1
|
Han J, Sheng T, Zhang Y, Cheng H, Gao J, Yu J, Gu Z. Bioresponsive Immunotherapeutic Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2209778. [PMID: 36639983 DOI: 10.1002/adma.202209778] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/31/2022] [Indexed: 06/17/2023]
Abstract
The human immune system is an interaction network of biological processes, and its dysfunction is closely associated with a wide array of diseases, such as cancer, infectious diseases, tissue damage, and autoimmune diseases. Manipulation of the immune response network in a desired and controlled fashion has been regarded as a promising strategy for maximizing immunotherapeutic efficacy and minimizing side effects. Integration of "smart" bioresponsive materials with immunoactive agents including small molecules, biomacromolecules, and cells can achieve on-demand release of agents at targeted sites to reduce overdose-related toxicity and alleviate off-target effects. This review highlights the design principles of bioresponsive immunotherapeutic materials and discusses the critical roles of controlled release of immunoactive agents from bioresponsive materials in recruiting, housing, and manipulating immune cells for evoking desired immune responses. Challenges and future directions from the perspective of clinical translation are also discussed.
Collapse
Affiliation(s)
- Jinpeng Han
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Tao Sheng
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuqi Zhang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hao Cheng
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Jianqing Gao
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Jicheng Yu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Zhen Gu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
2
|
Xie W, Xu Z. (Nano)biotechnological approaches in the treatment of cervical cancer: integration of engineering and biology. Front Immunol 2024; 15:1461894. [PMID: 39346915 PMCID: PMC11427397 DOI: 10.3389/fimmu.2024.1461894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/08/2024] [Indexed: 10/01/2024] Open
Abstract
Cervical cancer is one of the most malignant gynaecological tumors characterised with the aggressive behaviour of the tumor cells. In spite of the development of different strategies for the treatment of cervical cancer, the tumor cells have developed resistance to conventional therapeutics. On the other hand, nanoparticles have been recently applied for the treatment of human cancers through delivery of drugs and facilitate tumor suppression. The stimuli-sensitive nanostructures can improve the release of therapeutics at the tumor site. In the present review, the nanostructures for the treatment of cervical cancer are discussed. Nanostructures can deliver both chemotherapy drugs and natural compounds to increase anti-cancer activity and prevent drug resistance in cervical tumor. Moreover, the genetic tools such as siRNA can be delivered by nanoparticles to enhance their accumulation at tumor site. In order to enhance selectivity, the stimuli-responsive nanoparticles such as pH- and redox-responsive nanocarriers have been developed to suppress cervical tumor. Moreover, nanoparticles can induce photo-thermal and photodynamic therapy to accelerate cell death in cervical tumor. In addition, nanobiotechnology demonstrates tremendous potential in the treatment of cervical cancer, especially in the context of tumor immunotherapy. Overall, metal-, carbon-, lipid- and polymer-based nanostructures have been utilized in cervical cancer therapy. Finally, hydrogels have been developed as novel kinds of carriers to encapsulate therapeutics and improve anti-cancer activity.
Collapse
Affiliation(s)
| | - Zhengmei Xu
- Department of Gynecology, Affiliated Hengyang Hospital of Hunan Normal University &
Hengyang Central Hospital, Hengyang, China
| |
Collapse
|
3
|
Hartmeier PR, Ostrowski SM, Busch EE, Empey KM, Meng WS. Lymphatic distribution considerations for subunit vaccine design and development. Vaccine 2024; 42:2519-2529. [PMID: 38494411 DOI: 10.1016/j.vaccine.2024.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/30/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
Subunit vaccines are an important platform for controlling current and emerging infectious diseases. The lymph nodes are the primary site generating the humoral response and delivery of antigens to these sites is critical to effective immunization. Indeed, the duration of antigen exposure within the lymph node is correlated with the antibody response. While current licensed vaccines are typically given through the intramuscular route, injecting vaccines subcutaneously allows for direct access to lymphatic vessels and therefore can enhance the transfer of antigen to the lymph nodes. However, protein subunit antigen uptake into the lymph nodes is inefficient, and subunit vaccines require adjuvants to stimulate the initial immune response. Therefore, formulation strategies have been developed to enhance the exposure of subunit proteins and adjuvants to the lymph nodes by increasing lymphatic uptake or prolonging the retention at the injection site. Given that lymph node exposure is a crucial consideration in vaccine design, in depth analyses of the pharmacokinetics of antigens and adjuvants should be the focus of future preclinical and clinical studies. This review will provide an overview of formulation strategies for targeting the lymphatics and prolonging antigen exposure and will discuss pharmacokinetic evaluations which can be applied toward vaccine development.
Collapse
Affiliation(s)
- Paul R Hartmeier
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Sarah M Ostrowski
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA 15213, USA
| | - Emelia E Busch
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Kerry M Empey
- Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, PA 15213, USA; Department of Immunology, School of Medicine University of Pittsburgh, PA 15213, USA
| | - Wilson S Meng
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA 15219, USA.
