1
|
Wang J, Yang W, Li Y, Ma X, Xie Y, Zhou G, Liu S. Dual-Temperature/pH-Sensitive Hydrogels with Excellent Strength and Toughness Crosslinked Using Three Crosslinking Methods. Gels 2024; 10:480. [PMID: 39057503 PMCID: PMC11275505 DOI: 10.3390/gels10070480] [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: 06/29/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Hydrogels are widely used as excellent drug carriers in the field of biomedicine. However, their application in medicine is limited by their poor mechanical properties and softness. To improve the mechanical properties of hydrogels, a novel triple-network amphiphilic hydrogel with three overlapping crosslinking methods using a one-pot free-radical polymerization was synthesized in this study. Temperature-sensitive and pH-sensitive monomers were incorporated into the hydrogel to confer stimulus responsiveness, making the hydrogel stimuli-responsive. The successful synthesis of the hydrogel was confirmed using techniques, such as proton nuclear magnetic resonance spectroscopy (1H NMR), Fourier-transform infrared spectroscopy (FT-IR), and X-ray diffraction (XRD). In order to compare and analyze the properties of physically crosslinked hydrogels, physically-chemically double-crosslinked hydrogels, and physically-chemically clicked triple-crosslinked hydrogels, various tests were conducted on the gels' morphology, swelling behavior, thermal stability, mechanical properties, and drug loading capacity. The results indicate that the triple-crosslinked hydrogel maintains low swelling, high mechanical strength, and good thermal stability while not significantly compromising its drug delivery capability.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shouxin Liu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi’an 710119, China; (J.W.); (W.Y.); (Y.L.); (X.M.); (Y.X.); (G.Z.)
| |
Collapse
|
2
|
Desai N, Rana D, Salave S, Benival D, Khunt D, Prajapati BG. Achieving Endo/Lysosomal Escape Using Smart Nanosystems for Efficient Cellular Delivery. Molecules 2024; 29:3131. [PMID: 38999083 PMCID: PMC11243486 DOI: 10.3390/molecules29133131] [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: 06/06/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
The delivery of therapeutic agents faces significant hurdles posed by the endo-lysosomal pathway, a bottleneck that hampers clinical effectiveness. This comprehensive review addresses the urgent need to enhance cellular delivery mechanisms to overcome these obstacles. It focuses on the potential of smart nanomaterials, delving into their unique characteristics and mechanisms in detail. Special attention is given to their ability to strategically evade endosomal entrapment, thereby enhancing therapeutic efficacy. The manuscript thoroughly examines assays crucial for understanding endosomal escape and cellular uptake dynamics. By analyzing various assessment methods, we offer nuanced insights into these investigative approaches' multifaceted aspects. We meticulously analyze the use of smart nanocarriers, exploring diverse mechanisms such as pore formation, proton sponge effects, membrane destabilization, photochemical disruption, and the strategic use of endosomal escape agents. Each mechanism's effectiveness and potential application in mitigating endosomal entrapment are scrutinized. This paper provides a critical overview of the current landscape, emphasizing the need for advanced delivery systems to navigate the complexities of cellular uptake. Importantly, it underscores the transformative role of smart nanomaterials in revolutionizing cellular delivery strategies, leading to a paradigm shift towards improved therapeutic outcomes.
Collapse
Affiliation(s)
- Nimeet Desai
- Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India;
| | - Dhwani Rana
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Derajram Benival
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Dignesh Khunt
- School of Pharmacy, Gujarat Technological University, Gandhinagar 382027, Gujarat, India
| | - Bhupendra G. Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva 384012, Gujarat, India
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| |
Collapse
|
3
|
Baljon J, Kwiatkowski AJ, Pagendarm HM, Stone PT, Kumar A, Bharti V, Schulman JA, Becker KW, Roth EW, Christov PP, Joyce S, Wilson JT. A Cancer Nanovaccine for Co-Delivery of Peptide Neoantigens and Optimized Combinations of STING and TLR4 Agonists. ACS NANO 2024; 18:6845-6862. [PMID: 38386282 PMCID: PMC10919087 DOI: 10.1021/acsnano.3c04471] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/23/2024]
Abstract
Immune checkpoint blockade (ICB) has revolutionized cancer treatment and led to complete and durable responses, but only for a minority of patients. Resistance to ICB can largely be attributed to insufficient number and/or function of antitumor CD8+ T cells in the tumor microenvironment. Neoantigen targeted cancer vaccines can activate and expand the antitumor T cell repertoire, but historically, clinical responses have been poor because immunity against peptide antigens is typically weak, resulting in insufficient activation of CD8+ cytotoxic T cells. Herein, we describe a nanoparticle vaccine platform that can overcome these barriers in several ways. First, the vaccine can be reproducibly formulated using a scalable confined impingement jet mixing method to coload a variety of physicochemically diverse peptide antigens and multiple vaccine adjuvants into pH-responsive, vesicular nanoparticles that are monodisperse and less than 100 nm in diameter. Using this approach, we encapsulated synergistically acting adjuvants, cGAMP and monophosphoryl lipid A (MPLA), into the nanocarrier to induce a robust and tailored innate immune response that increased peptide antigen immunogenicity. We found that incorporating both adjuvants into the nanovaccine synergistically enhanced expression of dendritic cell costimulatory markers, pro-inflammatory cytokine secretion, and peptide antigen cross-presentation. Additionally, the nanoparticle delivery increased lymph node accumulation and uptake of peptide antigen by dendritic cells in the draining lymph node. Consequently, nanoparticle codelivery of peptide antigen, cGAMP, and MPLA enhanced the antigen-specific CD8+ T cell response and delayed tumor growth in several mouse models. Finally, the nanoparticle platform improved the efficacy of ICB immunotherapy in a murine colon carcinoma model. This work establishes a versatile nanoparticle vaccine platform for codelivery of peptide neoantigens and synergistic adjuvants to enhance responses to cancer vaccines.
Collapse
Affiliation(s)
- Jessalyn
J. Baljon
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Alexander J. Kwiatkowski
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Hayden M. Pagendarm
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Payton T. Stone
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Amrendra Kumar
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Vijaya Bharti
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jacob A. Schulman
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Kyle W. Becker
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Eric W. Roth
- Northwestern
University Atomic and Nanoscale Characterization Experimental (NUANCE)
Center, Northwestern University, Evanston, Illinois 60208, United States
| | - Plamen P. Christov
- Vanderbilt
Institute of Chemical Biology, Vanderbilt
University Medical Center, Nashville, Tennessee 37232, United States
| | - Sebastian Joyce
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department
of Veteran Affairs Tennessee Valley Healthcare System, Nashville, Tennessee 37212, United States
- Vanderbilt
Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt
Center for Immunobiology, Vanderbilt University
Medical Center, Nashville, Tennessee 37232, United States
| | - John T. Wilson
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Vanderbilt
Institute of Chemical Biology, Vanderbilt
University Medical Center, Nashville, Tennessee 37232, United States
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt
Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt
Center for Immunobiology, Vanderbilt University
Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt-Ingram
Cancer Center, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
4
|
Niu L, Miao Y, Cao Z, Wei T, Zhu J, Li M, Bai B, Chen L, Liu N, Pan F, Zhu J, Wang C, Yang Y, Chen Q. Minimalist Nanovaccine with Optimized Amphiphilic Copolymers for Cancer Immunotherapy. ACS NANO 2024; 18:3349-3361. [PMID: 38230639 DOI: 10.1021/acsnano.3c10174] [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: 01/18/2024]
Abstract
Cancer vaccines with the ability to elicit tumor-specific immune responses have attracted significant interest in cancer immunotherapy. A key challenge for effective cancer vaccines is the spatiotemporal codelivery of antigens and adjuvants. Herein, we synthesized a copolymer library containing nine poly(ethylene glycol) methyl ether methacrylate-co-butyl methacrylate-co-2-(azepan-1-yl)ethyl methacrylate (PEGMA-co-BMA-co-C7AMA) graft copolymers with designed proportions of different components to regulate their properties. Among these polymers, C-25, with a C7AMA:BMA ratio at 1.5:1 and PEG wt % of 25%, was screened as the most effective nanovaccine carrier with enhanced ability to induce mouse bone marrow-derived dendritic cell (BMDC) maturation. Additionally, RNA-sequencing (RNA-Seq) analysis revealed that C-25 could activate dendritic cells (DCs) through multisignaling pathways to trigger potent immune effects. Then, the screened C-25 was used to encapsulate the model peptide antigen, OVA257-280, to form nanovaccine C-25/OVA257-280. It was found that the C-25/OVA257-280 nanovaccine could effectively facilitate DC maturation and antigen cross-presentation without any other additional adjuvant and exhibited excellent prophylactic efficacy in the B16F10-OVA tumor model. Moreover, in combination with antiprogrammed cell death protein-ligand 1 (anti-PD-L1), the C-25/OVA257-280 nanovaccine could significantly delay the growth of pre-existing tumors. Therefore, this work developed a minimalist nanovaccine with a simple formulation and high efficiency in activating tumor-specific immune responses, showing great potential for further application in cancer immunotherapy.
Collapse
Affiliation(s)
- Le Niu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Yu Miao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Zhiqin Cao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Ting Wei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Jiafei Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Maoyi Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Boxiong Bai
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Linfu Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Nanhui Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Feng Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Junjie Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Cheng Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| |
Collapse
|
5
|
Pagendarm HM, Stone PT, Kimmel BR, Baljon JJ, Aziz MH, Pastora LE, Hubert L, Roth EW, Almunif S, Scott EA, Wilson JT. Engineering endosomolytic nanocarriers of diverse morphologies using confined impingement jet mixing. NANOSCALE 2023; 15:16016-16029. [PMID: 37753868 PMCID: PMC10568979 DOI: 10.1039/d3nr02874g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023]
Abstract
The clinical translation of many biomolecular therapeutics has been hindered by undesirable pharmacokinetic (PK) properties, inadequate membrane permeability, poor endosomal escape and cytosolic delivery, and/or susceptibility to degradation. Overcoming these challenges merits the development of nanoscale drug carriers (nanocarriers) to improve the delivery of therapeutic cargo. Herein, we implement a flash nanoprecipitation (FNP) approach to produce nanocarriers of diverse vesicular morphologies by using various molecular weight PEG-bl-DEAEMA-co-BMA (PEG-DB) polymers. We demonstrated that FNP can produce uniform (PDI < 0.1) particles after 5 impingements, and that by varying the copolymer hydrophilic mass fraction, FNP enables access to a diverse variety of nanoarchitectures including micelles, unilamellar vesicles (polymersomes), and multi-compartment vesicles (MCVs). We synthesized a library of 2 kDa PEG block copolymers, with DEAEMA-co-BMA second block molecular weights of 3, 6, 12, 15, 20, and 30 kDa. All formulations were both pH responsive, endosomolytic, and capable of loading and cytosolically delivering small negatively charged molecules - albeit to different degrees. Using a B16.F10 melanoma model, we showcased the therapeutic potential of a lead FNP formulated PEG-DB nanocarrier, encapsulating the cyclic dinucleotide (CDN) cGAMP to activate the stimulator of interferon genes (STING) pathway in a therapeutically relevant context. Collectively, these data demonstrate that an FNP process can be used to formulate pH-responsive nanocarriers of diverse morphologies using a PEG-DB polymer system. As FNP is an industrially scalable process, these data address the critical translational challenge of producing PEG-DB nanoparticles at scale. Furthermore, the diverse morphologies produced may specialize in the delivery of distinct biomolecular cargos for other therapeutic applications, implicating the therapeutic potential of this platform in an array of disease applications.
