1
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2025; 314:122856. [PMID: 39366184 DOI: 10.1016/j.biomaterials.2024.122856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Vaccine science, nanotechnology, and immunotherapy are at the forefront of cancer treatment strategies, each offering significant potential for enhancing tumor-specific immunity and establishing long-lasting immune memory to prevent tumor recurrence. Despite the promise of these personalized and precision-based anti-cancer approaches, challenges such as immunosuppression, suboptimal immune activation, and T-cell exhaustion continue to hinder their effectiveness. The limited clinical success of cancer vaccines often stems from difficulties in identifying effective antigens, efficiently targeting immune cells, lymphoid organs, and the tumor microenvironment, overcoming immune evasion, enhancing immunogenicity, and avoiding lysosomal degradation. However, numerous studies have demonstrated that integrating nanotechnology with immunotherapeutic strategies in vaccine development can overcome these challenges, leading to potent antitumor immune responses and significant progress in the field. This review highlights the critical components of cancer vaccine and nanovaccine strategies for immunomodulatory antitumor therapy. It covers general vaccine strategies, types of vaccines, antigen forms, nanovaccine platforms, challenges faced, potential solutions, and key findings from preclinical and clinical studies, along with future perspectives. To fully unlock the potential of cancer vaccines and nanovaccines, precise immunological monitoring during early-phase trials is essential. This approach will help identify and address obstacles, ultimately expanding the available options for patients who are resistant to conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tulasi Vithiananthan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Sun M, Shi T, Tuerhong S, Li M, Wang Q, Lu C, Zou L, Zheng Q, Wang Y, Du J, Li R, Liu B, Meng F. An Immunomodulator-Boosted Lactococcus Lactis Platform For Enhanced In Situ Tumor Vaccine. Adv Healthc Mater 2024; 13:e2401635. [PMID: 39054611 DOI: 10.1002/adhm.202401635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/15/2024] [Indexed: 07/27/2024]
Abstract
In situ vaccination is an attractive type of cancer immunotherapy, and methods of persistently dispersing immune agonists throughout the entire tumor are crucial for maximizing their therapeutic efficacy. Based on the probiotics usually used for dietary supplements, an immunomodulator-boosted Lactococcus lactis (IBL) strategy is developed to enhance the effectiveness of in situ vaccination with the immunomodulators. The intratumoral delivery of OX40 agonist and resiquimod-modified Lactococcus lactis (OR@Lac) facilitates local retention and persistent dispersion of immunomodulators, and dramatically modulates the key components of anti-tumor immune response. This novel vaccine activated dendritic cells and cytotoxic T lymphocytes in the tumor and tumor-draining lymph nodes, and ultimately significantly inhibited tumor growth and prolonged the survival rate of tumor-bearing mice. The combination of OR@Lac and ibrutinib, a myeloid-derived suppressor cell inhibitor, significantly alleviated or even completely inhibited tumor growth in tumor-bearing mice. In conclusion, IBL is a promising in situ tumor vaccine approach for clinical application and provides an inspiration for the delivery of other drugs.
Collapse
Affiliation(s)
- Mengna Sun
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Tianyu Shi
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Subiyinuer Tuerhong
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Mengru Li
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Qiaoli Wang
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Changchang Lu
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Lu Zou
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Qinghua Zheng
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital,Joint Institute of Nanjing Drum Tower Hospital for Life and Health, College of Life Science, Nanjing Normal University, Nanjing, 210008, China
| | - Yingxin Wang
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Juan Du
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Rutian Li
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Baorui Liu
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Fanyan Meng
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| |
Collapse
|
3
|
Gu P, Zhao Q, Zhu Y, Xu P, Zhao X, Wang X, Zhang T, Bao Y, Shi W. Chinese yam polysaccharide-loaded aluminium hydroxide nanoparticles used as vaccine adjuvant to induce potent humoral and cellular immune responses. Int J Biol Macromol 2024; 281:135914. [PMID: 39370063 DOI: 10.1016/j.ijbiomac.2024.135914] [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: 06/11/2024] [Revised: 08/31/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Due to their safety and efficacy, aluminium salts (Alum) are considered the most important adjuvants in human vaccines. However, Alum adjuvants are unable to elicit a cellular immune response, which is vital for the prevention of various chronic infectious diseases and cancers. Herein, we isolated and purified a water-soluble polysaccharide from Chinese yam, named CYP, which was primarily composed of →4)-α-D-Glcp-(1→, →4,6)-α-D-Glcp-(1→, and α-D-Glcp-(1→. Meanwhile, we prepared aluminium hydroxide nanoparticles (Al NPs) with a nanometer-scale size and thin stick-like shape. Being an immunostimulant, the CYP was then loaded onto the Al NPs to obtain a novel adjuvant delivery system (CYP-Al NPs) that enhances the immunostimulatory activity of CYP. Our findings showed that the CYP-Al NPs facilitated macrophages activation and promoted the antigen uptake by macrophages. The in vivo experiment showed that the CYP-Al NPs, as the adjuvant to ovalbumin, promoted the activation of dendritic cells and germinal center B cells in draining lymph nodes, induced a durable and strong antibody response, especially the Th1-type IgG2a antibody response, and improved the cytotoxic T lymphocytes response. These results demonstrated that the CYP-Al NPs could generate robust humoral and cellular responses, and has the great potential to serve as an adjuvant delivery system.
Collapse
Affiliation(s)
- Pengfei Gu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Qi Zhao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Yixuan Zhu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Panpan Xu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Xinghua Zhao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Xiao Wang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Tie Zhang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Yongzhan Bao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Wanyu Shi
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China.
| |
Collapse
|
4
|
Sharma A, Bhatia D. Programmable bionanomaterials for revolutionizing cancer immunotherapy. Biomater Sci 2024; 12:5415-5432. [PMID: 39291418 DOI: 10.1039/d4bm00815d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Cancer immunotherapy involves a cutting-edge method that utilizes the immune system to detect and eliminate cancer cells. It has shown substantial effectiveness in treating different types of cancer. As a result, its growing importance is due to its distinct benefits and potential for sustained recovery. However, the general deployment of this treatment is hindered by ongoing issues in maintaining minimal toxicity, high specificity, and prolonged effectiveness. Nanotechnology offers promising solutions to these challenges due to its notable attributes, including expansive precise surface areas, accurate ability to deliver drugs and controlled surface chemistry. This review explores the current advancements in the application of nanomaterials in cancer immunotherapy, focusing on three primary areas: monoclonal antibodies, therapeutic cancer vaccines, and adoptive cell treatment. In adoptive cell therapy, nanomaterials enhance the expansion and targeting capabilities of immune cells, such as T cells, thereby improving their ability to locate and destroy cancer cells. For therapeutic cancer vaccines, nanoparticles serve as delivery vehicles that protect antigens from degradation and enhance their uptake by antigen-presenting cells, boosting the immune response against cancer. Monoclonal antibodies benefit from nanotechnology through improved delivery mechanisms and reduced off-target effects, which increase their specificity and effectiveness. By highlighting the intersection of nanotechnology and immunotherapy, we aim to underscore the transformative potential of nanomaterials in enhancing the effectiveness and safety of cancer immunotherapies. Nanoparticles' ability to deliver drugs and biomolecules precisely to tumor sites reduces systemic toxicity and enhances therapeutic outcomes.
Collapse
Affiliation(s)
- Ayushi Sharma
- Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University, Mathura, Uttar Pradesh-281406, India.
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj 382355, Gandhinagar, India
| |
Collapse
|
5
|
Wells K, Liu T, Zhu L, Yang L. Immunomodulatory nanoparticles activate cytotoxic T cells for enhancement of the effect of cancer immunotherapy. NANOSCALE 2024; 16:17699-17722. [PMID: 39257225 DOI: 10.1039/d4nr01780c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Cancer immunotherapy represents a promising targeted treatment by leveraging the patient's immune system or adoptive transfer of active immune cells to selectively eliminate cancer cells. Despite notable clinical successes, conventional immunotherapies face significant challenges stemming from the poor infiltration of endogenous or adoptively transferred cytotoxic T cells in tumors, immunosuppressive tumor microenvironment and the immune evasion capability of cancer cells, leading to limited efficacy in many types of solid tumors. Overcoming these hurdles is essential to broaden the applicability of immunotherapies. Recent advances in nanotherapeutics have emerged as an innovative tool to overcome these challenges and enhance the therapeutic potential of tumor immunotherapy. The unique biochemical and biophysical properties of nanomaterials offer advantages in activation of immune cells in vitro for cell therapy, targeted delivery, and controlled release of immunomodulatory agents in vivo. Nanoparticles are excellent carriers for tumor associated antigens or neoantigen peptides for tumor vaccine, empowering activation of tumor specific T cell responses. By precisely delivering immunomodulatory agents to the tumor site, immunoactivating nanoparticles can promote tumor infiltration of endogenous T cells or adoptively transferred T cells into tumors, to overcoming delivery and biological barriers in the tumor microenvironment, augmenting the immune system's ability to recognize and eliminate cancer cells. This review provides an overview of the current advances in immunotherapeutic approaches utilizing nanotechnology. With a focus on discussions concerning strategies to enhance activity and efficacy of cytotoxic T cells and explore the intersection of engineering nanoparticles and immunomodulation aimed at bolstering T cell-mediated immune responses, we introduce various nanoparticle formulations designed to deliver therapeutic payloads, tumor antigens and immunomodulatory agents for T cell activation. Diverse mechanisms through which nanoparticle-based approaches influence T cell responses by improving antigen presentation, promoting immune cell trafficking, and reprogramming immunosuppressive tumor microenvironments to potentiate anti-tumor immunity are examined. Additionally, the synergistic potential of combining nanotherapeutics with existing immunotherapies, such as immune checkpoint inhibitors and adoptive T cell therapies is explored. In conclusion, this review highlights emerging research advances on activation of cytotoxic T cells using nanoparticle agents to support the promises and potential applications of nanoparticle-based immunomodulatory agents for cancer immunotherapy.
Collapse
Affiliation(s)
- Kory Wells
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tongrui Liu
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
| | - Lei Zhu
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
6
|
Salazar VA, Comenge J, Suárez-López R, Burger JA, Sanders RW, Bastús NG, Jaime C, Joseph-Munne J, Puntes V. Gold Nanoparticle Virus-like Particles Presenting SARS-CoV-2 Spike Protein: Synthesis, Biophysical Properties and Immunogenicity in BALB/c Mice. Vaccines (Basel) 2024; 12:829. [PMID: 39203954 PMCID: PMC11359663 DOI: 10.3390/vaccines12080829] [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: 06/14/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 09/03/2024] Open
Abstract
Gold nanoparticles (AuNPs) decorated with antigens have recently emerged as promising tools for vaccine development due to their innate ability to provide stability to antigens and modulate immune responses. In this study, we have engineered deactivated virus-like particles (VLPs) by precisely functionalizing gold cores with coronas comprising the full SARS-CoV-2 spike protein (S). Using BALB/c mice as a model, we investigated the immunogenicity of these S-AuNPs-VLPs. Our results demonstrate that S-AuNPs-VLPs consistently enhanced antigen-specific antibody responses compared to the S protein free in solution. This enhancement included higher binding antibody titers, higher neutralizing capacity of antibodies, and stronger T-cell responses. Compared to the mRNA COVID-19 vaccine, where the S protein is synthesized in situ, S-AuNPs-VLPs induced comparable binding and neutralizing antibody responses, but substantially superior T-cell responses. In conclusion, our study highlights the potential of conjugated AuNPs as an effective antigen-delivery system for protein-based vaccines targeting a broad spectrum of infectious diseases and other emergent viruses.