| |
Collapse
|
4
|
Lee ALZ, Balakrishnan N, Ng JY, Liu S, Ong ZY, Wang Y, Gao S, Yang YY. Injectable Hydrogels Prepared Using Novel Synthetic Short Peptides with Defined Structure and Gelatin as Scaffolds to Support Cell and Tissue Growth. Adv Healthc Mater 2024; 13:e2302786. [PMID: 37837308 DOI: 10.1002/adhm.202302786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/06/2023] [Indexed: 10/15/2023]
Abstract
Animal-derived basement-membrane matrices such as Geltrex are used to grow cells and tissues. Particularly, these are commonly applied to support tumor growth in animals for cancer research. However, a material derived from an animal source has an undefined composition, and may thus have unavoidable batch-to-batch variation in properties. To overcome these issues, a series of synthetic short peptides to form hydrogels is designed in combination with gelatin to promote cell adhesion and growth. The peptides have sequences of (X1Y1X2Y2)2 , where X1 and X2 are hydrophobic residues, while Y1 and Y2 are hydrophilic residues. The peptides spontaneously fold and self-assemble into a β-sheet secondary structure upon contact with salts, and then aggregate to form hydrophilic networks of hydrogels. Hybrid hydrogels formed by mixing the peptide IEVEIRVK (IVK8) with gelatin are injectable and enzymatically degradable. The hybrid hydrogels at optimal compositions support SW480 and HepG2 tumor spheroid growth in vitro as effectively as Geltrex. More importantly, the peptide/gelatin hydrogels support tumor growth in a SW480 human colorectal adenocarcinoma xenograft mouse model. Altogether, the results illustrate that the synthetic peptide/gelatin hybrid hydrogel is a promising scaffold that can be used to support cell and tissue growth both in vitro and in vivo.
Collapse
Affiliation(s)
- Ashlynn Ling Zhi Lee
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Nithiyaa Balakrishnan
- Bioprocessing Technology Institute, Agency for Science, Technology, and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Jian Yao Ng
- Bioprocessing Technology Institute, Agency for Science, Technology, and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Shaoqiong Liu
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Zhan Yuin Ong
- School of Physics and Astronomy, St. James's University of Leeds, Leeds, LS2 9JT, UK
- Leeds Institute of Medical Research, St. James's University of Leeds, Leeds, LS2 9JT, UK
| | - Yanming Wang
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Shujun Gao
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology, and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| |
Collapse
|
5
|
Cheng L, Yu J, Hao T, Wang W, Wei M, Li G. Advances in Polymeric Micelles: Responsive and Targeting Approaches for Cancer Immunotherapy in the Tumor Microenvironment. Pharmaceutics 2023; 15:2622. [PMID: 38004600 PMCID: PMC10675796 DOI: 10.3390/pharmaceutics15112622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/01/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
In recent years, to treat a diverse array of cancer forms, considerable advancements have been achieved in the field of cancer immunotherapies. However, these therapies encounter multiple challenges in clinical practice, such as high immune-mediated toxicity, insufficient accumulation in cancer tissues, and undesired off-target reactions. To tackle these limitations and enhance bioavailability, polymer micelles present potential solutions by enabling precise drug delivery to the target site, thus amplifying the effectiveness of immunotherapy. This review article offers an extensive survey of recent progress in cancer immunotherapy strategies utilizing micelles. These strategies include responsive and remodeling approaches to the tumor microenvironment (TME), modulation of immunosuppressive cells within the TME, enhancement of immune checkpoint inhibitors, utilization of cancer vaccine platforms, modulation of antigen presentation, manipulation of engineered T cells, and targeting other components of the TME. Subsequently, we delve into the present state and constraints linked to the clinical utilization of polymeric micelles. Collectively, polymer micelles demonstrate excellent prospects in tumor immunotherapy by effectively addressing the challenges associated with conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Lichun Cheng
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Jiankun Yu
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Tangna Hao
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| | - Wenshuo Wang
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Guiru Li
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| |
Collapse
|
6
|
Wang Z, Ye Q, Yu S, Akhavan B. Poly Ethylene Glycol (PEG)-Based Hydrogels for Drug Delivery in Cancer Therapy: A Comprehensive Review. Adv Healthc Mater 2023; 12:e2300105. [PMID: 37052256 PMCID: PMC11468892 DOI: 10.1002/adhm.202300105] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/08/2023] [Indexed: 04/14/2023]
Abstract
Hydrogel-based drug delivery systems (DDSs) can leverage therapeutically beneficial outcomes in cancer therapy. In this domain, polyethylene glycol (PEG) has become increasingly popular as a biomedical polymer and has found clinical use. Owing to their excellent biocompatibility, facile modifiability, and high drug encapsulation rate, PEG hydrogels have shown great promise as drug delivery platforms. Here, the progress in emerging novel designs of PEG-hydrogels as DDSs for anti-cancer therapy is reviewed and discussed, focusing on underpinning multiscale release mechanisms categorized under stimuli-responsive and non-responsive drug release. The responsive drug delivery approaches are discussed, and the underpinning release mechanisms are elucidated, covering the systems functioning based on either exogenous stimuli-response, such as photo- and magnetic-sensitive PEG hydrogels, or endogenous stimuli-response, such as enzyme-, pH-, reduction-, and temperature-sensitive PEG hydrogels. Special attention is paid to the commercial potential of PEG-based hydrogels in cancer therapy, highlighting the limitations that need to be addressed in future research for their clinical translation.
