1
|
Yang X, Li L, Li R, Li X, Li S, Su C, Liao H. Arginine N-glycosylation of melittin enhances its bacteriostatic activity and antiproliferative therapeutic index. Org Biomol Chem 2025; 23:4471-4479. [PMID: 40223544 DOI: 10.1039/d5ob00398a] [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: 04/15/2025]
Abstract
Melittin is a natural antimicrobial peptide isolated from bee venom, and the non-specific cytotoxicity and hemolytic activity severely limit its clinical application. Glycosylation of proteins is very common in physiological and biochemical processes and can modulate the interaction of proteins with their target. In this study, eight glycosyl groups were used to modify the arginine of melittin at sites 22 and/or 24, and single and double arginine N-glycosylated peptides were designed and synthesized. Among the acquired 24 glycopeptides, MLT-1c, MLT-3c, MLT-1f, MLT-3f, MLT-1g, and MLT-3h were found to possess higher helicity, while MLT-3c, MLT-3f and MLT-3h showed dramatically reduced hemolytic activity, especially MLT-3c, whose HC50 value is 199.3 μM. MLT-1a, MLT-3a and MLT-2c exhibited improved inhibitory activity against Puzza streptococcus, and the MIC was 4 μg mL-1. MLT-1e and MLT-2g have the strongest tolerance to trypsase, and MLT-3c has the highest therapeutic index. In general, rhamnosyl-modified melittin MLT-3c could be a potent agent for antibacterial and antitumor therapy with high stability and low hemolytic side effects.
Collapse
Affiliation(s)
- Xiantao Yang
- School of Pharmacy, Chengdu Medical College, 783 Xindu Avenue, Xindu District, Chengdu 610500, China.
| | - Linji Li
- School of Pharmacy, Chengdu Medical College, 783 Xindu Avenue, Xindu District, Chengdu 610500, China.
| | - Rong Li
- Pidu area center, Chengdu Institute of Food Inspection, 456 Yong'an West Rd., Ande Street, Pidu District, Chengdu 611730, China
| | - Xiang Li
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Shuna Li
- Department of Otorhinolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Rd., Shanghai, 200092, China.
| | - Chunli Su
- School of Public Health, Chengdu Medical College, 783 Xindu Avenue, Xindu District, Chengdu 610500, China.
| | - Hongli Liao
- School of Pharmacy, Chengdu Medical College, 783 Xindu Avenue, Xindu District, Chengdu 610500, China.
| |
Collapse
|
2
|
Levkovsky IO, Trachsel L, Murata H, Matyjaszewski K. Versatile and Controlled Synthesis of Degradable, Water-Soluble Bottlebrush Polymers with Poly(disulfide) Backbones Derived from α-Lipoic Acid. ACS Macro Lett 2025; 14:207-213. [PMID: 39899736 PMCID: PMC11841036 DOI: 10.1021/acsmacrolett.4c00839] [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: 12/21/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
Bottlebrush (BB) polymers, with their densely grafted side chains and unique architecture, are highly advantageous for drug delivery due to their high functional group density for drug conjugation, unimolecular nature, and enhanced biodistribution properties. These attributes enable extended blood circulation half-life, improved tumor tissue penetration, and high tumoral drug accumulation. However, the typically nondegradable, all-carbon backbones of most BB polymers limit their suitability for applications requiring controlled clearance and biodegradability. To address this, we developed degradable BB polymers with poly(disulfide) backbones synthesized via reversible addition-fragmentation chain transfer (RAFT) copolymerization of α-lipoic acid (LA), a renewable and readily available compound, with acrylate-based inimers. These copolymers feature degradable backbones and initiating sites for subsequent BB synthesis. Using an atom transfer radical polymerization (ATRP) grafting-from methodology, we synthesized BB polymers with relatively low dispersities (Đ = 1.30-1.53), high backbone degrees of polymerization (DPbb), and high molar masses (Mn,MALS = 650-2700 kg/mol). The easily cleavable disulfide bonds enabled backbone degradation under mild reducing conditions. Beyond hydrophilic BB with tri(ethylene glycol) methyl ether acrylate (TEGA) side chains, we synthesized BB with cationic, anionic, and zwitterionic side chains, demonstrating broad monomer compatibility. This scalable approach produces water-soluble, degradable BB polymers with tunable architectures and predictable molecular weights. By addressing the need for degradability in BB polymers, this work advances their potential for drug delivery, offering enhanced functionality, biocompatibility, and sustainability.
