1
|
Li Y, Wu Y, Gao S, Sun T, Jiang C. PROTAC delivery in tumor immunotherapy: Where are we and where are we going? J Control Release 2024; 378:116-144. [PMID: 39637991 DOI: 10.1016/j.jconrel.2024.11.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Immunotherapy has emerged as a pioneering therapeutic modality, particularly within the realm of oncology, where Chimeric Antigen Receptor T-cell (CAR-T) therapy has manifested significant efficacy in the treatment of hematological malignancies. Nonetheless, the extension of immunotherapy to solid tumors poses a considerable challenge. This challenge is largely attributed to the intrinsic "cold" characteristics of certain tumors, which are defined by scant T-cell infiltration and a diminished immune response. Additionally, the impediment is exacerbated by the elusive nature of numerous targets within the tumor microenvironment, notably those deemed "undruggable" by small molecule inhibitors. This scenario underscores an acute necessity for the inception of innovative therapeutic strategies aimed at countering the resistance mechanisms underlying immune evasion in cold tumors, thereby amplifying the efficacy of cancer immunotherapy. Among the promising strategies is the deployment of Proteolysis Targeting Chimeras (PROTACs), which facilitate the targeted degradation of proteins. PROTACs present unique advantages and have become indispensable in oncology. However, they concurrently grapple with challenges such as solubility issues, permeability barriers, and the classical Hook effect. Notably, advanced delivery systems have been instrumental in surmounting these obstacles. This review commences with an analysis of the factors contributing to the suboptimal responses to immunotherapy in cold tumors. Subsequently, it delivers a thorough synthesis of immunotherapeutic concepts tailored for these tumors, clarifying the integral role of PROTACs in their management and delineating the trajectory of PROTAC technology from bench-side investigation to clinical utilization, facilitated by drug delivery systems. Ultimately, the review extrapolates the prospective future of this approach, aspiring to present novel insights that could catalyze progress in immunotherapy for the treatment of cold tumors.
Collapse
Affiliation(s)
- Yiyang Li
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yike Wu
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Sihan Gao
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tao Sun
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; Quzhou Fudan Institute, Quzhou 324003, China.
| | - Chen Jiang
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; Department of Digestive Diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
2
|
Hu Y, Yu Q, Li X, Wang J, Guo L, Huang L, Gao W. Nanoformula Design for Inducing Non-Apoptotic Cell Death Regulation: A Powerful Booster for Cancer Immunotherapy. Adv Healthc Mater 2024:e2403493. [PMID: 39632361 DOI: 10.1002/adhm.202403493] [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: 11/03/2024] [Indexed: 12/07/2024]
Abstract
Cancer treatment has witnessed revolutionary advancements marked by the emergence of immunotherapy, specifically immune checkpoint blockade (ICB). However, the inherent low immunogenicity of tumor cells and the intricate immunosuppressive network within the tumor microenvironment (TME) pose significant challenges to the further development of immunotherapy. Nanotechnology has ushered in unprecedented opportunities and vast prospects for tumor immunotherapy. Nevertheless, traditional nano-formulations often rely on inducing apoptosis to kill cancer cells, which encounters the issue of immune silencing, hindering effective tumor immune activation. The non-apoptotic modes of regulated cell death (RCD), including pyroptosis, ferroptosis, autophagy, necroptosis, and cuproptosis, have gradually garnered attention. These non-apoptotic cell death pathways can induce effective immunogenic cell death (ICD), enhancing cancer immunotherapy. This review comprehensively explores advanced nano-formulation design strategies and their applications in enhancing cancer immunotherapy by promoting non-apoptotic RCD in recent years. It also discusses the potential advantages of these strategies in inducing tumor-specific non-apoptotic RCD. By deeply understanding the significance of non-apoptotic RCD in synergistic cancer immunotherapy, this article provides valuable insights for developing more advanced nano-delivery systems that can robustly induce highly immunogenic non-apoptotic modes, offering novel research and development avenues to address the clinical challenges encountered by immunotherapy represented by ICB.
Collapse
Affiliation(s)
- Yi Hu
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, P.R. China
| | - Qing Yu
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, P.R. China
| | - Xia Li
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, P.R. China
| | - Juan Wang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, P.R. China
| | - Lanping Guo
- National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Wenyuan Gao
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, P.R. China
| |
Collapse
|
3
|
Yuan Y, Nasri M, Manayi A, Zhang J, Wu C, Jeon TJ, Kang L. Sericin coats of silk fibres, a degumming waste or future material? Mater Today Bio 2024; 29:101306. [PMID: 39534681 PMCID: PMC11554926 DOI: 10.1016/j.mtbio.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Silk is a fibrous biopolymer with a recorded history in the textile industries for centuries. This fibre is constituted of two different proteins: fibroin and sericin, of which the latter accounting for approximately 20-30 % of the silk mass. Silk sericin (SSER) was previously considered as a waste by-product in silk fibroin extraction. SSER has recently garnered significant scientific interest due to its extensive biological and pharmacological properties. These include antioxidant effects, biocompatibility, low immunogenicity, controlled biodegradability, and the ability to induce cell proliferation. This review covers studies about various aspects of this emerging material, namely, its general morphology, specific structure, molecular weight, features of different layers, and gene sequences. The impact of different extraction methods and the application of extracted SSER based on molecular weight are discussed. Additionally, the characteristic functional groups in the amino acids of sericin facilitate its applications in regenerative medicine, wound healing, drug delivery, textile, environment, and energy, in various forms like hydrogels, films, scaffolds, and conduits. SSER-based materials offer great potentials for multi-functional applications in the upcoming decades, showcasing adaptability for various functional uses and promising future technological advancements.
Collapse
Affiliation(s)
- Yunong Yuan
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Pharmacy and Bank Building A15, Science Road, New South Wales 2006, Australia
- Sydney Nano Institute, University of Sydney, NSW, 2006, Australia
| | - Mohammad Nasri
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Pharmacy and Bank Building A15, Science Road, New South Wales 2006, Australia
| | - Azadeh Manayi
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Pharmacy and Bank Building A15, Science Road, New South Wales 2006, Australia
- Medicinal Plants Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Junying Zhang
- Department of TCMs Pharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
| | - Chunyong Wu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Tae-Joon Jeon
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Lifeng Kang
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Pharmacy and Bank Building A15, Science Road, New South Wales 2006, Australia
- Sydney Nano Institute, University of Sydney, NSW, 2006, Australia
| |
Collapse
|
4
|
Shin K, Suh HW, Suberi A, Whang CH, Ene M, Grundler J, Grun MK, Saltzman WM. Branching in poly(amine-co-ester) polyplexes impacts mRNA transfection. Biomaterials 2024; 311:122692. [PMID: 38986360 PMCID: PMC11298310 DOI: 10.1016/j.biomaterials.2024.122692] [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: 02/15/2024] [Revised: 05/23/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Branching is a key structural parameter of polymers, which can have profound impacts on physicochemical properties. It has been demonstrated that branching is a modulating factor for mRNA delivery and transfection using delivery vehicles built from cationic polymers, but the influence of polymer branching on mRNA delivery remains relatively underexplored compared to other polymer features such as monomer composition, hydrophobicity, pKa, or the type of terminal group. In this study, we examined the impact of branching on the physicochemical properties of poly(amine-co-esters) (PACE) and their efficiency in mRNA transfection in vivo and in vitro under various conditions. PACE polymers were synthesized with various degrees of branching ranging from 0 to 0.66, and their transfection efficiency was systemically evaluated. We observed that branching improves the stability of polyplexes but reduces the pH buffering capacity. Therefore, the degree of branching (DB) must be optimized in a delivery route specific manner due to differences in challenges faced by polyplexes in different physiological compartments. Through a systematic analysis of physicochemical properties and mRNA transfection in vivo and in vitro, this study highlights the influence of polymer branching on nucleic acid delivery.
Collapse
Affiliation(s)
- Kwangsoo Shin
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA; Department of Polymer Science & Engineering and Program in Environmental and Polymer Engineering, Inha University, Incheon, 22212, Republic of Korea
| | - Hee-Won Suh
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Alexandra Suberi
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Chang-Hee Whang
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Madalina Ene
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Julian Grundler
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA; Department of Chemistry, Yale University, New Haven, CT, 06511, USA
| | - Molly K Grun
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA; Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, 06511, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, 06510, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
5
|
Zuo K, Liu N, Zhou P, Zheng M, Wang L, Tang T, Yang Z, Chen L, Zhu X. Human serum albumin promotes interactions between HSA-IL-2 fusion protein and CD122 for enhancing immunotherapy. Biomed Pharmacother 2024; 181:117664. [PMID: 39522264 DOI: 10.1016/j.biopha.2024.117664] [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: 09/24/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Interleukin 2 (IL-2) is a multifunctional cytokine that is crucial for T-lymphocytes proliferation and differentiation. However, IL-2 binds to IL-2Rα (CD25) subunit preferentially and tends to stimulate regulatory T cells (Tregs), which express high-affinity trimeric receptors (IL-2Rαβγ), resulting in immunosuppressive effects. Therefore, development of methods that enhance IL-2/CD122 interactions and activate immune responses without affecting therapeutic efficacy of IL-2 may be desirable. In this work, we constructed a recombinant IL-2 fusion protein (HSA-IL-2), comprising human serum albumin (HSA) and IL-2, there was a new interaction interface between HSA domain and CD122 in HSA-IL-2 fusion protein predicted by AlphaFold2, and followed by determining binding affinity between HSA-IL-2 and CD122 through ForteBio's Bio-Layer Interferometry technology. Strikingly, HSA did promoted interactions between HSA-IL-2 fusion protein and CD122 compared with wild-type IL-2. In vivo experiments, HSA-IL-2 fusion protein had capacity to promote CD8+ T cells infiltration while reducing Treg cells infiltration for boosting immunotherapeutic efficacy. Furthermore, it facilitated synergistic therapeutic effect with α-PD-L1 to inhibit tumor growth. Overall, our research unveiled an enhanced binding affinity method underlying interactions between IL-2 and CD122 via fusing albumin, and propose a promising therapeutic strategy to facilitate IL-2 administration and broaden its clinical use.
Collapse
Affiliation(s)
- Kaiyue Zuo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; Li Song's Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Hainan University, Sanya 572000, China
| | - Naiyu Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; Li Song's Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Hainan University, Sanya 572000, China
| | - Peng Zhou
- School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Mengzhu Zheng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Lingjuan Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; Li Song's Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Hainan University, Sanya 572000, China
| | - Tingting Tang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; Li Song's Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Hainan University, Sanya 572000, China
| | - Zhanqun Yang
- Department of Pharmacy, Peking University Third Hospital Cancer Center, Peking University Third Hospital, Beijing 100191, China
| | - Long Chen
- Department of Pharmacy, Peking University Third Hospital Cancer Center, Peking University Third Hospital, Beijing 100191, China.
| | - Xinjie Zhu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; Li Song's Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Hainan University, Sanya 572000, China.
| |
Collapse
|
6
|
Firouzbakht A, De A, Gruebele M. Context-dependent effect of polyethylene glycol on the structure and dynamics of hirudin. Biophys J 2024:S0006-3495(24)02239-2. [PMID: 39600093 DOI: 10.1016/j.bpj.2024.11.3311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/30/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Hirudin is a bioactive small protein that binds thrombin to interrupt the blood clotting cascade. It contains an ordered and a disordered (IDR) region. Conjugating with polyethylene glycol (PEGylation) is an important modification of biopharmaceuticals to improve their lifetime and retention. Here, we studied by molecular dynamics (MD) simulation how hirudin P18 and its PEGylated variant differ in their structural flexibility depending on binding to thrombin and charge screening by NaCl. We also compare with glycated hirP18 and the hirV1 variant to assess effects of different polar attachments and sequence variability. First, we synthesized unlabeled and PEG-labeled hirP18 followed by an activity assay to ascertain that the peptide-PEG conjugate retains anticoagulant activity. Next, we carried 16 different microsecond MD simulations of the different proteins, bound and unbound, for 2 sequences and different salt conditions. Simulations were analyzed in terms of scaling exponents to study the effect of ionic strength on hirudin size and solvent-exposed surface area. We conclude that charge patterning of the sequence and the presence of arginine are 2 important features for how PEG interacts with the protein folded and intrinsically disordered regions. Specifically, PEG can screen end-to-end electrostatic interactions by "hiding" a positively charged region of hirudin, whereas hirV1 is less sticky than hirP18 due to different PEG-hirudin hydrophobic interactions and the presence of an arginine in hirP18. Conjugation with either PEG or a glycan significantly reduces solvent-exposed area of hirudin, but PEG interacts more efficiently with surface residues than does glycan due to its narrower chain that can fit in surface grooves, and alternation of polar (oxygen) and nonpolar (CH2-CH2) groups that interact favorably with charged and hydrophobic surface patches.