Collapse
Affiliation(s)
- Zihan Wang
- College of ChemistryNankai UniversityTianjin300071P. R. China
| | - Qinzhou Ye
- Sichuan Agricultural UniversitySichuan611130P. R. China
| | - Sheng Yu
- Chemical Synthesis and Pollution Control Key Laboratory of Sichuan ProvinceChina West Normal UniversityNanchong637000P. R. China
| | - Behnam Akhavan
- School of EngineeringUniversity of NewcastleCallaghanNSW2308Australia
- Hunter Medical Research Institute (HMRI)New Lambton HeightsNSW2305Australia
- School of PhysicsThe University of SydneySydneyNSW2006Australia
- School of Biomedical EngineeringThe University of SydneySydneyNSW2006Australia
- Sydney Nano InstituteThe University of SydneySydneyNSW2006Australia
| |
Collapse
|
7
|
Qiu E, Liu F. PLGA-based drug delivery systems in treating bone tumors. Front Bioeng Biotechnol 2023; 11:1199343. [PMID: 37324432 PMCID: PMC10267463 DOI: 10.3389/fbioe.2023.1199343] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Bone tumor has become a common disease that endangers human health. Surgical resection of bone tumors not only causes biomechanical defects of bone but also destroys the continuity and integrity of bone and cannot completely remove the local tumor cells. The remaining tumor cells in the lesion bring a hidden danger of local recurrence. To improve the chemotherapeutic effect and effectively clear tumor cells, traditional systemic chemotherapy often requires higher doses, and high doses of chemotherapeutic drugs inevitably cause a series of systemic toxic side effects, often intolerable to patients. PLGA-based drug delivery systems, such as nano delivery systems and scaffold-based local delivery systems, can help eliminate tumors and promote bone regeneration and therefore have more significant potential for application in bone tumor treatment. In this review, we summarize the research progress of PLGA nano drug delivery systems and PLGA scaffold-based local delivery systems in bone tumor treatment applications, expecting to provide a theoretical basis for developing novel bone tumor treatment strategies.
Collapse
Affiliation(s)
- Enduo Qiu
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | | |
Collapse
|
8
|
Rama B, Ribeiro AJ. Role of nanotechnology in the prolonged release of drugs by the subcutaneous route. Expert Opin Drug Deliv 2023; 20:559-577. [PMID: 37305971 DOI: 10.1080/17425247.2023.2214362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 05/11/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION Subcutaneous physiology is distinct from other parenteral routes that benefit the administration of prolonged-release formulations. A prolonged-release effect is particularly convenient for treating chronic diseases because it is associated with complex and often prolonged posologies. Therefore, drug-delivery systems focused on nanotechnology are proposed as alternatives that can overcome the limitations of current therapeutic regimens and improve therapeutic efficacy. AREAS COVERED This review presents an updated systematization of nanosystems, focusing on their applications in highly prevalent chronic diseases. Subcutaneous-delivered nanosystem-based therapies comprehensively summarize nanosystems, drugs, and diseases and their advantages, limitations, and strategies to increase their translation into clinical applications. An outline of the potential contribution of quality-by-design (QbD) and artificial intelligence (AI) to the pharmaceutical development of nanosystems is presented. EXPERT OPINION Although recent academic research and development (R&D) advances in the subcutaneous delivery of nanosystems have exhibited promising results, pharmaceutical industries and regulatory agencies need to catch up. The lack of standardized methodologies for analyzing in vitro data from nanosystems for subcutaneous administration and subsequent in vivo correlation limits their access to clinical trials. There is an urgent need for regulatory agencies to develop methods that faithfully mimic subcutaneous administration and specific guidelines for evaluating nanosystems.
Collapse
Affiliation(s)
- B Rama
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, Portugal
| | - A J Ribeiro
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, Portugal
- Genetics of Cognitive Disfunction, i3S, IBMC, Porto, Portugal
| |
Collapse
|
9
|
Affonso de Oliveira JF, Zhao Z, Xiang Y, Shin MD, Villaseñor KE, Deng X, Shukla S, Chen S, Steinmetz NF. COVID-19 vaccines based on viral nanoparticles displaying a conserved B-cell epitope show potent immunogenicity and a long-lasting antibody response. Front Microbiol 2023; 14:1117494. [PMID: 37152732 PMCID: PMC10157238 DOI: 10.3389/fmicb.2023.1117494] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/13/2023] [Indexed: 05/09/2023] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 sparked intensive research into the development of effective vaccines, 50 of which have been approved thus far, including the novel mRNA-based vaccines developed by Pfizer and Moderna. Although limiting the severity of the disease, the mRNA-based vaccines presented drawbacks, such as the cold chain requirement. Moreover, antibody levels generated by these vaccines decline significantly after 6 months. These vaccines deliver mRNA encoding the full-length spike (S) glycoprotein of SARS-CoV-2, but must be updated as new strains and variants of concern emerge, creating a demand for adjusted formulations and booster campaigns. To overcome these challenges, we have developed COVID-19 vaccine candidates based on the highly conserved SARS CoV-2, 809-826 B-cell peptide epitope (denoted 826) conjugated to cowpea mosaic virus (CPMV) nanoparticles and bacteriophage Qβ virus-like particles, both platforms have exceptional thermal stability and facilitate epitope delivery with inbuilt adjuvant activity. We evaluated two administration methods: subcutaneous injection and an implantable polymeric scaffold. Mice received a prime-boost regimen of 100 μg per dose (2 weeks apart) or a single dose of 200 μg administered as a liquid formulation, or a polymer implant. Antibody titers were evaluated longitudinally over 50 weeks. The vaccine candidates generally elicited an early Th2-biased immune response, which stimulates the production of SARS-CoV-2 neutralizing antibodies, followed by a switch to a Th1-biased response for most formulations. Exceptionally, vaccine candidate 826-CPMV (administered as prime-boost, soluble injection) elicited a balanced Th1/Th2 immune response, which is necessary to prevent pulmonary immunopathology associated with Th2 bias extremes. While the Qβ-based vaccine elicited overall higher antibody titers, the CPMV-induced antibodies had higher avidity. Regardless of the administration route and formulation, our vaccine candidates maintained high antibody titers for more than 50 weeks, confirming a potent and durable immune response against SARS-CoV-2 even after a single dose.