Collapse
Affiliation(s)
- Ivan O. Levkovsky
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Lucca Trachsel
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Hironobu Murata
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Krzysztof Matyjaszewski
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
3
|
Dave R, Mofford J, Hicks EA, Singh A, Sheardown H, Hoare T. Tuning mucoadhesion and mucopenetration in self-assembled poly(lactic acid)-block-poly(oligoethylene glycol methacrylate) block copolymer nanoparticles by controlling side-chain lengths. NANOSCALE 2025; 17:1417-1432. [PMID: 39620283 DOI: 10.1039/d4nr03805c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
The capacity to tune the degree of mucoadhesion and mucopenetration of nanoparticles is essential to improving drug bioavailability, transport, and efficacy at mucosal interfaces. Herein, self-assembled nanoparticles (NPs) fabricated from amphiphilic block copolymers of poly(lactic acid) (PLA) and poly(oligo(ethylene glycol) methacrylate) (POEGMA) with various side chain lengths (PLA-POEGMAn) are reported to facilitate tunable mucosal interactions. PLA-POEGMAn nanoparticles with long PEG side chain lengths (n = 20, or 40) demonstrated mucoadhesive properties based on rheological synergism, calorimetric tracking of mucin-nanoparticle interactions, and the formation of larger NP-mucin hybrid structures; in contrast, NPs fabricated from block copolymers with shorter PEG side chains (n = 2/8-9 or n = 8,9) showed poor mucoadhesion but penetrated through the mucin layer with significantly higher permeation rates (>80%). All NP formulations showed good cytocompatibility (viability > 70%) with human corneal epithelial cells in vitro and no detectable acute in vivo ocular irritation in Sprague-Dawley rats. Coupled with the capacity of the synthetic route to easily incorporate different brush lengths and/or different functional groups into the hydrophilic block, we anticipate this approach may offer a solution in applications in which balancing mucoadhesion and mucopenetration is critical for enabling effective drug delivery.
Collapse
Affiliation(s)
- Ridhdhi Dave
- Department of Chemical Engineering, McMaster University, 1280 Main St. W, Hamilton, Ontario, L8S 4L7, Canada.
| | - Jon Mofford
- Department of Chemical Engineering, McMaster University, 1280 Main St. W, Hamilton, Ontario, L8S 4L7, Canada.
| | - Emily Anne Hicks
- Department of Chemical Engineering, McMaster University, 1280 Main St. W, Hamilton, Ontario, L8S 4L7, Canada.
| | - Andrew Singh
- Department of Chemical Engineering, McMaster University, 1280 Main St. W, Hamilton, Ontario, L8S 4L7, Canada.
| | - Heather Sheardown
- Department of Chemical Engineering, McMaster University, 1280 Main St. W, Hamilton, Ontario, L8S 4L7, Canada.
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main St. W, Hamilton, Ontario, L8S 4L7, Canada.
| |
Collapse
|
4
|
Fang Y, Cai J, Fei F, Zhong T, Ren M, Wang D, Li Y, Zhang K. Targeting the Skin: The Study of a Bottlebrush Polymer-Antisense Oligonucleotide Conjugate in a Psoriasis Mouse Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403949. [PMID: 39140277 PMCID: PMC11581913 DOI: 10.1002/smll.202403949] [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: 05/17/2024] [Revised: 07/12/2024] [Indexed: 08/15/2024]
Abstract
The investigation of gene regulation therapeutics for the treatment of skin-related diseases is rarely explored in part due to inefficient systemic delivery. In this study, a bottlebrush polymer-antisense oligonucleotide (ASO) conjugate, termed pacDNA, designed to target IL-17 receptor A (IL-17RA), which is involved in psoriasis pathogenesis is presented. Systemic administration of pacDNA led to its accumulation in epidermis, dermis, and hypodermis of mouse skin, reduced IL-17RA gene expression in skin, and significantly reversed the development of imiquimod (IMQ)-induced psoriasis in a mouse model. These findings highlight the potential of the pacDNA as a promising nanoconstruct for systemic oligonucleotide delivery to the skin and for treating psoriasis and other skin-related disorders through systemic administration.