Collapse
Affiliation(s)
- Arash Firouzbakht
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Anomitra De
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Martin Gruebele
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois; Department of Physics, University of Illinois Urbana-Champaign, Urbana, Illinois; Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois; Carle-Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois.
| |
Collapse
|
7
|
Lin L, Liu H, Zhang D, Du L, Zhang H. Nanolevel Immunomodulators in Sepsis: Novel Roles, Current Perspectives, and Future Directions. Int J Nanomedicine 2024; 19:12529-12556. [PMID: 39606559 PMCID: PMC11600945 DOI: 10.2147/ijn.s496456] [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: 10/01/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Sepsis represents a profound challenge in critical care, characterized by a severe systemic inflammatory response which can lead to multi-organ failure and death. The intricate pathophysiology of sepsis involves an overwhelming immune reaction that disrupts normal host defense mechanisms, necessitating innovative approaches to modulation. Nanoscale immunomodulators, with their precision targeting and controlled release capabilities, have emerged as a potent solution to recalibrate immune responses in sepsis. This review explores the recent advancements in nanotechnology for sepsis management, emphasizing the integration of nanoparticulate systems to modulate immune function and inflammatory pathways. Discussions detail the development of the immune system, the distinct inflammatory responses triggered by sepsis, and the scientific principles underpinning nanoscale immunomodulation, including specific targeting mechanisms and delivery systems. The review highlights nanoformulation designs aimed at enhancing bioavailability, stability, and therapeutic efficacy, which shows promise in clinical settings by modulating key inflammatory pathways. Ultimately, this review synthesizes the current state of knowledge and projects future directions for research, underscoring the transformative potential of nanolevel immunomodulators for sepsis treatment through innovative technologies and therapeutic strategies.
Collapse
Affiliation(s)
- Liangkang Lin
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Hanyou Liu
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Dingshan Zhang
- Department of Intensive Care Unit, Public Health Clinical Center of Chengdu, Chengdu, People’s Republic of China
| | - Lijia Du
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| | - Haiyang Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
8
|
Ghosh D, Khan A, Bag S, Mallick AI, De P. Dual stimuli-responsive biotinylated polymer-drug conjugate for dual drug delivery. J Mater Chem B 2024; 12:11826-11840. [PMID: 39439369 DOI: 10.1039/d4tb01762e] [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: 10/25/2024]
Abstract
Stimuli-responsive nanoscale polymer-drug conjugates are one of the most promising alternatives in the realm of advanced therapeutics, rendering several characteristics such as spatio-temporal control over drug release, reduced off-target toxicity, enhanced bioavailability, and longer blood circulation time of the drug. Fostered by the aforementioned conceptualization, our quest to develop an ideal polymer-drug conjugate has originated the present investigation of developing a reactive oxygen species (ROS) and esterase-responsive self-assembled polymer-drug (chlorambucil, CBL) conjugate with biotin pendants (DP2) for cancer cell targeting, surrogating another antineoplastic drug, doxorubicin (DOX) via physical encapsulation (DP2@DOX). The ROS and esterase trigger not only released the covalently stitched CBL but also resulted in DOX release by dismantling the amphiphilic balance of the nanoaggregates. Biotinylation-mediated enhancement of cellular uptake of DP2@DOX was reflected in the synergistic anticancer activity of both the drugs (CBL and DOX) in HeLa cells (biotin receptor-positive cells) compared to HEK 293T cells (biotin receptor-negative cells). Furthermore, the selective internalization of the fluorophore-tagged DOX-loaded polymer (DP4@DOX) in HeLa cells compared to HEK 293T cells was confirmed by confocal microscopy and flow cytometry. In summary, the present investigation demonstrates a state-of-the-art self-assembled polymer-drug conjugate as a next-generation dual stimuli-responsive drug delivery vehicle.
Collapse
Affiliation(s)
- Desoshree Ghosh
- Polymer Research Centre and Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur - 741246, Nadia, West Bengal, India.
| | - Afruja Khan
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur - 741246, Nadia, West Bengal, India.
| | - Sagar Bag
- Polymer Research Centre and Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur - 741246, Nadia, West Bengal, India.
| | - Amirul Islam Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur - 741246, Nadia, West Bengal, India.
| | - Priyadarsi De
- Polymer Research Centre and Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur - 741246, Nadia, West Bengal, India.
| |
Collapse
|
9
|
Sakhaii P, Bohorc B, Olpp T, Mohnicke M, Rieke-Zapp J, Dhal PK. Radio frequency gradient enhanced diffusion-edited semi-solid state NMR spectroscopy for detailed structural characterization of chemically modified hyaluronic acid hydrogels. Sci Rep 2024; 14:28612. [PMID: 39562623 PMCID: PMC11577061 DOI: 10.1038/s41598-024-78731-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Applications of functionalized hyaluronic acid (HA) hydrogels for numerous biomedical applications requires their detailed structural characterization. Since these materials are prepared by multistep chemical modifications in the solid phase and not amenable to characterization by standard analytical tools, we employed high-resolution solid-state NMR spectroscopy to gain detailed insights into the structures of the functionalized HA hydrogels. Divinyl sulfone crosslinked HA hydrogels were converted into maleimide-functionalized hydrogels, which were subjected to chemoselective thiol-maleimide reaction using L-cysteine as the protein mimetic thiol reagent. To overcome challenges associated with obtaining high-resolution NMR spectra of crosslinked hydrogels (such as line broadening and overlapping of signals of the hydrogel with those of residual reagents and solvents used during multi-step reaction processes on insoluble polymer matrices), we devised a radio frequency mediated diffusion-edited semi solid-state NMR technique. This technique enabled us to record NMR spectra of hydrogels exclusively by effectively suppressing signals associated with low molecular weight impurities. Thus, it became possible to perform in-depth characterization of these chemically modified HA hydrogels including quantification of reaction outcome for each reaction step.
Collapse
Affiliation(s)
- Peyman Sakhaii
- Global CMC Development, Global R&D, Sanofi, Industrial Park Hoechst, D-65926, Frankfurt/Main, Germany.
| | - Bojan Bohorc
- Global CMC Development, Global R&D, Sanofi, Industrial Park Hoechst, D-65926, Frankfurt/Main, Germany
| | - Thomas Olpp
- Global CMC Development, Global R&D, Sanofi, Industrial Park Hoechst, D-65926, Frankfurt/Main, Germany
| | - Mandy Mohnicke
- Global CMC Development, Global R&D, Sanofi, Industrial Park Hoechst, D-65926, Frankfurt/Main, Germany
| | - Joerg Rieke-Zapp
- Manufacturing Science and Analytical Technology, Sanofi, Industrial Park Hoechst, D- 65926, Frankfurt/Main, Germany
| | - Pradeep K Dhal
- Global CMC Development, Global R&D, Sanofi, 350 Water Street, MA 02141, Cambridge, USA.
| |
Collapse
|
10
|
Gupta A, Kulkarni S, Soman S, Saha M, Kulkarni J, Rana K, Dhas N, Ayesha Farhana S, Kumar Tiyyagura P, Pandey A, Moorkoth S, Mutalik S. Breaking barriers in cancer management: The promising role of microsphere conjugates in cancer diagnosis and therapy. Int J Pharm 2024; 665:124687. [PMID: 39265846 DOI: 10.1016/j.ijpharm.2024.124687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/29/2024] [Accepted: 09/07/2024] [Indexed: 09/14/2024]
Abstract
Cancer is a significant worldwide health concern, and there is a demand for ongoing breakthroughs in treatment techniques. Microspheres are among the most studied drug delivery platforms for delivering cargo to a specified location over an extended period of time. They are biocompatible, biodegradable, and capable of surface modifications. Microspheres and their conjugates have emerged as potential cancer therapeutic options throughout the years. This review provides an in-depth look at the current advancements and applications of microspheres and their conjugates in cancer treatment. The review encompasses a wide array of conjugates, ranging from polymers such as ethyl cellulose and Eudragit to stimuli-responsive polymers, proteins, peptides, polysaccharides such as HA and chitosan, inorganic metals, aptamers, quantum dots (QDs), biomimetic conjugates, and radio conjugates designed for radioembolization. Conjugated microspheres precisely deliver chemotherapeutics to the intended target while achieving controlled drug release to prevent side effects. It offers a means of integrating several distinct therapeutic modalities (chemotherapy, photothermal therapy, photodynamic therapy, radiotherapy, immunotherapy, etc.) to provide synergistic effects during cancer treatment. This review offers insights into the prospects and evolving role of microspheres and their conjugates in the dynamic landscape of cancer therapy. This review provides a comprehensive resource for researchers and clinicians working towards advancements in cancer treatment through innovative applications in therapy and translational research.
Collapse
Affiliation(s)
- Ashutosh Gupta
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Sanjay Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Soji Soman
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Moumita Saha
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Jahnavi Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Komal Rana
- Manipal - Government of Karnataka Bioincubator, 3rd Floor, Advanced Research Centre, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Syeda Ayesha Farhana
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraidah, Qassim 51452, Saudi Arabia
| | - Pavan Kumar Tiyyagura
- Department of Chemical Engineering, Manipal Institute of Technology Manipal, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Abhijeet Pandey
- Global Drug Development/ Technical Research and Development, Novartis Healthcare Private Limited, Genome Valley, Hyderabad 500081, Telangana, India
| | - Sudheer Moorkoth
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
11
|
Boix-Montesinos P, Medel M, Malfanti A, Đorđević S, Masiá E, Charbonnier D, Carrascosa-Marco P, Armiñán A, Vicent MJ. Rational design of a poly-L-glutamic acid-based combination conjugate for hormone-responsive breast cancer treatment. J Control Release 2024; 375:193-208. [PMID: 39242032 DOI: 10.1016/j.jconrel.2024.09.002] [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: 06/17/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
Breast cancer represents the most prevalent tumor type worldwide, with hormone-responsive breast cancer the most common subtype. Despite the effectiveness of endocrine therapy, advanced disease forms represent an unmet clinical need. While drug combination therapies remain promising, differences in pharmacokinetic profiles result in suboptimal ratios of free drugs reaching tumors. We identified a synergistic combination of bisdemethoxycurcumin and exemestane through drug screening and rationally designed star-shaped poly-L-glutamic acid-based combination conjugates carrying these drugs conjugated through pH-responsive linkers for hormone-responsive breast cancer treatment. We synthesized/characterized single and combination conjugates with synergistic drug ratios/loadings. Physicochemical characterization/drug release kinetics studies suggested that lower drug loading prompted a less compact conjugate conformation that supported optimal release. Screening in monolayer and spheroid breast cancer cell cultures revealed that combination conjugates possessed enhanced cytotoxicity/synergism compared to physical mixtures of single-drug conjugates/free drugs; moreover, a combination conjugate with the lowest drug loading outperformed remaining conjugates. This candidate inhibited proliferation-associated signaling, reduced inflammatory chemokine/exosome levels, and promoted autophagy in spheroids; furthermore, it outperformed a physical mixture of single-drug conjugates/free drugs regarding cytotoxicity in patient-derived breast cancer organoids. Our findings highlight the importance of rational design and advanced in vitro models for the selection of polypeptide-based combination conjugates.