Collapse
Affiliation(s)
- Vivian A. Salazar
- Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain; (V.A.S.); (J.C.)
| | - Joan Comenge
- Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain; (V.A.S.); (J.C.)
- Networking Research Centre for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Rosa Suárez-López
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (R.S.-L.); (C.J.)
| | - Judith A. Burger
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, The Netherlands; (J.A.B.); (R.W.S.)
| | - Rogier W. Sanders
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, The Netherlands; (J.A.B.); (R.W.S.)
| | - Neus G. Bastús
- Networking Research Centre for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and BIST, Campus Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Carlos Jaime
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (R.S.-L.); (C.J.)
| | - Joan Joseph-Munne
- Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain; (V.A.S.); (J.C.)
- Department of Microbiology, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain
| | - Victor Puntes
- Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain; (V.A.S.); (J.C.)
- Networking Research Centre for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and BIST, Campus Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
7
|
Nankivell V, Vidanapathirana AK, Hoogendoorn A, Tan JTM, Verjans J, Psaltis PJ, Hutchinson MR, Gibson BC, Lu Y, Goldys E, Zheng G, Bursill CA. Targeting macrophages with multifunctional nanoparticles to detect and prevent atherosclerotic cardiovascular disease. Cardiovasc Res 2024; 120:819-838. [PMID: 38696700 PMCID: PMC11218693 DOI: 10.1093/cvr/cvae099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/29/2024] [Accepted: 04/02/2024] [Indexed: 05/04/2024] Open
Abstract
Despite the emergence of novel diagnostic, pharmacological, interventional, and prevention strategies, atherosclerotic cardiovascular disease remains a significant cause of morbidity and mortality. Nanoparticle (NP)-based platforms encompass diverse imaging, delivery, and pharmacological properties that provide novel opportunities for refining diagnostic and therapeutic interventions for atherosclerosis at the cellular and molecular levels. Macrophages play a critical role in atherosclerosis and therefore represent an important disease-related diagnostic and therapeutic target, especially given their inherent ability for passive and active NP uptake. In this review, we discuss an array of inorganic, carbon-based, and lipid-based NPs that provide magnetic, radiographic, and fluorescent imaging capabilities for a range of highly promising research and clinical applications in atherosclerosis. We discuss the design of NPs that target a range of macrophage-related functions such as lipoprotein oxidation, cholesterol efflux, vascular inflammation, and defective efferocytosis. We also provide examples of NP systems that were developed for other pathologies such as cancer and highlight their potential for repurposing in cardiovascular disease. Finally, we discuss the current state of play and the future of theranostic NPs. Whilst this is not without its challenges, the array of multifunctional capabilities that are possible in NP design ensures they will be part of the next frontier of exciting new therapies that simultaneously improve the accuracy of plaque diagnosis and more effectively reduce atherosclerosis with limited side effects.
Collapse
Affiliation(s)
- Victoria Nankivell
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Achini K Vidanapathirana
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Ayla Hoogendoorn
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Johan Verjans
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Peter J Psaltis
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Mark R Hutchinson
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Brant C Gibson
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- School of Science, RMIT University, Melbourne, Victoria, Australia
| | - Yiqing Lu
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- School of Engineering, Macquarie University, Sydney, NSW, Australia
| | - Ewa Goldys
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Graduate School of Biomedical Engineering, University of New South Wales, High Street, NSW, 2052, Australia
| | - Gang Zheng
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, M5G 1L7, Canada
| | - Christina A Bursill
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| |
Collapse
|
8
|
Abdelwahab WM, Le-Vinh B, Riffey A, Hicks L, Buhl C, Ettenger G, Jackson KJ, Weiss AM, Miller S, Ryter K, Evans JT, Burkhart DJ. Promotion of Th17 Polarized Immunity via Co-Delivery of Mincle Agonist and Tuberculosis Antigen Using Silica Nanoparticles. ACS APPLIED BIO MATERIALS 2024; 7:3877-3889. [PMID: 38832760 DOI: 10.1021/acsabm.4c00245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Adjuvants and immunomodulators that effectively drive a Th17-skewed immune response are not part of the standard vaccine toolkit. Vaccine adjuvants and delivery technologies that can induce Th17 or Th1/17 immunity and protection against bacterial pathogens, such as tuberculosis (TB), are urgently needed. Th17-polarized immune response can be induced using agonists that bind and activate C-type lectin receptors (CLRs) such as macrophage inducible C-type lectin (Mincle). A simple but effective strategy was developed for codelivering Mincle agonists with the recombinant Mycobacterium tuberculosis fusion antigen, M72, using tunable silica nanoparticles (SNP). Anionic bare SNP, hydrophobic phenyl-functionalized SNP (P-SNP), and cationic amine-functionalized SNP (A-SNP) of different sizes were coated with three synthetic Mincle agonists, UM-1024, UM-1052, and UM-1098, and evaluated for adjuvant activity in vitro and in vivo. The antigen and adjuvant were coadsorbed onto SNP via electrostatic and hydrophobic interactions, facilitating multivalent display and delivery to antigen presenting cells. The cationic A-SNP showed the highest coloading efficiency for the antigen and adjuvant. In addition, the UM-1098-adsorbed A-SNP formulation demonstrated slow-release kinetics in vitro, excellent stability over 12 months of storage, and strong IL-6 induction from human peripheral blood mononuclear cells. Co-adsorption of UM-1098 and M72 on A-SNP significantly improved antigen-specific humoral and Th17-polarized immune responses in vivo in BALB/c mice relative to the controls. Taken together, A-SNP is a promising platform for codelivery and proper presentation of adjuvants and antigens and provides the basis for their further development as a vaccine delivery platform for immunization against TB or other diseases for which Th17 immunity contributes to protection.
Collapse
Affiliation(s)
- Walid M Abdelwahab
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Bao Le-Vinh
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Alexander Riffey
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Linda Hicks
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Cassandra Buhl
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - George Ettenger
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Konner J Jackson
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - Adam M Weiss
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Shannon Miller
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - Kendal Ryter
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - Jay T Evans
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - David J Burkhart
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| |
Collapse
|
9
|
Sau S, Dey A, Pal P, Das B, Maity KK, Dash SK, Tamili DK, Das B. Immunomodulatory and immune-toxicological role of nanoparticles: Potential therapeutic applications. Int Immunopharmacol 2024; 135:112251. [PMID: 38781608 DOI: 10.1016/j.intimp.2024.112251] [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: 02/13/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
Nowadays, Nanoparticle-based immunotherapeutic research has invoked global interest due to their unique properties. The immune system is a shielding structure that defends living things from external threats. Before the use of any materials in drug design, it is essential to study the immunological response to avoid triggering undesirable immune responses in the body. This review tries to summarize the properties, various applications, and immunotherapeutic aspects of NP-induced immunomodulation relating to therapeutic development and toxicity in human health. The role of NPs in the immune system and their modulatory functions, resulting in immunosuppression or immunostimulation, exerts benefits or dangers depending on their compositions, sizes, surface chemistry, and so forth. After NPs enter into the body, they can interact with body fluid exposing, them to different body proteins to form protein corona particles and other bio-molecules (DNA, RNA, sugars, etc.), which may alter their bioactivity. Phagocytes are the first immune cells that can interact with foreign materials including nanoparticles. Immunostimulation and immunosuppression operate in two distinct manners. Overall, functionalized nanocarriers optimized various therapeutic implications by stimulating the host immune system and regulating the tranquility of the host immune system. Among others, toxicity and bio-clearance of nanomaterials are always prime concerns at the preclinical and clinical stages before final approval. The interaction of nanoparticles with immune cells causes direct cell damage via apoptosis and necroses as well as immune signaling pathways also become influenced.
Collapse
Affiliation(s)
- Somnath Sau
- Department of Physiology and Natural Science Research Center of Belda College Affiliated from Vidyasagar University, Belda College, Belda-721424, Paschim Medinipur, West Bengal, India; Department of Nutrition and Coastal Environmental Studies, Egra S.S.B. College Research Centre, Affiliated from Vidyasagar University, Egra-721429, Purba Medinipur, West Bengal, India
| | - Alo Dey
- Department of Physiology and Natural Science Research Center of Belda College Affiliated from Vidyasagar University, Belda College, Belda-721424, Paschim Medinipur, West Bengal, India
| | - Pritam Pal
- Department of Physiology and Natural Science Research Center of Belda College Affiliated from Vidyasagar University, Belda College, Belda-721424, Paschim Medinipur, West Bengal, India
| | - Bishal Das
- Department of Physiology and Natural Science Research Center of Belda College Affiliated from Vidyasagar University, Belda College, Belda-721424, Paschim Medinipur, West Bengal, India; Department of Physiology, Debra Thana Sahid Kshudiram Smriti Mahavidyalaya, Debra-721124, Paschim Medinipur, West Bengal, India
| | - Kankan Kumar Maity
- Department of Chemistry and Natural Science Research Center of Belda College Affiliated from Vidyasagar University, Belda College, Belda-721424, Paschim Medinipur, West Bengal, India
| | - Sandeep Kumar Dash
- Department of Physiology, University of Gour Banga, Malda 732103, West Bengal, India
| | - Dipak Kumar Tamili
- Department of Zoology and Coastal Environmental Studies, Egra S.S.B. College Research Centre, Affiliated from Vidyasagar University, Egra-721429, Purba Medinipur, West Bengal, India
| | - Balaram Das
- Department of Physiology and Natural Science Research Center of Belda College Affiliated from Vidyasagar University, Belda College, Belda-721424, Paschim Medinipur, West Bengal, India.
| |
Collapse
|
10
|
Wang Y, Li G, Su J, Liu Y, Zhang X, Zhang G, Wu Z, Li J, Zhang Y, Wang X, Yang Z, Wang R, Wang C, Wang L, Sun F, Zhao W, Wang X, Peng X, Shao K. Spatiotemporal Controllable Sono-Nanovaccines Driven by Free-Field Based Whole-Body Ultrasound for Personalized Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307920. [PMID: 38308196 PMCID: PMC11005707 DOI: 10.1002/advs.202307920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/10/2024] [Indexed: 02/04/2024]
Abstract
Therapeutic cancer vaccines fail to produce satisfactory outcomes against solid tumors since vaccine-induced anti-tumor immunity is significantly hampered by immunosuppression. Generating an in situ cancer vaccine targeting immunological cold tumor microenvironment (TME) appears attractive. Here, a type of free-field based whole-body ultrasound (US)-driven nanovaccines are constructed, named G5-CHC-R, by conjugating the sonosensitizer, Chenghai chlorin (CHC) and the immunomodulator, resiquimod (R848) on top of a super small-sized dendrimeric nanoscaffold. Once entering tumors, R848 can be cleaved from a hypoxia-sensitive linker, thus modifying the TME via converting macrophage phenotypes. The animals bearing orthotopic pancreatic cancer with intestinal metastasis and breast cancer with lung metastasis are treated with G5-CHC-R under a free-field based whole-body US system. Benefit from the deep penetration capacity and highly spatiotemporal selectiveness, G5-CHC-R triggered by US represented a superior alternative for noninvasive irradiation of deep-seated tumors and magnification of local immune responses via driving mass release of tumor antigens and "cold-warm-hot" three-state transformation of TME. In addition to irradiating primary tumors, a robust adaptive anti-tumor immunity is potentiated, leading to successful induction of systemic tumor suppression. The sono-nanovaccines with good biocompatibility posed wide applicability to a broad spectrum of tumors, revealing immeasurable potential for translational research in oncology.
