| |
Collapse
|
9
|
Voci S, Gagliardi A, Ambrosio N, Zannetti A, Cosco D. Lipid- and polymer-based formulations containing TNF-α inhibitors for the treatment of inflammatory bowel diseases. Drug Discov Today 2024; 29:104090. [PMID: 38977124 DOI: 10.1016/j.drudis.2024.104090] [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: 02/23/2024] [Revised: 06/10/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
Monoclonal antibodies inhibiting tumor necrosis factor-alpha (iTNF-α) have revolutionized the therapeutic regimen of inflammatory bowel disease, but their main drawback is the parenteral route of administration they require. An alternative approach lies in the delivery of these molecules to the area involved in the inflammatory process by means of innovative formulations able to promote their localization in affected tissues while also decreasing the number of administrations required. This review describes the advantages deriving from the use of lipid- and polymer-based systems containing iTNF-α, focusing on their physicochemical and technological properties and discussing the preclinical results obtained in vivo using rodent models of colitis.
Collapse
Affiliation(s)
- Silvia Voci
- Department of Health Sciences, University of Catanzaro 'Magna Græcia', 88100 Catanzaro, Italy
| | - Agnese Gagliardi
- Department of Health Sciences, University of Catanzaro 'Magna Græcia', 88100 Catanzaro, Italy
| | - Nicola Ambrosio
- Department of Health Sciences, University of Catanzaro 'Magna Græcia', 88100 Catanzaro, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging, National Research Council (IBB-CNR), Naples 80145, Italy
| | - Donato Cosco
- Department of Health Sciences, University of Catanzaro 'Magna Græcia', 88100 Catanzaro, Italy.
| |
Collapse
|
10
|
Cybulski M, Zaremba-Czogalla M, Trzaskowski B, Kubiszewski M, Tobiasz J, Jaromin A, Krzeczyński P, Gubernator J, Michalak O. The conjugates of 5'-deoxy-5-fluorocytidine and hydroxycinnamic acids - synthesis, anti-pancreatic cancer activity and molecular docking studies. RSC Adv 2024; 14:13129-13141. [PMID: 38655481 PMCID: PMC11036175 DOI: 10.1039/d4ra01683a] [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: 03/04/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024] Open
Abstract
New amide conjugates 1-6 of hydroxycinnamic acids (HCA) and 5'-deoxy-5-fluorocytidine (5-dFCR), the prodrug of 5-fluorouracil (5-FU), were synthesized and tested in vitro against pancreatic cancer lines (PDAC). The compounds showed slightly higher efficacy against primary BxPC-3 cells (IC50 values of 14-45 μM) than against metastatic AsPC-1 (IC50 values of 37-133 μM), and similar to that of 5-FU for both PDAC lines. Compound 1, which has a para-(acetyloxy)coumaroyl substituent, was found to be the most potent (IC50 = 14 μM) with a selectivity index of approximately 7 to normal dermal fibroblasts (IC50 = 96 μM). The potential pharmacological profiles were discussed on the basis of the ADME data. Docking to the carboxylesterase CES2 showed that the synthesized compounds have the ability to bind via hydrogen bonding between a specific acetate group of the sugar moiety and Ser228, which belongs to the catalytic triad that causes hydrolysis. Docking to albumin, a major transport protein in the circulatory system, revealed a strong interaction of the conjugates at the binding site which is native to warfarin and responsible for its transport in the body.