Collapse
Affiliation(s)
- Hayden M Pagendarm
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
| | - Payton T Stone
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Blaise R Kimmel
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Jessalyn J Baljon
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
| | - Mina H Aziz
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Neuroscience, Vanderbilt University, Nashville, TN 37235, USA
| | - Lucinda E Pastora
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Lauren Hubert
- Department of Chemical Engineering, The University of Rhode Island, Kingston, RI 02881, USA
| | - Eric W Roth
- NUANCE BioCryo, Northwestern University, Evanston, IL 60208, USA
| | - Sultan Almunif
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - John T Wilson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute for Infection Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
6
|
Feng S, Xie X, Liu J, Li A, Wang Q, Guo D, Li S, Li Y, Wang Z, Guo T, Zhou J, Tang DYY, Show PL. A potential paradigm in CRISPR/Cas systems delivery: at the crossroad of microalgal gene editing and algal-mediated nanoparticles. J Nanobiotechnology 2023; 21:370. [PMID: 37817254 PMCID: PMC10563294 DOI: 10.1186/s12951-023-02139-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/03/2023] [Indexed: 10/12/2023] Open
Abstract
Microalgae as the photosynthetic organisms offer enormous promise in a variety of industries, such as the generation of high-value byproducts, biofuels, pharmaceuticals, environmental remediation, and others. With the rapid advancement of gene editing technology, CRISPR/Cas system has evolved into an effective tool that revolutionised the genetic engineering of microalgae due to its robustness, high target specificity, and programmability. However, due to the lack of robust delivery system, the efficacy of gene editing is significantly impaired, limiting its application in microalgae. Nanomaterials have become a potential delivery platform for CRISPR/Cas systems due to their advantages of precise targeting, high stability, safety, and improved immune system. Notably, algal-mediated nanoparticles (AMNPs), especially the microalgae-derived nanoparticles, are appealing as a sustainable delivery platform because of their biocompatibility and low toxicity in a homologous relationship. In addition, living microalgae demonstrated effective and regulated distribution into specified areas as the biohybrid microrobots. This review extensively summarised the uses of CRISPR/Cas systems in microalgae and the recent developments of nanoparticle-based CRISPR/Cas delivery systems. A systematic description of the properties and uses of AMNPs, microalgae-derived nanoparticles, and microalgae microrobots has also been discussed. Finally, this review highlights the challenges and future research directions for the development of gene-edited microalgae.
Collapse
Affiliation(s)
- Shuying Feng
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China.
| | - Xin Xie
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Junjie Liu
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Aifang Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Qianqian Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Dandan Guo
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Shuxuan Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Yalan Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Zilong Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Tao Guo
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China.
| | - Jin Zhou
- Institute for Ocean Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China.
| | - Doris Ying Ying Tang
- Department of Chemical and Environmental Engineering, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Malaysia
| | - Pau Loke Show
- Department of Chemical Engineering, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
7
|
Grimme CJ, Hanson MG, Reineke TM. Enhanced ASO-Mediated Gene Silencing with Lipophilic pH-Responsive Micelles. Bioconjug Chem 2023. [PMID: 37384839 DOI: 10.1021/acs.bioconjchem.3c00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Herein, we examine the ASO-mediated gene silencing efficiency of pH-responsive micelles, by incorporating 2-(diisopropylamino)ethyl methacrylate (DIP) into the micelle core and comparing physical and biological properties with non-pH-responsive micelles. Additionally, the lipophilic effect of the micelle cores was examined in both types of micelles. Varying lipophilicity was achieved by varying alkyl monomer chain lengths─butyl (4), lauryl (12), and stearyl (18) methacrylate. Each of the micelles formed within our family offered the added benefit of well-defined and uniform templates for loading antisense oligonucleotide (ASO) payloads. Overall, the micelles followed previously established trends of outperforming their linear polymer (nonmicelle) analogs and ASO only control. More specifically, the highest performing micelles were the pH-responsive micelles with longer alkyl chains or higher lipophilicity─D-DIP+LMA and D-DIP+SMA (∼90% silencing). These two micelles demonstrated silencing efficiencies similar to Jet-PEI and Lipofectamine 2000 and caused lower toxicity than Lipofectamine 2000. The shortest alkyl chain pH-responsive micelle, D-DIP+BMA (64%), displayed strong gene silencing similar to that about that of its non-pH-responsive micelle, D-BMA (68%), and the pH-responsive micelle without an alkyl chain incorporated, D-DIP (59%). This work illuminates a minimum alkyl chain length dependence to allow gene silencing within our micelle family. However, including only longer alkyl chains into the micelle core without the pH-responsive unit DIP had a hindering effect, thus demonstrating the requirement of the DIP unit when including longer alkyl chain lengths. This work demonstrates the exemplary gene silencing efficiencies of polymeric micelles and uncovers the relationship between pH responsiveness and performance with lipophilic polymer micelles for enhancing ASO-mediated gene silencing.
Collapse
Affiliation(s)
- Christian J Grimme
- Department of Chemical Engineering & Materials Science, University of Minnesota, 421 Washington Avenue SE, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
8
|
Feng W, Chittò M, Moriarty TF, Li G, Wang X. Targeted Drug Delivery Systems for Eliminating Intracellular Bacteria. Macromol Biosci 2023; 23:e2200311. [PMID: 36189899 DOI: 10.1002/mabi.202200311] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/08/2022] [Indexed: 01/19/2023]
Abstract
The intracellular survival of pathogenic bacteria requires a range of survival strategies and virulence factors. These infections are a significant clinical challenge, wherein treatment frequently fails because of poor antibiotic penetration, stability, and retention in host cells. Drug delivery systems (DDSs) are promising tools to overcome these shortcomings and enhance the efficacy of antibiotic therapy. In this review, the classification and the mechanisms of intracellular bacterial persistence are elaborated. Furthermore, the systematic design strategies applied to DDSs to eliminate intracellular bacteria are also described, and the strategies used for internalization, intracellular activation, bacterial targeting, and immune enhancement are highlighted. Finally, this overview provides guidance for constructing functionalized DDSs to effectively eliminate intracellular bacteria.
Collapse
Affiliation(s)
- Wenli Feng
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.,AO Research Institute Davos, Davos, 7270, Switzerland
| | - Marco Chittò
- AO Research Institute Davos, Davos, 7270, Switzerland
| | | | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| |
Collapse
|
9
|
Bhattacharya K, Kundu M, Das S, Samanta S, Roy SS, Mandal M, Singha NK. Glycopolymer Decorated pH-Dependent Ratiometric Fluorescent Probe Based on Förster Resonance Energy Transfer for the Detection of Cancer Cells. Macromol Rapid Commun 2023; 44:e2200594. [PMID: 36302094 DOI: 10.1002/marc.202200594] [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: 07/05/2022] [Revised: 10/17/2022] [Indexed: 01/26/2023]
Abstract
Development of fluorescent imaging probes is an important topic of research for the early diagnosis of cancer. Based on the difference between the cellular environment of tumor cells and normal cells, several "smart" fluorescent probes have been developed. In this work, a glycopolymer functionalized Förster resonance energy transfer (FRET) based fluorescent sensor is developed, which can monitor the pH change in cellular system. One-pot sequential reversible addition-fragmentation chain transfer (RAFT)polymerization technique is employed to synthesize fluorescent active triblock glycopolymer that can undergo FRET change on the variation of pH. A FRET pair, fluorescein o-acrylate (FA) and 7-amino-4-methylcoumarin (AMC) is linked via a pH-responsive polymer poly [2-(diisopropylamino)ethyl methacrylate] (PDPAEMA), which can undergo reversible swelling/deswelling under acidic/neutral condition. The presence of glycopolymer segment provides stability, water solubility, and specificity toward cancer cells. The cellular FRET experiments on cancer cells (MDA MB 231) and normal cells (3T3 fibroblast cells) demonstrate that the material is capable of distinguishing cells as a function of pH change.
Collapse
Affiliation(s)
- Koushik Bhattacharya
- Rubber Technology Centre, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Moumita Kundu
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Subhayan Das
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Sarthik Samanta
- Rubber Technology Centre, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Sib Sankar Roy
- Indian Institute of Chemical Biology, 4, Raja Subodh Chandra Mallick Rd, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Mahitosh Mandal
- School of Nano Science and Technology, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Nikhil K Singha
- Rubber Technology Centre, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India.,School of Nano Science and Technology, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| |
Collapse
|
10
|
Bellato F, Feola S, Dalla Verde G, Bellio G, Pirazzini M, Salmaso S, Caliceti P, Cerullo V, Mastrotto F. Mannosylated Polycations Target CD206 + Antigen-Presenting Cells and Mediate T-Cell-Specific Activation in Cancer Vaccination. Biomacromolecules 2022; 23:5148-5163. [PMID: 36394394 DOI: 10.1021/acs.biomac.2c00993] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunotherapy is deemed one of the most powerful therapeutic approaches to treat cancer. However, limited response and tumor specificity are still major challenges to address. Herein, mannosylated polycations targeting mannose receptor- are developed as vectors for plasmid DNA (pDNA)-based vaccines to improve selective delivery of genetic material to antigen-presenting cells and enhance immune cell activation. Three diblock glycopolycations (M15A12, M29A25, and M58A45) and two triblock copolymers (M29A29B9 and M62A52B32) are generated by using mannose (M), agmatine (A), and butyl (B) derivatives to target CD206, complex nucleic acids, and favor the endosomal escape, respectively. All glycopolycations efficiently complex pDNA at N/P ratios <5, protecting the pDNA from degradation in a physiological milieu. M58A45 and M62A52B32 complexed with plasmid encoding for antigenic ovalbumin (pOVA) trigger the immune activation of cultured dendritic cells, which present the SIINFEKL antigenic peptide via specific major histocompatibility complex-I. Importantly, administration of M58A45/pOVA elicits SIINFEKL-specific T-cell response in C56BL/6 mice bearing the melanoma tumor model B16-OVA, well in line with a reduction in tumor growth. These results qualify mannosylation as an efficient strategy to target immune cells in cancer vaccination and emphasize the potential of these glycopolycations as effective delivery vehicles for nucleic acids.