Collapse
Affiliation(s)
- Yang Wang
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Guangzhe Li
- State Key Laboratory of Fine ChemicalsDepartment of PharmacySchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Jianlong Su
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Yiming Liu
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Xiaomai Zhang
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Guanyi Zhang
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Zhihao Wu
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Jinrong Li
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Yuxuan Zhang
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Xu Wang
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Zejia Yang
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Ruimin Wang
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Chengdong Wang
- Nuclear MedicineFirst Affiliated Hospital of Dalian Medical UniversityDalian116021China
| | - Liu Wang
- State Key Laboratory of Fine ChemicalsDepartment of PharmacySchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Fangfang Sun
- Nuclear MedicineFirst Affiliated Hospital of Dalian Medical UniversityDalian116021China
| | - Weijie Zhao
- State Key Laboratory of Fine ChemicalsDepartment of PharmacySchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Xuejian Wang
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalian116021China
| | - Xiaojun Peng
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| | - Kun Shao
- State Key Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of TechnologyDalian116024China
| |
Collapse
|
11
|
Tang Y, Li L. The Application of Nanovaccines in Autoimmune Diseases. Int J Nanomedicine 2024; 19:367-388. [PMID: 38229706 PMCID: PMC10790641 DOI: 10.2147/ijn.s440612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/05/2024] [Indexed: 01/18/2024] Open
Abstract
Autoimmune diseases are diseases caused by the body's chronic immune responses to self-antigens and attacks on the host's own cells, tissues and organs. The dysfunction of innate immunity and adaptive immunity leads to the destruction of autoimmune tolerance, which is the most basic factor leading to pathogenesis. The optimal strategy for autoimmune diseases is to modify the host immune system to restore tolerance. The ideal effect of therapeutic autoimmune diseases is to eliminate the autoantigen-specific spontaneous immune response without interfering with the immune response against other antigens. Therapeutic nanovaccines that produce immune tolerance conform to this principle. Nanomaterials provide a platform for antigen loading and modification due to their unique physical and chemical properties. Nanovaccines based on nanomaterial technology can simultaneously enable antigens and adjuvants to be absorbed by immune cells and induce rapid and durable immunity. Nanovaccines have the advantages of being able to be designed and loaded and of better protecting antigens from premature degradation. Nanovaccines also have the ability to target specific tissues or cells through optimized design. We review the latest research progress of nanovaccines for autoimmune diseases and the design strategies of nanovaccines to promote the development of more effective nanovaccines for autoimmune diseases.
Collapse
Affiliation(s)
- Yuhong Tang
- Department of Dermatology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Lili Li
- Department of Dermatology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| |
Collapse
|
12
|
Zhou Z, Lian Y, Zhu L, Zhang H, Li Z, Wang M. Platinum Nanoparticles Prevent the Resistance of Pseudomonas aeruginosa to Ciprofloxacin and Imipenem: Mechanism Insights. ACS NANO 2023; 17:24685-24695. [PMID: 38048441 DOI: 10.1021/acsnano.3c04167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
Metal nanoparticles (MNPs) have recently gained extensive attention due to their broad-spectrum prospect, particularly in biomedical application. Here, we reveal that long-term exposure to platinum nanoparticles (Pt NPs) increases the susceptibility of Pseudomonas aeruginosa PAO1 to imipenem and ciprofloxacin. We exposed PAO1 to Pt NPs (a series of doses, varying from 0.125 to 35 μg/mL) for 60 days and characterized the evolved strains (ES) and compared with wild type (WT) to understand the mechanism of heightened sensitivity. We found that overexpression of oprD and downregulation of mexEF-oprN facilitate the intracellular accumulation of antibiotic, thus increasing susceptibility. Furthermore, loss-of-function mutations were discovered in regulators lasR and mexT. Cloning intact lasR from wild-type (WT) into ES slightly improves imipenem resistance. Strikingly, cloning mexT from WT into ES reverts the imipenem and ciprofloxacin resistance to the original level. Briefly, the increase of membrane permeability controlled by mexT made PAO1 greatly susceptible to imipenem and ciprofloxacin, and the decrease of quorum sensing mediated by lasR made PAO1 slightly susceptible to imipenem. Overall, these results reveal an antibiotic susceptibility mechanism from prolonged exposure to MNPs, which provides a promising approach to prevent antibiotic resistance.
Collapse
Affiliation(s)
- Zhiruo Zhou
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Yulu Lian
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Lin Zhu
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Haibo Zhang
- China National Demonstration Center for Experimental Chemistry, Wuhan University, Wuhan 430072, China
| | - Zhangqiang Li
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Meizhen Wang
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
- Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, Hangzhou 310012, China
| |
Collapse
|
13
|
Zhuang F, Xiang H, Huang B, Chen Y. Ultrasound-Triggered Cascade Amplification of Nanotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303158. [PMID: 37222084 DOI: 10.1002/adma.202303158] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/14/2023] [Indexed: 05/25/2023]
Abstract
Ultrasound (US)-triggered cascade amplification of nanotherapies has attracted considerable attention as an effective strategy for cancer treatment. With the remarkable advances in materials chemistry and nanotechnology, a large number of well-designed nanosystems have emerged that incorporate presupposed cascade amplification processes and can be activated to trigger therapies such as chemotherapy, immunotherapy, and ferroptosis, under exogenous US stimulation or specific substances generated by US actuation, to maximize antitumor efficacy and minimize detrimental effects. Therefore, summarizing the corresponding nanotherapies and applications based on US-triggered cascade amplification is essential. This review comprehensively summarizes and highlights the recent advances in the design of intelligent modalities, consisting of unique components, distinctive properties, and specific cascade processes. These ingenious strategies confer unparalleled potential to nanotherapies based on ultrasound-triggered cascade amplification and provide superior controllability, thus overcoming the unmet requirements of precision medicine and personalized treatment. Finally, the challenges and prospects of this emerging strategy are discussed and it is expected to encourage more innovative ideas and promote their further development.
Collapse
Affiliation(s)
- Fan Zhuang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, Shanghai, 200032, P. R. China
- Institute of Medical Ultrasound and Engineering, Fudan University, Shanghai, 200032, P. R. China
| | - Huijing Xiang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Beijian Huang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, Shanghai, 200032, P. R. China
- Institute of Medical Ultrasound and Engineering, Fudan University, Shanghai, 200032, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
14
|
Liu Y, Pan C, Wang K, Guo Y, Sun Y, Li X, Sun P, Wu J, Wang H, Zhu L. Preparation of a Klebsiella pneumoniae conjugate nanovaccine using glycol-engineered Escherichia coli. Microb Cell Fact 2023; 22:95. [PMID: 37149632 PMCID: PMC10163571 DOI: 10.1186/s12934-023-02099-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/17/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Engineered strains of Escherichia coli have been used to produce bioconjugate vaccines using Protein Glycan Coupling Technology (PGCT). Nanovaccines have also entered the vaccine development arena with advances in nanotechnology and have been significantly developed, but chassis cells for conjugate nanovaccines have not been reported. RESULTS To facilitate nanovaccine preparation, a generic recombinant protein (SpyCather4573) was used as the acceptor protein for O-linked glycosyltransferase PglL, and a glycol-engineered Escherichia coli strain with these two key components (SC4573 and PglL) integrated in its genome was developed in this study. The targeted glycoproteins with antigenic polysaccharides produced by our bacterial chassis can be spontaneously bound to proteinous nanocarriers with surface exposed SpyTag in vitro to form conjugate nanovaccines. To improve the yields of the targeted glycoprotein, a series of gene cluster deletion experiments was carried out, and the results showed that the deletion of the yfdGHI gene cluster increased the expression of glycoproteins. Using the updated system, to the best of our knowledge, we report for the first time the successful preparation of an effective Klebsiella pneumoniae O1 conjugate nanovaccine (KPO1-VLP), with antibody titers between 4 and 5 (Log10) after triple immunization and up to 100% protection against virulent strain challenge. CONCLUSIONS Our results define a convenient and reliable framework for bacterial glycoprotein vaccine preparation that is flexible and versatile, and the genomic stability of the engineered chassis cells promises a wide range of applications for biosynthetic glycobiology research.
Collapse
Affiliation(s)
- Yan Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Kangfeng Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
- College of Life Science, Hebei University, Baoding, 071002, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - YanGe Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiang Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Peng Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jun Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Hengliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China.
| |
Collapse
|
15
|
Singh R, Srivastava P, Manna PP. Chemokine-targeted nanoparticles: stimulation of the immune system in cancer immunotherapy. EXPLORATION OF IMMUNOLOGY 2023:123-147. [DOI: 10.37349/ei.2023.00093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/24/2023] [Indexed: 01/06/2025]
Abstract
Surgery, chemotherapy, radiation therapy, and immunotherapy are potential therapeutic choices for many malignant and metastatic cancers. Despite adverse side effects and pain, surgery and chemotherapy continue to be the most common cancer treatments. However, patients treated with immunotherapy had better cancer control than those who got other treatments. There are two methods to activate immunological pathways: systemically and locally. To modify the tumor microenvironment (TME), the former uses systemic cytokine/chemokine (CK) delivery, whilst the latter uses immunological checkpoints or small molecule inhibitors. Organic and inorganic nanomaterials (NMs) enhanced the efficacy of cancer immunotherapy. NMs can transmit drugs, peptides, antigens, antibodies, whole cell membranes, etc. Surface-modified NMs precisely target and enter the tissues. The inner core of surface-modified NMs is composed of chemicals with limited bioavailability and biocompatibility, resulting in prolonged blood retention and decreased renal clearance. These platforms hinder or prevent many immune cell activities and modify the TME, enhancing the efficiency of cancer immunotherapy. By inhibiting CK/CK receptor signaling, cell migration and other immune responses could be controlled. Developing CK-targeted nanoparticles (NPs) that inhibit CK signaling or take advantage of the ligand-receptor connection is possible. Surface chemical modification of NMs with CKs or specific peptides has several medicinal applications, including tissue-specific drug delivery and limited cell migration in cancer-afflicted conditions. This review covers current developments in the role of different groups of CK-loaded NP in tumor therapy targeting immune cells and cancer. It also covers the role of NP targeting CK signaling which aids in immunogenic cell death (ICD) and induction of antitumor immunity. In addition, CK gene silencing and its capacity to prevent cancer metastasis as well as inhibition of immune cell migration to modulate the TME are discussed.