Collapse
Affiliation(s)
| | - Zhongchao Zhao
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| | - Yi Xiang
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Matthew D. Shin
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | | | - Xinyi Deng
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Sourabh Shukla
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Shaochen Chen
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, United States
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
- Center for Engineering in Cancer, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
10
|
Meng R, Zhu H, Deng P, Li M, Ji Q, He H, Jin L, Wang B. Research progress on albumin-based hydrogels: Properties, preparation methods, types and its application for antitumor-drug delivery and tissue engineering. Front Bioeng Biotechnol 2023; 11:1137145. [PMID: 37113668 PMCID: PMC10127125 DOI: 10.3389/fbioe.2023.1137145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Albumin is derived from blood plasma and is the most abundant protein in blood plasma, which has good mechanical properties, biocompatibility and degradability, so albumin is an ideal biomaterial for biomedical applications, and drug-carriers based on albumin can better reduce the cytotoxicity of drug. Currently, there are numerous reviews summarizing the research progress on drug-loaded albumin molecules or nanoparticles. In comparison, the study of albumin-based hydrogels is a relatively small area of research, and few articles have systematically summarized the research progress of albumin-based hydrogels, especially for drug delivery and tissue engineering. Thus, this review summarizes the functional features and preparation methods of albumin-based hydrogels, different types of albumin-based hydrogels and their applications in antitumor drugs, tissue regeneration engineering, etc. Also, potential directions for future research on albumin-based hydrogels are discussed.
Collapse
Affiliation(s)
- Run Meng
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Huimin Zhu
- Sheyang County Comprehensive Inspection and Testing Center, Yancheng, China
| | - Peiying Deng
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Minghui Li
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Qingzhi Ji
- School of Pharmacy, Yancheng Teachers’ University, Yancheng, China
| | - Hao He
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
- *Correspondence: Liang Jin, ; Bochu Wang,
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
- *Correspondence: Liang Jin, ; Bochu Wang,
| |
Collapse
|
11
|
Shukla A, Maiti P. Nanomedicine and versatile therapies for cancer treatment. MedComm (Beijing) 2022; 3:e163. [PMID: 35992969 PMCID: PMC9386439 DOI: 10.1002/mco2.163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 12/19/2022] Open
Abstract
The higher prevalence of cancer is related to high rates of mortality and morbidity worldwide. By virtue of the properties of matter at the nanoscale, nanomedicine is proven to be a powerful tool to develop innovative drug carriers with greater efficacies and fewer side effects than conventional therapies. In this review, different nanocarriers for controlled drug release and their routes of administration have been discussed in detail, especially for cancer treatment. Special emphasis has been given on the design of drug delivery vehicles for sustained release and specific application methods for targeted delivery to the affected areas. Different polymeric vehicles designed for the delivery of chemotherapeutics have been discussed, including graft copolymers, liposomes, hydrogels, dendrimers, micelles, and nanoparticles. Furthermore, the effect of dimensional properties on chemotherapy is vividly described. Another integral section of the review focuses on the modes of administration of nanomedicines and emerging therapies, such as photothermal, photodynamic, immunotherapy, chemodynamic, and gas therapy, for cancer treatment. The properties, therapeutic value, advantages, and limitations of these nanomedicines are highlighted, with a focus on their increased performance versus conventional molecular anticancer therapies.
Collapse
Affiliation(s)
- Aparna Shukla
- School of Materials Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| | - Pralay Maiti
- School of Materials Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| |
Collapse
|
12
|
Fan R, Cheng Y, Wang R, Zhang T, Zhang H, Li J, Song S, Zheng A. Thermosensitive Hydrogels and Advances in Their Application in Disease Therapy. Polymers (Basel) 2022; 14:polym14122379. [PMID: 35745954 PMCID: PMC9227257 DOI: 10.3390/polym14122379] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 01/27/2023] Open
Abstract
Thermosensitive hydrogels, having unique sol–gel transition properties, have recently received special research attention. These hydrogels exhibit a phase transition near body temperature. This feature is the key to their applications in human medicine. In addition, hydrogels can quickly gel at the application site with simple temperature stimulation and without additional organic solvents, cross-linking agents, or external equipment, and the loaded drugs can be retained locally to improve the local drug concentration and avoid unexpected toxicity or side effects caused by systemic administration. All of these features have led to thermosensitive hydrogels being some of the most promising and practical drug delivery systems. In this paper, we review thermosensitive hydrogel materials with biomedical application potential, including natural and synthetic materials. We describe their structural characteristics and gelation mechanism and briefly summarize the mechanism of drug release from thermosensitive hydrogels. Our focus in this review was to summarize the application of thermosensitive hydrogels in disease treatment, including the postoperative recurrence of tumors, the delivery of vaccines, the prevention of postoperative adhesions, the treatment of nervous system diseases via nasal brain targeting, wound healing, and osteoarthritis treatment.
Collapse
Affiliation(s)
- Ranran Fan
- School of Pharmacy, Bengbu Medical College, Anhui 233030, China;
| | - Yi Cheng
- College of Pharmacy, Yanbian University, Jilin 133002, China;
| | - Rongrong Wang
- School of Pharmacy, North China University of Science and Technology, Hebei 063210, China;
| | - Ting Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China;
| | - Hui Zhang
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100850, China;
- Correspondence: (H.Z.); (J.L.); (S.S.)
| | - Jianchun Li
- School of Pharmacy, Bengbu Medical College, Anhui 233030, China;
- Correspondence: (H.Z.); (J.L.); (S.S.)
| | - Shenghan Song
- Department of Vascular Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
- Correspondence: (H.Z.); (J.L.); (S.S.)