Collapse
Affiliation(s)
- Federica Bellato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| | - Sara Feola
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy, Helsinki University, Viikinkaari 5E, 00790Helsinki, Finland.,iCAN Digital Precision Cancer Medicine Flagship, FI-00014Helsinki, Finland
| | - Gloria Dalla Verde
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| | - Greta Bellio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| | - Marco Pirazzini
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131Padova, Italy
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| | - Vincenzo Cerullo
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy, Helsinki University, Viikinkaari 5E, 00790Helsinki, Finland.,iCAN Digital Precision Cancer Medicine Flagship, FI-00014Helsinki, Finland
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| |
Collapse
|
11
|
Rennick JJ, Nowell CJ, Pouton CW, Johnston APR. Resolving subcellular pH with a quantitative fluorescent lifetime biosensor. Nat Commun 2022; 13:6023. [PMID: 36224168 PMCID: PMC9556823 DOI: 10.1038/s41467-022-33348-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Changes in sub-cellular pH play a key role in metabolism, membrane transport, and triggering cargo release from therapeutic delivery systems. Most methods to measure pH rely on intensity changes of pH sensitive fluorophores, however, these measurements are hampered by high uncertainty in the inferred pH and the need for multiple fluorophores. To address this, here we combine pH dependant fluorescent lifetime imaging microscopy (pHLIM) with deep learning to accurately quantify sub-cellular pH in individual vesicles. We engineer the pH sensitive protein mApple to localise in the cytosol, endosomes, and lysosomes, and demonstrate that pHLIM can rapidly detect pH changes induced by drugs such as bafilomycin A1 and chloroquine. We also demonstrate that polyethylenimine (a common transfection reagent) does not exhibit a proton sponge effect and had no measurable impact on the pH of endocytic vesicles. pHLIM is a simple and quantitative method that will help to understand drug action and disease progression.
Collapse
Affiliation(s)
- Joshua J Rennick
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Cameron J Nowell
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
| |
Collapse
|
12
|
Xiao X, Teng F, Shi C, Chen J, Wu S, Wang B, Meng X, Essiet Imeh A, Li W. Polymeric nanoparticles—Promising carriers for cancer therapy. Front Bioeng Biotechnol 2022; 10:1024143. [PMID: 36277396 PMCID: PMC9585261 DOI: 10.3389/fbioe.2022.1024143] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/16/2022] [Indexed: 12/03/2022] Open
Abstract
Polymeric nanoparticles (NPs) play an important role in controlled cancer drug delivery. Anticancer drugs can be conjugated or encapsulated by polymeric nanocarriers, which are known as polymeric nanomedicine. Polymeric nanomedicine has shown its potential in providing sustained release of drugs with reduced cytotoxicity and modified tumor retention, but until now, few delivery systems loading drugs have been able to meet clinical demands, so more efforts are needed. This research reviews the current state of the cancer drug-loading system by exhibiting a series of published articles that highlight the novelty and functions from a variety of different architectures including micelles, liposomes, dendrimers, polymersomes, hydrogels, and metal–organic frameworks. These may contribute to the development of useful polymeric NPs to achieve different therapeutic purposes.
Collapse
Affiliation(s)
- Xiao Xiao
- School of Pharmacy, Jilin Medical University, Jilin, China
| | - Fei Teng
- School of Pharmacy, Jilin Medical University, Jilin, China
| | - Changkuo Shi
- School of Pharmacy, Jilin Medical University, Jilin, China
| | - Junyu Chen
- School of Pharmacy, Jilin Medical University, Jilin, China
| | - Shuqing Wu
- School of Pharmacy, Jilin Medical University, Jilin, China
| | - Bao Wang
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, China
| | - Xiang Meng
- School of Pharmacy, Jilin Medical University, Jilin, China
| | | | - Wenliang Li
- School of Pharmacy, Jilin Medical University, Jilin, China
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, China
- *Correspondence: Wenliang Li,
| |
Collapse
|
13
|
Wang Y, Hu D, Chang X, Zhu Y. Temperature-Driven Reversible Shape Transformation of Polymeric Nanoparticles from Emulsion Confined Coassembly of Block Copolymers and Poly( N-isopropylacrylamide). Macromolecules 2022. [DOI: 10.1021/acs.macromol.2c00893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yaping Wang
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou 311121, Zhejiang, People’s Republic of China
| | - Dengwen Hu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou 311121, Zhejiang, People’s Republic of China
| | - Xiaohua Chang
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou 311121, Zhejiang, People’s Republic of China
| | - Yutian Zhu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou 311121, Zhejiang, People’s Republic of China
| |
Collapse
|
14
|
Yan Y, Zhang G, Wu C, Ren Q, Liu X, Huang F, Cao Y, Ye W. Structural Exploration of Polycationic Nanoparticles for siRNA Delivery. ACS Biomater Sci Eng 2022; 8:1964-1974. [PMID: 35380797 DOI: 10.1021/acsbiomaterials.2c00196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
RNA interference (RNAi) is a promising approach to the treatment of genetic diseases by the specific knockdown of target genes. Functional polymers are potential vehicles for the effective delivery of vulnerable small interfering RNA (siRNA), which is required for the broad application of RNAi-based therapeutics. The development of methods for the facile modulation of chemical structures of polymeric carriers and an elucidation of detailed delivery mechanisms remain important areas of research. In this paper, we synthesized a series of methacrylate-based polymers with controllable structures and narrow distributions by atom transfer radical polymerization using various combinations of cationic monomers (2-dimethylaminoethyl methacrylate, 2-diethylaminoethyl methacrylate, and 2-dibutylaminoethyl methacrylate) and hydrophobic monomers (2-butyl methacrylate (BMA), cyclohexyl methacrylate, and 2-ethylhexyl methacrylate). These polymers exhibited varying hydrophobicities, charge densities, and pKa values, enabling the discovery of effective carriers for siRNA by in vitro delivery assays. For the polymers with BMA segments, 50% of cationic segments were beneficial to the formation of siRNA nanoparticles (NPs) and the in vitro delivery of siRNA. The optimal ratio varied for different combinations of cationic and hydrophobic segments. In particular, 20k PMB 0.5, PME 0.5, and PEB 1.0 showed >75% luciferase knockdown. Efficacious delivery was dependent on high siRNA binding, the small size of NPs, and balanced hydrophobicity and charge density. Cellular uptake and endosomal escape experiments indicated that carboxybetaine modification of 20k PMB 0.5 did not remarkably affect the internalization of corresponding NPs after incubation for 6 h but significantly reduced the endosomal escape of NPs, which leads to the notable decrease in delivery efficacy of polymers. These results provide insights into the mechanism of polymer-based siRNA delivery and may inspire the development of novel polymeric carriers.
Collapse
Affiliation(s)
- Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Guangliang Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Chengfan Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Qidi Ren
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xiaomin Liu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Fangqian Huang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yi Cao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Wenbo Ye
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
15
|
Roy P, Saha S, Chakraborty J. Looking into the possibilities of cure of the type 2 diabetes mellitus by nanoparticle-based RNAi and CRISPR-Cas9 system: A review. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
16
|
Wang Z, Liu X, Duan Y, Huang Y. Infection microenvironment-related antibacterial nanotherapeutic strategies. Biomaterials 2021; 280:121249. [PMID: 34801252 DOI: 10.1016/j.biomaterials.2021.121249] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 11/04/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022]
Abstract
The emergence and spread of antibiotic resistance is one of the biggest challenges in public health. There is an urgent need to discover novel agents against the occurrence of multidrug-resistant bacteria, such as methicillin-resistant Staphylococcus aureus and vancomycin-resistant enterococci. The drug-resistant pathogens are able to grow and persist in infected sites, including biofilms, phagosomes, or phagolysosomes, which are more difficult to eradicate than planktonic ones and also foster the development of drug resistance. For years, various nano-antibacterial agents have been developed in the forms of antibiotic nanocarriers. Inorganic nanoparticles with intrinsic antibacterial activity and inert nanoparticles assisted by external stimuli, including heat, photon, magnetism, or sound, have also been discovered. Many of these strategies are designed to target the unique microenvironment of bacterial infections, which have shown potent antibacterial effects in vitro and in vivo. This review summarizes ongoing efforts on antibacterial nanotherapeutic strategies related to bacterial infection microenvironments, including targeted antibacterial therapy and responsive antibiotic delivery systems. Several grand challenges and future directions for the development and translation of effective nano-antibacterial agents are also discussed. The development of innovative nano-antibacterial agents could provide powerful weapons against drug-resistant bacteria in systemic or local bacterial infections in the foreseeable future.
Collapse
Affiliation(s)
- Zhe Wang
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Xingyun Liu
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Yanwen Duan
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, Hunan, 410013, China; Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug Discover, Changsha, Hunan, 410011, China; National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha, Hunan, 410011, China.
| | - Yong Huang
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, Hunan, 410013, China; National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha, Hunan, 410011, China.
| |
Collapse
|
17
|
Nandi D, Shivrayan M, Gao J, Krishna J, Das R, Liu B, Thanyumanavan S, Kulkarni A. Core Hydrophobicity of Supramolecular Nanoparticles Induces NLRP3 Inflammasome Activation. ACS APPLIED MATERIALS & INTERFACES 2021; 13:45300-45314. [PMID: 34543013 PMCID: PMC8761361 DOI: 10.1021/acsami.1c14082] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Designer nanomaterials capable of delivering immunomodulators to specific immune cells have been extensively studied. However, emerging evidence suggests that several of these nanomaterials can nonspecifically activate NLRP3 inflammasomes, an intracellular multiprotein complex controlling various immune cell functions, leading to undesirable effects. To understand what nanoparticle attributes activate inflammasomes, we designed a multiparametric polymer supramolecular nanoparticle system to modulate various surface and core nanoparticle-associated molecular patterns (NAMPs), one at a time. We also investigated several underlying signaling pathways, including lysosomal rupture-cathepsin B maturation and calcium flux-mitochondrial ROS production, to gain mechanistic insights into NAMPs-mediated inflammasome activation. Here, we report that out of the four NAMPs tested, core hydrophobicity strongly activates and positively correlates with the NLRP3 assembly compared to surface charge, core rigidity, and surface hydrophobicity. Moreover, we demonstrate different signaling inclinations and kinetics followed by differential core hydrophobicity patterns with the most hydrophobic ones exhibiting both lysosomal rupture and calcium influx early on. Altogether, this study will help design the next generation of polymeric nanomaterials for specific regulation of inflammasome activation, aiding efficient immunotherapy and vaccine delivery.