Collapse
Affiliation(s)
- Ranjeet Singh
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Prateek Srivastava
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India;Current address: Postdoctoral Fellow, Ben-Gurion University of the Negev, Beersheba 8410501, Israel
| | - Partha Pratim Manna
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
16
|
Li S, Zhang MY, Yuan J, Zhang YX. Nano-vaccines for gene delivery against HIV-1 infection. Expert Rev Vaccines 2023; 22:315-326. [PMID: 36945780 DOI: 10.1080/14760584.2023.2193266] [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] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Over the last four decades, human immunodeficiency virus type 1 (HIV-1) infection has been a major public health concern. It is acknowledged that an effective vaccine remains the best hope for eliminating the HIV-1 pandemic. The prophylaxis of HIV-1 infection remains a central theme because of the absence of an available HIV-1 vaccine. The incapability of conventional delivery strategies to induce potent immunity is a crucial task to overcome and ultimately lead to a major obstacle in HIV-1 vaccine research. AREAS COVERED The literature search was conducted in the following databases: PubMed, Web of Science, and Embase. Nano-platforms based vaccines have proven prophylaxis of various diseases for effectively activating the immune system. Nano-vaccines, including non-viral and viral vectored nano-vaccines, are in a position to improve the effectiveness of HIV-1 antigen delivery and enhance the innate and adaptive immune responses against HIV-1. Compared to traditional vaccination strategies, genetic immunization can elicit a long-term immune response to provide protective immunity for HIV-1 prevention. EXPERT OPINION The research progress on nano-vaccines for gene delivery against HIV-1 was discussed. The vaccine strategies based on nano-platforms that are being applied to stimulate effective HIV-1-specific cellular and humoral immune responses were particularly emphasized.
Collapse
Affiliation(s)
- Shuang Li
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Meng-Yue Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jie Yuan
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yi-Xuan Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| |
Collapse
|
17
|
Liu Y, Zhang M, Wang X, Yang F, Cao Z, Wang L, Liu J. Dressing Bacteria With a Hybrid Immunoactive Nanosurface to Elicit Dual Anticancer and Antiviral Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210949. [PMID: 36564893 DOI: 10.1002/adma.202210949] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Indexed: 06/17/2023]
Abstract
Approaches capable of simultaneously treating cancer and protecting susceptible patients from lethal infections such as coronavirus disease 2019, are highly desirable but prove to be difficult. Here, dressing bacteria with a hybrid immunoactive nanosurface is reported to elicit dual anticancer and antiviral immunity. A combination of a checkpoint blocking antibody and a virus-specific antigen is covalently conjugated to polydopamine nanoparticles, which can be anchored onto bacterial surface, by a one-step in situ polymerization of dopamine under a cell-friendly condition. By virtue of the ability to colonize and penetrate deep tumor tissue, dressed bacteria enable sustained release and expanded exposure of carried immunoactivators to stimulate immune cells. In addition to a carrier role, bacteria are able to further provoke innate immunity due to the native immunogenicity of the pathogen-associated molecular patterns. Immunization with dressed bacteria promotes the maturation, and activation of antigen-presenting cells, which induces robust humoral and cellular immune responses in tumor-bearing mice. As evidenced by efficient production of viral-antigen-specific immunoglobulin G antibody in serum and significantly suppressed tumor growth in different models, dressing bacteria with a hybrid immunoactive nanosurface paves an avenue to prepare next-generation therapeutics for synergistic treatment and prevention.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mengmeng Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xinyue Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Fengmin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhenping Cao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lu Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jinyao Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
18
|
Chen H, Li Y, Li L, Yang Z, Wen Z, Liu L, Liu H, Chen Y. Carrier-free subunit nanovaccine amplifies immune responses against tumors and viral infections. Acta Biomater 2023; 158:525-534. [PMID: 36572250 DOI: 10.1016/j.actbio.2022.12.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/09/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022]
Abstract
Codelivering subunit antigens and Toll-like receptor (TLR) molecular adjuvants via nanocarriers can stimulate potent innate and specific immune responses. Simple and effective nanovaccines fabrication is crucial for application. However, most nanovaccines were fabricated by introducing additional delivery materials, increasing safety risk, cost and processing complexity. Herein, a carrier-free nanovaccine was facilely prepared using a TLR1/TLR2 adjuvant, Diprovocim, rich in benzene rings that could interact with aromatic residues in subunit antigens through π-π stacking without additional materials. The carrier-free nanovaccines with a narrow size distribution could target lymph nodes (LNs) after intravenous injection to mice. The carrier-free nanovaccines based on ovalbumin (OVA) can stimulate strong antibody titers and CD4+ and CD8+ T cell immune responses in mice, and it synergized with anti-PD1 showing a potent tumor suppression in B16F10-OVA tumor model of mice. Furthermore, the carrier-free nanovaccine with glycoprotein E (gE), a glycoprotein of the varicella-zoster virus (VZV), also showed potent humoral and cellular immune responses. Therefore, using subunit proteins to support Diprovocim by π-π stacking provides a new approach for the preparation and application of novel vaccines for tumor therapy and prevention of infectious diseases. STATEMENT OF SIGNIFICANCE: Codelivering subunit antigens and adjuvants via nanocarriers stimulate potent innate and specific immune responses. However, existing delivery materials for fabricating nanovaccines will inevitably increase the cost of preparation, controllability, process complexity and safety assessment. Therefore, this study easily prepared carrier-free nanovaccines using the benzene ring-rich TLR1/TLR2 adjuvant Diprovocim, which can interact with aromatic residues in subunit antigens via π-π stacking without additional materials. The carrier-free nanovaccines of OVA demonstrated a potent tumor inhibition in treating melanoma in combination with anti-PD1. And the nanovaccines of gE stimulated a strong antibody titer and cellular immune response for herpes zoster. Thus, the present study provides a new approach for the preparation of subunit vaccines to combat various cancers and virus infections.
Collapse
Affiliation(s)
- Haolin Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yuhui Li
- Department of Pathology, Sun Yat-sen Memorial Hospital, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen University, Guangzhou, 510275, China
| | - Liyan Li
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275, China
| | - Zeyu Yang
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275, China
| | - Zhenfu Wen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275, China
| | - Lixin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275, China;; State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Hong Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275, China;.
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275, China;; State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
19
|
Yi Y, Yu M, Li W, Zhu D, Mei L, Ou M. Vaccine-like nanomedicine for cancer immunotherapy. J Control Release 2023; 355:760-778. [PMID: 36822241 DOI: 10.1016/j.jconrel.2023.02.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023]
Abstract
The successful clinical application of immune checkpoint blockade (ICB) and chimeric antigen receptor T cells (CAR-T) therapeutics has attracted extensive attention to immunotherapy, however, their drawbacks such as limited specificity, persistence and toxicity haven't met the high expectations on efficient cancer treatments. Therapeutic cancer vaccines which instruct the immune system to capture tumor specific antigens, generate long-term immune memory and specifically eliminate cancer cells gradually become the most promising strategies to eradicate tumor. However, the disadvantages of some existing vaccines such as weak immunogenicity and in vivo instability have restricted their development. Nanotechnology has been recently incorporated into vaccine fabrication and exhibited promising results for cancer immunotherapy. Nanoparticles promote the stability of vaccines, as well as enhance antigen recognition and presentation owing to their nanometer size which promotes internalization of antigens by phagocytic cells. The surface modification with targeting units further permits the delivery of vaccines to specific cells. Meanwhile, nanocarriers with adjuvant effect can improve the efficacy of vaccines. In addition to classic vaccines composed of antigens and adjuvants, the nanoparticle-mediated chemotherapy, radiotherapy and certain other therapeutics could induce the release of tumor antigens in situ, which therefore effectively simulate antitumor immune responses. Such vaccine-like nanomedicine not only kills primary tumors, but also prevents tumor recurrence and helps eliminate metastatic tumors. Herein, we introduce recent developments in nanoparticle-based delivery systems for antigen delivery and in situ antitumor vaccination. We will also discuss the remaining opportunities and challenges of nanovaccine in clinical translation towards cancer treatment.
Collapse
Affiliation(s)
- Yunfei Yi
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China; School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Meitong Ou
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
20
|
Jiang H, Liu R, Wang L, Wang X, Zhang M, Lin S, Cao Z, Wu F, Liu Y, Liu J. Chiral-Selective Antigen-Presentation by Supramolecular Chiral Polymer Micelles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208157. [PMID: 36398497 DOI: 10.1002/adma.202208157] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/14/2022] [Indexed: 06/16/2023]
Abstract
Chirality is ubiquitous in biological systems, which is closely related to biological functions, life processes, and even the pathogenesis of diseases. However, the interface between the chirality of synthetic materials and organisms, particularly the immune system, remains poorly understood. Here, supramolecular chiral polymer micelles (SCPMs) are prepared by complexing antigenic proteins with chiral amino acid-modified polyethyleneimine. The introduction of chirality not only reduces the toxicity of cationic polymer, but also benefits cell uptake and antigen presentation. Especially, D-chirality presents the lowest cytotoxicity, while promoting the highest expression level of costimulatory molecules on dendritic cells compared to L-chirality and achirality. The superiority of D-chirality to stimulate dendritic cell maturation is supported by immunization with D-SCPMs, which achieves significant antigen-specific proliferation of T cells in the spleen, lymph nodes, and tumor of mice. Chirality-mediated antigen processing and presentation are demonstrated by D-SCPMs self-assembled from chiral alkaline histidine or neutral phenylalanine modified polyethyleneimine and tumor associated ovalbumin or severe acute respiratory syndrome coronavirus 2 spike 1 antigenic protein. Immunoactivation enabled by D-chirality opens a window to prepare potent nanotherapeutics for disease prevention and treatment.