Collapse
Affiliation(s)
- Yang Fang
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | - Jiansong Cai
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | - Feng Fei
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | - Tongtong Zhong
- Bouvé College of Health Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Mengqi Ren
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | - Dali Wang
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | - Yao Li
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Ke Zhang
- Departments of Chemistry and Chemical Biology, Chemical Engineering, and Bioengineering, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
5
|
Laurindo LF, de Lima EP, Laurindo LF, Rodrigues VD, Chagas EFB, de Alvares Goulart R, Araújo AC, Guiguer EL, Pomini KT, Rici REG, Maria DA, Direito R, Barbalho SM. The therapeutic potential of bee venom-derived Apamin and Melittin conjugates in cancer treatment: A systematic review. Pharmacol Res 2024; 209:107430. [PMID: 39332751 DOI: 10.1016/j.phrs.2024.107430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024]
Abstract
The therapeutic potential of bee venom-derived peptides, particularly apamin and melittin, in cancer treatment has garnered significant attention as a promising avenue for advancing oncology. This systematic review examines preclinical studies highlighting the emerging role of these peptides in enhancing cancer therapies. Melittin and apamin, when conjugated with other therapeutic agents or formulated into novel delivery systems, have demonstrated improved efficacy in targeting tumor cells. Key findings indicate that melittin-based conjugates, such as polyethylene glycol (PEG)ylated versions, show potential in enhancing therapeutic outcomes and minimizing toxicity across various cancer models. Similarly, apamin-conjugated formulations have improved the efficacy of established anti-cancer drugs, contributing to enhanced targeting and reduced systemic toxicity. These developments underscore a growing interest in leveraging bee venom-derived peptides as adjuncts in cancer therapy. The integration of these peptides into treatment regimens offers a promising strategy to address current limitations in cancer treatment, such as drug resistance and off-target effects. However, comprehensive validation through clinical trials is essential to confirm their safety and effectiveness in human patients. This review highlights the global emergence of bee venom-derived peptides in cancer treatment, advocating for continued research and development to fully realize their therapeutic potential.
Collapse
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília, São Paulo 17519-030, Brazil; Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil; Department of Administration, Associate Degree in Hospital Management, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil
| | - Enzo Pereira de Lima
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil
| | - Lívia Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, São Paulo 15090-000, Brazil
| | - Victória Dogani Rodrigues
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília, São Paulo 17519-030, Brazil
| | - Eduardo Federighi Baisi Chagas
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil
| | - Ricardo de Alvares Goulart
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil; Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília, São Paulo 17500-000, Brazil
| | - Karina Torres Pomini
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil
| | - Rose Eli Grassi Rici
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil; Graduate Program in Anatomy of Domestic and Wild Animals, College of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-220, Brazil
| | - Durvanei Augusto Maria
- Development and innovation Laboratory, Butantan Institute, São Paulo, São Paulo 05585-000, Brazil
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines, Universidade de Lisboa (iMed.ULisboa), Av. Prof. Gama Pinto, Lisbon 1649-003, Portugal.