Collapse
Affiliation(s)
- Marcin Cybulski
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute Rydygiera 8 01-793 Warsaw Poland +48 453 056 175 +48 453 056 177
| | - Magdalena Zaremba-Czogalla
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw Fryderyka Joliot-Curie 14a 50-383 Wroclaw Poland
| | - Bartosz Trzaskowski
- Chemical and Biological Systems Simulation Laboratory, Center of New Technologies, University of Warsaw Banacha 2c 02-097 Warsaw Poland
| | - Marek Kubiszewski
- Pharmaceutical Analysis Laboratory, Łukasiewicz Research Network-Industrial Chemistry Institute Rydygiera 8 01-793 Warsaw Poland
| | - Joanna Tobiasz
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute Rydygiera 8 01-793 Warsaw Poland +48 453 056 175 +48 453 056 177
| | - Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw Fryderyka Joliot-Curie 14a 50-383 Wroclaw Poland
| | - Piotr Krzeczyński
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute Rydygiera 8 01-793 Warsaw Poland +48 453 056 175 +48 453 056 177
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw Fryderyka Joliot-Curie 14a 50-383 Wroclaw Poland
| | - Olga Michalak
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute Rydygiera 8 01-793 Warsaw Poland +48 453 056 175 +48 453 056 177
| |
Collapse
|
11
|
Moles E, Chang DW, Mansfeld FM, Duly A, Kimpton K, Logan A, Howard CB, Thurecht KJ, Kavallaris M. EGFR Targeting of Liposomal Doxorubicin Improves Recognition and Suppression of Non-Small Cell Lung Cancer. Int J Nanomedicine 2024; 19:3623-3639. [PMID: 38660023 PMCID: PMC11042481 DOI: 10.2147/ijn.s450534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/23/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction Despite improvements in chemotherapy and molecularly targeted therapies, the life expectancy of patients with advanced non-small cell lung cancer (NSCLC) remains less than 1 year. There is thus a major global need to advance new treatment strategies that are more effective for NSCLC. Drug delivery using liposomal particles has shown success at improving the biodistribution and bioavailability of chemotherapy. Nevertheless, liposomal drugs lack selectivity for the cancer cells and have a limited ability to penetrate the tumor site, which severely limits their therapeutic potential. Epidermal growth factor receptor (EGFR) is overexpressed in NSCLC tumors in about 80% of patients, thus representing a promising NSCLC-specific target for redirecting liposome-embedded chemotherapy to the tumor site. Methods Herein, we investigated the targeting of PEGylated liposomal doxorubicin (Caelyx), a powerful off-the-shelf antitumoral liposomal drug, to EGFR as a therapeutic strategy to improve the specific delivery and intratumoral accumulation of chemotherapy in NSCLC. EGFR-targeting of Caelyx was enabled through its complexing with a polyethylene glycol (PEG)/EGFR bispecific antibody fragment. Tumor targeting and therapeutic potency of our treatment approach were investigated in vitro using a panel of NSCLC cell lines and 3D tumoroid models, and in vivo in a cell line-derived tumor xenograft model. Results Combining Caelyx with our bispecific antibody generated uniform EGFR-targeted particles with improved binding and cytotoxic efficacy toward NSCLC cells. Effects were exclusive to cancer cells expressing EGFR, and increments in efficacy positively correlated with EGFR density on the cancer cell surface. The approach demonstrated increased penetration within 3D spheroids and was effective at targeting and suppressing the growth of NSCLC tumors in vivo while reducing drug delivery to the heart. Conclusion EGFR targeting represents a successful approach to enhance the selectivity and therapeutic potency of liposomal chemotherapy toward NSCLC.
Collapse
Affiliation(s)
- Ernest Moles
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, 2052, Australia
- UNSW Australian Centre for Nanomedicine, Faculty of Engineering, UNSW, Sydney, NSW, 2052, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW, Sydney, NSW, 2052, Australia
- UNSW RNA Institute, Faculty of Science, UNSW, Sydney, NSW, 2052, Australia
| | - David W Chang
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, 2052, Australia
- UNSW Australian Centre for Nanomedicine, Faculty of Engineering, UNSW, Sydney, NSW, 2052, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW, Sydney, NSW, 2052, Australia
| | - Friederike M Mansfeld
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, 2052, Australia
- UNSW Australian Centre for Nanomedicine, Faculty of Engineering, UNSW, Sydney, NSW, 2052, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW, Sydney, NSW, 2052, Australia
| | - Alastair Duly
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, 2052, Australia
- UNSW Australian Centre for Nanomedicine, Faculty of Engineering, UNSW, Sydney, NSW, 2052, Australia
| | - Kathleen Kimpton
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, 2052, Australia
| | - Amy Logan
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, 2052, Australia
- UNSW Australian Centre for Nanomedicine, Faculty of Engineering, UNSW, Sydney, NSW, 2052, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW, Sydney, NSW, 2052, Australia