Collapse
Affiliation(s)
- Dipika Nandi
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Manisha Shivrayan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Jingjing Gao
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Jithu Krishna
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Ritam Das
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Bin Liu
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - S. Thanyumanavan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| |
Collapse
|
18
|
Day RA, Sletten EM. Experimental Perspectives on Direct Visualization of Endosomal Rupture. Chembiochem 2021; 22:3277-3282. [PMID: 34519410 DOI: 10.1002/cbic.202100379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/10/2021] [Indexed: 11/05/2022]
Abstract
Endosomal escape continues to be a limiting factor in the therapeutic use of nanomaterials. Assays to visualize endosomal escape often do not decouple the endosomal/lysosomal disruption from the release of payload into the cytosol. Here, we discuss three approaches to directly probe endosomal/lysosomal rupture: calcein dye dilution, lysosome size quantification and endosome/lysosome membrane integrity visualized with a genetically engineered cell line. We apply the three assays to endosomes/lysosomes ruptured via osmotic pressure and photochemical internalization.
Collapse
Affiliation(s)
- Rachael A Day
- Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California, 90095, USA
| | - Ellen M Sletten
- Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California, 90095, USA
| |
Collapse
|
19
|
Escaping the endosome: assessing cellular trafficking mechanisms of non-viral vehicles. J Control Release 2021; 335:465-480. [PMID: 34077782 DOI: 10.1016/j.jconrel.2021.05.038] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022]
Abstract
Non-viral vehicles hold therapeutic promise in advancing the delivery of a variety of cargos in vitro and in vivo, including small molecule drugs, biologics, and especially nucleic acids. However, their efficacy at the cellular level is limited by several delivery barriers, with endolysosomal degradation being most significant. The entrapment of vehicles and their cargo in the acidified endosome prevents access to the cytosol, nucleus, and other subcellular compartments. Understanding the factors that contribute to uptake and intracellular trafficking, especially endosomal entrapment and release, is key to overcoming delivery obstacles within cells. In this review, we summarize and compare experimental techniques for assessing the extent of endosomal escape of a variety of non-viral vehicles and describe proposed escape mechanisms for different classes of lipid-, polymer-, and peptide-based delivery agents. Based on this evaluation, we present forward-looking strategies utilizing information gained from mechanistic studies to inform the rational design of efficient delivery vehicles.
Collapse
|
20
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
21
|
Richter F, Mapfumo P, Martin L, Solomun JI, Hausig F, Frietsch JJ, Ernst T, Hoeppener S, Brendel JC, Traeger A. Improved gene delivery to K-562 leukemia cells by lipoic acid modified block copolymer micelles. J Nanobiotechnology 2021; 19:70. [PMID: 33676500 PMCID: PMC7936509 DOI: 10.1186/s12951-021-00801-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/09/2021] [Indexed: 12/23/2022] Open
Abstract
Although there has been substantial progress in the research field of gene delivery, there are some challenges remaining, e.g. there are still cell types such as primary cells and suspension cells (immune cells) known to be difficult to transfect. Cationic polymers have gained increasing attention due to their ability to bind, condense and mask genetic material, being amenable to scale up and highly variable in their composition. In addition, they can be combined with further monomers exhibiting desired biological and chemical properties, such as antioxidative, pH- and redox-responsive or biocompatible features. By introduction of hydrophobic monomers, in particular as block copolymers, cationic micelles can be formed possessing an improved chance of transfection in otherwise challenging cells. In this study, the antioxidant biomolecule lipoic acid, which can also be used as crosslinker, was incorporated into the hydrophobic block of a diblock copolymer, poly{[2-(dimethylamino)ethyl methacrylate]101-b-[n-(butyl methacrylate)124-co-(lipoic acid methacrylate)22]} (P(DMAEMA101-b-[nBMA124-co-LAMA22])), synthesized by RAFT polymerization and assembled into micelles (LAMA-mic). These micelles were investigated regarding their pDNA binding, cytotoxicity mechanisms and transfection efficiency in K-562 and HEK293T cells, the former representing a difficult to transfect, suspension leukemia cell line. The LAMA-mic exhibited low cytotoxicity at applied concentrations but demonstrated superior transfection efficiency in HEK293T and especially K-562 cells. In-depth studies on the transfection mechanism revealed that transfection efficiency in K-562 cells does not depend on the specific oncogenic fusion gene BCR-ABL alone. It is independent of the cellular uptake of polymer-pDNA complexes but correlates with the endosomal escape of the LAMA-mic. A comparison of the transfection efficiency of the LAMA-mic with structurally comparable micelles without lipoic acid showed that lipoic acid is not solely responsible for the superior transfection efficiency of the LAMA-mic. More likely, a synergistic effect of the antioxidative lipoic acid and the micellar architecture was identified. Therefore, the incorporation of lipoic acid into the core of hydrophobic-cationic micelles represents a promising tailor-made transfer strategy, which can potentially be beneficial for other difficult to transfect cell types.
Collapse
Affiliation(s)
- Friederike Richter
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Prosper Mapfumo
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Liam Martin
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Jana I Solomun
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Franziska Hausig
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Jochen J Frietsch
- Klinik für Innere Medizin II, Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Thomas Ernst
- Klinik für Innere Medizin II, Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Johannes C Brendel
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany.
| |
Collapse
|
22
|
Munson MJ, O'Driscoll G, Silva AM, Lázaro-Ibáñez E, Gallud A, Wilson JT, Collén A, Esbjörner EK, Sabirsh A. A high-throughput Galectin-9 imaging assay for quantifying nanoparticle uptake, endosomal escape and functional RNA delivery. Commun Biol 2021; 4:211. [PMID: 33594247 PMCID: PMC7887203 DOI: 10.1038/s42003-021-01728-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
RNA-based therapies have great potential to treat many undruggable human diseases. However, their efficacy, in particular for mRNA, remains hampered by poor cellular delivery and limited endosomal escape. Development and optimisation of delivery vectors, such as lipid nanoparticles (LNPs), are impeded by limited screening methods to probe the intracellular processing of LNPs in sufficient detail. We have developed a high-throughput imaging-based endosomal escape assay utilising a Galectin-9 reporter and fluorescently labelled mRNA to probe correlations between nanoparticle-mediated uptake, endosomal escape frequency, and mRNA translation. Furthermore, this assay has been integrated within a screening platform for optimisation of lipid nanoparticle formulations. We show that Galectin-9 recruitment is a robust, quantitative reporter of endosomal escape events induced by different mRNA delivery nanoparticles and small molecules. We identify nanoparticles with superior escape properties and demonstrate cell line variances in endosomal escape response, highlighting the need for fine-tuning of delivery formulations for specific applications.
Collapse
Affiliation(s)
- Michael J Munson
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Gwen O'Driscoll
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Andreia M Silva
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Elisa Lázaro-Ibáñez
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Audrey Gallud
- Division of Chemical and Biomolecular Engineering, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Anna Collén
- Projects, Research and Early Development, Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Elin K Esbjörner
- Division of Chemical and Biomolecular Engineering, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Alan Sabirsh
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
23
|
Ofridam F, Tarhini M, Lebaz N, Gagnière É, Mangin D, Elaissari A. pH
‐sensitive polymers: Classification and some fine potential applications. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5230] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Fabrice Ofridam
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Mohamad Tarhini
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, ISA UMR 5280 Villeurbanne France
| | - Noureddine Lebaz
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Émilie Gagnière
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Denis Mangin
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Abdelhamid Elaissari
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, ISA UMR 5280 Villeurbanne France
| |
Collapse
|
24
|
Solomun JI, Cinar G, Mapfumo P, Richter F, Moek E, Hausig F, Martin L, Hoeppener S, Nischang I, Traeger A. Solely aqueous formulation of hydrophobic cationic polymers for efficient gene delivery. Int J Pharm 2021; 593:120080. [PMID: 33246046 DOI: 10.1016/j.ijpharm.2020.120080] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/05/2020] [Accepted: 11/08/2020] [Indexed: 01/01/2023]
Abstract
Cationic polymers are promising gene delivery vectors due to their ability to bind and protect genetic material. The introduction of hydrophobic moieties into cationic polymers can further improve the vector efficiency, but common formulations of hydrophobic polymers involve harsh conditions such as organic solvents, impairing intactness and loading efficiency of the genetic material. In this study, a mild, aqueous formulation method for the encapsulation of high amounts of genetic material is presented. A well-defined pH-responsive hydrophobic copolymer, i.e. poly((n-butylmethacrylate)-co-(methylmethacrylate)-co-(2-(dimethylamino) ethylmethacrylate)), (PBMD) was synthesized by reversible addition fragmentation chain transfer (RAFT) polymerization. Exploiting the pH-dependent solubility behavior of the polymer, stable pDNA loaded nanoparticles were prepared and characterized using analytical ultracentrifugation (AUC), cryo-transmission electron microscopy (cryo-TEM) and dynamic light scattering (DLS). This novel formulation approach showed high transfection efficiencies in HEK293T cells, while requiring 5- to 10-fold less pDNA compared to linear polyethylenimine (LPEI), in particular at short incubation times and in serum-containing media. Furthermore, the formulation was successfully adopted for siRNA and mRNA encapsulation and the commercially approved polymer Eudragit® E(PO/100). Overall, the aqueous formulation approach, accompanied by a tailor-made hydrophobic polymer and detailed physicochemical and application studies, led to improved gene delivery vectors with high potential for further applications.