Collapse
Affiliation(s)
- Hejin Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Rui Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lu Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xinyue Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mengmeng Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Sisi Lin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhenping Cao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Feng Wu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jinyao Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
21
|
Polyelectrolyte Coating of Ferumoxytol Differentially Impacts the Labeling of Inflammatory and Steady-State Dendritic Cell Subtypes. Biomedicines 2022; 10:biomedicines10123137. [PMID: 36551893 PMCID: PMC9776020 DOI: 10.3390/biomedicines10123137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Engineered magnetic nanoparticles (MNPs) are emerging as advanced tools for medical applications. The coating of MNPs using polyelectrolytes (PEs) is a versatile means to tailor MNP properties and is used to optimize MNP functionality. Dendritic cells (DCs) are critical regulators of adaptive immune responses. Functionally distinct DC subsets exist, either under steady-state or inflammatory conditions, which are explored for the specific treatment of various diseases, such as cancer, autoimmunity, and transplant rejection. Here, the impact of the PE coating of ferumoxytol for uptake into both inflammatory and steady-state DCs and the cellular responses to MNP labeling is addressed. Labeling efficiency by uncoated and PE-coated ferumoxytol is highly variable in different DC subsets, and PE coating significantly improves the labeling of steady-state DCs. Uncoated ferumoxytol results in increased cytotoxicity of steady-state DCs after labeling, which is abolished by the PE coating, while no increased cell death is observed in inflammatory DCs. Furthermore, uncoated and PE-coated ferumoxytol appear immunologically inert in inflammatory DCs, but they induce activation of steady-state DCs. These results show that the PE coating of MNPs can be applied to endow particles with desired properties for enhanced uptake and cell type-specific responses in distinct target DC populations.
Collapse
|
22
|
Dai H, Fan Q, Wang C. Recent applications of immunomodulatory biomaterials for disease immunotherapy. EXPLORATION (BEIJING, CHINA) 2022; 2:20210157. [PMID: 37324799 PMCID: PMC10191059 DOI: 10.1002/exp.20210157] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/25/2022] [Indexed: 06/16/2023]
Abstract
Immunotherapy is used to regulate systemic hyperactivation or hypoactivation to treat various diseases. Biomaterial-based immunotherapy systems can improve therapeutic effects through targeted drug delivery, immunoengineering, etc. However, the immunomodulatory effects of biomaterials themselves cannot be neglected. In this review, we outline biomaterials with immunomodulatory functions discovered in recent years and their applications in disease treatment. These biomaterials can treat inflammation, tumors, or autoimmune diseases by regulating immune cell function, exerting enzyme-like activity, neutralizing cytokines, etc. The prospects and challenges of biomaterial-based modulation of immunotherapy are also discussed.
Collapse
Affiliation(s)
- Huaxing Dai
- Institute of Functional Nano & Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐Based Functional Materials & DevicesSoochow University199 Ren'ai RoadSuzhouJiangsuChina
| | - Qin Fan
- Key Laboratory for Organic Electronics & Information Displays (KLOEID)Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM) and School of Materials Science and EngineeringNanjing University of Posts & TelecommunicationsNanjingChina
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐Based Functional Materials & DevicesSoochow University199 Ren'ai RoadSuzhouJiangsuChina
| |
Collapse
|
23
|
Wang Q, Dong Z, Lou F, Yin Y, Zhang J, Wen H, Lu T, Wang Y. Phenylboronic ester-modified polymeric nanoparticles for promoting TRP2 peptide antigen delivery in cancer immunotherapy. Drug Deliv 2022; 29:2029-2043. [PMID: 35766157 PMCID: PMC9248950 DOI: 10.1080/10717544.2022.2086941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022] Open
Abstract
The tremendous development of peptide-based cancer vaccine has attracted incremental interest as a powerful approach in cancer management, prevention and treatment. As successful as tumor vaccine has been, major challenges associated with achieving efficient immune response against cancer are (1) drainage to and retention in lymph nodes; (2) uptake by dendritic cells (DCs); (3) activation of DCs. In order to overcome these barriers, here we construct PBE-modified TRP2 nanovaccine, which comprises TRP2 peptide tumor antigen and diblock copolymer PEG-b-PAsp grafted with phenylboronic ester (PBE). We confirmed that this TRP2 nanovaccine can be effectively trapped into lymph node, uptake by dendritic cells and induce DC maturation, relying on increased negative charge, ROS response and pH response. Consistently, this vehicle loaded with TRP2 peptide could boost the strongest T cell immune response against melanoma in vivo and potentiate antitumor efficacy both in tumor prevention and tumor treatment without any exogenous adjuvant. Furthermore, the TRP2 nanovaccine can suppress the tumor growth and prolong animal survival time, which may result from its synergistic effect of inhibiting tumor immunosuppression and increasing cytotoxic lymphocyte (CTL) response. Hence this type of PBE-modified nanovaccine would be widely used as a simple, safe and robust platform to deliver other antigen in cancer immunotherapy.
Collapse
Affiliation(s)
- Qiyan Wang
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
- Immunology Research program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan, USA
| | - Zhipeng Dong
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fangning Lou
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yunxue Yin
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jiahao Zhang
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Hanning Wen
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Tao Lu
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yue Wang
- Key Laboratory of Biomedical Functional Materials, School of Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
24
|
Wu Z, Setyawati MI, Lim HK, Ng KW, Tay CY. Nanoparticle-induced chemoresistance: the emerging modulatory effects of engineered nanomaterials on human intestinal cancer cell redox metabolic adaptation. NANOSCALE 2022; 14:14491-14507. [PMID: 36106385 DOI: 10.1039/d2nr03893e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The widespread use of engineered nanomaterials (ENMs) in food products necessitates the understanding of their impact on the gastrointestinal tract (GIT). Herein, we screened several representative food-borne comparator ENMs (i.e. ZnO, SiO2 and TiO2 nanoparticles (NPs)) and report that human colon cancer cells can insidiously exploit ZnO NP-induced adaptive response to acquire resistance against several chemotherapeutic drugs. By employing a conditioning and challenge treatment regime, we demonstrate that repeated exposure to a non-toxic dose of ZnO NPs (20 μM) could dampen the efficacy of cisplatin, paclitaxel and doxorubicin by 10-50% in monolayer culture and 3D spheroids of human colon adenocarcinoma cells. Structure-activity relationship studies revealed a complex interplay between nanoparticle surface chemistry and cell type in determining the chemoresistance-inducing effect, with silica coated ZnO NPs having a negligible influence on the anticancer treatment. Mechanistically, we showed that the pro-survival paracrine signaling was potentiated and propagated by a subset of ZnO NP "stressed" (Zn2++/ROS+) cells to the surrounding "bystander" (Zn2++/ROS-) cells. Transcriptome profiling, bioinformatics analysis and siRNA gene knockdown experiments revealed the nuclear factor erythroid 2-related factor 2 (Nrf2) as the key modulator of the ZnO NP-induced drug resistance. Our findings suggest that a ROS-inducing ENM can emerge as a nano-stressor, capable of regulating the chemosensitivity of colon cancer cells.
Collapse
Affiliation(s)
- Zhuoran Wu
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore.
| | - Magdiel Inggrid Setyawati
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore.
| | - Hong Kit Lim
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore.
| | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore.
- Environmental Chemistry and Materials Centre, Nanyang Environment & Water Research Institute, 1 Cleantech Loop, CleanTech One, Singapore 637141, Singapore
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Chor Yong Tay
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore.
- Environmental Chemistry and Materials Centre, Nanyang Environment & Water Research Institute, 1 Cleantech Loop, CleanTech One, Singapore 637141, Singapore
| |
Collapse
|
25
|
Wu Z, Chan B, Low J, Chu JJH, Hey HWD, Tay A. Microbial resistance to nanotechnologies: An important but understudied consideration using antimicrobial nanotechnologies in orthopaedic implants. Bioact Mater 2022; 16:249-270. [PMID: 35415290 PMCID: PMC8965851 DOI: 10.1016/j.bioactmat.2022.02.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
Microbial resistance to current antibiotics therapies is a major cause of implant failure and adverse clinical outcomes in orthopaedic surgery. Recent developments in advanced antimicrobial nanotechnologies provide numerous opportunities to effective remove resistant bacteria and prevent resistance from occurring through unique mechanisms. With tunable physicochemical properties, nanomaterials can be designed to be bactericidal, antifouling, immunomodulating, and capable of delivering antibacterial compounds to the infection region with spatiotemporal accuracy. Despite its substantial advancement, an important, but under-explored area, is potential microbial resistance to nanomaterials and how this can impact the clinical use of antimicrobial nanotechnologies. This review aims to provide a better understanding of nanomaterial-associated microbial resistance to accelerate bench-to-bedside translations of emerging nanotechnologies for effective control of implant associated infections.
Collapse
Affiliation(s)
- Zhuoran Wu
- Institute of Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Brian Chan
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Jessalyn Low
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Justin Jang Hann Chu
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117545, Singapore
- Infectious Disease Programme, Yong Loo Lin School of Medicine, National University of Singapore, 117547, Singapore
- Institute of Molecular and Cell Biology, 35 Agency for Science, Technology and Research, 138673, Singapore
| | - Hwee Weng Dennis Hey
- National University Health System, National University of Singapore, 119228, Singapore
| | - Andy Tay
- Institute of Health Innovation & Technology, National University of Singapore, 117599, Singapore
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Tissue Engineering Programme, National University of Singapore, 117510, Singapore
| |
Collapse
|
26
|
Ko CN, Zang S, Zhou Y, Zhong Z, Yang C. Nanocarriers for effective delivery: modulation of innate immunity for the management of infections and the associated complications. J Nanobiotechnology 2022; 20:380. [PMID: 35986268 PMCID: PMC9388998 DOI: 10.1186/s12951-022-01582-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/01/2022] [Indexed: 12/24/2022] Open
Abstract
Innate immunity is the first line of defense against invading pathogens. Innate immune cells can recognize invading pathogens through recognizing pathogen-associated molecular patterns (PAMPs) via pattern recognition receptors (PRRs). The recognition of PAMPs by PRRs triggers immune defense mechanisms and the secretion of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6. However, sustained and overwhelming activation of immune system may disrupt immune homeostasis and contribute to inflammatory disorders. Immunomodulators targeting PRRs may be beneficial to treat infectious diseases and their associated complications. However, therapeutic performances of immunomodulators can be negatively affected by (1) high immune-mediated toxicity, (2) poor solubility and (3) bioactivity loss after long circulation. Recently, nanocarriers have emerged as a very promising tool to overcome these obstacles owning to their unique properties such as sustained circulation, desired bio-distribution, and preferred pharmacokinetic and pharmacodynamic profiles. In this review, we aim to provide an up-to-date overview on the strategies and applications of nanocarrier-assisted innate immune modulation for the management of infections and their associated complications. We first summarize examples of important innate immune modulators. The types of nanomaterials available for drug delivery, as well as their applications for the delivery of immunomodulatory drugs and vaccine adjuvants are also discussed.