Collapse
Affiliation(s)
- Paz Boix-Montesinos
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - María Medel
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain
| | - Alessio Malfanti
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| | - Snežana Đorđević
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain
| | - Esther Masiá
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain; Screening Platform, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - David Charbonnier
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Screening Platform, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), IISCIII and CIEMAT, Madrid, Spain
| | - Paula Carrascosa-Marco
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - Ana Armiñán
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain.
| | - María J Vicent
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain; Screening Platform, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| |
Collapse
|
12
|
Chen Y, Xue T, Chen C, Jang S, Braun PV, Cheng J, Evans CM. Helical peptide structure improves conductivity and stability of solid electrolytes. NATURE MATERIALS 2024; 23:1539-1546. [PMID: 39107570 DOI: 10.1038/s41563-024-01966-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 07/03/2024] [Indexed: 10/09/2024]
Abstract
Ion transport is essential to energy storage, cellular signalling and desalination. Polymers have been explored for decades as solid-state electrolytes by either adding salt to polar polymers or tethering ions to the backbone to create less flammable and more robust systems. New design paradigms are needed to advance the performance of solid polymer electrolytes beyond conventional systems. Here the role of a helical secondary structure is shown to greatly enhance the conductivity of solvent-free polymer electrolytes using cationic polypeptides with a mobile anion. Longer helices lead to higher conductivity, and random coil peptides show substantially lower conductivity. The macrodipole of the helix increases with peptide length, leading to larger dielectric constants. The hydrogen bonding of the helix also imparts thermal and electrochemical stability, while allowing for facile dissolution back to monomer in acid. Peptide polymer electrolytes present a promising platform for the design of next-generation ion-transporting materials.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Materials Science and Engineering, Materials Research Laboratory, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Tianrui Xue
- Department of Materials Science and Engineering, Materials Research Laboratory, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Chen Chen
- Department of Materials Science and Engineering, Materials Research Laboratory, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Seongon Jang
- Department of Materials Science and Engineering, Materials Research Laboratory, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Paul V Braun
- Department of Materials Science and Engineering, Materials Research Laboratory, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jianjun Cheng
- Department of Materials Science and Engineering, Materials Research Laboratory, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
- School of Engineering, Westlake University, Hangzhou, China
| | - Christopher M Evans
- Department of Materials Science and Engineering, Materials Research Laboratory, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
13
|
Wang Y, Shi J, Xin M, Kahkoska AR, Wang J, Gu Z. Cell-drug conjugates. Nat Biomed Eng 2024; 8:1347-1365. [PMID: 38951139 PMCID: PMC11646559 DOI: 10.1038/s41551-024-01230-6] [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: 01/10/2022] [Accepted: 05/01/2024] [Indexed: 07/03/2024]
Abstract
By combining living cells with therapeutics, cell-drug conjugates can potentiate the functions of both components, particularly for applications in drug delivery and therapy. The conjugates can be designed to persist in the bloodstream, undergo chemotaxis, evade surveillance by the immune system, proliferate, or maintain or transform their cellular phenotypes. In this Review, we discuss strategies for the design of cell-drug conjugates with specific functions, the techniques for their preparation, and their applications in the treatment of cancers, autoimmune diseases and other pathologies. We also discuss the translational challenges and opportunities of this class of drug-delivery systems and therapeutics.
Collapse
Affiliation(s)
- Yanfang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Jiaqi Shi
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Minhang Xin
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Anna R Kahkoska
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jinqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Hangzhou, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
14
|
Banerjee T, Dan K, Ghosh S. pH-Responsive self-assembled polymer-photosensitizer conjugate for activable photodynamic therapy. NANOSCALE 2024; 16:19756-19762. [PMID: 39373067 DOI: 10.1039/d4nr03249g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
This paper reports synthesis, aqueous self-assembly and relevance of the pH-triggered activable photodynamic therapy of amphiphilic polyurethane (P1S) functionalized with a heavy-atom free organic photosensitizer. Condensation polymerization between 1,4-diisocyanatobutane and two different dihydroxy monomers (one having a pendant hydrophilic wedge and the other having 1,3-dihydroxypropan-2-one with a reactive carbonyl group) in the presence of a measured amount of (S)-2-methylbutan-1-ol (chain-stopper) and DABCO catalyst produces a reactive pre-polymer P1. Hydrazide-functionalized thionated-naphthalenemonoimide (NMIS), which acts as a photosensitizer, reacted with the carbonyl-functionality of P1 to obtain the desired polymer-photosensitizer conjugate P1S in which the dye was attached to the polymer backbone via an acid-labile hydrazone linker. In water, P1S adopted an intra-chain H-bonding stabilized folded structure, which further assembled to produce a polymersome structure (Dh ≈ 200 nm), in which the hydrophobic membrane consists of aggregated NMIS and trialkoxy-benzene chromophores, as evident from UV/vis, CD and small-angle X-ray diffraction studies. In the aggregated state, NMIS loses its reactive oxygen species (ROS) generation ability and remains in a dormant state. However, under acidic conditions (pH 5.5), the photosensitizer is detached (presumably because of the cleavage of the hydrazone linker) and regains its full ROS-generation activity under photoirradiation, as evidenced from the standard DCFH assay. To test the possibility of such pH-activable intra-cellular ROS generation, P1S was treated with HeLa cells, as it is known that cancer cells are more acidic than normal cells. Indeed, photoirradiation-induced intra-cellular ROS generation was evident by the DCFH assay, resulting in efficient cell killing.
Collapse
Affiliation(s)
- Tanushri Banerjee
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata, 700032, India.
| | - Krishna Dan
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata, 700032, India.
| | - Suhrit Ghosh
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata, 700032, India.
| |
Collapse
|
15
|
Martin J, Michaelis M, Petrović S, Lehnen AC, Müllers Y, Wendler P, Möller HM, Hartlieb M, Glebe U. Application of Sortase-Mediated Ligation for the Synthesis of Block Copolymers and Protein-Polymer Conjugates. Macromol Biosci 2024:e2400316. [PMID: 39360589 DOI: 10.1002/mabi.202400316] [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: 07/05/2024] [Revised: 09/18/2024] [Indexed: 10/04/2024]
Abstract
Sortase-mediated ligation (SML) has become a powerful tool for site-specific protein modification. However, sortase A (SrtA) suffers from low catalytic efficiency and mediates an equilibrium reaction. Therefore, ligations with large macromolecules may be challenging. Here, the synthesis of polymeric building blocks for sortase-mediated ligation constituting peptide-polymers with either the recognition sequence for sortase A (LPX1TGX2) or its nucleophilic counterpart (Gx) is demonstrated. The peptide-polymers are synthesized by solid-phase peptide synthesis followed by photo-iniferter (PI) reversible addition-fragmentation chain-transfer (RAFT) polymerization of various monomers. The building blocks are subsequently utilized to investigate possibilities and limitations when using macromolecules in SML. In particular, diblock copolymers are obtained even when using the orthogonal building blocks in equimolar ratio by exploiting a technique to shift the reaction equilibrium. However, ligations of two polymers can not be achieved when the degree of polymerization exceeds 100. Subsequently, C-terminal protein-polymer conjugates are synthesized. Several polymers are utilized that can replace the omnipresent polyethylene glycol (PEG) in future therapeutics. The conjugation is exemplified with a nanobody that is known for efficient neutralization of SARS-CoV-2. The study demonstrates a universal approach to polymer-LPX1TGX2 and Gx-polymer building blocks and gives insight into their application in SML.
Collapse
Affiliation(s)
- Johannes Martin
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, Potsdam-Golm, Germany
- Fraunhofer Institute for Applied Polymer Research IAP, Geiselbergstr. 69, 14476, Potsdam-Golm, Germany
| | - Marcus Michaelis
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, Potsdam-Golm, Germany
| | - Saša Petrović
- Department of Biochemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, Potsdam-Golm, Germany
| | - Anne-Catherine Lehnen
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, Potsdam-Golm, Germany
- Fraunhofer Institute for Applied Polymer Research IAP, Geiselbergstr. 69, 14476, Potsdam-Golm, Germany
| | - Yannic Müllers
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, Potsdam-Golm, Germany
- Fraunhofer Institute for Applied Polymer Research IAP, Geiselbergstr. 69, 14476, Potsdam-Golm, Germany
| | - Petra Wendler
- Department of Biochemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, Potsdam-Golm, Germany
| | - Heiko M Möller
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, Potsdam-Golm, Germany
| | - Matthias Hartlieb
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, Potsdam-Golm, Germany
- Fraunhofer Institute for Applied Polymer Research IAP, Geiselbergstr. 69, 14476, Potsdam-Golm, Germany
| | - Ulrich Glebe
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, Potsdam-Golm, Germany
- Fraunhofer Institute for Applied Polymer Research IAP, Geiselbergstr. 69, 14476, Potsdam-Golm, Germany
| |
Collapse
|
16
|
López-Espinosa J, Park P, Holcomb M, Godin B, Villapol S. Nanotechnology-driven therapies for neurodegenerative diseases: a comprehensive review. Ther Deliv 2024; 15:997-1024. [PMID: 39297726 PMCID: PMC11583628 DOI: 10.1080/20415990.2024.2401307] [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: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 11/22/2024] Open
Abstract
Neurological diseases, characterized by neuroinflammation and neurodegeneration, impose a significant global burden, contributing to substantial morbidity, disability and mortality. A common feature of these disorders, including stroke, traumatic brain injury and Alzheimer's disease, is the impairment of the blood-brain barrier (BBB), a critical structure for maintaining brain homeostasis. The compromised BBB in neurodegenerative conditions poses a significant challenge for effective treatment, as it allows harmful substances to accumulate in the brain. Nanomedicine offers a promising approach to overcoming this barrier, with nanoparticles (NPs) engineered to deliver therapeutic agents directly to affected brain regions. This review explores the classification and design of NPs, divided into organic and inorganic categories and further categorized based on their chemical and physical properties. These characteristics influence the ability of NPs to carry and release therapeutic agents, target specific tissues and ensure appropriate clearance from the body. The review emphasizes the potential of NPs to enhance the diagnosis and treatment of neurodegenerative diseases through targeted delivery, improved drug bioavailability and real-time therapeutic efficacy monitoring. By addressing the challenges of the compromised BBB and targeting inflammatory biomarkers, NPs represent a cutting-edge strategy in managing neurological disorders, promising better patient outcomes.
Collapse
Affiliation(s)
- Jessica López-Espinosa
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
- School of Medicine and Health Sciences of Tecnológico de Monterrey, Guadalajara, México
| | - Peter Park
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
| | - Morgan Holcomb
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TXUSA
- Department of Obstetrics & Gynecology, Houston Methodist Hospital, Houston, TXUSA
- Department of Obstetrics & Gynecology, Weill Cornell Medicine College, New York, NYUSA
- Department of Biomedical Engineering, Texas A&M University, College Station, TXUSA
| | - Sonia Villapol
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
- Department of Neuroscience in Neurological Surgery, Weill Cornell Medical College, New York, NY USA
| |
Collapse
|
17
|
Swansiger AK, Crittenden CM, Chan SA, Yang SH, Kou D, Prell JS, Chen B. Streamlining LC-MS Characterization of Pharmaceutical Polymers by Fourier-Transform-Based Deconvolution and Macromolecular Mass Defect Analysis. Anal Chem 2024; 96:14715-14719. [PMID: 39229957 DOI: 10.1021/acs.analchem.4c02174] [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: 09/05/2024]
Abstract
Polymer conjugation has risen in importance over the past three decades as a means of increasing the in vivo half-life of biotherapeutics, with benefits including better stability, greater drug efficacy, and lower toxicity. However, the intrinsic variability of polymer synthesis results in products with broad distributions in chain length and branching structure, complicating quality control for successful functionalization and downstream conjugation. Frequently, a combination of several analytical techniques is required for comprehensive characterization. While liquid chromatography-mass spectrometry (LC-MS) is a powerful platform that can provide detailed molecular features of polymers, the mass spectra are inherently challenging to interpret due to high mass polydispersity and overlapping charge distributions. Here, by leveraging Fourier transform-based deconvolution and macromolecular mass defect analysis, we demonstrate a new way to streamline pharmaceutical polymer analysis, shedding light on polymer size, composition, branching, and end-group functionalization with the capability for reaction monitoring.
Collapse
Affiliation(s)
- Andrew K Swansiger
- Department of Chemistry and Biochemistry, 1253 University of Oregon, Eugene, Oregon 97403-1253, United States
| | - Christopher M Crittenden
- Synthetic Molecule Analytical Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Simon A Chan
- Synthetic Molecule Analytical Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Samuel H Yang
- Synthetic Molecule Analytical Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Dawen Kou
- Synthetic Molecule Analytical Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - James S Prell
- Department of Chemistry and Biochemistry, 1253 University of Oregon, Eugene, Oregon 97403-1253, United States
- Materials Science Institute, 1252 University of Oregon, Eugene, Oregon 97403-1252, United States
| | - Bifan Chen
- Synthetic Molecule Analytical Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
18
|
Bourang S, Noruzpour M, Jahanbakhsh Godekahriz S, Ebrahimi HAC, Amani A, Asghari Zakaria R, Yaghoubi H. Application of nanoparticles in breast cancer treatment: a systematic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6459-6505. [PMID: 38700795 DOI: 10.1007/s00210-024-03082-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/02/2024] [Indexed: 09/25/2024]
Abstract
It is estimated that cancer is the second leading cause of death worldwide. The primary or secondary cause of cancer-related mortality for women is breast cancer. The main treatment method for different types of cancer is chemotherapy with drugs. Because of less water solubility of chemotherapy drugs or their inability to pass through membranes, their body absorbs them inadequately, which lowers the treatment's effectiveness. Drug specificity and pharmacokinetics can be changed by nanotechnology using nanoparticles. Instead, targeted drug delivery allows medications to be delivered to the targeted sites. In this review, we focused on nanoparticles as carriers in targeted drug delivery, their characteristics, structure, and the previous studies related to breast cancer. It was shown that nanoparticles could reduce the negative effects of chemotherapy drugs while increasing their effectiveness. Lipid-based nanocarriers demonstrated notable results in this instance, and some products that are undergoing various stages of clinical trials are among the examples. Nanoparticles based on metal or polymers demonstrated a comparable level of efficacy. With the number of cancer cases rising globally, many researchers are now looking into novel treatment approaches, particularly the use of nanotechnology and nanoparticles in the treatment of cancer. In order to help clinicians, this article aimed to gather more information about various areas of nanoparticle application in breast cancer therapy, such as modifying their synthesis and physicochemical characterization. It also sought to gain a deeper understanding of the mechanisms underlying the interactions between nanoparticles and biologically normal or infected tissues.