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil; Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília, São Paulo 17500-000, Brazil; UNIMAR Charity Hospital, Universidade de Marília (UNIMAR), Marília, São Paulo 17525-902, Brazil
| |
Collapse
|
6
|
Cui Z, Zhou Z, Sun Z, Duan J, Liu R, Qi C, Yan C. Melittin and phospholipase A2: Promising anti-cancer candidates from bee venom. Biomed Pharmacother 2024; 179:117385. [PMID: 39241571 DOI: 10.1016/j.biopha.2024.117385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024] Open
Abstract
As the research on cancer-related treatment deepens, integrating traditional therapies with emerging interventions reveals new therapeutic possibilities. Melittin and phospholipase A2, the primary anti-cancer components of bee venom, are currently gaining increasing attention. This article reviews the various formulations of melittin in cancer therapy and its potential applications in clinical treatments. The reviewed formulations include melittin analogs, hydrogels, adenoviruses, fusion toxins, fusion peptides/proteins, conjugates, liposomes, and nanoparticles. The article also explored the collaborative therapeutic effects of melittin with natural products, synthetic drugs, radiotherapy, and gene expression regulatory strategies. Phospholipase A2 plays a key role in bee venom anti-cancer strategy due to its unique biological activity. Using an extensive literature review and the latest scientific results, this paper explores the current state and challenges of this field, with the aim to provide new perspectives that guide future research and potential clinical applications. This will further promote the application of bee venom in cancer therapy.
Collapse
Affiliation(s)
- Ziyan Cui
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Medical University, Shijiazhuang, China
| | - Zegao Zhou
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Medical University, Shijiazhuang, China
| | - Ziyan Sun
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Medical University, Shijiazhuang, China
| | - Jiayue Duan
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Medical University, Shijiazhuang, China
| | - Runtian Liu
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Medical University, Shijiazhuang, China
| | - Cheng Qi
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Medical University, Shijiazhuang, China
| | - Changqing Yan
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
7
|
Zhang T, Bai L, You R, Yang M, Chen Q, Cheng Y, Qian Z, Wang Y, Liu Y. Homologous-targeting biomimetic nanoparticles co-loaded with melittin and a photosensitizer for the combination therapy of triple negative breast cancer. J Mater Chem B 2024; 12:5465-5478. [PMID: 38742364 DOI: 10.1039/d3tb02919k] [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: 05/16/2024]
Abstract
Melittin (Mel) is considered a promising candidate drug for the treatment of triple negative breast cancer (TNBC) due to its various antitumor effects. However, its clinical application is hampered by notable limitations, including hemolytic activity, rapid clearance, and a lack of tumor selectivity. Here, we designed novel biomimetic nanoparticles based on homologous tumor cell membranes and poly(lactic-co-glycolic acid) (PLGA)/poly(beta-aminoester) (PBAE), denoted MDM@TPP, which efficiently coloaded the cytolytic peptide Mel and the photosensitizer mTHPC. Both in vitro and in vivo, the MDM@TPP nanoparticles effectively mitigated the acute toxicity of melittin and exhibited strong TNBC targeting ability due to the homologous targeting effect of the tumor cell membrane. Under laser irradiation, the MDM@TPP nanoparticles showed excellent photodynamic performance and thus accelerated the release of Mel by disrupting cell membrane integrity. Moreover, Mel combined with photodynamic therapy (PDT) can synergistically kill tumor cells and induce significant immunogenic cell death, thereby stimulating the maturation of dendritic cells (DCs). In 4T1 tumor-bearing mice, MDM@TPP nanoparticles effectively inhibited the growth and metastasis of primary tumors and finally prevented tumor recurrence by improving the immune response.