Collapse
|
27
|
From vaccines to nanovaccines: A promising strategy to revolutionize rheumatoid arthritis treatment. J Control Release 2022; 350:107-121. [PMID: 35977582 DOI: 10.1016/j.jconrel.2022.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Rheumatoid arthritis (RA) is a joint-related autoimmune disease that is difficult to cure. Most therapeutics act to alleviate the symptoms but not correct the causes of RA. Novel strategies that specifically target the causes are highly needed for RA management. Currently, early interruption of RA is increasingly suggested but the corresponding therapeutics are not available. Vaccines that have shown great success to combat infection, cancer, degenerative diseases, autoimmune diseases, etc. are ideal candidates for a new generation of anti-RA therapeutics to correct the causes and prevent RA or interrupt RA in early phases. Anti-RA vaccines can be divided into two major categories. One is to induce neutralizing antibodies and the other is to induce antigen-specific immune tolerance. The vaccines are inherently linked to nanotechnology because they usually need a biomacromolecule or carrier to provoke sufficient immune responses. In the past decade, designed nanocarriers such as nanoparticles, liposomes, nanoemulsion, etc., have been applied to optimize the vaccines for autoimmune disease treatment. Nanotechnology endows vaccines with a higher biostability, tunable in vivo behavior, better targeting, co-delivery with stimulatory agents, regulatory effects on immune responses, etc. In this review, unmet medical needs for RA treatment and anti-RA vaccinology are first introduced. The development of anti-RA therapies from vaccines to nanovaccines are then reviewed and perspectives on how nanotechnology promotes vaccine development and advancement are finally provided. In addition, challenges for anti-RA vaccine development are summarized and advantages of nanovaccines are analyzed. In conclusion, nanovaccines will be a promising strategy to revolutionize the treatment of RA by correcting the causes in an early phase of RA.
Collapse
|
28
|
Ortega DD, Pavlakovich N, Shon YS. Effects of lipid bilayer encapsulation and lipid composition on the catalytic activity and colloidal stability of hydrophobic palladium nanoparticles in water. RSC Adv 2022; 12:21866-21874. [PMID: 36043067 PMCID: PMC9361304 DOI: 10.1039/d2ra03974e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/25/2022] [Indexed: 11/21/2022] Open
Abstract
This article shows the preparation of a lipid-nanoparticle assembly (LNA) which contains hydrophobic palladium nanoparticles (PdNPs) within the hydrophobic regions of the liposomal micelles. To understand the colloidal stability and catalytic activity of LNAs, the structure–property relationships of LNAs are investigated by manipulating the lipid composition and reaction temperature. The studies of LNAs using dynamic light scattering (DLS), differential scanning calorimetry (DSC), and transmission electron microscopy (TEM) show decreased colloidal stability with the incorporation of PdNPs compared to their counterpart 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) liposomes without PdNPs. LNAs with PdNPs catalyze the hydrogenation of 1-octene and its isomers to octane under one atm hydrogen gas and at room temperature within 24 h. The kinetic studies show that the isomerization of 1-octene to 2-octene occurs more favorably in the early stage of the reactions, which is followed by the subsequent hydrogenation of all octene isomers. The studies on temperature effects indicate that there is a significant increase in conversion yield of substrates when the reaction temperature increases from 22 to 37 °C, which correspond to room temperature and biological temperature, respectively. Phase transition of DSPC-PdNP LNAs from gel to liquid crystalline phase changing the fluidity of the bilayer is proposed to be the main reason for dramatic increases in the catalytic activity of the LNAs. It is also found that the rate of hydrogenation is dependent on the lipid composition of LNAs with the presence of cholesterol having a negative influence on the catalytic activity of LNAs while increasing their colloidal stability. Hydrophobic micellization effect and dynamic lipid bilayer–substrate interactions enhance the catalytic activity of hydrophobic Pd nanoparticles embedded in liposomal assemblies.![]()
Collapse
Affiliation(s)
- Dominick D Ortega
- Department of Chemistry and Biochemistry, California State University, Long Beach 1250 Bellflower Blvd. Long Beach CA 90840 Unites States
| | - Nicholas Pavlakovich
- Department of Chemistry and Biochemistry, California State University, Long Beach 1250 Bellflower Blvd. Long Beach CA 90840 Unites States
| | - Young-Seok Shon
- Department of Chemistry and Biochemistry, California State University, Long Beach 1250 Bellflower Blvd. Long Beach CA 90840 Unites States
| |
Collapse
|
29
|
Huang Z, Callmann CE, Wang S, Vasher MK, Evangelopoulos M, Petrosko SH, Mirkin CA. Rational Vaccinology: Harnessing Nanoscale Chemical Design for Cancer Immunotherapy. ACS CENTRAL SCIENCE 2022; 8:692-704. [PMID: 35756370 PMCID: PMC9228553 DOI: 10.1021/acscentsci.2c00227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Indexed: 05/12/2023]
Abstract
Cancer immunotherapy is a powerful treatment strategy that mobilizes the immune system to fight disease. Cancer vaccination is one form of cancer immunotherapy, where spatiotemporal control of the delivery of tumor-specific antigens, adjuvants, and/or cytokines has been key to successfully activating the immune system. Nanoscale materials that take advantage of chemistry to control the nanoscale structural arrangement, composition, and release of immunostimulatory components have shown significant promise in this regard. In this Outlook, we examine how the nanoscale structure, chemistry, and composition of immunostimulatory compounds can be modulated to maximize immune response and mitigate off-target effects, focusing on spherical nucleic acids as a model system. Furthermore, we emphasize how chemistry and materials science are driving the rational design and development of next-generation cancer vaccines. Finally, we identify gaps in the field that should be addressed moving forward and outline future directions to galvanize researchers from multiple disciplines to help realize the full potential of this form of cancer immunotherapy through chemistry and rational vaccinology.
Collapse
Affiliation(s)
- Ziyin Huang
- Department
of Materials Science and Engineering, International Institute for Nanotechnology, Department of Chemistry, Interdisciplinary
Biological Sciences Graduate Program, andDepartment of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Cassandra E. Callmann
- Department
of Materials Science and Engineering, International Institute for Nanotechnology, Department of Chemistry, Interdisciplinary
Biological Sciences Graduate Program, andDepartment of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Shuya Wang
- Department
of Materials Science and Engineering, International Institute for Nanotechnology, Department of Chemistry, Interdisciplinary
Biological Sciences Graduate Program, andDepartment of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Matthew K. Vasher
- Department
of Materials Science and Engineering, International Institute for Nanotechnology, Department of Chemistry, Interdisciplinary
Biological Sciences Graduate Program, andDepartment of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Michael Evangelopoulos
- Department
of Materials Science and Engineering, International Institute for Nanotechnology, Department of Chemistry, Interdisciplinary
Biological Sciences Graduate Program, andDepartment of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Sarah Hurst Petrosko
- Department
of Materials Science and Engineering, International Institute for Nanotechnology, Department of Chemistry, Interdisciplinary
Biological Sciences Graduate Program, andDepartment of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Chad A. Mirkin
- Department
of Materials Science and Engineering, International Institute for Nanotechnology, Department of Chemistry, Interdisciplinary
Biological Sciences Graduate Program, andDepartment of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
30
|
Abstract
A favorable outcome of the COVID-19 crisis might be achieved with massive vaccination. The proposed vaccines contain several different vaccine active principles (VAP), such as inactivated virus, antigen, mRNA, and DNA, which are associated with either standard adjuvants or nanomaterials (NM) such as liposomes in Moderna's and BioNTech/Pfizer's vaccines. COVID-19 vaccine adjuvants may be chosen among liposomes or other types of NM composed for example of graphene oxide, carbon nanotubes, micelles, exosomes, membrane vesicles, polymers, or metallic NM, taking inspiration from cancer nano-vaccines, whose adjuvants may share some of their properties with those of viral vaccines. The mechanisms of action of nano-adjuvants are based on the facilitation by NM of targeting certain regions of immune interest such as the mucus, lymph nodes, and zones of infection or blood irrigation, the possible modulation of the type of attachment of the VAP to NM, in particular VAP positioning on the NM external surface to favor VAP presentation to antigen presenting cells (APC) or VAP encapsulation within NM to prevent VAP degradation, and the possibility to adjust the nature of the immune response by tuning the physico-chemical properties of NM such as their size, surface charge, or composition. The use of NM as adjuvants or the presence of nano-dimensions in COVID-19 vaccines does not only have the potential to improve the vaccine benefit/risk ratio, but also to reduce the dose of vaccine necessary to reach full efficacy. It could therefore ease the overall spread of COVID-19 vaccines within a sufficiently large portion of the world population to exit the current crisis.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, 75005 Paris, France. .,Nanobacterie SARL, 36 Boulevard Flandrin, 75116, Paris, France.,Institute of Anatomy, UZH University of Zurich, Instiute of Anatomy, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|
31
|
S K, S R, S H. Gold nanoparticles: a novel paradigm for targeted drug delivery. INORG NANO-MET CHEM 2022. [DOI: 10.1080/24701556.2021.2025077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Kamalavarshini S
- School of Life Sciences, B.S. Abdur Rahman Crescent institute of Science and Technology, Chennai, India
| | - Ranjani S
- School of Life Sciences, B.S. Abdur Rahman Crescent institute of Science and Technology, Chennai, India
| | - Hemalatha S
- School of Life Sciences, B.S. Abdur Rahman Crescent institute of Science and Technology, Chennai, India
| |
Collapse
|
32
|
Mi XJ, Xu XY, Choi HS, Kim H, Cho IH, Yi TH, Kim YJ. The Immune-Enhancing Properties of Hwanglyeonhaedok-Tang-Mediated Biosynthesized Gold Nanoparticles in Macrophages and Splenocytes. Int J Nanomedicine 2022; 17:477-494. [PMID: 35125869 PMCID: PMC8812323 DOI: 10.2147/ijn.s338334] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/05/2021] [Indexed: 12/19/2022] Open
Abstract
Background Despite great advances in the field of immunotherapy, there is still a need for novel and effective immunostimulants to overcome challenges, such as instability and autoinflammatory toxicity, associated with conventional immunostimulants. Nanotechnology provides the possibility to overcome these challenges. The well-known classical Chinese formula, Hwanglyeonhaedok-tang (HHT) has been widely used to treat immune-related diseases in clinical practice. Methods We developed novel gold nanoparticles (AuNPs) utilizing one-pot synthesis with the herbal formula-HHT. The optimal conditions for HHT-AuNP biosynthesis were established, and physicochemical properties of the optimized HHT-AuNPs were identified using various spectrometric and microscopic techniques. Bio-TEM analysis revealed that HHT-AuNPs were highly engulfed within RAW264.7 cells without inducing cytotoxicity. The effect of HHT-AuNPs on immunostimulatory activity was evaluated in innate and adaptive immune cells (RAW264.7 macrophages and ICR mice splenocytes) using qRT-PCR, immunoblotting, and ELISA. Results The HHT-AuNPs remarkably increased the nitric oxide (NO) and immune-related cytokines production by activating the mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) signaling pathways in RAW264.7 cells. Furthermore, HHT-AuNPs exerted immunostimulatory effects on mouse splenocytes by priming T/B-cells and macrophages. Discussion The present study is the first to demonstrate that HHT-AuNPs could be utilized as immunostimulators to activate both innate and adaptive immune systems. These results provide a foundation for the application of traditional Chinese medicinal formulae in the field of nanomedicine.