Collapse
Affiliation(s)
- Shima Bourang
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mehran Noruzpour
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Sodabeh Jahanbakhsh Godekahriz
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hossein Ali Ca Ebrahimi
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Amin Amani
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Rasool Asghari Zakaria
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hashem Yaghoubi
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| |
Collapse
|
19
|
Kopeček J. Hydrophilic biomaterials: From crosslinked and self-assembled hydrogels to polymer-drug conjugates and drug-free macromolecular therapeutics. J Control Release 2024; 373:1-22. [PMID: 38734315 PMCID: PMC11384549 DOI: 10.1016/j.jconrel.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024]
Abstract
This "Magnum Opus" accentuates my lifelong belief that the future of science is in the interdisciplinary approach to hypotheses formulation and problem solving. Inspired by the invention of hydrogels and soft contact lenses by my mentors, my six decades of research have continuously proceeded from the synthesis of biocompatible hydrogels to the development of polymer-drug conjugates, then generation of drug-free macromolecular therapeutics (DFMT) and finally to multi-antigen T cell hybridizers (MATCH). This interdisciplinary journey was inspiring; the lifetime feeling that one is a beginner in some aspects of the research is a driving force that keeps the enthusiasm high. Also, I wanted to illustrate that systematic research in one wide area can be a life-time effort without the need to jump to areas that are temporarily en-vogue. In addition to generating general scientific knowledge, hydrogels from my laboratory have been transferred to the clinic, polymer-drug conjugates to clinical trials, and drug-free macromolecular systems have an excellent potential for personalizing patient therapies. There is a limit to life but no limit to imagination. I anticipate that systematic basic research will contribute to the expansion of our knowledge and create a foundation for the design of new paradigms based on the comprehension of mechanisms of physiological processes. The emerging novel platform technologies in biomaterial-based devices and implants as well as in personalized nanomedicines will ultimately impact clinical practice.
Collapse
Affiliation(s)
- Jindřich Kopeček
- Center for Controlled Chemical Delivery, Department of Molecular Pharmaceutics, Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
20
|
Peng X, Fang J, Lou C, Yang L, Shan S, Wang Z, Chen Y, Li H, Li X. Engineered nanoparticles for precise targeted drug delivery and enhanced therapeutic efficacy in cancer immunotherapy. Acta Pharm Sin B 2024; 14:3432-3456. [PMID: 39220871 PMCID: PMC11365410 DOI: 10.1016/j.apsb.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 09/04/2024] Open
Abstract
The advent of cancer immunotherapy has imparted a transformative impact on cancer treatment paradigms by harnessing the power of the immune system. However, the challenge of practical and precise targeting of malignant cells persists. To address this, engineered nanoparticles (NPs) have emerged as a promising solution for enhancing targeted drug delivery in immunotherapeutic interventions, owing to their small size, low immunogenicity, and ease of surface modification. This comprehensive review delves into contemporary research at the nexus of NP engineering and immunotherapy, encompassing an extensive spectrum of NP morphologies and strategies tailored toward optimizing tumor targeting and augmenting therapeutic effectiveness. Moreover, it underscores the mechanisms that NPs leverage to bypass the numerous obstacles encountered in immunotherapeutic regimens and probes into the combined potential of NPs when co-administered with both established and novel immunotherapeutic modalities. Finally, the review evaluates the existing limitations of NPs as drug delivery platforms in immunotherapy, which could shape the path for future advancements in this promising field.
Collapse
Affiliation(s)
- Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Jianjun Fang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Chuyuan Lou
- Department of Ophthalmology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an 710004, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Shaobo Shan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 10050, China
| | - Zixian Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, China
| | - Yutong Chen
- Department of Pathology, Medical College, Jinan University, Guangzhou 510632, China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Xuexin Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm SE-17177, Sweden
| |
Collapse
|
21
|
Fan L, Tong W, Wei A, Mu X. Progress of proteolysis-targeting chimeras (PROTACs) delivery system in tumor treatment. Int J Biol Macromol 2024; 275:133680. [PMID: 38971291 DOI: 10.1016/j.ijbiomac.2024.133680] [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: 05/03/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) can use the intrinsic protein degradation system in cells to degrade pathogenic target proteins, and are currently a revolutionary frontier of development strategy for tumor treatment with small molecules. However, the poor water solubility, low cellular permeability, and off-target side effects of most PROTACs have prevented them from passing the preclinical research stage of drug development. This requires the use of appropriate delivery systems to overcome these challenging hurdles and ensure precise delivery of PROTACs towards the tumor site. Therefore, the combination of PROTACs and multifunctional delivery systems will open up new research directions for targeted degradation of tumor proteins. In this review, we systematically reviewed the design principles and the most recent advances of various PROTACs delivery systems. Moreover, the constructive strategies for developing multifunctional PROTACs delivery systems were proposed comprehensively. This review aims to deepen the understanding of PROTACs drugs and promote the further development of PROTACs delivery system.
Collapse
Affiliation(s)
- Lianlian Fan
- Department of Pharmacy, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Weifang Tong
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130021, China
| | - Anhui Wei
- Jilin University School of Pharmaceutical Sciences, Changchun 130021, China
| | - Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
22
|
Xue T, Li Y, Torre M, Shao R, Han Y, Chen S, Lee D, Kohane DS. Polymeric Prodrugs using Dynamic Covalent Chemistry for Prolonged Local Anesthesia. Angew Chem Int Ed Engl 2024; 63:e202406158. [PMID: 38885607 PMCID: PMC11337095 DOI: 10.1002/anie.202406158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Indexed: 06/20/2024]
Abstract
Depot-type drug delivery systems are designed to deliver drugs at an effective rate over an extended period. Minimizing initial "burst" can also be important, especially with drugs causing systemic toxicity. Both goals are challenging with small hydrophilic molecules. The delivery of molecules such as the ultrapotent local anesthetic tetrodotoxin (TTX) exemplifies both challenges. Toxicity can be mitigated by conjugating TTX to polymers with ester bonds, but the slow ester hydrolysis can result in subtherapeutic TTX release. Here, we developed a prodrug strategy, based on dynamic covalent chemistry utilizing a reversible reaction between the diol TTX and phenylboronic acids. These polymeric prodrugs exhibited TTX encapsulation efficiencies exceeding 90 % and the resulting polymeric nanoparticles showed a range of TTX release rates. In vivo injection of the TTX polymeric prodrugs at the sciatic nerve reduced TTX systemic toxicity and produced nerve block lasting 9.7±2.0 h, in comparison to 1.6±0.6 h from free TTX. This approach could also be used to co-deliver the diol dexamethasone, which prolonged nerve block to 21.8±5.1 h. This work emphasized the usefulness of dynamic covalent chemistry for depot-type drug delivery systems with slow and effective drug release kinetics.
Collapse
Affiliation(s)
- Tianrui Xue
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School Boston, Massachusetts, 02115, United States
| | - Yang Li
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School Boston, Massachusetts, 02115, United States
| | - Matthew Torre
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School Boston, Massachusetts, 02115, United States
| | - Rachelle Shao
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School Boston, Massachusetts, 02115, United States
| | - Yiyuan Han
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School Boston, Massachusetts, 02115, United States
| | - Shuanglong Chen
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School Boston, Massachusetts, 02115, United States
| | - Daniel Lee
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School Boston, Massachusetts, 02115, United States
| | - Daniel S. Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School Boston, Massachusetts, 02115, United States
| |
Collapse
|
23
|
Zhai Z, Niu J, Xu L, Xu J. Advanced Application of Polymer Nanocarriers in Delivery of Active Ingredients from Traditional Chinese Medicines. Molecules 2024; 29:3520. [PMID: 39124924 PMCID: PMC11314021 DOI: 10.3390/molecules29153520] [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: 07/10/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Active ingredients from Traditional Chinese Medicines (TCMs) have been a cornerstone of healthcare for millennia, offering a rich source of bioactive compounds with therapeutic potential. However, the clinical application of TCMs is often limited by challenges such as poor solubility, low bioavailability, and variable pharmacokinetics. To address these issues, the development of advanced polymer nanocarriers has emerged as a promising strategy for the delivery of TCMs. This review focuses on the introduction of common active ingredients from TCMs and the recent advancements in the design and application of polymer nanocarriers for enhancing the efficacy and safety of TCMs. We begin by discussing the unique properties of TCMs and the inherent challenges associated with their delivery. We then delve into the types of polymeric nanocarriers, including polymer micelles, polymer vesicles, polymer hydrogels, and polymer drug conjugates, highlighting their application in the delivery of active ingredients from TCMs. The main body of the review presents a comprehensive analysis of the state-of-the-art nanocarrier systems and introduces the impact of these nanocarriers on the solubility, stability, and bioavailability of TCM components. On the basis of this, we provide an outlook on the future directions of polymer nanocarriers in TCM delivery. This review underscores the transformative potential of polymer nanocarriers in revolutionizing TCM delivery, offering a pathway to harness the full therapeutic potential of TCMs while ensuring safety and efficacy in a modern medical context.
Collapse
Affiliation(s)
- Zhiyuan Zhai
- School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China
| | - Jianda Niu
- School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China
| | - Liguo Xu
- College of Light Chemical Industry and Materials Engineering, Shunde Polytechnic, Foshan 528333, China
| | - Jinbao Xu
- School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
24
|
Liu Y, Li C, Yang X, Yang B, Fu Q. Stimuli-responsive polymer-based nanosystems for cardiovascular disease theranostics. Biomater Sci 2024; 12:3805-3825. [PMID: 38967109 DOI: 10.1039/d4bm00415a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Stimulus-responsive polymers have found widespread use in biomedicine due to their ability to alter their own structure in response to various stimuli, including internal factors such as pH, reactive oxygen species (ROS), and enzymes, as well as external factors like light. In the context of atherosclerotic cardiovascular diseases (CVDs), stimulus-response polymers have been extensively employed for the preparation of smart nanocarriers that can deliver therapeutic and diagnostic drugs specifically to inflammatory lesions. Compared with traditional drug delivery systems, stimulus-responsive nanosystems offer higher sensitivity, greater versatility, wider applicability, and enhanced biosafety. Recent research has made significant contributions towards designing stimulus-responsive polymer nanosystems for CVDs diagnosis and treatment. This review summarizes recent advances in this field by classifying stimulus-responsive polymer nanocarriers according to different responsiveness types and describing numerous stimuli relevant to these materials. Additionally, we discuss various applications of stimulus-responsive polymer nanomaterials in CVDs theranostics. We hope that this review will provide valuable insights into optimizing the design of stimulus-response polymers for accelerating their clinical application in diagnosing and treating CVDs.
Collapse
Affiliation(s)
- Yuying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Congcong Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Bin Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
25
|
Yu G, Ye Z, Yuan Y, Wang X, Li T, Wang Y, Wang Y, Yan J. Recent Advancements in Biomaterials for Chimeric Antigen Receptor T Cell Immunotherapy. Biomater Res 2024; 28:0045. [PMID: 39011521 PMCID: PMC11246982 DOI: 10.34133/bmr.0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/13/2024] [Indexed: 07/17/2024] Open
Abstract
Cellular immunotherapy is an innovative cancer treatment method that utilizes the patient's own immune system to combat tumor cells effectively. Currently, the mainstream therapeutic approaches include chimeric antigen receptor T cell (CAR-T) therapy, T cell receptor gene-modified T cell therapy and chimeric antigen receptor natural killer-cell therapy with CAR-T therapy mostly advanced. Nonetheless, the conventional manufacturing process of this therapy has shortcomings in each step that call for improvement. Marked efforts have been invested for its enhancement while notable progresses achieved in the realm of biomaterials application. With CAR-T therapy as a prime example, the aim of this review is to comprehensively discuss the various biomaterials used in cell immunotherapy, their roles in regulating immune cells, and their potential for breakthroughs in cancer treatment from gene transduction to efficacy enhancement. This article additionally addressed widely adopted animal models for efficacy evaluating.