Collapse
Affiliation(s)
- Tao Zhang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin 300192, China
| | - Liya Bai
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Ran You
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Meng Yang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Qian Chen
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Yuanyuan Cheng
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Zhanyin Qian
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Yinsong Wang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Yuanyuan Liu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
8
|
Saha B, Lee JH, Kwon I, Chung H. Site-Specific Conjugation of Bottlebrush Polymers to Therapeutic Protein via Bioorthogonal Chemistry. Biomacromolecules 2024; 25:3200-3211. [PMID: 38591457 DOI: 10.1021/acs.biomac.4c00359] [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: 04/10/2024]
Abstract
Achieving efficient and site-specific conjugation of therapeutic protein to polymer is crucial to augment their applicability in the realms of biomedicine by improving their stability and enzymatic activity. In this study, we exploited tetrazine bioorthogonal chemistry to achieve the site-specific conjugation of bottlebrush polymers to urate oxidase (UOX), a therapeutic protein for gout treatment. An azido-functionalized zwitterionic bottlebrush polymer (N3-ZBP) using a "grafting-from" strategy involving RAFT and ATRP methods was synthesized, and a trans-cyclooctene (TCO) moiety was introduced at the polymer end through the strain-promoted azide-alkyne click (SPAAC) reaction. The subsequent coupling between TCO-incorporated bottlebrush polymer and tetrazine-labeled UOX using a fast and safe bioorthogonal reaction, inverse electron demand Diels-Alder (IEDDA), led to the formation of UOX-ZBP conjugates with a 52% yield. Importantly, the enzymatic activity of UOX remained unaffected following polymer conjugation, suggesting a minimal change in the folded structure of UOX. Moreover, UOX-ZBP conjugates exhibited enhanced proteolytic resistance and reduced antibody binding, compared to UOX-wild type. Overall, the present findings reveal an efficient and straightforward route for synthesizing protein-bottlebrush polymer conjugates without compromising the enzymatic activity while substantially reducing proteolytic degradation and antibody binding.
Collapse
Affiliation(s)
- Biswajit Saha
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
| | - Jae Hun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Inchan Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Hoyong Chung
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
| |
Collapse
|
9
|
Chen P, Wei Y, Sun T, Lin J, Zhang K. Enabling safer, more potent oligonucleotide therapeutics with bottlebrush polymer conjugates. J Control Release 2024; 366:44-51. [PMID: 38145661 PMCID: PMC10922259 DOI: 10.1016/j.jconrel.2023.12.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
Oligonucleotide therapeutics have the unique ability to address traditionally undruggable targets through various target engagement pathways. However, despite advances in chemically modified oligonucleotides and carrier-assisted delivery systems such as lipid nanoparticles and protein/peptide conjugates, the development of oligonucleotide drugs is still plagued with lackluster potency, narrow therapeutic window, poor delivery to non-liver target sites, and/or high potential for toxicity and unwanted immune system activation. In this perspective, we discuss an unconventional delivery solution based upon bottlebrush polymers, which overcomes many key challenges in oligonucleotide drug development. We address the molecular basis of the polymer's ability to enhance tissue bioavailability and drug potency, reduce side effects, and suppress anti-carrier immunity. Furthermore, we discuss the potential of the technology in advancing oligonucleotide-based therapies for non-liver targets.
Collapse
Affiliation(s)
- Peiru Chen
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Yun Wei
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Tingyu Sun
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Jiachen Lin
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Ke Zhang
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA; Department of Chemical Engineering and Bioengineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Fang Y, Cai J, Ren M, Zhong T, Wang D, Zhang K. Inhalable Bottlebrush Polymer Bioconjugates as Vectors for Efficient Pulmonary Delivery of Oligonucleotides. ACS NANO 2024; 18:592-599. [PMID: 38147573 PMCID: PMC10786149 DOI: 10.1021/acsnano.3c08660] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
Antisense oligonucleotides hold therapeutic promise for various lung disorders, but their efficacy is limited by suboptimal delivery. To address this challenge, we explored the use of inhaled bottlebrush polymer-DNA conjugates, named pacDNA, as a delivery strategy. Inhaled pacDNA exhibits superior mucus penetration, achieving a uniform and sustained lung distribution in mice. Targeting the 5' splice site of an aberrant enhanced green fluorescence protein (EGFP) pre-mRNA in EGFP-654 mice, inhaled pacDNA more efficiently corrects splicing than a B-peptide conjugate and restores EGFP expression in the lung. Additionally, in an orthotopic NCI-H358 non-small-cell lung tumor mouse model, inhaled pacDNA targeting wild-type KRAS mRNA effectively suppresses KRAS expression and inhibits lung tumor growth, requiring a substantially lower dosage compared to intravenously injected pacDNA. These findings demonstrate the potential of bottlebrush polymer-DNA conjugates as a promising agent for enhanced oligonucleotide therapy in the lung and advancing the treatment landscape for lung disorders.