| | - Aiping Zheng
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100850, China;
| |
Collapse
|
13
|
Ali F, Khan I, Chen J, Akhtar K, Bakhsh EM, Khan SB. Emerging Fabrication Strategies of Hydrogels and Its Applications. Gels 2022; 8:gels8040205. [PMID: 35448106 PMCID: PMC9024659 DOI: 10.3390/gels8040205] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/04/2022] [Accepted: 03/15/2022] [Indexed: 12/19/2022] Open
Abstract
Recently, hydrogels have been investigated for the controlled release of bioactive molecules, such as for living cell encapsulation and matrices. Due to their remote controllability and quick response, hydrogels are widely used for various applications, including drug delivery. The rate and extent to which the drugs reach their targets are highly dependent on the carriers used in drug delivery systems; therefore the demand for biodegradable and intelligent carriers is progressively increasing. The biodegradable nature of hydrogel has created much interest for its use in drug delivery systems. The first part of this review focuses on emerging fabrication strategies of hydrogel, including physical and chemical cross-linking, as well as radiation cross-linking. The second part describes the applications of hydrogels in various fields, including drug delivery systems. In the end, an overview of the application of hydrogels prepared from several natural polymers in drug delivery is presented.
Collapse
Affiliation(s)
- Fayaz Ali
- Department of Chemistry, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (F.A.); (K.A.); (E.M.B.)
- Centre of Excellence for Advance Materials Research, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - Imran Khan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science & Technology Avenida Wai Long, Taipa, Macau 999078, China;
| | - Jianmin Chen
- School of Pharmacy and Medical Technology, Putian University, No. 1133 Xueyuan Zhong Jie, Putian 351100, China
- Correspondence: (J.C.); (S.B.K.)
| | - Kalsoom Akhtar
- Department of Chemistry, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (F.A.); (K.A.); (E.M.B.)
| | - Esraa M. Bakhsh
- Department of Chemistry, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (F.A.); (K.A.); (E.M.B.)
| | - Sher Bahadar Khan
- Department of Chemistry, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (F.A.); (K.A.); (E.M.B.)
- Centre of Excellence for Advance Materials Research, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
- Correspondence: (J.C.); (S.B.K.)
| |
Collapse
|
14
|
Lamrayah M, Phelip C, Coiffier C, Lacroix C, Willemin T, Trimaille T, Verrier B. A Polylactide-Based Micellar Adjuvant Improves the Intensity and Quality of Immune Response. Pharmaceutics 2022; 14:pharmaceutics14010107. [PMID: 35057003 PMCID: PMC8778782 DOI: 10.3390/pharmaceutics14010107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 11/21/2022] Open
Abstract
Micelles from amphiphilic polylactide-block-poly(N-acryloxysuccinimide-co-N-vinylpyrrolidone) (PLA-b-P(NAS-co-NVP)) block copolymers of 105 nm in size were characterized and evaluated in a vaccine context. The micelles were non-toxic in vitro (both in dendritic cells and HeLa cells). In vitro fluorescence experiments combined with in vivo fluorescence tomography imaging, through micelle loading with the DiR near infrared probe, suggested an efficient uptake of the micelles by the immune cells. The antigenic protein p24 of the HIV-1 was successfully coupled on the micelles using the reactive N-succinimidyl ester groups on the micelle corona, as shown by SDS-PAGE analyses. The antigenicity of the coupled antigen was preserved and even improved, as assessed by the immuno-enzymatic (ELISA) test. Then, the performances of the micelles in immunization were investigated and compared to different p24-coated PLA nanoparticles, as well as Alum and MF59 gold standards, following a standardized HIV-1 immunization protocol in mice. The humoral response intensity (IgG titers) was substantially similar between the PLA micelles and all other adjuvants over an extended time range (one year). More interestingly, this immune response induced by PLA micelles was qualitatively higher than the gold standards and PLA nanoparticles analogs, expressed through an increasing avidity index over time (>60% at day 365). Taken together, these results demonstrate the potential of such small-sized micellar systems for vaccine delivery.
Collapse
Affiliation(s)
- Myriam Lamrayah
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
- Correspondence: (M.L.); (T.T.)
| | - Capucine Phelip
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Céline Coiffier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Céline Lacroix
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Thibaut Willemin
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Thomas Trimaille
- Laboratoire Ingénierie des Matériaux Polymères (IMP), CNRS UMR 5223, Université Lyon 1, Université de Lyon, 69622 Villeurbanne, France
- Correspondence: (M.L.); (T.T.)
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| |
Collapse
|
15
|
Roth GA, Picece VCTM, Ou BS, Luo W, Pulendran B, Appel EA. Designing spatial and temporal control of vaccine responses. NATURE REVIEWS. MATERIALS 2022; 7:174-195. [PMID: 34603749 PMCID: PMC8477997 DOI: 10.1038/s41578-021-00372-2] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 05/02/2023]
Abstract
Vaccines are the key technology to combat existing and emerging infectious diseases. However, increasing the potency, quality and durability of the vaccine response remains a challenge. As our knowledge of the immune system deepens, it becomes clear that vaccine components must be in the right place at the right time to orchestrate a potent and durable response. Material platforms, such as nanoparticles, hydrogels and microneedles, can be engineered to spatially and temporally control the interactions of vaccine components with immune cells. Materials-based vaccination strategies can augment the immune response by improving innate immune cell activation, creating local inflammatory niches, targeting lymph node delivery and controlling the time frame of vaccine delivery, with the goal of inducing enhanced memory immunity to protect against future infections. In this Review, we highlight the biological mechanisms underlying strong humoral and cell-mediated immune responses and explore materials design strategies to manipulate and control these mechanisms.