Collapse
Affiliation(s)
- Gaoyu Yu
- School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Zhichao Ye
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine,
Zhejiang University, Hangzhou 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Yuyang Yuan
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine,
Zhejiang University, Hangzhou 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
- Department of Translational Medicine & Clinical Research, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Xiaofeng Wang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital,
Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang Province, China
| | - Tianyu Li
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
- Department of Translational Medicine & Clinical Research, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Yi Wang
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Yifan Wang
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine,
Zhejiang University, Hangzhou 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
- Department of Translational Medicine & Clinical Research, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| | - Jianing Yan
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine,
Zhejiang University, Hangzhou 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou 310028, China
| |
Collapse
|
26
|
Guo H, Mi P. Polymer-drug and polymer-protein conjugated nanocarriers: Design, drug delivery, imaging, therapy, and clinical applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1988. [PMID: 39109479 DOI: 10.1002/wnan.1988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 01/06/2025]
Abstract
Polymer-drug conjugates and polymer-protein conjugates have been pivotal in the realm of drug delivery systems for over half a century. These polymeric drugs are characterized by the conjugation of therapeutic molecules or functional moieties to polymers, enabling a range of benefits including extended circulation times, targeted delivery, controlled release, and decreased immunogenicity. This review delves into recent advancements and challenges in the clinical translations and preclinical studies of polymer-drug conjugates and polymer-protein conjugates. The design principles and functionalization strategies crucial for the development of these polymeric drugs were explored followed by the review of structural properties and characteristics of various polymer carriers. This review also identifies significant obstacles in the clinical translation of polymer-drug conjugates and provides insights into the directions for their future development. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Haochen Guo
- Department of Radiology, Huaxi MR Research Center (HMRRC), and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Peng Mi
- Department of Radiology, Huaxi MR Research Center (HMRRC), and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Van Guyse JFR, Abbasi S, Toh K, Nagorna Z, Li J, Dirisala A, Quader S, Uchida S, Kataoka K. Facile Generation of Heterotelechelic Poly(2-Oxazoline)s Towards Accelerated Exploration of Poly(2-Oxazoline)-Based Nanomedicine. Angew Chem Int Ed Engl 2024; 63:e202404972. [PMID: 38651732 DOI: 10.1002/anie.202404972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 04/25/2024]
Abstract
Controlling the end-groups of biocompatible polymers is crucial for enabling polymer-based therapeutics and nanomedicine. Typically, end-group diversification is a challenging and time-consuming endeavor, especially for polymers prepared via ionic polymerization mechanisms with limited functional group tolerance. In this study, we present a facile end-group diversification approach for poly(2-oxazoline)s (POx), enabling quick and reliable production of heterotelechelic polymers to facilitate POxylation. The approach relies on the careful tuning of reaction parameters to establish differential reactivity of a pentafluorobenzyl initiator fragment and the living oxazolinium chain-end, allowing the selective introduction of N-, S-, O-nucleophiles via the termination of the polymerization, and a consecutive nucleophilic para-fluoro substitution. The value of this approach for the accelerated development of nanomedicine is demonstrated through the synthesis of well-defined lipid-polymer conjugates and POx-polypeptide block-copolymers, which are well-suited for drug and gene delivery. Furthermore, we investigated the application of a lipid-POx conjugate for the formulation and delivery of mRNA-loaded lipid nanoparticles for immunization against the SARS-COV-2 virus, underscoring the value of POx as a biocompatible polymer platform.
Collapse
Affiliation(s)
- Joachim F R Van Guyse
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, 210-0821, Kawasaki, Japan
- Present Adresses: S. A., Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 21231, Baltimore, MD, USA
- Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Saed Abbasi
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, 210-0821, Kawasaki, Japan
- Present Adresses: S. A., Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 21231, Baltimore, MD, USA
| | - Kazuko Toh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, 210-0821, Kawasaki, Japan
- Present Adresses: S. A., Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 21231, Baltimore, MD, USA
| | - Zlata Nagorna
- Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Junjie Li
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, 210-0821, Kawasaki, Japan
- Present Adresses: S. A., Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 21231, Baltimore, MD, USA
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, 819-0395, Fukuoka, Japan
| | - Anjaneyulu Dirisala
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, 210-0821, Kawasaki, Japan
- Present Adresses: S. A., Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 21231, Baltimore, MD, USA
| | - Sabina Quader
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, 210-0821, Kawasaki, Japan
- Present Adresses: S. A., Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 21231, Baltimore, MD, USA
| | - Satoshi Uchida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, 210-0821, Kawasaki, Japan
- Present Adresses: S. A., Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 21231, Baltimore, MD, USA
- Department of Medical, Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 606-0823, Kyoto, Japan
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 113-8510, Tokyo, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, 210-0821, Kawasaki, Japan
- Present Adresses: S. A., Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 21231, Baltimore, MD, USA
| |
Collapse
|
28
|
Guerassimoff L, Ferrere M, Bossion A, Nicolas J. Stimuli-sensitive polymer prodrug nanocarriers by reversible-deactivation radical polymerization. Chem Soc Rev 2024; 53:6511-6567. [PMID: 38775004 PMCID: PMC11181997 DOI: 10.1039/d2cs01060g] [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: 12/27/2023] [Indexed: 06/18/2024]
Abstract
Polymer prodrugs are based on the covalent linkage of therapeutic molecules to a polymer structure which avoids the problems and limitations commonly encountered with traditional drug-loaded nanocarriers in which drugs are just physically entrapped (e.g., burst release, poor drug loadings). In the past few years, reversible-deactivation radical polymerization (RDRP) techniques have been extensively used to design tailor-made polymer prodrug nanocarriers. This synthesis strategy has received a lot of attention due to the possibility of fine tuning their structural parameters (e.g., polymer nature and macromolecular characteristics, linker nature, physico-chemical properties, functionalization, etc.), to achieve optimized drug delivery and therapeutic efficacy. In particular, adjusting the nature of the drug-polymer linker has enabled the easy synthesis of stimuli-responsive polymer prodrugs for efficient spatiotemporal drug release. In this context, this review article will give an overview of the different stimuli-sensitive polymer prodrug structures designed by RDRP techniques, with a strong focus on the synthesis strategies, the macromolecular architectures and in particular the drug-polymer linker, which governs the drug release kinetics and eventually the therapeutic effect. Their biological evaluations will also be discussed.
Collapse
Affiliation(s)
- Léa Guerassimoff
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France.
| | - Marianne Ferrere
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France.
| | - Amaury Bossion
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France.
| | - Julien Nicolas
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France.
| |
Collapse
|
29
|
Akpo E, Colin C, Perrin A, Cambedouzou J, Cornu D. Encapsulation of Active Substances in Natural Polymer Coatings. MATERIALS (BASEL, SWITZERLAND) 2024; 17:2774. [PMID: 38894037 PMCID: PMC11173946 DOI: 10.3390/ma17112774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Already used in the food, pharmaceutical, cosmetic, and agrochemical industries, encapsulation is a strategy used to protect active ingredients from external degradation factors and to control their release kinetics. Various encapsulation techniques have been studied, both to optimise the level of protection with respect to the nature of the aggressor and to favour a release mechanism between diffusion of the active compounds and degradation of the barrier material. Biopolymers are of particular interest as wall materials because of their biocompatibility, biodegradability, and non-toxicity. By forming a stable hydrogel around the drug, they provide a 'smart' barrier whose behaviour can change in response to environmental conditions. After a comprehensive description of the concept of encapsulation and the main technologies used to achieve encapsulation, including micro- and nano-gels, the mechanisms of controlled release of active compounds are presented. A panorama of natural polymers as wall materials is then presented, highlighting the main results associated with each polymer and attempting to identify the most cost-effective and suitable methods in terms of the encapsulated drug.
Collapse
Affiliation(s)
| | | | | | - Julien Cambedouzou
- IEM, Université de Montpellier, CNRS, ENSCM, F-34095 Montpellier, France
| | - David Cornu
- IEM, Université de Montpellier, CNRS, ENSCM, F-34095 Montpellier, France
| |
Collapse
|
30
|
Liu Y, Wang Y, Zhang J, Peng Q, Wang X, Xiao X, Shi K. Nanotherapeutics targeting autophagy regulation for improved cancer therapy. Acta Pharm Sin B 2024; 14:2447-2474. [PMID: 38828133 PMCID: PMC11143539 DOI: 10.1016/j.apsb.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/29/2023] [Accepted: 01/29/2024] [Indexed: 06/05/2024] Open
Abstract
The clinical efficacy of current cancer therapies falls short, and there is a pressing demand to integrate new targets with conventional therapies. Autophagy, a highly conserved self-degradation process, has received considerable attention as an emerging therapeutic target for cancer. With the rapid development of nanomedicine, nanomaterials have been widely utilized in cancer therapy due to their unrivaled delivery performance. Hence, considering the potential benefits of integrating autophagy and nanotechnology in cancer therapy, we outline the latest advances in autophagy-based nanotherapeutics. Based on a brief background related to autophagy and nanotherapeutics and their impact on tumor progression, the feasibility of autophagy-based nanotherapeutics for cancer treatment is demonstrated. Further, emerging nanotherapeutics developed to modulate autophagy are reviewed from the perspective of cell signaling pathways, including modulation of the mammalian target of rapamycin (mTOR) pathway, autophagy-related (ATG) and its complex expression, reactive oxygen species (ROS) and mitophagy, interference with autophagosome-lysosome fusion, and inhibition of hypoxia-mediated autophagy. In addition, combination therapies in which nano-autophagy modulation is combined with chemotherapy, phototherapy, and immunotherapy are also described. Finally, the prospects and challenges of autophagy-based nanotherapeutics for efficient cancer treatment are envisioned.
Collapse
Affiliation(s)
- Yunmeng Liu
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Yaxin Wang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Jincheng Zhang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Qikai Peng
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xingdong Wang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xiyue Xiao
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Kai Shi
- College of Pharmacy, Nankai University, Tianjin 300350, China
| |
Collapse
|
31
|
Jan N, Shah H, Khan S, Nasar F, Madni A, Badshah SF, Ali A, Bostanudin MF. Old drug, new tricks: polymer-based nanoscale systems for effective cytarabine delivery. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3565-3584. [PMID: 38015258 DOI: 10.1007/s00210-023-02865-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
Cytarabine, an antimetabolite antineoplastic agent, has been utilized to treat various cancers. However, because of its short half-life, low stability, and limited bioavailability, achieving an optimal plasma concentration requires continuous intravenous administration, which can lead to toxicity in normal cells and tissues. Addressing these limitations is crucial to optimize the therapeutic efficacy of cytarabine while minimizing its adverse effects. The use of novel drug delivery systems, such as polymer-based nanocarriers have emerged as promising vehicles for targeted drug delivery due to their unique properties, including high stability, biocompatibility, and tunable release kinetics. In this review, we examine the application of various polymer-based nanocarriers, including polymeric nanoparticles, polymeric micelles, dendrimers, polymer-drug conjugates, and nano-hydrogels, for the delivery of cytarabine. The article highlights the limitations of conventional cytarabine administration which often lead to suboptimal therapeutic outcomes and systemic toxicity. The rationale for using polymer-based nanocarriers is discussed, highlighting their ability to overcome challenges by providing controlled drug release, improved stability, and enhanced targeting capabilities. In summary, this review offers a valuable resource for drug delivery scientists by providing insights into the design principles, formulation strategies, and potential applications of polymer-based nanocarriers that can enhance the therapeutic efficacy of cytarabine.
Collapse
Affiliation(s)
- Nasrullah Jan
- Akson College of Pharmacy, Mirpur University of Science and Technology (MUST), Mirpur, 10250, Azad Kashmir, Pakistan.