Collapse
Affiliation(s)
- Yang Fang
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Jiansong Cai
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Mengqi Ren
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Tongtong Zhong
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Dali Wang
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Ke Zhang
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
11
|
Yu X, Jia S, Yu S, Chen Y, Zhang C, Chen H, Dai Y. Recent advances in melittin-based nanoparticles for antitumor treatment: from mechanisms to targeted delivery strategies. J Nanobiotechnology 2023; 21:454. [PMID: 38017537 PMCID: PMC10685715 DOI: 10.1186/s12951-023-02223-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/19/2023] [Indexed: 11/30/2023] Open
Abstract
As a naturally occurring cytolytic peptide, melittin (MLT) not only exhibits a potent direct tumor cell-killing effect but also possesses various immunomodulatory functions. MLT shows minimal chances for developing resistance and has been recognized as a promising broad-spectrum antitumor drug because of this unique dual mechanism of action. However, MLT still displays obvious toxic side effects during treatment, such as nonspecific cytolytic activity, hemolytic toxicity, coagulation disorders, and allergic reactions, seriously hampering its broad clinical applications. With thorough research on antitumor mechanisms and the rapid development of nanotechnology, significant effort has been devoted to shielding against toxicity and achieving tumor-directed drug delivery to improve the therapeutic efficacy of MLT. Herein, we mainly summarize the potential antitumor mechanisms of MLT and recent progress in the targeted delivery strategies for tumor therapy, such as passive targeting, active targeting and stimulus-responsive targeting. Additionally, we also highlight the prospects and challenges of realizing the full potential of MLT in the field of tumor therapy. By exploring the antitumor molecular mechanisms and delivery strategies of MLT, this comprehensive review may inspire new ideas for tumor multimechanism synergistic therapy.
Collapse
Affiliation(s)
- Xiang Yu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou, China.
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Haikou, China.
| | - Siyu Jia
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Shi Yu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Yaohui Chen
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Chengwei Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Haidan Chen
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China.
| | - Yanfeng Dai
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou, China.
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Haikou, China.
| |
Collapse
|
12
|
Bahreyni A, Liu H, Mohamud Y, Xue YC, Fan YM, Zhang YL, Luo H. A combination of genetically engineered oncolytic virus and melittin-CpG for cancer viro-chemo-immunotherapy. BMC Med 2023; 21:193. [PMID: 37226233 DOI: 10.1186/s12916-023-02901-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Immunotherapy has emerged as an efficient therapeutic approach for cancer management. However, stimulation of host immune system against cancer cells often fails to achieve promising clinical outcomes mainly owing to the immunosuppressive characteristics of the tumor microenvironment (TME). Combination therapeutics that can trigger sustained immunogenic cell death (ICD) have provided new opportunities for cancer treatment. METHODS In this study, we designed and applied an ICD inducer regimen, including a genetically engineered oncolytic virus (miRNA-modified coxsackieviruses B3, miR-CVB3), a pore-forming lytic peptide (melittin, found in bee venom), and a synthetic toll-like receptor 9 ligand (CpG oligodeoxynucleotides), for breast cancer and melanoma treatment. We compared the anti-tumor efficacy of miR-CVB3 and CpG-melittin (CpGMel) alone and in combination (miR-CVB3 + CpGMel) and investigated possible mechanisms involved. RESULTS We demonstrated that miR-CVB3 + CpGMel had no major impact on viral growth, while enhancing the cellular uptake of CpGMel in vitro. We further showed that combination therapy led to significant increases in tumor cell death and release of damage-associated molecular patterns compared with individual treatment. In vivo studies in 4T1 tumor-bearing Balb/c mice revealed that both primary and distant tumors were significantly suppressed, and the survival rate was significantly prolonged after administration of miR-CVB3 + CpGMel compared with single treatment. This anti-tumor effect was accompanied by increased ICD and immune cell infiltration into the TME. Safety analysis showed no significant pathological abnormalities in Balb/c mice. Furthermore, the developed therapeutic regimen also demonstrated a great anti-tumor activity in B16F10 melanoma tumor-bearing C57BL/6 J mice. CONCLUSIONS Overall, our findings indicate that although single treatment using miR-CVB3 or CpGMel can efficiently delay tumor growth, combining oncolytic virus-based therapy can generate even stronger anti-tumor immunity, leading to a greater reduction in tumor size.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC, V6Z 1Y6, Canada
| | - Huitao Liu
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, V6Z 1Y6, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC, V6Z 1Y6, Canada
| | - Yuan Chao Xue
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC, V6Z 1Y6, Canada
| | - Yiyun Michelle Fan
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Endowment Lands, Canada
| | - Yizhuo Lyanne Zhang
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Endowment Lands, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada.
- Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC, V6Z 1Y6, Canada.
| |
Collapse
|
13
|
Su X, Feng Y, Shi H, Wang F, Wang Z, Hou S, Song X, Yang J, Liu L. A hydrogel dressing with tunable critical temperature and photothermal modulating melittin release for multiply antibacterial treatment. Int J Biol Macromol 2023; 239:124272. [PMID: 37001785 DOI: 10.1016/j.ijbiomac.2023.124272] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023]
Abstract
It is imperative to develop an antibiotic-free and long-term effective strategy for treating chronic wound infections due to the long-term utilization of antibiotics easily causing drug resistance. Herein, we fabricated a novel poly-N-isopropylacrylamide (PNIPAM)/polyacrylamide (PAM) coupling thermosensitive hydrogel integrating 1D lysozyme nanofiber doped with CuS nanoparticles (CuS/PP) and loading antibacterial peptide melittin (M) (CuS/PP-M) for combating chronic wound infection via photothermal modulating the release of melittin. For the CuS/PP-M hydrogel, the copolymerization of PNIPAM and PAM allows the lower critical solution temperature (LCST) higher than the body temperature, effectively hindering the spontaneous release of melittin when contacts the infected wound, while the integration of LNF/CuS nanofibers provides a stable photothermal treatment for triggering the release of melittin. As a result, the CuS/PP-M hydrogel exhibits synergistically enhanced effect on killing both Gram-positive and Gram-negative bacteria, which maintains more than 99 % bactericidal efficiency, even displays a long-term and multiply antibacterial performance by photothermal modulating melittin release. Moreover, the CuS/PP-M hydrogel presents both high antibacterial activity and excellent wound healing performance in the mouse wound model, thereby benefiting the chronic wound healing.
Collapse
Affiliation(s)
- Xianhao Su
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang 202013, China
| | - Yonghai Feng
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang 202013, China.
| | - Hui Shi
- School of Medicine, Jiangsu University, Zhenjiang 202013, China
| | - Fenghua Wang
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang 202013, China
| | - Zengkai Wang
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang 202013, China
| | - Shuai Hou
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang 202013, China
| | - Xiaolu Song
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang 202013, China
| | - Juan Yang
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang 202013, China
| | - Lei Liu
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang 202013, China.
| |
Collapse
|
14
|
Pacheco C, Baião A, Ding T, Cui W, Sarmento B. Recent advances in long-acting drug delivery systems for anticancer drug. Adv Drug Deliv Rev 2023; 194:114724. [PMID: 36746307 DOI: 10.1016/j.addr.2023.114724] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/20/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
The use of systemic anticancer chemotherapy is intrinsically limited by its toxicity. Whether dealing with small molecules or biopharmaceuticals, after systemic administration, small doses fail to reach effective intratumoral concentrations, while high doses with significant tumor inhibition effects may also drive the death of healthy cells, endangering the patients. Therefore, strategies based on drug delivery systems (DDSs) for avoiding the systemic toxicity have been designed. Due to their ability to protect drugs from early elimination and control drug release, DDSs can foster tumor exposure to anticancer therapeutics by extending their circulation time or steadily releasing drugs into the tumor sites. However, approval of tailored DDSs systems for clinical use is minimal as the safety and the in vivo activity still need to be ameliorated by manipulating their physicochemical characteristics. During the last few years, several strategies have been described to improve their safety, stability, and fine-tune pharmaceuticals release kinetics. Herein, we reviewed the main DDSs, namely polymeric conjugates, nano or microparticles, hydrogels, and microneedles, explored for long-acting anticancer treatments, highlighting recently proposed modifications and their potential advantages for different anticancer therapies. Additionally, important limitations of long-acting anticancer therapies and future technology directions were also covered.