Collapse
Affiliation(s)
- Jana I Solomun
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Gizem Cinar
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Prosper Mapfumo
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Friederike Richter
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Elisabeth Moek
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Franziska Hausig
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Liam Martin
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Ivo Nischang
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Anja Traeger
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany.
| |
Collapse
|
25
|
Stimuli-responsive polymersomes of poly [2-(dimethylamino) ethyl methacrylate]-b-polystyrene. Polym Bull (Berl) 2021. [DOI: 10.1007/s00289-020-03533-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
26
|
Nguyen DC, Shae D, Pagendarm HM, Becker KW, Wehbe M, Kilchrist KV, Pastora LE, Palmer CR, Seber P, Christov PP, Duvall CL, Wilson JT. Amphiphilic Polyelectrolyte Graft Copolymers Enhance the Activity of Cyclic Dinucleotide STING Agonists. Adv Healthc Mater 2021; 10:e2001056. [PMID: 33225632 PMCID: PMC7856189 DOI: 10.1002/adhm.202001056] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/08/2020] [Indexed: 11/10/2022]
Abstract
Cyclic dinucleotide (CDN) agonists of stimulator of interferon genes (STING) hold great therapeutic potential, but their activity is hindered by poor drug-like properties that restrict cytosolic bioavailability. Here, this challenge is addressed through the synthesis and evaluation of a novel series of PEGMA-co-DEAEMA-co-BMA copolymers with pH-responsive, membrane-destabilizing activity to enhance intracellular delivery of the CDN, cGAMP. Copolymers are synthesized with PEGMA of two different molecular weights (300 and 950 Da) and over a range of PEG mass fraction and polymer molecular weight, and relationships between copolymer structure, self-assembly, endosomal escape, and cGAMP activity are elucidated. A subset of polymers that self-assembled into 50-800 nm nanoparticles is identified, which can be loaded with cGAMP via a simple mixing strategy, resulting in significantly enhanced immunostimulatory activity. Increased cGAMP activity is found to be highly correlated with the capacity of carriers to enhance intracellular CDN uptake and to promote endosomal destabilization, findings that establish efficient cytosolic delivery as a criterion for CDN carriers. Additionally, it is demonstrated that a lead CDN carrier formulation can enhance STING activation in vivo in a model of intratumoral immunotherapy. Collectively, these investigations demonstrate the utility of PEGMA-co-DEAEMA-co-BMA copolymers as carriers for CDNs and potentially other cytosolically-acting drug cargo.
Collapse
Affiliation(s)
- D. Chuong Nguyen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
- The SyBBURE Searle Undergraduate Research Program, Vanderbilt University, Nashville, TN, 37235, USA
| | - Daniel Shae
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Hayden M. Pagendarm
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kyle W. Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Mohamed Wehbe
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kameron V. Kilchrist
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Lucinda E. Pastora
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Christian R. Palmer
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Pedro Seber
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Plamen P. Christov
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - John T. Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN 37232
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
27
|
Jiang X, Abedi K, Shi J. Polymeric nanoparticles for RNA delivery. REFERENCE MODULE IN MATERIALS SCIENCE AND MATERIALS ENGINEERING 2021. [PMCID: PMC8568333 DOI: 10.1016/b978-0-12-822425-0.00017-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
As exemplified by recent clinical approval of RNA drugs including the latest COVID-19 mRNA vaccines, RNA therapy has demonstrated great promise as an emerging medicine. Central to the success of RNA therapy is the delivery of RNA molecules into the right cells at the right location. While the clinical success of nanotechnology in RNA therapy has been limited to lipid-based nanoparticles currently, polymers, due to their tunability and robustness, have also evolved as a class of promising material for the delivery of various therapeutics including RNAs. This article overviews different types of polymers used in RNA delivery and the methods for the formulation of polymeric nanoparticles and highlights recent progress of polymeric nanoparticle-based RNA therapy.
Collapse
|
28
|
Jacobson ME, Becker KW, Palmer CR, Pastora LE, Fletcher RB, Collins KA, Fedorova O, Duvall CL, Pyle AM, Wilson JT. Structural Optimization of Polymeric Carriers to Enhance the Immunostimulatory Activity of Molecularly Defined RIG-I Agonists. ACS CENTRAL SCIENCE 2020; 6:2008-2022. [PMID: 33274278 PMCID: PMC7706089 DOI: 10.1021/acscentsci.0c00568] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Indexed: 05/03/2023]
Abstract
RNA ligands of retinoic acid-inducible gene I (RIG-I) hold significant promise as antiviral agents, vaccine adjuvants, and cancer immunotherapeutics, but their efficacy is hindered by inefficient intracellular delivery to the cytosol where RIG-I is localized. Here, we address this challenge through the synthesis and evaluation of a library of polymeric carriers rationally designed to promote the endosomal escape of 5'-triphosphate RNA (3pRNA) RIG-I agonists. We synthesized a series of PEG-block-(DMAEMA-co-A n MA) polymers, where A n MA is an alkyl methacrylate monomer ranging from n = 2-12 carbons, of variable composition, and examined effects of polymer structure on the intracellular delivery of 3pRNA. Through in vitro screening of 30 polymers, we identified four lead carriers (4-50, 6-40, 8-40, and 10-40, where the first number refers to the alkyl chain length and the second number refers to the percentage of hydrophobic monomer) that packaged 3pRNA into ∼100-nm-diameter particles and significantly enhanced its immunostimulatory activity in multiple cell types. In doing so, these studies also revealed an interplay between alkyl chain length and monomer composition in balancing RNA loading, pH-responsive properties, and endosomal escape, studies that establish new structure-activity relationships for polymeric delivery of 3pRNA and other nucleic acid therapeutics. Importantly, lead carriers enabled intravenous administration of 3pRNA in mice, resulting in increased RIG-I activation as measured by increased levels of IFN-α in serum and elevated expression of Ifnb1 and Cxcl10 in major clearance organs, effects that were dependent on polymer composition. Collectively, these studies have yielded novel polymeric carriers designed and optimized specifically to enhance the delivery and activity of 3pRNA with potential to advance the clinical development of RIG-I agonists.
Collapse
Affiliation(s)
- Max E. Jacobson
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Kyle W. Becker
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Christian R. Palmer
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Lucinda E. Pastora
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - R. Brock Fletcher
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Kathryn A. Collins
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Olga Fedorova
- Department
of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
| | - Craig L. Duvall
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Anna M. Pyle
- Department
of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
- Department
of Chemistry, Howard Hughes Medical Institute, Yale University, New Haven, Connecticut 06511, United States
| | - John. T. Wilson
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Vanderbilt
Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt
Center for Immunobiology, Vanderbilt University
Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt
Institute of Chemical Biology, Vanderbilt
University, Nashville, Tennessee 37232, United States
- Vanderbilt
Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt-Ingram
Cancer Center, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
29
|
Synthesis of poly(diethylaminoethyl methacrylate-co-2,2,6,6-tetramethyl-4-piperidyl methacrylate)s and their segmental motion study. Colloid Polym Sci 2020. [DOI: 10.1007/s00396-020-04717-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
30
|
Wang X, Liu G, Chen N, Wu J, Zhang J, Qian Y, Zhang L, Zhou D, Yu Y. Angiopep2-Conjugated Star-Shaped Polyprodrug Amphiphiles for Simultaneous Glioma-Targeting Therapy and MR Imaging. ACS APPLIED MATERIALS & INTERFACES 2020; 12:12143-12154. [PMID: 32078286 DOI: 10.1021/acsami.0c00509] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The development of valuable theranostic agents for overcoming the blood-brain barrier (BBB) to achieve efficient imaging-guided glioma-targeting delivery of therapeutics remains a great challenge for personalized glioma therapy. We herein developed a novel functional star-shaped polyprodrug amphiphile (denoted as CPP-2) via a combination of successive reversible addition-fragmentation chain transfer (RAFT) polymerization and click functionalization. In a diluted solution, the star amphiphile existed as structurally stable unimolecular micelles, containing hydrophobic cores conjugated with reduction-responsive camptothecin prodrugs Camptothecin (CPT) prodrug monomer (CPTM) and a tertiary amine monomer (2-(diethylamine) ethyl methacrylate, DEA) and hydrophilic oligo-(ethylene glycol) monomethyl ether methacrylat (OEGMA) outer coronas covalently decorated with dual-targeting moieties Angiopep2 (ANG) and small magnetic resonance imaging (MRI) contrast agents DOTA-Gd. In vitro and in vivo data in this study demonstrated that the ANG-modified micelles were capable of efficiently penetrating the BBB and delivering loaded cargoes such as CPT and Gd3+ contrast agents to glioma cells, leading to a considerably enhanced t1 relaxivity as well as antiglioma efficacy. Simultaneously, the targeted antiglioma efficacy and noninvasive MR imaging for a visualized therapy were realized. These collective findings augured well for the star polyprodrug amphiphiles to be utilized as a novel theranostic platform for clinical application in glioma therapy.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Guhuan Liu
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Ni Chen
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Jing Wu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Jingjing Zhang
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Yinfeng Qian
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Lei Zhang
- Department of Pharmaceutics, School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| | - Dandan Zhou
- Department of Pharmaceutics, School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| | - Yongqiang Yu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| |
Collapse
|
31
|
Yang DC, Eldredge AC, Hickey JC, Muradyan H, Guan Z. Multivalent Peptide-Functionalized Bioreducible Polymers for Cellular Delivery of Various RNAs. Biomacromolecules 2020; 21:1613-1624. [DOI: 10.1021/acs.biomac.0c00211] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Dong-Chu Yang
- Department of Chemistry, University of California, 1102 Natural
Sciences 2, Irvine, California 92697-2025, United States
| | - Alexander C. Eldredge
- Department of Chemistry, University of California, 1102 Natural
Sciences 2, Irvine, California 92697-2025, United States
| | - James C. Hickey
- Department of Chemistry, University of California, 1102 Natural
Sciences 2, Irvine, California 92697-2025, United States
| | - Hurik Muradyan
- Department of Chemistry, University of California, 1102 Natural
Sciences 2, Irvine, California 92697-2025, United States
| | - Zhibin Guan
- Department of Chemistry, University of California, 1102 Natural
Sciences 2, Irvine, California 92697-2025, United States
| |
Collapse
|
32
|
Jiang Y, Lu Q, Wang Y, Xu E, Ho A, Singh P, Wang Y, Jiang Z, Yang F, Tietjen GT, Cresswell P, Saltzman WM. Quantitating Endosomal Escape of a Library of Polymers for mRNA Delivery. NANO LETTERS 2020; 20:1117-1123. [PMID: 32003222 PMCID: PMC7195212 DOI: 10.1021/acs.nanolett.9b04426] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Endosomal escape is a key step for intracellular drug delivery of nucleic acids, but reliable and sensitive methods for its quantitation remain an unmet need. In order to rationally optimize the mRNA transfection efficiency of a library of polymeric materials, we designed a deactivated Renilla luciferase-derived molecular probe whose activity can be restored only in the cytosol. This probe can be coencapsulated with mRNA in the same delivery vehicle, thereby accurately measuring its endosomal escape efficiency. We examined a library of poly(amine-co-ester) (PACE) polymers with different end groups using this probe and observed a strong correlation between endosomal escape and transfection efficiency (R2 = 0.9334). In addition, we found that mRNA encapsulation efficiency and endosomal escape, but not uptake, were determinant factors for transfection efficiency. The polymers with high endosomal escape/transfection efficiency in vitro also showed good transfection efficiency in vivo, and mRNA expression was primarily observed in spleens after intravenous delivery. Together, our study suggests that the luciferase probe can be used as an effective tool to quantitate endosomal escape, which is essential for rational optimization of intracellular drug delivery systems.