- UNSW RNA Institute, Faculty of Science, UNSW, Sydney, NSW, 2052, Australia
| | - Christopher B Howard
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD, 4072, Australia
- Centre for Advanced Imaging, ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Maria Kavallaris
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, 2052, Australia
- UNSW Australian Centre for Nanomedicine, Faculty of Engineering, UNSW, Sydney, NSW, 2052, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW, Sydney, NSW, 2052, Australia
- UNSW RNA Institute, Faculty of Science, UNSW, Sydney, NSW, 2052, Australia
| |
Collapse
|
12
|
Bunyatova U, Dogan M, Tekin E, Ferhanoğlu O. Ultra-stable nano-micro bubbles in a biocompatible medium for safe delivery of anti-cancer drugs. Sci Rep 2024; 14:5321. [PMID: 38438442 PMCID: PMC10912087 DOI: 10.1038/s41598-024-55654-w] [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: 09/07/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024] Open
Abstract
We conducted a series of experimental investigations to generate laser-stimulated millimeter bubbles (MBs) around silver nanoparticles (AgNPs) and thoroughly examined the mechanism of bubble formation within this nanocomposite system. One crucial aspect we explored was the lifetime and kinetics of these bubbles, given that bubbles generated by plasmonic nanoparticles are known to be transient with short durations. Surprisingly, our findings revealed that the achieved lifetime of these MBs extended beyond seven days. This impressive longevity far surpasses what has been reported in the existing literature. Further analysis of the experimental data uncovered a significant correlation between bubble volume and its lifetime. Smaller bubbles demonstrated longer lifetimes compared to larger ones, which provided valuable insights for future applications. The experimental results not only confirmed the validity of our model and simulations but also highlighted essential characteristics, including extended lifetime, matching absorption coefficients, adherence to physical boundary conditions, and agreement with simulated system parameters. Notably, we generated these MBs around functionalized AgNPs in a biocompatible nanocomposite medium by utilizing low-power light excitation. By readily binding potent cancer drugs to AgNPs through simple physical mixing, these medications can be securely encapsulated within bubbles and precisely guided to targeted locations within the human body. This capability to deliver drugs directly to the tumor site, while minimizing contact with healthy tissues, can lead to improved treatment outcomes and reduced side effects, significantly enhancing the quality of life for cancer patients.
Collapse
Affiliation(s)
- Ulviye Bunyatova
- Biomedical Engineering Department, Engineering Facility, Baskent University, Ankara, Turkey.
| | - Mustafa Dogan
- Department of Control and Automation Engineering, Faculty of Electrical-Electronics Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Engincan Tekin
- Department of Electronics and Communications Engineering, Faculty of Electrical-Electronics Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Onur Ferhanoğlu
- Department of Electronics and Communications Engineering, Faculty of Electrical-Electronics Engineering, Istanbul Technical University, Istanbul, Turkey
| |
Collapse
|
13
|
Cybulski M, Sidoryk K, Zaremba-Czogalla M, Trzaskowski B, Kubiszewski M, Tobiasz J, Jaromin A, Michalak O. The Conjugates of Indolo[2,3- b]quinoline as Anti-Pancreatic Cancer Agents: Design, Synthesis, Molecular Docking and Biological Evaluations. Int J Mol Sci 2024; 25:2573. [PMID: 38473820 DOI: 10.3390/ijms25052573] [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: 01/29/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
New amide conjugates of hydroxycinnamic acids (HCAs) and the known antineoplastic 5,11-dimethyl-5H-indolo[2,3-b]quinoline (DiMIQ), an analog of the natural alkaloid neocryptolepine, were synthesized and tested in vitro for anticancer activity. The compound 9-[((2-hydroxy)cinnamoyl)amino]-5,11-dimethyl-5H-indolo[2,3-b]quinoline (2), which contains the ortho-coumaric acid fragment, demonstrated dose-dependent effectiveness against both normal BxPC-3 and metastatic AsPC-1 pancreatic cancer cells. The IC50 values for AsPC-1 and BxPC-3 were 336.5 nM and 347.5 nM, respectively, with a selectivity index of approximately 5 for both pancreatic cancer cells compared to normal dermal fibroblasts. Conjugate 2 did not exhibit any hemolytic activity against human erythrocytes at the tested concentration. Computational studies were performed to predict the pharmacokinetic profile and potential mechanism of action of the synthesized conjugates. These studies focused on the ADME properties of the conjugates and their interactions with DNA, as well as DNA-topoisomerase alpha and beta complexes. All of the conjugates studied showed approximately one order of magnitude stronger binding to DNA compared to the reference DiMIQ, and approximately two orders of magnitude stronger binding to the topoisomerase II-DNA complex compared to DiMIQ. Conjugate 2 was predicted to have the strongest binding to the enzyme-DNA complex, with a Ki value of 2.8 nM.