Collapse
Affiliation(s)
- Xiao-Jie Mi
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, 17104, Gyeonggi-do, Republic of Korea
| | - Xing Yue Xu
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, 17104, Gyeonggi-do, Republic of Korea
| | - Han Sol Choi
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, 17104, Gyeonggi-do, Republic of Korea
| | - Hoon Kim
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, 17104, Gyeonggi-do, Republic of Korea
| | - Ik Hyun Cho
- Department of Science in Korean Medicine and Brain Korea 21 Plus Program, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Tae-Hoo Yi
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, 17104, Gyeonggi-do, Republic of Korea
| | - Yeon-Ju Kim
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, 17104, Gyeonggi-do, Republic of Korea
- Correspondence: Yeon-Ju Kim; Ik Hyun Cho Tel +82-31-201-5634Fax +82-31-204-8116 Email ;
| |
Collapse
|
33
|
Suzuki K, Yoshizaki Y, Horii K, Murase N, Kuzuya A, Ohya Y. Preparation of hyaluronic acid-coated polymeric micelles for nasal vaccine delivery. Biomater Sci 2022; 10:1920-1928. [DOI: 10.1039/d1bm01985f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hyaluronic acid (HA)-coated biodegradable polymeric micelles were developed as nanoparticulate vaccine delivery systems to establish an effective nasal vaccine. We previously reported HA-coated micelles prepared by forming a polyion complex...
Collapse
|
34
|
Wang L, Li G, Cao L, Dong Y, Wang Y, Wang S, Li Y, Guo X, Zhang Y, Sun F, Du X, Su J, Li Q, Peng X, Shao K, Zhao W. An ultrasound-driven immune-boosting molecular machine for systemic tumor suppression. SCIENCE ADVANCES 2021; 7:eabj4796. [PMID: 34669472 PMCID: PMC8528430 DOI: 10.1126/sciadv.abj4796] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/30/2021] [Indexed: 06/13/2023]
Abstract
Exploring facile and effective therapeutic modalities for synergistically controlling primary tumor and metastasis remains a pressing clinical need. Sonodynamic therapy (SDT) offers the possibility of noninvasively eradicating local solid tumors, but lacks antimetastatic activity because of its limited ability in generating systemic antitumor effect. Here, we exploited a previously unidentified ultrasound-driven “molecular machine,” DYSP-C34 (C34 for short), with multiple attractive features, emerging from preferential tumor accumulation, potent ultrasound-triggered cytotoxicity, and intrinsic immune-boosting capacity. Driven by the ultrasound, C34 functioned not only as a tumor cell killing reagent but also as an immune booster that could potentiate robust adaptive antitumor immunity by directly stimulating dendritic cells, resulting in the eradication of the primary solid tumor along with the inhibition of metastasis. This molecular machine, C34, rendered great promise to achieve systemic treatment against cancer via unimolecule-mediated SDT.
Collapse
Affiliation(s)
- Liu Wang
- State Key Laboratory of Fine Chemicals, Department of Pharmacy, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Guangzhe Li
- State Key Laboratory of Fine Chemicals, Department of Pharmacy, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Lei Cao
- State Key Laboratory of Fine Chemicals, Department of Pharmacy, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yi Dong
- State Key Laboratory of Fine Chemicals, Department of Pharmacy, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yang Wang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shisheng Wang
- State Key Laboratory of Fine Chemicals, Department of Pharmacy, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yueqing Li
- State Key Laboratory of Fine Chemicals, Department of Pharmacy, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Xiuhan Guo
- State Key Laboratory of Fine Chemicals, Department of Pharmacy, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yi Zhang
- State Key Laboratory of Fine Chemicals, Department of Pharmacy, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Fangfang Sun
- Nuclear Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Xuemei Du
- Nuclear Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Jiangan Su
- EEC Biotech Co. Ltd, Guangzhou 510070, China
| | - Qing Li
- EEC Biotech Co. Ltd, Guangzhou 510070, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Kun Shao
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Weijie Zhao
- State Key Laboratory of Fine Chemicals, Department of Pharmacy, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
35
|
Xu Y, Tan Y, Ma X, Jin X, Tian Y, Li M. Photodynamic Therapy with Tumor Cell Discrimination through RNA-Targeting Ability of Photosensitizer. Molecules 2021; 26:5990. [PMID: 34641533 PMCID: PMC8512109 DOI: 10.3390/molecules26195990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/19/2022] Open
Abstract
Photodynamic therapy (PDT) represents an effective treatment to cure cancer. The targeting ability of the photosensitizer is of utmost importance. Photosensitizers that discriminate cancer cells can avoid the killing of normal cells and improve PDT efficacy. However, the design and synthesis of photosensitizers conjugated with a recognition unit of cancer cell markers is complex and may not effectively target cancer. Considering that the total RNA content in cancer cells is commonly higher than in normal cells, this study has developed the photosensitizer QICY with RNA-targeting abilities for the discrimination of cancer cells. QICY was specifically located in cancer cells rather than normal cells due to their stronger electrostatic interactions with RNA, thereby further improving the PDT effects on the cancer cells. After intravenous injection into mice bearing a xenograft tumor, QICY accumulated into the tumor location through the enhanced permeability and retention effect, automatically targeted cancer cells under the control of RNA, and inhibited tumor growth under 630 nm laser irradiation without obvious side effects. This intelligent photosensitizer with RNA-targeting ability not only simplifies the design and synthesis of cancer-cell-targeting photosensitizers but also paves the way for the further development of highly efficient PDTs.
Collapse
Affiliation(s)
- Yuan Xu
- College of Marine Technology and Environment, Dalian Ocean University, Dalian 116023, China;
- Key Laboratory of Environment Controlled Aquaculture, Ministry of Education, Dalian 116023, China
| | - Yang Tan
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116023, China; (Y.T.); (X.M.); (X.J.)
| | - Xiuqin Ma
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116023, China; (Y.T.); (X.M.); (X.J.)
| | - Xiaoyi Jin
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116023, China; (Y.T.); (X.M.); (X.J.)
| | - Ye Tian
- College of Marine Technology and Environment, Dalian Ocean University, Dalian 116023, China;
- Key Laboratory of Environment Controlled Aquaculture, Ministry of Education, Dalian 116023, China
| | - Miao Li
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116023, China; (Y.T.); (X.M.); (X.J.)
| |
Collapse
|
36
|
Gao S, Wang Y, Li Y, Xiao D, Lin Y, Chen Y, Cai X. Tetrahedral Framework Nucleic Acids Reestablish Immune Tolerance and Restore Saliva Secretion in a Sjögren's Syndrome Mouse Model. ACS APPLIED MATERIALS & INTERFACES 2021; 13:42543-42553. [PMID: 34477358 DOI: 10.1021/acsami.1c14861] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
As one of the most frequent autoimmune diseases, Sjogren's syndrome (SS) is characterized by overactive lymphocytic infiltration in the exocrine glands, with ensuing dry mouth and dry eyes. Unfortunately, so far, there are no appropriate therapies without causing overall immunosuppression. Tetrahedral framework nucleic acids (tFNAs) were regarded as promising nanoscale materials whose immunomodulatory capabilities have already been verified. Herein, we reveal, for the first time, that tFNAs were utilized to treat SS in female nonobese diabetic (NOD) mice, the animal model used for SS. We proved a 250 nM tFNA treatment was successful in suppressing inflammation and stimulating saliva secretion in NOD mice. Specialised proteins for the secretory function and structure of acinar cells in submandibular glands (SMGs) were restored. It has been the permanent goal for SS treatment to establish immune tolerance and stop disease development. Surprisingly, tFNA treatment guided T cells toward regulatory T cells (Tregs), while suppressing T helper (Th) cell responses. Th cells include Th1, Th17, and follicular helper T (Tfh) cells. Tregs are highly significant in immune tolerance. Inducing Tregs is a promising approach to reestablish immune tolerance. Comparable results were also observed in B cell responses. Reductions in the percentage of germinal center (GC) B cells and plasma cells were detected, and a marked increase in the percentage of regulatory B cells (Bregs) was also noticed. The mechanisms of inducing Tregs may associated with cytokine changes. Changes of T cell subsets, especially changes of Tfh, may influence the differentiation of B cells accordingly. Collectively, our results demonstrated the immunomodulatory capacities of tFNAs once again, which may provide a novel, safe, and effective option for the treatment of SS and other autoimmune diseases.
Collapse
Affiliation(s)
- Shaojingya Gao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yun Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yanjing Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Yu Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
37
|
Gold nanorods enhance different immune cells and allow for efficient targeting of CD4+ Foxp3+ Tregulatory cells. PLoS One 2021; 16:e0241882. [PMID: 34460818 PMCID: PMC8404976 DOI: 10.1371/journal.pone.0241882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 07/27/2021] [Indexed: 01/22/2023] Open
Abstract
Gold nanoparticles (AuNPs) hold great promise in nanomedicine, yet their successful clinical translation has not been realized. Some challenges include effective AuNP targeting and delivery to improve modulation of immune cells of interest while limiting potential adverse effects. In order to overcome these challenges, we must fully understand how AuNPs impact different immune cell subsets, particularly within the dendritic cell and T cell compartments. Herein, we show that polyethylene glycol coated (PEG) gold nanorods (AuNRs) and PEG AuNRs covered with a thin layer of silver (AuNR/Ag) may enhance the immune response towards immune suppression or activation. We also studied the ability to enhance CD4+ Foxp3+ Tregs in vitro using AuNRs functionalized with interleukin 2 (IL2), a cytokine that is important in Treg development and homeostasis. Our results indicate that AuNRs enhance different immune cells and that NP composition matters in immune targeting. This knowledge will help us understand how to better design AuNRs to target and enhance the immune system.