Collapse
Affiliation(s)
- Yuhang Jiang
- Department of Biomedical Engineering , Yale University , New Haven , Connecticut 06511 , United States
| | - Qiao Lu
- Department of Immunobiology , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Yongheng Wang
- Department of Biomedical Engineering , Yale University , New Haven , Connecticut 06511 , United States
| | - Emily Xu
- Department of Biomedical Engineering , Yale University , New Haven , Connecticut 06511 , United States
| | - Alison Ho
- Department of Biomedical Engineering , Yale University , New Haven , Connecticut 06511 , United States
| | - Priya Singh
- Department of Biomedical Engineering , Yale University , New Haven , Connecticut 06511 , United States
| | - Yifei Wang
- Department of Biomedical Engineering , Yale University , New Haven , Connecticut 06511 , United States
| | - Zhaozhong Jiang
- Department of Biomedical Engineering , Yale University , New Haven , Connecticut 06511 , United States
| | - Fan Yang
- Department of Biomedical Engineering , Yale University , New Haven , Connecticut 06511 , United States
| | - Gregory T Tietjen
- Department of Biomedical Engineering , Yale University , New Haven , Connecticut 06511 , United States
- Department of Surgery , Yale School of Medicine , New Haven , Connecticut 06520 , United States
| | - Peter Cresswell
- Department of Immunobiology , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - W Mark Saltzman
- Department of Biomedical Engineering , Yale University , New Haven , Connecticut 06511 , United States
- Department of Chemical & Environmental Engineering , Yale University , New Haven , Connecticut 06511 , United States
- Department of Cellular & Molecular Physiology , Yale School of Medicine , New Haven , Connecticut 06510 , United States
| |
Collapse
|
33
|
Hou Z, Ren M, Wang K, Yang Y, Xu J, Zhu J. Deformable Block Copolymer Microparticles by Controllable Localization of pH-Responsive Nanoparticles. Macromolecules 2019. [DOI: 10.1021/acs.macromol.9b01936] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Zaiyan Hou
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage, Ministry of Education (HUST), School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Min Ren
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage, Ministry of Education (HUST), School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Ke Wang
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage, Ministry of Education (HUST), School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Yi Yang
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage, Ministry of Education (HUST), School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Jiangping Xu
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage, Ministry of Education (HUST), School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Jintao Zhu
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage, Ministry of Education (HUST), School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| |
Collapse
|
34
|
Ibrahim A, Twizeyimana E, Lu N, Ke W, Mukerabigwi JF, Mohammed F, Japir AAWMM, Ge Z. Reduction-Responsive Polymer Prodrug Micelles with Enhanced Endosomal Escape Capability for Efficient Intracellular Translocation and Drug Release. ACS APPLIED BIO MATERIALS 2019; 2:5099-5109. [DOI: 10.1021/acsabm.9b00769] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Alhadi Ibrahim
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Etienne Twizeyimana
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Nannan Lu
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
- Department of Oncology, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Wendong Ke
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Jean Felix Mukerabigwi
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
- Department of Applied Chemistry, College of Science and Technology, University of Rwanda, Kigali, Rwanda
| | - Fathelrahman Mohammed
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Abd Al-Wali Mohammed M. Japir
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Zhishen Ge
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| |
Collapse
|
35
|
Ulkoski D, Bak A, Wilson JT, Krishnamurthy VR. Recent advances in polymeric materials for the delivery of RNA therapeutics. Expert Opin Drug Deliv 2019; 16:1149-1167. [PMID: 31498013 DOI: 10.1080/17425247.2019.1663822] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: The delivery of nucleic acid therapeutics through non-viral carriers face multiple biological barriers that reduce their therapeutic efficiency. Despite great progress, there remains a significant technological gap that continues to limit clinical translation of these nanocarriers. A number of polymeric materials are being exploited to efficiently deliver nucleic acids and achieve therapeutic effects. Areas covered: We discuss the recent advances in the polymeric materials for the delivery of nucleic acid therapeutics. We examine the use of common polymer architectures and highlight the challenges that exist for their development from bench side to clinic. We also provide an overview of the most notable improvements made to circumvent such challenges, including structural modification and stimuli-responsive approaches, for safe and effective nucleic acid delivery. Expert opinion: It has become apparent that a universal carrier that follows 'one-size' fits all model cannot be expected for delivery of all nucleic acid therapeutics. Carriers need to be designed to exhibit sensitivity and specificity toward individual targets diseases/indications, and relevant subcellular compartments, each of which possess their own unique challenges. The ability to devise synthetic methods that control the molecular architecture enables the future development that allow for the construction of 'intelligent' designs.
Collapse
Affiliation(s)
- David Ulkoski
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca , Boston , USA
| | - Annette Bak
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca , Gothenburg , Sweden
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University , Nashville , TN , USA
| | | |
Collapse
|
36
|
Shae D, Baljon JJ, Wehbe M, Becker KW, Sheehy TL, Wilson JT. At the bench: Engineering the next generation of cancer vaccines. J Leukoc Biol 2019; 108:1435-1453. [PMID: 31430398 DOI: 10.1002/jlb.5bt0119-016r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 06/29/2019] [Accepted: 07/25/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer vaccines hold promise as an immunotherapeutic modality based on their potential to generate tumor antigen-specific T cell responses and long-lived antitumor responses capable of combating metastatic disease and recurrence. However, cancer vaccines have historically failed to deliver significant therapeutic benefit in the clinic, which we maintain is due in part to drug delivery challenges that have limited vaccine immunogenicity and efficacy. In this review, we examine some of the known and putative failure mechanisms of common first-generation clinical cancer vaccines, and describe how the rational design of materials engineered for vaccine delivery and immunomodulation can address these shortcomings. First, we outline vaccine design principles for augmenting cellular immunity to tumor antigens and describe how well-engineered materials can improve vaccine efficacy, highlighting recent innovations in vaccine delivery technology that are primed for integration into neoantigen vaccine development pipelines. We also discuss the importance of sequencing, timing, and kinetics in mounting effective immune responses to cancer vaccines, and highlight examples of materials that potentiate antitumor immunity through spatiotemporal control of immunomodulation. Furthermore, we describe several engineering strategies for improving outcomes of in situ cancer vaccines, which leverage local, intratumoral delivery to stimulate systemic immunity. Finally, we highlight recent innovations leveraging nanotechnology for increasing the immunogenicity of the tumor microenvironment (TME), which is critical to enhancing tumor infiltration and function of T cells elicited in response to cancer vaccines. These immunoengineering strategies and tools complement ongoing advances in cancer vaccines as they reemerge as an important component of the immunotherapeutic armamentarium.
Collapse
Affiliation(s)
- Daniel Shae
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Jessalyn J Baljon
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Mohamed Wehbe
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Kyle W Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Taylor L Sheehy
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - John Tanner Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
37
|
Chu PY, Tsai SC, Ko HY, Wu CC, Lin YH. Co-Delivery of Natural Compounds with a Dual-Targeted Nanoparticle Delivery System for Improving Synergistic Therapy in an Orthotopic Tumor Model. ACS APPLIED MATERIALS & INTERFACES 2019; 11:23880-23892. [PMID: 31192580 DOI: 10.1021/acsami.9b06155] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Various natural compounds including epigallocatechin gallate (EGCG) and curcumin (CU) have potential in developing anticancer therapy. However, their clinical use is commonly limited by instability and low tissue distribution. EGCG and CU combined treatment can improve the efficacy with synergistic effects. To improve the synergistic effect and overcome the limitations of low tissue distribution, we applied a dual cancer-targeted nanoparticle system to co-deliver EGCG and CU. Nanoparticles were composed of hyaluronic acid, fucoidan, and poly(ethylene glycol)-gelatin to encapsulate EGCG and CU. Furthermore, a dual targeting system was established with hyaluronic acid and fucoidan, which were used as agents for targeting CD44 on prostate cancer cells and P-selectin in tumor vasculature, respectively. Their effect and efficacy were investigated in prostate cancer cells and a orthotopic prostate tumor model. The EGCG/CU-loaded nanoparticles bound to prostate cancer cells, which were uptaken more into cells, leading to a better anticancer efficiency compared to the EGCG/CU combination solution. In addition, the releases of EGCG and CU were regulated by their pH value that avoided the premature release. In mice, treatment of the cancer-targeted EGCG/CU-loaded nanoparticles significantly attenuated the orthotopic tumor growth without inducing organ injuries. Overall, the dual-targeted nanoparticle system for the co-delivery of EGCG and CU greatly improved its synergistic effect in cancer therapy, indicating its great potential in developing treatments for prostate cancer therapy.