Collapse
Affiliation(s)
- Marcin Cybulski
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute, 01-793 Warsaw, Poland
| | - Katarzyna Sidoryk
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute, 01-793 Warsaw, Poland
| | - Magdalena Zaremba-Czogalla
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland
| | - Bartosz Trzaskowski
- Chemical and Biological Systems Simulation Lab, Center of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Marek Kubiszewski
- Pharmaceutical Analysis Laboratory, Łukasiewicz Research Network-Industrial Chemistry Institute, 01-793 Warsaw, Poland
| | - Joanna Tobiasz
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute, 01-793 Warsaw, Poland
| | - Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland
| | - Olga Michalak
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute, 01-793 Warsaw, Poland
| |
Collapse
|
14
|
Prange CJ, Hu X, Tang L. Smart chemistry for traceless release of anticancer therapeutics. Biomaterials 2023; 303:122353. [PMID: 37925794 DOI: 10.1016/j.biomaterials.2023.122353] [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: 07/11/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023]
Abstract
In the design of delivery strategies for anticancer therapeutics, the controlled release of intact cargo at the destined tumor and metastasis locations is of particular importance. To this end, stimuli-responsive chemical linkers have been extensively investigated owing to their ability to respond to tumor-specific physiological stimuli, such as lowered pH, altered redox conditions, increased radical oxygen species and pathological enzymatic activities. To prevent premature action and off-target effects, anticancer therapeutics are chemically modified to be transiently inactivated, a strategy known as prodrug development. Prodrugs are reactivated upon stimuli-dependent release at the sites of interest. As most drugs and therapeutic proteins have the optimal activity when released from carriers in their native and original forms, traceless release mechanisms are increasingly investigated. In this review, we summarize the chemical toolkit for developing innovative traceless prodrug strategies for stimuli-responsive drug delivery and discuss the applications of these chemical modifications in anticancer treatment including cancer immunotherapy.
Collapse
Affiliation(s)
- Céline Jasmin Prange
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland; Institute of Chemical Sciences and Engineering, EPFL, Lausanne, CH-1015, Switzerland
| | - Xile Hu
- Institute of Chemical Sciences and Engineering, EPFL, Lausanne, CH-1015, Switzerland.
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland; Institute of Materials Science & Engineering, EPFL, Lausanne, CH-1015, Switzerland.
| |
Collapse
|
15
|
Kaspute G, Arunagiri BD, Alexander R, Ramanavicius A, Samukaite-Bubniene U. Development of Essential Oil Delivery Systems by 'Click Chemistry' Methods: Possible Ways to Manage Duchenne Muscular Dystrophy. MATERIALS (BASEL, SWITZERLAND) 2023; 16:6537. [PMID: 37834674 PMCID: PMC10573547 DOI: 10.3390/ma16196537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023]
Abstract
Recently, rare diseases have received attention due to the need for improvement in diagnosed patients' and their families' lives. Duchenne muscular dystrophy (DMD) is a rare, severe, progressive, muscle-wasting disease. Today, the therapeutic standard for treating DMD is corticosteroids, which cause serious adverse side effects. Nutraceuticals, e.g., herbal extracts or essential oils (EOs), are possible active substances to develop new drug delivery systems to improve DMD patients' lives. New drug delivery systems lead to new drug effects, improved safety and accuracy, and new therapies for rare diseases. Herbal extracts and EOs combined with click chemistry can lead to the development of safer treatments for DMD. In this review, we focus on the need for novel drug delivery systems using EOs as the therapy for DMD and the potential use of click chemistry for drug delivery systems. New EO complex drug delivery systems may offer a new approach for improving muscle conditions and mental health issues associated with DMD. However, further research should identify the potential of these systems in the context of DMD. In this review, we discuss possibilities for applying EOs to DMD before implementing expensive research in a theoretical way.
Collapse
Affiliation(s)
- Greta Kaspute
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekis av. 3, LT-10257 Vilnius, Lithuania;
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania; (B.D.A.); (R.A.)
| | - Bharani Dharan Arunagiri
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania; (B.D.A.); (R.A.)
| | - Rakshana Alexander
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania; (B.D.A.); (R.A.)
| | - Arunas Ramanavicius
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekis av. 3, LT-10257 Vilnius, Lithuania;
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania; (B.D.A.); (R.A.)
| | - Urte Samukaite-Bubniene
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekis av. 3, LT-10257 Vilnius, Lithuania;
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania; (B.D.A.); (R.A.)