- Department of Pharmacy, The University of Chenab, Gujrat, 50700, Punjab, Pakistan.
| | - Hassan Shah
- Department of Pharmacy, The University of Chenab, Gujrat, 50700, Punjab, Pakistan
| | - Safiullah Khan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Punjab, Pakistan
- Cadson College of Pharmacy, Kharian, 50090, Punjab, Pakistan
| | - Faiza Nasar
- Akson College of Pharmacy, Mirpur University of Science and Technology (MUST), Mirpur, 10250, Azad Kashmir, Pakistan
| | - Asadullah Madni
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Punjab, Pakistan
| | - Syed Faisal Badshah
- Department of Pharmacy, Faculty of Medical and Health Sciences, University of Poonch, Rawalakot, 12350, Azad Kashmir, Pakistan
| | - Ahsan Ali
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Punjab, Pakistan
| | - Mohammad F Bostanudin
- College of Pharmacy, Al Ain University, 112612, Abu Dhabi, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, 112612, Abu Dhabi, United Arab Emirates
| |
Collapse
|
32
|
Wei F, Zheng H, Gao C, Tian J, Gou J, Hamouda HI, Xue C. In Situ Preparation of Star-Shaped Protein-"Smart" Polymer Conjugates with pH and Thermo-Dual Responsibility for Bacterial Separation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38817042 DOI: 10.1021/acs.jafc.3c09129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
To achieve effective separation and enrichment of bacteria, a novel synthetic scheme was developed to synthesize star-style boronate-functionalized copolymers with excellent hydrophilicity and temperature and pH responsiveness. A hydrophilic copolymer brush was synthesized by combining surface-initiated atom-transfer radical polymerization with amide reaction using bovine serum albumin as the core. The copolymer brush was further modified by introducing and immobilizing fluorophenylboronic acids through an amide reaction, resulting in the formation of boronate affinity material BSA@poly(NIPAm-co-AGE)@DFFPBA. The morphology and organic content of BSA@poly(NIPAm-co-AGE)@DFFPBA were systematically characterized. The BSA-derived composites demonstrated a strong binding capacity to both Gram-positive and Gram-negative bacteria. The binding capabilities of the affinity composite to Staphylococcus aureus and Salmonella spp. were 195.8 × 1010 CFU/g and 79.2 × 1010 CFU/g, respectively, which indicates that the novel composite exhibits a high binding capability to bacteria and shows a particularly more significant binding capacity toward Gram-positive bacteria. The bacterial binding of BSA@poly(NIPAm-co-AGE)@DFFPBA can be effectively altered by adjusting the pH and temperature. This study demonstrated that the star-shaped affinity composite had the potential to serve as an affinity material for the rapid separation and enrichment of bacteria in complex samples.
Collapse
Affiliation(s)
- Fayi Wei
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science & Engineering, Ocean University of China, Qingdao 266404, China
- Qingdao Institute of Marine Bioresources for Nutrition & Health Innovation, Qingdao 266109, China
| | - Hongwei Zheng
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science & Engineering, Ocean University of China, Qingdao 266404, China
- Qingdao Institute of Marine Bioresources for Nutrition & Health Innovation, Qingdao 266109, China
| | - Chao Gao
- Technology Center of Qingdao Customs, Qingdao 266003, China
| | - Jiaojiao Tian
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science & Engineering, Ocean University of China, Qingdao 266404, China
| | - Jinpeng Gou
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science & Engineering, Ocean University of China, Qingdao 266404, China
| | - Hamed I Hamouda
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science & Engineering, Ocean University of China, Qingdao 266404, China
| | - Changhu Xue
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science & Engineering, Ocean University of China, Qingdao 266404, China
- Qingdao Institute of Marine Bioresources for Nutrition & Health Innovation, Qingdao 266109, China
- Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
33
|
Rezhdo A, Hershman RL, Van Deventer JA. Design, Construction, and Validation of a Yeast-Displayed Chemically Expanded Antibody Library. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596443. [PMID: 38853888 PMCID: PMC11160716 DOI: 10.1101/2024.05.29.596443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
In vitro display technologies, exemplified by phage and yeast display, have emerged as powerful platforms for antibody discovery and engineering. However, the identification of antibodies that disrupt target functions beyond binding remains a challenge. In particular, there are very few strategies that support identification and engineering of either protein-based irreversible binders or inhibitory enzyme binders. Expanding the range of chemistries in antibody libraries has the potential to lead to efficient discovery of function-disrupting antibodies. In this work, we describe a yeast display-based platform for the discovery of chemically diversified antibodies. We constructed a billion-member antibody library that supports the presentation of a range of chemistries within antibody variable domains via noncanonical amino acid (ncAA) incorporation and subsequent bioorthogonal click chemistry conjugations. Use of a polyspecific orthogonal translation system enables introduction of chemical groups with various properties, including photo-reactive, proximity-reactive, and click chemistry-enabled functional groups for library screening. We established conjugation conditions that facilitate modification of the full library, demonstrating the feasibility of sorting the full billion-member library in "protein-small molecule hybrid" format in future work. Here, we conducted initial library screens after introducing O-(2-bromoethyl)tyrosine (OBeY), a weakly electrophilic ncAA capable of undergoing proximity-induced crosslinking to a target. Enrichments against donkey IgG and protein tyrosine phosphatase 1B (PTP1B) each led to the identification of several OBeY-substituted clones that bind to the targets of interest. Flow cytometry analysis on the yeast surface confirmed higher retention of binding for OBeY-substituted clones compared to clones substituted with ncAAs lacking electrophilic side chains after denaturation. However, subsequent crosslinking experiments in solution with ncAA-substituted clones yielded inconclusive results, suggesting that weakly reactive OBeY side chain is not sufficient to drive robust crosslinking in the clones isolated here. Nonetheless, this work establishes a multi-modal, chemically expanded antibody library and demonstrates the feasibility of conducting discovery campaigns in chemically expanded format. This versatile platform offers new opportunities for identifying and characterizing antibodies with properties beyond what is accessible with the canonical amino acids, potentially enabling discovery of new classes of reagents, diagnostics, and even therapeutic leads.
Collapse
Affiliation(s)
- Arlinda Rezhdo
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, USA
| | - Rebecca L. Hershman
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, USA
| | - James A. Van Deventer
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, USA
- Biomedical Engineering Department, Tufts University, Medford, Massachusetts 02155, USA
| |
Collapse
|
34
|
Geng WC, Jiang ZT, Chen SL, Guo DS. Supramolecular interaction in the action of drug delivery systems. Chem Sci 2024; 15:7811-7823. [PMID: 38817563 PMCID: PMC11134347 DOI: 10.1039/d3sc04585d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/27/2024] [Indexed: 06/01/2024] Open
Abstract
Complex diseases and diverse clinical needs necessitate drug delivery systems (DDSs), yet the current performance of DDSs is far from ideal. Supramolecular interactions play a pivotal role in various aspects of drug delivery, encompassing biocompatibility, drug loading, stability, crossing biological barriers, targeting, and controlled release. Nevertheless, despite having some understanding of the role of supramolecular interactions in drug delivery, their incorporation is frequently overlooked in the design and development of DDSs. This perspective provides a brief analysis of the involved supramolecular interactions in the action of drug delivery, with a primary emphasis on the DDSs employed in the clinic, mainly liposomes and polymers, and recognized phenomena in research, such as the protein corona. The supramolecular interactions implicated in various aspects of drug delivery systems, including biocompatibility, drug loading, stability, spatiotemporal distribution, and controlled release, were individually analyzed and discussed. This perspective aims to trigger a comprehensive and systematic consideration of supramolecular interactions in the further development of DDSs. Supramolecular interactions embody the true essence of the interplay between the majority of DDSs and biological systems.
Collapse
Affiliation(s)
- Wen-Chao Geng
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Ze-Tao Jiang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Shi-Lin Chen
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| |
Collapse
|
35
|
Lu K, Shen X, Shi Y, He Z, Zhang D, Zhou M. Biodegradable polyester copolymers: synthesis based on the Biginelli reaction, characterization, and evaluation of their application properties. RSC Adv 2024; 14:17440-17447. [PMID: 38813120 PMCID: PMC11135155 DOI: 10.1039/d4ra02002b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/11/2024] [Indexed: 05/31/2024] Open
Abstract
The Biginelli reaction, a three-component cyclocondensation reaction, is an important member of the multicomponent reaction (MCR) family. In this study, we conducted end-group modifications on a variety of biodegradable polyesters, including poly(1,4-butylene adipate) (PBA), poly(ε-caprolactone) (PCL), polylactic acid (PLA), and poly(p-dioxanone) (PPDO), based on the precursor polyethylene glycol (PEG). By combining two polymers through the Biginelli multi-component reaction, four new biodegradable polyester copolymers, namely DHPM-PBA, DHPM-PCL, DHPM-PLA, and DHPM-PPDO, were formed. These Biginelli reactions demonstrated exceptional completeness, validating the efficiency of the synthesis strategy. Although the introduction of various polyesters lead to different properties, such as crystallinity and cytotoxicity, the newly synthesized 3,4-dihydro-2(H)-pyrimidinone compounds (DHPMs) exhibit enhanced hydrophilicity and can self-assemble in water and N,N-dimethylformamide (DMF) solution to form micelles with a controllable size. Furthermore, DHPM-PPDO promotes cellular growth and has potential applications in wound healing and tissue engineering. In conclusion, this method demonstrates great universality and methodological significance and offers insights into the medical applications of polyethylene glycol.
Collapse
Affiliation(s)
- Kai Lu
- College of Materials Science and Engineering, Zhejiang University of Technology Hangzhou Zhejiang 310014 China
| | - Xinyi Shen
- College of Materials Science and Engineering, Zhejiang University of Technology Hangzhou Zhejiang 310014 China
| | - Yunhai Shi
- College of Materials Science and Engineering, Zhejiang University of Technology Hangzhou Zhejiang 310014 China
| | - Zejian He
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University Hangzhou Zhejiang 310027 China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center Hangzhou Zhejiang 311215 China
| | - Dahong Zhang
- Department of Urology, Zhejiang Provincial People's Hospital Hangzhou Zhejiang 310014 China
| | - Mi Zhou
- College of Materials Science and Engineering, Zhejiang University of Technology Hangzhou Zhejiang 310014 China
| |
Collapse
|
36
|
Zhang Z, Lv X, Mu X, Zhao M, Wang S, Ke C, Ding S, Zhou D, Wang M, Zeng R. In-situ noncovalent interaction of ammonium ion enabled C-H bond functionalization of polyethylene glycols. Nat Commun 2024; 15:4445. [PMID: 38789453 PMCID: PMC11126569 DOI: 10.1038/s41467-024-48584-8] [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: 11/23/2023] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
The noncovalent interactions of ammonium ion with multidentate oxygen-based host has never been reported as a reacting center in catalytic reactions. In this work, we report a reactivity enhancement process enabled by non-covalent interaction of ammonium ion, achieving the C-H functionalization of polyethylene glycols with acrylates by utilizing photoinduced co-catalysis of iridium and quinuclidine. A broad scope of alkenes can be tolerated without observing significant degradation. Moreover, this cyano-free condition respectively allows the incorporation of bioactive molecules and the PEGylation of dithiothreitol-treated bovine serum albumin, showing great potentials in drug delivery and protein modification. DFT calculations disclose that the formed α-carbon radical adjacent to oxygen-atom is reduced directly by iridium before acrylate addition. And preliminary mechanistic experiments reveal that the noncovalent interaction of PEG chain with the formed quinuclidinium species plays a unique role as a catalytic site by facilitating the proton transfer and ultimately enabling the transformation efficiently.
Collapse
Affiliation(s)
- Zongnan Zhang
- School of Chemistry & School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Xueli Lv
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, P. R. China
| | - Xin Mu
- School of Chemistry & School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Mengyao Zhao
- College of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, P. R. China
| | - Sichang Wang
- College of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, P. R. China
| | - Congyu Ke
- College of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, P. R. China
| | - Shujiang Ding
- School of Chemistry & School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China.
| | - Dezhong Zhou
- School of Chemistry & School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China.
| | - Minyan Wang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, P. R. China.
| | - Rong Zeng
- School of Chemistry & School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China.
| |
Collapse
|
37
|
Xiang Y, Zhao Y, Cheng T, Sun S, Wang J, Pei R. Implantable Neural Microelectrodes: How to Reduce Immune Response. ACS Biomater Sci Eng 2024; 10:2762-2783. [PMID: 38591141 DOI: 10.1021/acsbiomaterials.4c00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Implantable neural microelectrodes exhibit the great ability to accurately capture the electrophysiological signals from individual neurons with exceptional submillisecond precision, holding tremendous potential for advancing brain science research, as well as offering promising avenues for neurological disease therapy. Although significant advancements have been made in the channel and density of implantable neural microelectrodes, challenges persist in extending the stable recording duration of these microelectrodes. The enduring stability of implanted electrode signals is primarily influenced by the chronic immune response triggered by the slight movement of the electrode within the neural tissue. The intensity of this immune response increases with a higher bending stiffness of the electrode. This Review thoroughly analyzes the sequential reactions evoked by implanted electrodes in the brain and highlights strategies aimed at mitigating chronic immune responses. Minimizing immune response mainly includes designing the microelectrode structure, selecting flexible materials, surface modification, and controlling drug release. The purpose of this paper is to provide valuable references and ideas for reducing the immune response of implantable neural microelectrodes and stimulate their further exploration in the field of brain science.