Collapse
|
38
|
Baljon JJ, Dandy A, Wang-Bishop L, Wehbe M, Jacobson ME, Wilson JT. The efficiency of cytosolic drug delivery using pH-responsive endosomolytic polymers does not correlate with activation of the NLRP3 inflammasome. Biomater Sci 2019; 7:1888-1897. [PMID: 30843539 PMCID: PMC6478565 DOI: 10.1039/c8bm01643g] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Inefficient cytosolic delivery has limited the development of many promising biomacromolecular drugs, a long-standing challenge that has prompted extensive development of drug carriers that facilitate endosomal escape. Although many such carriers have shown considerable promise for cytosolic delivery of a diversity of therapeutics, the rupture or destabilization of endo/lysosomal membranes has also been associated with activation of the inflammasome with attendant risk of inflammation and toxicity. In this study, we investigated relationships between pH-dependent membrane destabilization, cytosolic drug delivery, and inflammasome activation using a series of well-defined poly[(ethylene glycol)-block-[(2-(dimethylamino)ethyl methacrylate)-co-(butyl methacrylate)] copolymers of variable second block composition and pH-responsive properties. We found that polymers that demonstrated the most potent membrane-destabilizing activity at early endosomal pH values in an erythrocyte hemolysis assay were most efficient at delivery of siRNA, yet tended to be associated with the least amount of NOD-like related protein 3 (NLRP3) inflammasome activation. By contrast, polymers that displayed minimal hemolysis activity and poor siRNA knockdown, and instead mediated lysosomal rupture likely due to a proton sponge mechanism, strongly induced NLPR3 inflammasome activation in a caspase- and cathepsin-dependent manner. Collectively, these findings reinforce the importance of early endosomal escape in minimizing inflammasome activation and also demonstrate the ability to tune the degree inflammasome activation via control of polymer structure with potential implications for design of vaccine adjuvants and immunotherapeutics.
Collapse
|
39
|
Lee J, Ku KH, Park CH, Lee YJ, Yun H, Kim BJ. Shape and Color Switchable Block Copolymer Particles by Temperature and pH Dual Responses. ACS NANO 2019; 13:4230-4237. [PMID: 30856312 DOI: 10.1021/acsnano.8b09276] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Herein, we report a simple and robust strategy for preparing dual-responsive shape-switchable block copolymer (BCP) particles, which respond to subtle temperature and pH changes near physiological conditions (i.e., human body temperature and neutral pH). The shape transition of polystyrene- b-poly(4-vinylpyridine) BCP particles between lens and football shapes occurs in very narrow temperature and pH ranges: no temperature-based transition for pH 6.0, 40-50 °C transition for pH 6.5, and 25-35 °C for pH 7.0. To achieve these shape transitions, temperature/pH-responsive polymer surfactants of poly( N-(2-(diethylamino)ethyl)acrylamide- r- N-isopropylacrylamide) are designed to induce dramatic changes in relative solubility and their location in response to temperature and pH changes near physiological conditions. In addition, the BCP particles exhibit reversible shape-transforming behavior according to orthogonal temperature and pH changes. Colorimetric measurements of temperature and pH changes are enabled by shape-transforming properties combined with selective positioning of dyes, suggesting promising potential for these particles in clinical and biomedical applications.
Collapse
Affiliation(s)
- Junhyuk Lee
- Department of Chemical and Biomolecular Engineering , Korea Advanced Institute of Science and Technology , Daejeon 34141 , Republic of Korea
| | - Kang Hee Ku
- Department of Chemical and Biomolecular Engineering , Korea Advanced Institute of Science and Technology , Daejeon 34141 , Republic of Korea
| | - Chan Ho Park
- Department of Chemical and Biomolecular Engineering , Korea Advanced Institute of Science and Technology , Daejeon 34141 , Republic of Korea
| | - Young Jun Lee
- Department of Chemical and Biomolecular Engineering , Korea Advanced Institute of Science and Technology , Daejeon 34141 , Republic of Korea
| | - Hongseok Yun
- Department of Chemical and Biomolecular Engineering , Korea Advanced Institute of Science and Technology , Daejeon 34141 , Republic of Korea
| | - Bumjoon J Kim
- Department of Chemical and Biomolecular Engineering , Korea Advanced Institute of Science and Technology , Daejeon 34141 , Republic of Korea
| |
Collapse
|
40
|
Raza A, Rasheed T, Nabeel F, Hayat U, Bilal M, Iqbal HMN. Endogenous and Exogenous Stimuli-Responsive Drug Delivery Systems for Programmed Site-Specific Release. Molecules 2019; 24:1117. [PMID: 30901827 PMCID: PMC6470858 DOI: 10.3390/molecules24061117] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 02/05/2023] Open
Abstract
In this study, we reviewed state-of-the-art endogenous-based and exogenous-based stimuli-responsive drug delivery systems (DDS) for programmed site-specific release to overcome the drawbacks of conventional therapeutic modalities. This particular work focuses on the smart chemistry and mechanism of action aspects of several types of stimuli-responsive polymeric carriers that play a crucial role in extracellular and intracellular sections of diseased tissues or cells. With ever increasing scientific knowledge and awareness, research is underway around the globe to design new types of stimuli (external/internal) responsive polymeric carriers for biotechnological applications at large and biomedical and/or pharmaceutical applications, in particular. Both external/internal and even dual/multi-responsive behavior of polymeric carriers is considered an essential element of engineering so-called 'smart' DDS, which controls the effective and efficient dose loading, sustained release, individual variability, and targeted permeability in a sophisticated manner. So far, an array of DDS has been proposed, developed, and implemented. For instance, redox, pH, temperature, photo/light, magnetic, ultrasound, and electrical responsive DDS and/or all in all dual/dual/multi-responsive DDS (combination or two or more from any of the above). Despite the massive advancement in DDS arena, there are still many challenging concerns that remain to be addressed to cover the research gap. In this context, herein, an effort has been made to highlight those concerning issues to cover up the literature gap. Thus, the emphasis was given to the drug release mechanism and applications of endogenous and exogenous based stimuli-responsive DDS in the clinical settings.
Collapse
Affiliation(s)
- Ali Raza
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Tahir Rasheed
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Faran Nabeel
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Uzma Hayat
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey CP 64849, Mexico.
| |
Collapse
|
41
|
Affiliation(s)
- Jeffrey A Hubbell
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA.
| | - Melody A Swartz
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
42
|
Shae D, Becker KW, Christov P, Yun DS, Lytton-Jean AKR, Sevimli S, Ascano M, Kelley M, Johnson DB, Balko JM, Wilson JT. Endosomolytic polymersomes increase the activity of cyclic dinucleotide STING agonists to enhance cancer immunotherapy. NATURE NANOTECHNOLOGY 2019; 14:269-278. [PMID: 30664751 PMCID: PMC6402974 DOI: 10.1038/s41565-018-0342-5] [Citation(s) in RCA: 438] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/26/2018] [Indexed: 05/17/2023]
Abstract
Cyclic dinucleotide (CDN) agonists of stimulator of interferon genes (STING) are a promising class of immunotherapeutics that activate innate immunity to increase tumour immunogenicity. However, the efficacy of CDNs is limited by drug delivery barriers, including poor cellular targeting, rapid clearance and inefficient transport to the cytosol where STING is localized. Here, we describe STING-activating nanoparticles (STING-NPs)-rationally designed polymersomes for enhanced cytosolic delivery of the endogenous CDN ligand for STING, 2'3' cyclic guanosine monophosphate-adenosine monophosphate (cGAMP). STING-NPs increase the biological potency of cGAMP, enhance STING signalling in the tumour microenvironment and sentinel lymph node, and convert immunosuppressive tumours to immunogenic, tumoricidal microenvironments. This leads to enhanced therapeutic efficacy of cGAMP, inhibition of tumour growth, increased rates of long-term survival, improved response to immune checkpoint blockade and induction of immunological memory that protects against tumour rechallenge. We validate STING-NPs in freshly isolated human melanoma tissue, highlighting their potential to improve clinical outcomes of immunotherapy.
Collapse
Affiliation(s)
- Daniel Shae
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kyle W Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Plamen Christov
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - Dong Soo Yun
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Abigail K R Lytton-Jean
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sema Sevimli
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Manuel Ascano
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mark Kelley
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Douglas B Johnson
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Justin M Balko
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
43
|
Cupic KI, Rennick JJ, Johnston APR, Such GK. Controlling endosomal escape using nanoparticle composition: current progress and future perspectives. Nanomedicine (Lond) 2019; 14:215-223. [DOI: 10.2217/nnm-2018-0326] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Polymer nanoparticles offer significant benefits for improving delivery of biological therapeutics such as DNA and proteins, as they allow the cargo to be protected until it is delivered to a target cell. However, there are still challenges with achieving efficient delivery to the optimal cellular region. One significant roadblock is escape of nanoparticles from within the endosomal/lysosomal compartments into the cytosol. Here, we review the recent advances in understanding endosomal escape of polymer nanoparticles. We also discuss the current progress on investigating how nanoparticle structure can control endosomal escape. It is important to understand the fundamental biological processes that govern endosomal escape in order to design more effective therapeutic delivery systems.
Collapse
Affiliation(s)
- Kristofer I Cupic
- The School of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Joshua J Rennick
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Angus PR Johnston
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Georgina K Such
- The School of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
44
|
Tang H, Zhao W, Yu J, Li Y, Zhao C. Recent Development of pH-Responsive Polymers for Cancer Nanomedicine. Molecules 2018; 24:E4. [PMID: 30577475 PMCID: PMC6337262 DOI: 10.3390/molecules24010004] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer remains a leading cause of death worldwide with more than 10 million new cases every year. Tumor-targeted nanomedicines have shown substantial improvements of the therapeutic index of anticancer agents, addressing the deficiencies of conventional chemotherapy, and have had a tremendous growth over past several decades. Due to the pathophysiological characteristics that almost all tumor tissues have lower pH in comparison to normal healthy tissues, among various tumor-targeted nanomaterials, pH-responsive polymeric materials have been one of the most prevalent approaches for cancer diagnosis and treatment. In this review, we summarized the types of pH-responsive polymers, describing their chemical structures and pH-response mechanisms; we illustrated the structure-property relationships of pH-responsive polymers and introduced the approaches to regulating their pH-responsive behaviors; we also highlighted the most representative applications of pH-responsive polymers in cancer imaging and therapy. This review article aims to provide general guidelines for the rational design of more effective pH-responsive nanomaterials for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Houliang Tang
- Department of Chemistry, Southern Methodist University, 3215 Daniel Avenue, Dallas, TX 75275, USA.
| | - Weilong Zhao
- Global Research IT, Merck & Co., Inc., Boston, MA 02210, USA.
| | - Jinming Yu
- Department of Chemical and Biological Engineering, the University of Alabama, Tuscaloosa, AL 35487, USA.
| | - Yang Li
- Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| | - Chao Zhao
- Department of Chemical and Biological Engineering, the University of Alabama, Tuscaloosa, AL 35487, USA.
| |
Collapse
|
45
|
Smith SA, Selby LI, Johnston APR, Such GK. The Endosomal Escape of Nanoparticles: Toward More Efficient Cellular Delivery. Bioconjug Chem 2018; 30:263-272. [PMID: 30452233 DOI: 10.1021/acs.bioconjchem.8b00732] [Citation(s) in RCA: 408] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Many emerging therapies rely on the delivery of biological cargo into the cytosol. Nanoparticle delivery systems hold great potential to deliver these therapeutics but are hindered by entrapment and subsequent degradation in acidic compartments of the endo/lysosomal pathway. Engineering polymeric delivery systems that are able to escape the endosome has significant potential to address this issue. However, the development of safe and effective delivery systems that can reliably deliver cargo to the cytosol is still a challenge. Greater understanding of the properties that govern endosomal escape and how it can be quantified is important for the development of more efficient nanoparticle delivery systems. This Topical Review highlights the current understanding of the mechanisms by which nanoparticles escape the endosome, and the emerging techniques to improve the quantification of endosomal escape.