Collapse
Affiliation(s)
- Gillie A. Roth
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Vittoria C. T. M. Picece
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Ben S. Ou
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Wei Luo
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA USA
| | - Eric A. Appel
- Department of Bioengineering, Stanford University, Stanford, CA USA
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Paediatrics — Endocrinology, Stanford University School of Medicine, Stanford, CA USA
| |
Collapse
|
16
|
Production and Immunogenicity of a Tag-Free Recombinant Chimera Based on PfMSP-1 and PfMSP-3 Using Alhydrogel and Dipeptide-Based Hydrogels. Vaccines (Basel) 2021; 9:vaccines9070782. [PMID: 34358198 PMCID: PMC8310097 DOI: 10.3390/vaccines9070782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
A fusion chimeric vaccine comprising multiple protective domains of different blood-stage Plasmodium falciparum antigens is perhaps necessary for widening the protective immune responses and reducing the morbidity caused by the disease. Here we continue to build upon the prior work of developing a recombinant fusion chimera protein, His-tagged PfMSP-Fu24, by producing it as a tag-free recombinant protein. In this study, tag-free recombinant PfMSPFu24 (rFu24) was expressed in Escherichia coli, and the soluble protein was purified using a three-step purification involving ammonium sulphate precipitation followed by 2-step ion exchange chromatography procedures and shown that it was highly immunogenic with the human-compatible adjuvant Alhydrogel. We further investigated two dipeptides, phenylalanine-α, β-dehydrophenylalanine (FΔF) and Leucine-α, β-dehydrophenylalanine (LΔF) based hydrogels as effective delivery platforms for rFu24. These dipeptides self-assembled spontaneously to form a highly stable hydrogel under physiological conditions. rFu24 was efficiently entrapped in both the F∆F and L∆F hydrogels, and the three-dimensional (3D) mesh-like structures of the hydrogels remained intact after the entrapment of the antigen. The two hydrogels significantly stimulated rFu24-specific antibody titers, and the sera from the immunized mice showed an invasion inhibitory activity comparable to that of Alhydrogel. Easily synthesized dipeptide hydrogels can be used as an effective antigen delivery platform to induce immune responses.
Collapse
|
17
|
Luzuriaga MA, Shahrivarkevishahi A, Herbert FC, Wijesundara YH, Gassensmith JJ. Biomaterials and nanomaterials for sustained release vaccine delivery. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1735. [PMID: 34180608 DOI: 10.1002/wnan.1735] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/03/2021] [Accepted: 06/04/2021] [Indexed: 12/17/2022]
Abstract
Vaccines are considered one of the most significant medical advancements in human history, as they have prevented hundreds of millions of deaths since their discovery; however, modern travel permits disease spread at unprecedented rates, and vaccine shortcomings like thermal sensitivity and required booster shots have been made evident by the COVID-19 pandemic. Approaches to overcoming these issues appear promising via the integration of vaccine technology with biomaterials, which offer sustained-release properties and preserve proteins, prevent conformational changes, and enable storage at room temperature. Sustained release and thermal stabilization of therapeutic biomacromolecules is an emerging area that integrates material science, chemistry, immunology, nanotechnology, and pathology to investigate different biocompatible materials. Biomaterials, including natural sugar polymers, synthetic polyesters produced from biologically derived monomers, hydrogel blends, protein-polymer blends, and metal-organic frameworks, have emerged as early players in the field. This overview will focus on significant advances of sustained release biomaterial in the context of vaccines against infectious disease and the progress made towards thermally stable "single-shot" formulations. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease.
Collapse
Affiliation(s)
- Michael A Luzuriaga
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Division of Medical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Fabian C Herbert
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardon, Texas, USA
| | - Yalini H Wijesundara
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardon, Texas, USA
| | - Jeremiah J Gassensmith
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardon, Texas, USA.,Department of Bioengineering, The University of Texas at Dallas, Richardon, Texas, USA
| |
Collapse
|
18
|
Chopra H, Singh I, Kumar S, Bhattacharya T, Rahman MH, Akter R, Kabir MT. Comprehensive Review on Hydrogels. Curr Drug Deliv 2021; 19:658-675. [PMID: 34077344 DOI: 10.2174/1567201818666210601155558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/26/2021] [Accepted: 04/05/2021] [Indexed: 11/22/2022]
Abstract
The conventional drug delivery systems have a long list of issues of repeated dosing and toxicity arising due to it. The hydrogels are the answer to them and offer a result that minimizes such activities and optimizes therapeutic benefits. The hydrogels proffer tunable properties that can withstand degradation, metabolism, and controlled release moieties. Some of the areas of applications of hydrogels involve wound healing, ocular systems, vaginal gels, scaffolds for tissue, bone engineering, etc. They consist of about 90% of the water that makes them suitable bio-mimic moiety. Here, we present a birds-eye view of various perspectives of hydrogels, along with their applications.