Collapse
Affiliation(s)
- Ying Xiang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yuewu Zhao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Tingting Cheng
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Shengkai Sun
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jine Wang
- Jiangxi Institute of Nanotechnology, Nanchang 330200, China
- College of Medicine and Nursing, Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou 253023, China
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
38
|
Beach M, Nayanathara U, Gao Y, Zhang C, Xiong Y, Wang Y, Such GK. Polymeric Nanoparticles for Drug Delivery. Chem Rev 2024; 124:5505-5616. [PMID: 38626459 PMCID: PMC11086401 DOI: 10.1021/acs.chemrev.3c00705] [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] [Indexed: 04/18/2024]
Abstract
The recent emergence of nanomedicine has revolutionized the therapeutic landscape and necessitated the creation of more sophisticated drug delivery systems. Polymeric nanoparticles sit at the forefront of numerous promising drug delivery designs, due to their unmatched control over physiochemical properties such as size, shape, architecture, charge, and surface functionality. Furthermore, polymeric nanoparticles have the ability to navigate various biological barriers to precisely target specific sites within the body, encapsulate a diverse range of therapeutic cargo and efficiently release this cargo in response to internal and external stimuli. However, despite these remarkable advantages, the presence of polymeric nanoparticles in wider clinical application is minimal. This review will provide a comprehensive understanding of polymeric nanoparticles as drug delivery vehicles. The biological barriers affecting drug delivery will be outlined first, followed by a comprehensive description of the various nanoparticle designs and preparation methods, beginning with the polymers on which they are based. The review will meticulously explore the current performance of polymeric nanoparticles against a myriad of diseases including cancer, viral and bacterial infections, before finally evaluating the advantages and crucial challenges that will determine their wider clinical potential in the decades to come.
Collapse
Affiliation(s)
- Maximilian
A. Beach
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Umeka Nayanathara
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yanting Gao
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Changhe Zhang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yijun Xiong
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yufu Wang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Georgina K. Such
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
39
|
Zhao J, Li X, Ma T, Chang B, Zhang B, Fang J. Glutathione-triggered prodrugs: Design strategies, potential applications, and perspectives. Med Res Rev 2024; 44:1013-1054. [PMID: 38140851 DOI: 10.1002/med.22007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/20/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
The burgeoning prodrug strategy offers a promising avenue toward improving the efficacy and specificity of cytotoxic drugs. Elevated intracellular levels of glutathione (GSH) have been regarded as a hallmark of tumor cells and characteristic feature of the tumor microenvironment. Considering the pivotal involvement of elevated GSH in the tumorigenic process, a diverse repertoire of GSH-triggered prodrugs has been developed for cancer therapy, facilitating the attenuation of deleterious side effects associated with conventional chemotherapeutic agents and/or the attainment of more efficacious therapeutic outcomes. These prodrug formulations encompass a spectrum of architectures, spanning from small molecules to polymer-based and organic-inorganic nanomaterial constructs. Although the GSH-triggered prodrugs have been gaining increasing interests, a comprehensive review of the advancements made in the field is still lacking. To fill the existing lacuna, this review undertakes a retrospective analysis of noteworthy research endeavors, based on a categorization of these molecules by their diverse recognition units (i.e., disulfides, diselenides, Michael acceptors, and sulfonamides/sulfonates). This review also focuses on explaining the distinct benefits of employing various chemical architecture strategies in the design of these prodrug agents. Furthermore, we highlight the potential for synergistic functionality by incorporating multiple-targeting conjugates, theranostic entities, and combinational treatment modalities, all of which rely on the GSH-triggering. Overall, an extensive overview of the emerging field is presented in this review, highlighting the obstacles and opportunities that lie ahead. Our overarching goal is to furnish methodological guidance for the development of more efficacious GSH-triggered prodrugs in the future. By assessing the pros and cons of current GSH-triggered prodrugs, we expect that this review will be a handful reference for prodrug design, and would provide a guidance for improving the properties of prodrugs and discovering novel trigger scaffolds for constructing GSH-triggered prodrugs.
Collapse
Affiliation(s)
- Jintao Zhao
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Xinming Li
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Tao Ma
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Bingbing Chang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Jianguo Fang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
40
|
Guerassimoff L, Ferrere M, Van Herck S, Dehissi S, Nicolas V, De Geest BG, Nicolas J. Thermosensitive polymer prodrug nanoparticles prepared by an all-aqueous nanoprecipitation process and application to combination therapy. J Control Release 2024; 369:376-393. [PMID: 38554772 DOI: 10.1016/j.jconrel.2024.03.049] [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: 12/18/2023] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
Despite their great versatility and ease of functionalization, most polymer-based nanocarriers intended for use in drug delivery often face serious limitations that can prevent their clinical translation, such as uncontrolled drug release and off-target toxicity, which mainly originate from the burst release phenomenon. In addition, residual solvents from the formulation process can induce toxicity, alter the physico-chemical and biological properties and can strongly impair further pharmaceutical development. To address these issues, we report polymer prodrug nanoparticles, which are prepared without organic solvents via an all-aqueous formulation process, and provide sustained drug release. This was achieved by the "drug-initiated" synthesis of well-defined copolymer prodrugs exhibiting a lower critical solution temperature (LCST) and based on the anticancer drug gemcitabine (Gem). After screening for different structural parameters, prodrugs based on amphiphilic diblock copolymers were formulated into stable nanoparticles by all-aqueous nanoprecipitation, with rather narrow particle size distribution and average diameters in the 50-80 nm range. They exhibited sustained Gem release in human serum and acetate buffer, rapid cellular uptake and significant cytotoxicity on A549 and Mia PaCa-2 cancer cells. We also demonstrated the versatility of this approach by formulating Gem-based polymer prodrug nanoparticles loaded with doxorubicin (Dox) for combination therapy. The dual-drug nanoparticles exhibited sustained release of Gem in human serum and acidic release of Dox under accelerated pathophysiological conditions. Importantly, they also induced a synergistic effect on triple-negative breast cancer line MDA-MB-231, which is a relevant cell line to this combination.
Collapse
Affiliation(s)
- Léa Guerassimoff
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay 91400, France
| | - Marianne Ferrere
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay 91400, France
| | - Simon Van Herck
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Samy Dehissi
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay 91400, France
| | - Valérie Nicolas
- Institut Paris-Saclay d'Innovation Thérapeutique (IPSIT), UMS IPSIT Université Paris-Saclay US 31 INSERM, UMS 3679 CNRS, Microscopy Facility, Orsay 91400, France
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Julien Nicolas
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay 91400, France.
| |
Collapse
|
41
|
Zhang H, Wang J, Wu R, Zheng B, Sang Y, Wang B, Song L, Hu Y, Ma X. Self-Supplied Reactive Oxygen Species-Responsive Mitoxantrone Polyprodrug for Chemosensitization-Enhanced Chemotherapy under Moderate Hyperthermia. Adv Healthc Mater 2024; 13:e2303631. [PMID: 38278138 DOI: 10.1002/adhm.202303631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/11/2023] [Indexed: 01/28/2024]
Abstract
Currently, the secondary development and modification of clinical drugs has become one of the research priorities. Researchers have developed a variety of TME-responsive nanomedicine carriers to solve certain clinical problems. Unfortunately, endogenous stimuli such as reactive oxygen species (ROS), as an important prerequisite for effective therapeutic efficacy, are not enough to achieve the expected drug release process, therefore, it is difficult to achieve a continuous and efficient treatment process. Herein, a self-supply ROS-responsive cascade polyprodrug (PMTO) is designed. The encapsulation of the chemotherapy drug mitoxantrone (MTO) in a polymer backbone could effectively reduce systemic toxicity when transported in vivo. After PMTO is degraded by endogenous ROS of the TME, another part of the polyprodrug backbone becomes cinnamaldehyde (CA), which can further enhance intracellular ROS, thereby achieving a sustained drug release process. Meanwhile, due to the disruption of the intracellular redox environment, the efficacy of chemotherapy drugs is enhanced. Finally, the anticancer treatment efficacy is further enhanced due to the mild hyperthermia effect of PMTO. In conclusion, the designed PMTO demonstrates remarkable antitumor efficacy, effectively addressing the limitations associated with MTO.
Collapse
Affiliation(s)
- Hongjie Zhang
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Jing Wang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| | - Ruiying Wu
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| | - Benyan Zheng
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Yanxiang Sang
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Bibo Wang
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Lei Song
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Yuan Hu
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Xiaopeng Ma
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| |
Collapse
|
42
|
Bera S, Bej R, Kanjilal P, Sinha S, Ghosh S. Bioreducible Amphiphilic Hyperbranched Polymer-Drug Conjugate for Intracellular Drug Delivery. Bioconjug Chem 2024; 35:480-488. [PMID: 38514383 DOI: 10.1021/acs.bioconjchem.4c00006] [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: 03/23/2024]
Abstract
This paper reports synthesis of a bioreducible hyperbranched (HB) polymer by A2+B3 approach from commercially available dithiothreitol (DTT) (A2) and an easily accessible trifunctional monomer (B3) containing three reactive pyridyl-disulfide groups. Highly efficient thiol-activated disulfide exchange reaction leads to the formation of the HB polymer (Mw = 21000; Đ = 2.3) with bioreducible disulfide linkages in the backbone and two different functional groups, namely, hydroxyl and pyridyl-disulfide in the core and periphery, respectively, of the HB-polymer. Postpolymerization functionalization of the hydroxyl-groups with camptothecin (CPT), a topoisomerase inhibitor and known anticancer drug, followed by replacing the terminal pyridyl-disulfide groups with oligo-oxyethylene-thiol resulted in easy access to an amphiphilic HB polydisulfide-CPT conjugate (P1) with a very high drug loading content of ∼40%. P1 aggregated in water (above ∼10 μg/mL) producing drug-loaded nanoparticles (Dh ∼ 135 nm), which showed highly efficient glutathione (GSH)-triggered release of the active CPT. Mass spectrometry analysis of the GSH-treated P1 showed the presence of the active CPT drug as well as a cyclic monothiocarbonate product, which underpins the cascade-degradation mechanism involving GSH-triggered cleavage of the labile disulfide linkage, followed by intramolecular nucleophilic attack by the in situ generated thiol to the neighboring carbonate linkage, resulting in release of the active CPT drug. The P1 nanoparticle showed excellent cellular uptake as tested by confocal fluorescence microscopy in HeLa cells by predominantly endocytosis mechanism, resulting in highly efficient cell killing (IC50 ∼ 0.6 μg/mL) as evident from the results of the MTT assay, as well as the apoptosis assay. Comparative studies with an analogous linear polymer-CPT conjugate showed much superior intracellular drug delivery potency of the hyperbranched polymer.
Collapse
Affiliation(s)
- Sukanya Bera
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Raju Bej
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Pintu Kanjilal
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Satyaki Sinha
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Suhrit Ghosh
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
- Technical Research Center (TRC),Indian Association for the Cultivation of Science, Kolkata 700032, India
| |
Collapse
|
43
|
Ji P, Jin XK, Deng XC, Zhang SM, Liang JL, Li QR, Chen WH, Zhang XZ. Metabolic Regulation-Mediated Reversion of the Tumor Immunosuppressive Microenvironment for Potentiating Cooperative Metabolic Therapy and Immunotherapy. NANO LETTERS 2024; 24:4691-4701. [PMID: 38588212 DOI: 10.1021/acs.nanolett.4c01307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Tumor cells exhibit heightened glucose (Glu) consumption and increased lactic acid (LA) production, resulting in the formation of an immunosuppressive tumor microenvironment (TME) that facilitates malignant proliferation and metastasis. In this study, we meticulously engineer an antitumor nanoplatform, denoted as ZLGCR, by incorporating glucose oxidase, LA oxidase, and CpG oligodeoxynucleotide into zeolitic imidazolate framework-8 that is camouflaged with a red blood cell membrane. Significantly, ZLGCR-mediated consumption of Glu and LA not only amplifies the effectiveness of metabolic therapy but also reverses the immunosuppressive TME, thereby enhancing the therapeutic outcomes of CpG-mediated antitumor immunotherapy. It is particularly important that the synergistic effect of metabolic therapy and immunotherapy is further augmented when combined with immune checkpoint blockade therapy. Consequently, this engineered antitumor nanoplatform will achieve a cooperative tumor-suppressive outcome through the modulation of metabolism and immune responses within the TME.