Collapse
Affiliation(s)
- Samuel A Smith
- The School of Chemistry , The University of Melbourne , Parkville , Victoria , Australia , 3010
| | - Laura I Selby
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences , Monash University , Parkville , Victoria , Australia , 3052
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences , Monash University , Parkville , Victoria , Australia , 3052
| | - Georgina K Such
- The School of Chemistry , The University of Melbourne , Parkville , Victoria , Australia , 3010
| |
Collapse
|
46
|
Ferreira Soares DC, Oda CMR, Monteiro LOF, de Barros ALB, Tebaldi ML. Responsive polymer conjugates for drug delivery applications: recent advances in bioconjugation methodologies. J Drug Target 2018; 27:355-366. [PMID: 30010436 DOI: 10.1080/1061186x.2018.1499747] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | - Caroline Mari Ramos Oda
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Andre Luis Branco de Barros
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
47
|
Słyk E, Roth R, Bryk P. Microscopic density functional theory for monolayers of diblock copolymers. J Chem Phys 2018; 149:064902. [PMID: 30111154 DOI: 10.1063/1.5039522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We propose density functional theory for diblock copolymers in two dimensions. Our theoretical framework is based on Wertheim's first order thermodynamic perturbation theory. Using the proposed approach, we investigate the structure and phase behavior of monolayers of symmetric diblock copolymers. We find that the phase behavior of symmetric diblock copolymer monolayers is similar to that in 3D. This includes the scaling of the equilibrium lamellar width with chain length. We find that the topology of the resulting phase diagrams depends on the chain length and the unlike segment interaction incompatibility and involves either one, two, or three triple points (one of them being the peritectic point). We expect that a similar phase behavior could be obtained for monolayers of colloidal suspensions with carefully tuned interparticle interactions.
Collapse
Affiliation(s)
- Edyta Słyk
- Department for the Modeling of Physico-Chemical Processes, Maria Curie-Skłodowska University, 20-031 Lublin, Poland
| | - Roland Roth
- Institute for Theoretical Physics, University of Tübingen, Auf der Morgenstelle 14, 72076 Tübingen, Germany
| | - Paweł Bryk
- Department for the Modeling of Physico-Chemical Processes, Maria Curie-Skłodowska University, 20-031 Lublin, Poland
| |
Collapse
|
48
|
Cohen N, Ziv E, Vasilyev G, Convertine A, Levi-Kalisman Y, Yerushalmi-Rozen R. Micellization of a di-block copolymer in ethylene glycol and its utilization for suspension of carbonaceous nanostructures. J Appl Polym Sci 2018. [DOI: 10.1002/app.46518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Neta Cohen
- Department of Chemical Engineering; The Ben-Gurion University of the Negev; Beer-Sheva 84105 Israel
| | - Efrat Ziv
- Department of Chemical Engineering; The Ben-Gurion University of the Negev; Beer-Sheva 84105 Israel
| | - Gleb Vasilyev
- NanoEngineering Group, Faculty of Mechanical Engineering; Technion-Israel Institute of Technology; Haifa 32000 Israel
| | - Anthony Convertine
- Department of Bioengineering; University of Washington; Seattle Washington 98195
| | - Yael Levi-Kalisman
- The Center for Nanoscience and Nanotechnology; The Hebrew University of Jerusalem; Jerusalem 91904 Israel
- The Institute of Life Sciences; The Hebrew University of Jerusalem; Jerusalem 91904 Israel
| | - Rachel Yerushalmi-Rozen
- Department of Chemical Engineering; The Ben-Gurion University of the Negev; Beer-Sheva 84105 Israel
- The Ilse Katz Institute for Nanoscience and Technology; The Ben-Gurion University of the Negev; Beer-Sheva 84105 Israel
| |
Collapse
|
49
|
Su FY, Chen J, Son HN, Kelly AM, Convertine AJ, Ratner DM, Stayton PS. Polymer-augmented liposomes enhancing antibiotic delivery against intracellular infections. Biomater Sci 2018; 6:1976-1985. [PMID: 29850694 PMCID: PMC6195317 DOI: 10.1039/c8bm00282g] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pulmonary intracellular infections, such as tuberculosis, anthrax, and tularemia, have remained a significant challenge to conventional antibiotic therapy. Ineffective antibiotic treatment of these infections can lead not only to undesired side effects, but also to the emergence of antibiotic resistance. Aminoglycosides (e.g., streptomycin) have long been part of the therapeutic regiment for many pulmonary intracellular infections. Their bioavailability for intracellular bacterial pools, however, is limited by poor membrane permeability and rapid elimination. To address this challenge, polymer-augmented liposomes (PALs) were developed to provide improved cytosolic delivery of streptomycin to alveolar macrophages, an important host cell for intracellular pathogens. A multifunctional diblock copolymer was engineered to functionalize PALs with carbohydrate-mediated targeting, pH-responsive drug release, and endosomal release activity with a single functional polymer that replaces the pegylated lipid component to simplify the liposome formulation. The pH-sensing functionality enabled PALs to provide enhanced release of streptomycin under endosomal pH conditions (70% release in 6 hours) with limited release at physiological pH 7.4 (16%). The membrane-destabilizing activity connected to endosomal release was characterized in a hemolysis assay and PALs displayed a sharp pH profile across the endosomal pH development target range. The direct connection of this membrane-destabilizing pH profile to model drug release was demonstrated in an established pyranine/p-xylene bispyridinium dibromide (DPX) fluorescence dequenching assay. PALs displayed similar sharp pH-responsive release, whereas PEGylated control liposomes did not, and similar profiles were then shown for streptomycin release. The mannose-targeting capability of the PALs was also demonstrated with 2.5 times higher internalization compared to non-targeted PEGylated liposomes. Finally, the streptomycin-loaded PALs were shown to have a significantly improved intracellular antibacterial activity in a Francisella-macrophage co-culture model, compared with free streptomycin or streptomycin delivered by control PEGylated liposomes (13× and 16×, respectively). This study suggests the potential of PALs as a useful platform to deliver antibiotics for the treatment of intracellular macrophage infections.
Collapse
Affiliation(s)
- Fang-Yi Su
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Jasmin Chen
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Hye-Nam Son
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Abby M. Kelly
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | | | - Daniel M. Ratner
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Patrick S. Stayton
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
50
|
Shi XJ, Qiu YY, Yu H, Liu C, Yuan YX, Yin PH, Liu T. Increasing the anticancer performance of bufalin (BUF) by introducing an endosome-escaping polymer and tumor-targeting peptide in the design of a polymeric prodrug. Colloids Surf B Biointerfaces 2018; 166:224-234. [PMID: 29602078 DOI: 10.1016/j.colsurfb.2018.03.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 03/15/2018] [Accepted: 03/17/2018] [Indexed: 12/26/2022]
Abstract
A well-defined multifunctional brush-type polymeric prodrug covalently linked with an anticancer drug (bufalin, BUF), a tumor-targeting peptide (RGD), and an endosome-escaping polymer, poly(N,N-diethylaminoethyl methacrylate-co-butyl methacrylate (P(DEA-co-BMA)), was developed. Its anticancer performance against colon cancer was investigated in vitro and in vivo. Reversible addition-fragmentation transfer (RAFT) polymerization of oligo(ethylene glycol) monomethyl ether methacrylate (OEGMA), 2-((3-(tert-butoxy)-3-oxopropyl)thio)ethyl methacrylate (BSTMA), and 2-(2-bromoisobutyryloxy)ethylmethacrylate (BIEM) afforded the multifunctional random copolymer, P(OEGMA-co-BSTMA-co-BIEM), in which hydrophilic POEGMA can stabilize nanoparticles in water, PBSTMA can be converted into carboxyl groups, and PBIEM can be employed as a macromolecular atom radical transfer polymerization (ATRP) initiator. The ATRP of DEA and BMA using P(OEGMA-co-BSTMA-co-BIEM) as a macromolecular ATRP initiator led to the formation of the pH-responsive brush-type copolymer, P(OEGMA-co-BSTMA)-g-P(DEA-co- BMA). After hydrolysis by trifluoroacetic acid and post-functionalization the final polymeric prodrug, P(OEGMA-co-BUF-co-RGD)-g-P(DEA-co-BMA), was obtained with a drug content of ∼7.8 wt%. P(OEGMA-co-BUF-co-RGD)-g-P(DEA-co-BMA) can be assembled into nanoparticles (BUF- NP-RGD) in aqueous solution with a diameter of 148.4 ± 0.7 nm and a zeta potential of -7.6 ± 0.4 mV. BUF-NP-RGD exhibited controlled drug release in the presence of esterase. Additionally, P(OEGMA-co- BSMA)-g-P(DEA-co-BMA) showed a significant hemolysis effect at a pH comparable to that of endosomes/lysosomes. Cell viability and a tumor-bearing nude mouse model were employed to evaluate the anticancer efficacy of BUF-NP-RGD. It was revealed that BUF-NP-RGD showed improved anticancer performance compared with that of free BUF both in vitro and in vivo. Histological and immunochemical analysis further demonstrated that BUF-NP-RGD exhibited improved cell apoptosis, angiogenesis inhibition, and an anti-proliferation effect.
Collapse
Affiliation(s)
- Xiao-Jing Shi
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Yan-Yan Qiu
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Hui Yu
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Cheng Liu
- Centralab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Yu-Xia Yuan
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Pei-Hao Yin
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Tao Liu
- Centralab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| |
Collapse
|