Collapse
Affiliation(s)
- Hitesh Chopra
- Department of Pharmaceutics, Chitkara College of Pharmacy, Chitkara University, Rajpura-140401, Patiala, Punjab, India
| | - Inderbir Singh
- Department of Pharmaceutics, Chitkara College of Pharmacy, Chitkara University, Rajpura-140401, Patiala, Punjab, India
| | - Sandeep Kumar
- Department of Pharmaceutics, ASBASJSM College of Pharmacy, Bela-140111, Ropar, Punjab, India
| | | | - Md Habibur Rahman
- Department of Pharmacy, Jagannath University, Sadarghat, Dhaka-1100. Bangladesh
| | - Rokeya Akter
- Department of Pharmacy, Southeast University, Banani, Dhaka-1213. Bangladesh
| | - Md Tanvir Kabir
- Department of Pharmacy, Brac University, 66 Mohakhali, Dhaka 1212. Bangladesh
| |
Collapse
|
19
|
Li B, Wang W, Song W, Zhao Z, Tan Q, Zhao Z, Tang L, Zhu T, Yin J, Bai J, Dong X, Tan S, Hu Q, Tang BZ, Huang X. Antiviral and Anti‐Inflammatory Treatment with Multifunctional Alveolar Macrophage‐Like Nanoparticles in a Surrogate Mouse Model of COVID‐19. ADVANCED SCIENCE 2021; 8:2003556. [PMCID: PMC8209923 DOI: 10.1002/advs.202003556] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The pandemic of coronavirus disease 2019 (COVID‐19) is continually worsening. Clinical treatment for COVID‐19 remains primarily supportive with no specific medicines or regimens. Here, the development of multifunctional alveolar macrophage (AM)‐like nanoparticles (NPs) with photothermal inactivation capability for COVID‐19 treatment is reported. The NPs, made by wrapping polymeric cores with AM membranes, display the same surface receptors as AMs, including the coronavirus receptor and multiple cytokine receptors. By acting as AM decoys, the NPs block coronavirus from host cell entry and absorb various proinflammatory cytokines, thus achieving combined antiviral and anti‐inflammatory treatment. To enhance the antiviral efficiency, an efficient photothermal material based on aggregation‐induced emission luminogens is doped into the NPs for virus photothermal disruption under near‐infrared (NIR) irradiation. In a surrogate mouse model of COVID‐19 caused by murine coronavirus, treatment with multifunctional AM‐like NPs with NIR irradiation decreases virus burden and cytokine levels, reduces lung damage and inflammation, and confers a significant survival advantage to the infected mice. Crucially, this therapeutic strategy may be clinically applied for the treatment of COVID‐19 at early stage through atomization inhalation of the NPs followed by NIR irradiation of the respiratory tract, thus alleviating infection progression and reducing transmission risk.
Collapse
Affiliation(s)
- Bin Li
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Wei Wang
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of EducationXizang Minzu UniversityXianyangShaanxi712082China
| | - Weifeng Song
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Zheng Zhao
- Department of ChemistryThe Hong Kong University of Science and TechnologyClear Water BayKowloonHong Kong999077China
| | - Qingqin Tan
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Zhaoyan Zhao
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Lantian Tang
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Tianchuan Zhu
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Jialing Yin
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Jun Bai
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Xin Dong
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Siyi Tan
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Qunying Hu
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of EducationXizang Minzu UniversityXianyangShaanxi712082China
| | - Ben Zhong Tang
- Department of ChemistryThe Hong Kong University of Science and TechnologyClear Water BayKowloonHong Kong999077China
| | - Xi Huang
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| |
Collapse
|
20
|
Mo C, Xiang L, Chen Y. Advances in Injectable and Self-healing Polysaccharide Hydrogel Based on the Schiff Base Reaction. Macromol Rapid Commun 2021; 42:e2100025. [PMID: 33876841 DOI: 10.1002/marc.202100025] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/13/2021] [Indexed: 12/17/2022]
Abstract
Injectable hydrogel possesses great application potential in disease treatment and tissue engineering, but damage to gel often occurs due to the squeezing pressure from injection devices and the mechanical forces from limb movement, and leads to the rapid degradation of gel matrix and the leakage of the load material. The self-healing injectable hydrogels can overcome these drawbacks via automatically repairing gel structural defects and restoring gel function. The polysaccharide hydrogels constructed through the Schiff base reaction own advantages including simple fabrication, injectability, and self-healing under physiological conditions, and therefore have drawn extensive attention and investigation recently. In this short review, the preparation and self-healing properties of the polysaccharide hydrogels that is established on the Schiff base reaction are focused on and their biological applications in drug delivery and cell therapy are discussed.
Collapse
Affiliation(s)
- Chunxiang Mo
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, Hunan, 421001, China.,School of Pharmaceutical Science, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, 421001, China
| | - Li Xiang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, Hunan, 421001, China.,School of Pharmaceutical Science, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, 421001, China
| | - Yuping Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, Hunan, 421001, China.,School of Pharmaceutical Science, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
21
|
Vaccine implants: current status and recent advancements. Emerg Top Life Sci 2020; 4:319-330. [DOI: 10.1042/etls20200164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/29/2020] [Accepted: 11/05/2020] [Indexed: 01/29/2023]
Abstract
Implants have long been used in the field of drug delivery as controlled release vehicles and are now being investigated as single-shot vaccine technologies. Implants have shown great promise, minimizing the need for multiple immunizations while stimulating potent immune responses with reduced doses of vaccine. Synchronous release of vaccine components from implants over an appropriate period of time is important in order to avoid issues including immune tolerance, sequestration or deletion. Traditionally, implants require surgical implantation and removal, which can be a barrier to their widespread use. Degradable and in situ implants are now being developed that can be administered using minimally invasive subcutaneous or intramuscular injection techniques. Injectable hydrogels remain the most commonly studied approach for sustained vaccine delivery due to their ease of administration and tunable degradation properties. Despite exciting advancements in the field of vaccine implants, few technologies have progressed to clinical trials. To increase the likelihood of clinical translation of vaccine implants, strategic testing of disease-relevant antigens in appropriate species is essential. In this review, the significance of vaccine implants and the different types of implants being developed to deliver vaccines are discussed.