Collapse
Affiliation(s)
- Ping Ji
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Institute of Precision Medicine Peking University Shenzhen Hospital, Shenzhen 518036, P. R. China
| | - Xiao-Kang Jin
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xin-Chen Deng
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Shi-Man Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Jun-Long Liang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Qian-Ru Li
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| |
Collapse
|
44
|
Zhang D, Chen Y, Hao M, Xia Y. Putting Hybrid Nanomaterials to Work for Biomedical Applications. Angew Chem Int Ed Engl 2024; 63:e202319567. [PMID: 38429227 PMCID: PMC11478030 DOI: 10.1002/anie.202319567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/03/2024]
Abstract
Hybrid nanomaterials have found use in many biomedical applications. This article provides a comprehensive review of the principles, techniques, and recent advancements in the design and fabrication of hybrid nanomaterials for biomedicine. We begin with an introduction to the general concept of material hybridization, followed by a discussion of how this approach leads to materials with additional functionality and enhanced performance. We then highlight hybrid nanomaterials in the forms of nanostructures, nanocomposites, metal-organic frameworks, and biohybrids, including their fabrication methods. We also showcase the use of hybrid nanomaterials to advance biomedical engineering in the context of nanomedicine, regenerative medicine, diagnostics, theranostics, and biomanufacturing. Finally, we offer perspectives on challenges and opportunities.
Collapse
Affiliation(s)
- Dong Zhang
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332 (USA)
| | - Yidan Chen
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332 (USA)
| | - Min Hao
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332 (USA)
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332 (USA); School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332 (USA)
| |
Collapse
|
45
|
He X, Li J, Liang X, Mao W, Deng X, Qin M, Su H, Wu H. An all-in-one tetrazine reagent for cysteine-selective labeling and bioorthogonal activable prodrug construction. Nat Commun 2024; 15:2831. [PMID: 38565562 PMCID: PMC10987521 DOI: 10.1038/s41467-024-47188-6] [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: 09/23/2023] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
The prodrug design strategy offers a potent solution for improving therapeutic index and expanding drug targets. However, current prodrug activation designs are mainly responsive to endogenous stimuli, resulting in unintended drug release and systemic toxicity. In this study, we introduce 3-vinyl-6-oxymethyl-tetrazine (voTz) as an all-in-one reagent for modular preparation of tetrazine-caged prodrugs and chemoselective labeling peptides to produce bioorthogonal activable peptide-prodrug conjugates. These stable prodrugs can selectively bind to target cells, facilitating cellular uptake. Subsequent bioorthogonal cleavage reactions trigger prodrug activation, significantly boosting potency against tumor cells while maintaining exceptional off-target safety for normal cells. In vivo studies demonstrate the therapeutic efficacy and safety of this prodrug design approach. Given the broad applicability of functional groups and labeling versatility with voTz, we foresee that this strategy will offer a versatile solution to enhance the therapeutic range of cytotoxic agents and facilitate the development of bioorthogonal activatable biopharmaceuticals and biomaterials.
Collapse
Affiliation(s)
- Xinyu He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xinxin Liang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wuyu Mao
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xinglong Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan University, Chengdu, China
| | - Meng Qin
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Su
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Haoxing Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan University, Chengdu, China.
| |
Collapse
|
46
|
Xu X, Zhang J, Wang T, Li J, Rong Y, Wang Y, Bai C, Yan Q, Ran X, Wang Y, Zhang T, Sun J, Jiang Q. Emerging non-antibody‒drug conjugates (non-ADCs) therapeutics of toxins for cancer treatment. Acta Pharm Sin B 2024; 14:1542-1559. [PMID: 38572098 PMCID: PMC10985036 DOI: 10.1016/j.apsb.2023.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/31/2023] [Accepted: 11/23/2023] [Indexed: 04/05/2024] Open
Abstract
The non-selective cytotoxicity of toxins limits the clinical relevance of the toxins. In recent years, toxins have been widely used as warheads for antibody‒drug conjugates (ADCs) due to their efficient killing activity against various cancer cells. Although ADCs confer certain targeting properties to the toxins, low drug loading capacity, possible immunogenicity, and other drawbacks also limit the potential application of ADCs. Recently, non-ADC delivery strategies for toxins have been extensively investigated. To further understand the application of toxins in anti-tumor, this paper provided an overview of prodrugs, nanodrug delivery systems, and biomimetic drug delivery systems. In addition, toxins and their combination strategies with other therapies were discussed. Finally, the prospect and challenge of toxins in cancer treatment were also summarized.
Collapse
Affiliation(s)
- Xiaolan Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiaming Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tao Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jing Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yukang Rong
- School of Education, University of Nottingham, Nottingham NG7 2RD, UK
| | - Yanfang Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chenxia Bai
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qing Yan
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaohua Ran
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yingli Wang
- Department of Pharmacy, Linyi People's Hospital, Shandong University, Linyi 276000, China
| | - Tianhong Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qikun Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| |
Collapse
|
47
|
Dixit T, Dave N, Basu K, Sonawane P, Gawas T, Ravindran S. Nano-radiopharmaceuticals as therapeutic agents. Front Med (Lausanne) 2024; 11:1355058. [PMID: 38560384 PMCID: PMC10978739 DOI: 10.3389/fmed.2024.1355058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
In recent years, there has been an increased interest in exploring the potential synergy between nanotechnology and nuclear medicine. The application of radioactive isotopes, commonly referred to as radiopharmaceuticals, is recognized in nuclear medicine for diagnosing and treating various diseases. Unlike conventional pharmaceutical agents, radiopharmaceuticals are designed to work without any pharmacological impact on the body. Nevertheless, the radiation dosage employed in radiopharmaceuticals is often sufficiently high to elicit adverse effects associated with radiation exposure. Exploiting their capacity for selective accumulation on specific organ targets, radiopharmaceuticals have utility in treating diverse disorders. The incorporation of nanosystems may additionally augment the targeting capability of radiopharmaceuticals, leveraging their distinct pharmacokinetic characteristics. Conversely, radionuclides could be used in research to assess nanosystems pharmacologically. However, more investigation is needed to verify the safety and effectiveness of radiopharmaceutical applications mediated by nanosystems. The use of nano-radiopharmaceuticals as therapeutic agents to treat various illnesses and disorders is majorly covered in this review. The targeted approach to cancer therapy and various types of nanotools for nano-radiopharmaceutical delivery, is also covered in this article.
Collapse
Affiliation(s)
| | | | | | | | | | - Selvan Ravindran
- Symbiosis School of Biological Sciences, Faculty of Medical and Health Sciences, Symbiosis International (Deemed University), Lavale, Pune, India
| |
Collapse
|
48
|
Xie H, Zhang C. Potential of the nanoplatform and PROTAC interface to achieve targeted protein degradation through the Ubiquitin-Proteasome system. Eur J Med Chem 2024; 267:116168. [PMID: 38310686 DOI: 10.1016/j.ejmech.2024.116168] [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: 11/03/2023] [Revised: 01/11/2024] [Accepted: 01/21/2024] [Indexed: 02/06/2024]
Abstract
In eukaryotic cells, the ubiquitin-proteasome system (UPS) plays a crucial role in selectively breaking down specific proteins. The ability of the UPS to target proteins effectively and expedite their removal has significantly contributed to the evolution of UPS-based targeted protein degradation (TPD) strategies. In particular, proteolysis targeting chimeras (PROTACs) are an immensely promising tool due to their high efficiency, extensive target range, and negligible drug resistance. This breakthrough has overcome the limitations posed by traditionally "non-druggable" proteins. However, their high molecular weight and constrained solubility impede the delivery of PROTACs. Fortunately, the field of nanomedicine has experienced significant growth, enabling the delivery of PROTACs through nanoscale drug-delivery systems, which effectively improves the stability, solubility, drug distribution, tissue-specific accumulation, and stimulus-responsive release of PROTACs. This article reviews the mechanism of action attributed to PROTACs and their potential implications for clinical applications. Moreover, we present strategies involving nanoplatforms for the effective delivery of PROTACs and evaluate recent advances in targeting nanoplatforms to the UPS. Ultimately, an assessment is conducted to determine the feasibility of utilizing PROTACs and nanoplatforms for UPS-based TPD. The primary aim of this review is to provide innovative, reliable solutions to overcome the current challenges obstructing the effective use of PROTACs in the management of cancer, neurodegenerative diseases, and metabolic syndrome. Therefore, this is a promising technology for improving the treatment status of major diseases.
Collapse
Affiliation(s)
- Hanshu Xie
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Chao Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
49
|
Ghassemi Z, Leach JB. Impact of Confinement within a Hydrogel Mesh on Protein Thermodynamic Stability and Aggregation Kinetics. Mol Pharm 2024; 21:1137-1148. [PMID: 38277273 DOI: 10.1021/acs.molpharmaceut.3c00677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
Though protein stability and aggregation have been well characterized in dilute solutions, the influence of a confining environment that exists (e.g., in intercellular and tissue spaces and therapeutic formulations) on the protein structure is largely unknown. Herein, the effects of confinement on stability and aggregation were explored for proteins of different sizes, stability, and hydrophobicity when encapsulated in hydrophilic poly(ethylene glycol) hydrogels. Denaturation curves show linear correlations between confinement size (mesh size) and thermodynamic stability, i.e., unfolding free energy and surface area accessible for solvation (represented by m-value). Two clusters of protein types are identifiable from these correlations; the clusters are defined by differences in protein stability, surface area, and aggregation propensity. Proteins with higher stability, larger surface area, and lower aggregation propensity (e.g., lysozyme and myoglobin) are less affected by the confinement imposed by mesh size than proteins with lower stability, smaller surface area, and higher aggregation propensity (e.g., growth hormone and aldehyde dehydrogenase). According to aggregation kinetics measured by thioflavin T fluorescence, confinement in smaller mesh sizes resulted in slower aggregation rates than that in larger mesh sizes. Compared to that in buffer solution, the confinement of a hydrophobic protein (e.g., human insulin) in the hydrogels accelerates protein aggregation. Conversely, the confinement of a hydrophilic protein (e.g., human amylin) in the hydrogels decelerates or prevents aggregation, with the rates of aggregation inversely proportional to mesh size. These findings provide new insights into protein conformational stability in confined microenvironments relevant to various cellular, tissue, and therapeutics scenarios.
Collapse
Affiliation(s)
- Zahra Ghassemi
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, ECS 314, 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
| | - Jennie B Leach
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, ECS 314, 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
| |
Collapse
|
50
|
Singh D, Sharma Y, Dheer D, Shankar R. Stimuli responsiveness of recent biomacromolecular systems (concept to market): A review. Int J Biol Macromol 2024; 261:129901. [PMID: 38316328 DOI: 10.1016/j.ijbiomac.2024.129901] [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: 10/13/2023] [Revised: 01/08/2024] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
Stimuli responsive delivery systems, also known as smart/intelligent drug delivery systems, are specialized delivery vehicles designed to provide spatiotemporal control over drug release at target sites in various diseased conditions, including tumor, inflammation and many others. Recent advances in the design and development of a wide variety of stimuli-responsive (pH, redox, enzyme, temperature) materials have resulted in their widespread use in drug delivery and tissue engineering. The aim of this review is to provide an insight of recent nanoparticulate drug delivery systems including polymeric nanoparticles, dendrimers, lipid-based nanoparticles and the design of new polymer-drug conjugates (PDCs), with a major emphasis on natural along with synthetic commercial polymers used in their construction. Special focus has been placed on stimuli-responsive polymeric materials, their preparation methods, and the design of novel single and multiple stimuli-responsive materials that can provide controlled drug release in response a specific stimulus. These stimuli-sensitive drug nanoparticulate systems have exhibited varying degrees of substitution with enhanced in vitro/in vivo release. However, in an attempt to further increase drug release, new dual and multi-stimuli based natural polymeric nanocarriers have been investigated which respond to a mixture of two or more signals and are awaiting clinical trials. The translation of biopolymeric directed stimuli-sensitive drug delivery systems in clinic demands a thorough knowledge of its mechanism and drug release pattern in order to produce affordable and patient friendly products.
Collapse
Affiliation(s)
- Davinder Singh
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States.
| | - Yashika Sharma
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Divya Dheer
- Chitkara University School of Pharmacy, Chitkara University, Baddi 174103, Himachal Pradesh, India; Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali 140306, Punjab, India.
| | - Ravi Shankar
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|