| |
Collapse
|
16
|
Husni P, Lim C, Taek Oh K. Tumor microenvironment stimuli-responsive lipid-drug conjugates for cancer treatment. Int J Pharm 2023; 639:122942. [PMID: 37037397 DOI: 10.1016/j.ijpharm.2023.122942] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/12/2023]
Abstract
Lipid drug conjugates (LDCs) have attracted considerable attention in the fields of drug delivery and pharmacology due to their ability to target specific cells, increase drug solubility, reduce toxicity, and improve therapeutic efficacy. These unique features make LDCs promising candidates for the treatment cancer, inflammation, and infectious diseases. In fact, by choosing specific linkers between the lipid and drug molecules, stimuli-responsive LDCs can be designed to target cancer cells based on the unique properties of the tumor microenvironment. Despite the fact that many reviews have described LDCs, few articles have focused on tumor microenvironmental stimuli-responsive LDCs for cancer treatment. Therefore, the key elements of these types of LDCs in cancer treatment will be outlined and discussed in this paper. Our paper goes into detail on the concepts and benefits of LDCs, the various types of tumor microenvironment stimuli-responsive LDCs (such as pH, redox, enzyme, or reactive oxygen species-responsive LDCs), and the current status of LDCs in clinical trials.
Collapse
Affiliation(s)
- Patihul Husni
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, 221, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; College of Pharmacy, Chung-Ang University, 221, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Chaemin Lim
- College of Pharmacy, Chung-Ang University, 221, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea.
| | - Kyung Taek Oh
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, 221, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; College of Pharmacy, Chung-Ang University, 221, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea.
| |
Collapse
|
17
|
Yao P, Zhang Y, Zhang S, Wei X, Liu Y, Du C, Hu M, Feng C, Li J, Zhao F, Li C, Li Z, Du L. Knowledge atlas of antibody-drug conjugates on CiteSpace and clinical trial visualization analysis. Front Oncol 2023; 12:1039882. [PMID: 36686767 PMCID: PMC9850101 DOI: 10.3389/fonc.2022.1039882] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
Objective Antibody-drugs conjugates (ADCs) are novel drugs with highly targeted and tumor-killing abilities and developing rapidly. This study aimed to evaluate drug discovery and clinical trials of and explore the hotspots and frontiers from 2012 to 2022 using bibliometric methods. Methods Publications on ADCs were retrieved between 2012 and 2022 from Web of Science (WoS) and analyzed with CiteSpace 6.1.R2 software for the time, region, journals, institutions, etc. Clinical trials were downloaded from clinical trial.org and visualized with Excel software. Results A total of 696 publications were obtained and 187 drug trials were retrieved. Since 2012, research on ADCs has increased year by year. Since 2020, ADC-related research has increased dramatically, with the number of relevant annual publications exceeding 100 for the first time. The United States is the most authoritative and superior country and region in the field of ADCs. The University of Texas MD Anderson Cancer Center is the most authoritative institution in this field. Research on ADCs includes two clinical trials and one review, which are the most influential references. Clinical trials of ADCs are currently focused on phase I and phase II. Comprehensive statistics and analysis of the published literature and clinical trials in the field of ADCs, have shown that the most studied drug is brentuximab vedotin (BV), the most popular target is human epidermal growth factor receptor 2 (HER2), and breast cancer may become the main trend and hotspot for ADCs indications in recent years. Conclusion Antibody-drug conjugates have become the focus of targeted therapies in the field of oncology. The innovation of technology and combination application strategy will become the main trend and hotspots in the future.
Collapse
Affiliation(s)
- Peizhuo Yao
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yinbin Zhang
- School of Chemistry, Xi’an Jiaotong University, Xi’an, China,*Correspondence: Yinbin Zhang, ; Shuqun Zhang,
| | - Shuqun Zhang
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China,*Correspondence: Yinbin Zhang, ; Shuqun Zhang,
| | - Xinyu Wei
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yanbin Liu
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chong Du
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Mingyou Hu
- School of Chemistry, Xi’an Jiaotong University, Xi’an, China
| | - Cong Feng
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jia Li
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Fang Zhao
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chaofan Li
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zhen Li
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lisha Du
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
18
|
Yamazaki S, Matsuda Y. Tag‐Free Enzymatic Modification for Antibody−Drug Conjugate Production. ChemistrySelect 2022. [DOI: 10.1002/slct.202203753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
| | - Yutaka Matsuda
- Ajinomoto Bio-Pharma Services 11040 Roselle Street San Diego CA 92121 United States
| |
Collapse
|