Collapse
Affiliation(s)
- Catarina Pacheco
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Ana Baião
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Tao Ding
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal; Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China.
| |
Collapse
|
15
|
Wang R, Xiao P, Yu B, Sun Y, Li J, Zhang L, Jiang X, Wu W. Fluorination Effects on the Drug Delivery Property of Cylindrical Polymer Brushes. ACS APPLIED BIO MATERIALS 2022; 5:5924-5932. [PMID: 36417709 DOI: 10.1021/acsabm.2c00870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Fluorination has been widely applied to improving the properties of small-molecule drugs. However, relatively little is known about the effects of fluorination on the drug delivery property of nanomaterials. In this paper, we synthesized a fluoroalkane-modified cylindrical polymer brush (CPB) BCPB-F and an alkane-modified analogue BCPB-H. Doxorubicin (DOX) was used as a model drug and was loaded onto the CPBs through a pH-responsive acylhydrazone linkage. High drug loading and good water solubility were achieved. The in vitro and in vivo experiments suggested that fluorination played an important role in improving the cellular uptake, blood circulation, tissue permeability, and tumor targeting ability of CPBs. Due to these superiorities, the DOX-loaded BCPB-F exhibited excellent antitumor efficacy and eradicated the tumors of mice after five-dose treatments. The well-defined structures of the drug-free and drug-loaded CPBs guaranteed the accuracy of the results. This work demonstrates that fluorination is a promising strategy to improve the overall properties of nanomedicines.
Collapse
Affiliation(s)
- Ruonan Wang
- Department of Polymer Science & Engineering, State Key Laboratory of Analytical Chemistry for Life Science, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Panpan Xiao
- Department of Polymer Science & Engineering, State Key Laboratory of Analytical Chemistry for Life Science, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Bo Yu
- Department of Polymer Science & Engineering, State Key Laboratory of Analytical Chemistry for Life Science, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Ying Sun
- Department of Polymer Science & Engineering, State Key Laboratory of Analytical Chemistry for Life Science, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Jia Li
- Department of Polymer Science & Engineering, State Key Laboratory of Analytical Chemistry for Life Science, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Ling'e Zhang
- Department of Polymer Science & Engineering, State Key Laboratory of Analytical Chemistry for Life Science, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.,Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, P. R. China
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, State Key Laboratory of Analytical Chemistry for Life Science, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Wei Wu
- Department of Polymer Science & Engineering, State Key Laboratory of Analytical Chemistry for Life Science, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
16
|
Müllner M. Molecular polymer bottlebrushes in nanomedicine: therapeutic and diagnostic applications. Chem Commun (Camb) 2022; 58:5683-5716. [PMID: 35445672 DOI: 10.1039/d2cc01601j] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Molecular polymer bottlebrushes are densely grafted, individual macromolecules with nanoscale proportions. The last decade has seen an increased focus on this material class, especially in nanomedicine and for biomedical applications. This Feature Article provides an overview of major developments in this area to highlight the many opportunities that these polymer architectures bring to nano-bio research. The article covers aspects of bottlebrush synthesis and summarises their use in drug and gene delivery, imaging, as theranostics and as prototype materials to correlate nanoparticle structure and composition to biological function and behaviour. Areas for future research in this area are discussed.
Collapse
Affiliation(s)
- Markus Müllner
- Key Centre for Polymers and Colloids, School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia. .,The University of Sydney Nano Institute (Sydney Nano), Sydney, NSW 2006, Australia
| |
Collapse
|