Collapse
|
22
|
Lee ALZ, Yang C, Gao S, Wang Y, Hedrick JL, Yang YY. Biodegradable Cationic Polycarbonates as Vaccine Adjuvants. ACS APPLIED MATERIALS & INTERFACES 2020; 12:52285-52297. [PMID: 33179910 DOI: 10.1021/acsami.0c09649] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this study, biodegradable cationic polycarbonate and polylactide block copolymers were synthesized and successfully used as novel vaccine adjuvants to provide enhanced anticancer immunity. The polymers formed nanoparticles with the model vaccine, ovalbumin (OVA), and the immunostimulant toll-like receptor 3 agonist poly(I:C) (a synthetic analog of the double-stranded RNA). Higher uptake of poly(I:C) by the bone marrow-derived dendritic cells and macrophages and OVA by dendritic cells was observed when delivered using the polymer adjuvant. In vivo experiments showed that these nanoparticles remained longer in the subcutaneous injection site as compared to OVA alone and led to higher production of anti-OVA specific antibodies with prolonged immunostimulation. When OVA was combined with poly(I:C) that was either co-entrapped in the same particles or as separate particles, a comparable level of anti-OVA IgG1 antibodies and interleukin-6 (IL-6) was produced in mouse blood plasma, and a similar level of cytotoxic T lymphocyte (CTL) response in mice was stimulated as compared to OVA/Alum particles. Furthermore, tumor rejection in the mice that were vaccinated for 9 months with the formulations containing the polymer adjuvant was stronger than the other treatment groups without the polymer. Notably, the cationic polycarbonates were not associated with any adverse in vivo effects. Thus, these biodegradable polymers may be promising substitutes for aluminum-based adjuvants in vaccine formulations.
Collapse
Affiliation(s)
- Ashlynn L Z Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Chuan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Shujun Gao
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
- NanoBio Lab, 31 Biopolis Way, #09-01 The Nanos, Singapore 138669, Singapore
| | - Yanming Wang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - James L Hedrick
- IBM Almaden Research Center, 650 Harry Road, San Jose, California 95120 United States
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| |
Collapse
|
23
|
A Sacrificial PLA Block Mediated Route to Injectable and Degradable PNIPAAm-Based Hydrogels. Polymers (Basel) 2020; 12:polym12040925. [PMID: 32316376 PMCID: PMC7240404 DOI: 10.3390/polym12040925] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/06/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Thermoresponsive poly(N-isopropylacrylamide) (PNIPAAm)-based injectable hydrogels represent highly attractive materials in tissue engineering and drug/vaccine delivery but face the problem of long-term bioaccumulation due to non-degradability. In this context, we developed an amphiphilic poly(D,L-lactide)-b-poly(NIPAAm-co-polyethylene glycol methacrylate) (PLA-b-P(NIPAAm-co-PEGMA)) copolymer architecture, through a combination of ring-opening and nitroxide-mediated polymerizations, undergoing gelation in aqueous solution near 30 °C. Complete hydrogel mass loss was observed under physiological conditions after few days upon PLA hydrolysis. This was due to the inability of the resulting P(NIPAAm-co-PEGMA) segment, that contains sufficiently high PEG content, to gel. The copolymer was shown to be non-toxic on dendritic cells. These results thus provide a new way to engineer safe PNIPAAm-based injectable hydrogels with PNIPAAm-reduced content and a degradable feature.
Collapse
|
24
|
Zhang P, Zhou L, Wang L, Dong Q. A Novel Nanofiber Hydrogel Loaded with Platelet-Rich Plasma Promotes Wound Healing Through Enhancing the Survival of Fibroblasts. Med Sci Monit 2019; 25:8712-8721. [PMID: 31739310 PMCID: PMC6880629 DOI: 10.12659/msm.919812] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background Hydrogels are ideal biological carriers in vivo and have been widely used in the treatment of wound healing through loading with or without bioactive substances. Platelet-rich plasma (PRP) is purified from autologous plasma and has known curative efficacy for wound healing. The combined efficacy of shorten poly-N-acetyl glucosamine (sNAG) hydrogels and PRP in the treatment of wound healing has not been previously assessed. Material/Methods The cytotoxic and proliferative effects of PRP on fibroblasts were detected using Cell Counting Kit-8 assays and flow cytometry. The levels of cyclin D1 and cyclin D3 were assessed to evaluate cell proliferation. Protein expression was assessed by western blot analysis. Adenosine levels were assessed by enzyme-linked immunosorbent assay. Cell apoptosis was assessed by flow cytometry and western blot analysis. Rat wound models were performed, and the effects of PRP, single hydrogels, and sNAG hydrogels loaded with PRP were respectively detected through the assessment of wound closure. Hematoxylin eosin staining was used to measure the depth and width of regenerative scars. Results Our results demonstrated that PRP promotes fibroblast proliferation and inhibits apoptosis. PRP contains abundant levels of adenosine, which has a positive role on fibroblast function, whilst the inhibition of adenosine A2A receptors impairs the efficacy of PRP. sNAG hydrogels loaded with PRP showed curative efficacy during wound healing in mice. Mice treated with hydrogels loaded with PRP showed high levels of regeneration with scarless healing. Conclusions Our results indicate that sNAG hydrogels loaded with PRP promote wound healing. The pro-proliferative, and anti-apoptotic effects of the fibroblasts are mediated by the activating A2A receptor in response to elevated adenosine levels.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (mainland)
| | - Long Zhou
- Department of Orthopedics, Suzhou Science and Technology Town Hospital, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu, China (mainland)
| | - Lei Wang
- Department of Orthopedics, Suzhou Science and Technology Town Hospital, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu, China (mainland)
| | - Qirong Dong
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (mainland)
| |
Collapse
|