Collapse
|
38
|
Pang G, Liu Y, Wang Y, Wang Y, Wang F, Zhao J, Zhang LW. Endotoxin contamination in ovalbumin as viewed from a nano-immunotherapy perspective. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1747. [PMID: 34374214 DOI: 10.1002/wnan.1747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/18/2022]
Abstract
Ovalbumin (OVA) is a model antigen commonly incorporated in smartly designed nanoparticles for delivery into antigen-presenting cells (APC), aiming to investigate the immune activity and therapeutic efficacy of nanoparticles that contain immunoregulatory compounds. However, the immunoresponse observed in nano-immunotherapy may unexpectedly arise from endotoxin impurity of OVA in the nanoparticles. Literature review shows that most researchers did not notice the importance of endotoxin-free OVA when used in nano-immunotherapy studies. Concentration at as low as 5 μg/ml OVA from Sigma-Aldrich (contains 0.625 ng/ml endotoxin) was able to activate APC such as dendritic cells and macrophages. Here, we proposed that the endotoxin impurity in OVA or the finished nanoproducts should be determined by both Limulus Amebocyte Lysate (LAL) and cell-based assay, to ensure the endotoxin-free quality of the nanoparticles. The endotoxin in OVA can be removed by endotoxin removal column and phase separation methods and endotoxin-free OVA can be purchased. This perspective alerts the researchers of endotoxin impurity of OVA that may transfer into the finished nanoparticles and introduce an unfavorable immunoregulatory function with false-positive results. OVA with minimal endotoxin level should be used in nano-immunotherapy studies to accurately reflect the true effects of nanoparticles on the immune system. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Guibin Pang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yun Liu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yangyun Wang
- Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Yong Wang
- Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Fujun Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Leshuai W Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
39
|
Li H, Sui X, Wang Z, Fu H, Wang Z, Yuan M, Liu S, Wang G, Guo Q. A new antisarcoma strategy: multisubtype heat shock protein/peptide immunotherapy combined with PD-L1 immunological checkpoint inhibitors. Clin Transl Oncol 2021; 23:1688-1704. [PMID: 33792840 PMCID: PMC8238772 DOI: 10.1007/s12094-021-02570-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 02/07/2021] [Indexed: 12/11/2022]
Abstract
Osteosarcoma, a common malignant tumor in orthopedics, often has a very poor prognosis after lung metastasis. Immunotherapy has not achieved much progress in the treatment because of the characteristics of solid tumors and immune environment of osteosarcoma. The tumor environment is rather essential for sarcoma treatment. Our previous study demonstrated that heat shock proteins could be used as antitumor vaccines by carrying tumor antigen peptides, and we hypothesize that an anti-osteosarcoma effect may be increased with an immune check point inhibitor (PD-L1 inhibitor) as a combination treatment strategy. The present study prepared a multisubtype mixed heat shock protein osteosarcoma vaccine (mHSP/peptide vaccine) and concluded that the mHSP/peptide vaccine was more effective than a single subtype heat shock protein, like Grp94. Therefore, we used the mHSP/peptide vaccine in combination with a PD-L1 inhibitor to treat osteosarcoma, and the deterioration of osteosarcoma was effectively hampered. The mechanism of combined therapy was investigated, and AKT expression participates with sarcoma lung metastasis. This study proposed an antisarcoma strategy via stimulation of the immune system as a further alternative approach for sarcoma treatment and elucidated the mechanism of combined therapy.
Collapse
Affiliation(s)
- H. Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853 China
- Changzhi Second People’s Hospital, Changzhi, 046000 Shanxi China
| | - X. Sui
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853 China
| | - Z. Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853 China
| | - H. Fu
- School of Medicine, Nankai University, Tianjin, 300071 China
| | - Z. Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853 China
| | - M. Yuan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853 China
| | - S. Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853 China
| | - G. Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, 030001 Shanxi China
| | - Q. Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853 China
| |
Collapse
|
40
|
Zhang B, Su Y, Zhou J, Zheng Y, Zhu D. Toward a Better Regeneration through Implant-Mediated Immunomodulation: Harnessing the Immune Responses. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100446. [PMID: 34117732 PMCID: PMC8373114 DOI: 10.1002/advs.202100446] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/08/2021] [Indexed: 05/06/2023]
Abstract
Tissue repair/regeneration, after implantation or injury, involves comprehensive physiological processes wherein immune responses play a crucial role to enable tissue restoration, amidst the immune cells early-stage response to tissue damages. These cells break down extracellular matrix, clear debris, and secret cytokines to orchestrate regeneration. However, the immune response can also lead to abnormal tissue healing or scar formation if not well directed. This review first introduces the general immune response post injury, with focus on the major immune cells including neutrophils, macrophages, and T cells. Next, a variety of implant-mediated immunomodulation strategies to regulate immune response through physical, chemical, and biological cues are discussed. At last, various scaffold-facilitated regenerations of different tissue types, such as, bone, cartilage, blood vessel, and nerve system, by harnessing the immunomodulation are presented. Therefore, the most recent data in biomaterials and immunomodulation is presented here in a bid to shape expert perspectives, inspire researchers to go in new directions, and drive development of future strategies focusing on targeted, sequential, and dynamic immunomodulation elicited by implants.
Collapse
Affiliation(s)
- Ben Zhang
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Yingchao Su
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Juncen Zhou
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Yufeng Zheng
- Department of Materials Science and EngineeringCollege of EngineeringPeking UniversityBeijing100871China
| | - Donghui Zhu
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| |
Collapse
|
41
|
Cortez‐Jugo C, Czuba‐Wojnilowicz E, Tan A, Caruso F. A Focus on "Bio" in Bio-Nanoscience: The Impact of Biological Factors on Nanomaterial Interactions. Adv Healthc Mater 2021; 10:e2100574. [PMID: 34170631 DOI: 10.1002/adhm.202100574] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/18/2021] [Indexed: 12/17/2022]
Abstract
Bio-nanoscience research encompasses studies on the interactions of nanomaterials with biological structures or what is commonly referred to as the biointerface. Fundamental studies on the influence of nanomaterial properties, including size, shape, composition, and charge, on the interaction with the biointerface have been central in bio-nanoscience to assess nanomaterial efficacy and safety for a range of biomedical applications. However, the state of the cells, tissues, or biological models can also influence the behavior of nanomaterials at the biointerface and their intracellular processing. Focusing on the "bio" in bio-nano, this review discusses the impact of biological properties at the cellular, tissue, and whole organism level that influences nanomaterial behavior, including cell type, cell cycle, tumor physiology, and disease states. Understanding how the biological factors can be addressed or exploited to enhance nanomaterial accumulation and uptake can guide the design of better and suitable models to improve the outcomes of materials in nanomedicine.
Collapse
Affiliation(s)
- Christina Cortez‐Jugo
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Ewa Czuba‐Wojnilowicz
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Abigail Tan
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| |
Collapse
|
42
|
Peng Z, Wu J, Wang K, Li X, Sun P, Zhang L, Huang J, Liu Y, Hua X, Yu Y, Pan C, Wang H, Zhu L. Production of a Promising Biosynthetic Self-Assembled Nanoconjugate Vaccine against Klebsiella Pneumoniae Serotype O2 in a General Escherichia Coli Host. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100549. [PMID: 34032027 PMCID: PMC8292882 DOI: 10.1002/advs.202100549] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/09/2021] [Indexed: 05/09/2023]
Abstract
Klebsiella pneumoniae has emerged as a severe opportunistic pathogen with multiple drug resistances. Finding effective vaccines against this pathogen is urgent. Although O-polysaccharides (OPS) of K. pneumoniae are suitable antigens for the preparation of vaccines given their low levels of diversity, the low immunogenicity (especially serotype O2) limit their application. In this study, a general Escherichia coli host system is developed to produce a nanoscale conjugate vaccine against K. pneumoniae using the Nano-B5 self-assembly platform. The experimental data illustrate that this nanoconjugate vaccine can induce an efficient humoral immune response in draining lymph nodes (dLNs) and elicit high titers of the IgG antibody against bacterial lipopolysaccharide (LPS). The ideal prophylactic effects of these nanoconjugate vaccines are further demonstrated in mouse models of both systemic and pulmonary infection. These results demonstrate that OPS with low immunogenicity can be changed into an effective antigen, indicating that other haptens may be applicable to this strategy in the future. To the knowledge, this is the first study to produce biosynthetic nanoconjugate vaccines against K. pneumoniae in E. coli, and this strategy can be applied to the development of other vaccines against pathogenic bacteria.
Collapse
Affiliation(s)
- Zhehui Peng
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of BiotechnologyNo. 20, Dongda Street, Fengtai DistrictBeijing100071P. R. China
| | - Jun Wu
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of BiotechnologyNo. 20, Dongda Street, Fengtai DistrictBeijing100071P. R. China
| | - Kangfeng Wang
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of BiotechnologyNo. 20, Dongda Street, Fengtai DistrictBeijing100071P. R. China
| | - Xin Li
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of BiotechnologyNo. 20, Dongda Street, Fengtai DistrictBeijing100071P. R. China
| | - Peng Sun
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of BiotechnologyNo. 20, Dongda Street, Fengtai DistrictBeijing100071P. R. China
| | - Lulu Zhang
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of BiotechnologyNo. 20, Dongda Street, Fengtai DistrictBeijing100071P. R. China
| | - Jing Huang
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of BiotechnologyNo. 20, Dongda Street, Fengtai DistrictBeijing100071P. R. China
| | - Yan Liu
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of BiotechnologyNo. 20, Dongda Street, Fengtai DistrictBeijing100071P. R. China
| | - Xiaoting Hua
- Department of Infectious DiseasesSir Run Run Shaw HospitalCollege of MedicineZhejiang University866 Yuhangtang RdHangzhou310058P. R. China
| | - Yunsong Yu
- Department of Infectious DiseasesSir Run Run Shaw HospitalCollege of MedicineZhejiang University866 Yuhangtang RdHangzhou310058P. R. China
| | - Chao Pan
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of BiotechnologyNo. 20, Dongda Street, Fengtai DistrictBeijing100071P. R. China
| | - Hengliang Wang
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of BiotechnologyNo. 20, Dongda Street, Fengtai DistrictBeijing100071P. R. China
| | - Li Zhu
- State Key Laboratory of Pathogen and BiosecurityBeijing Institute of BiotechnologyNo. 20, Dongda Street, Fengtai DistrictBeijing100071P. R. China
| |
Collapse
|
43
|
Qin L, Zhang H, Zhou Y, Umeshappa CS, Gao H. Nanovaccine-Based Strategies to Overcome Challenges in the Whole Vaccination Cascade for Tumor Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006000. [PMID: 33768693 DOI: 10.1002/smll.202006000] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/26/2020] [Indexed: 06/12/2023]
Abstract
Nanovaccine-based immunotherapy (NBI) has received greater attention recently for its potential to prime tumor-specific immunity and establish a long-term immune memory that prevents tumor recurrence. Despite encouraging results in the recent studies, there are still numerous challenges to be tackled for eliciting potent antitumor immunity using NBI strategies. Based on the principles that govern immune response, here it is proposed that these challenges need to be addressed at the five critical cascading events: Loading tumor-specific antigens by nanoscale drug delivery systems (L); Draining tumor antigens to lymph nodes (D); Internalization by dendritic cells (DCs) (I); Maturation of DCs by costimulatory signaling (M); and Presenting tumor-peptide-major histocompatibility complexes to T cells (P) (LDIMP cascade in short). This review provides a detailed and objective overview of emerging NBI strategies to improve the efficacy of nanovaccines in each step of the LDIMP cascade. It is concluded that the balance between each step must be optimized by delicate designing and modification of nanovaccines and by combining with complementary approaches to provide a synergistic immunity in the fight against cancer.
Collapse
Affiliation(s)
- Lin Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Chongqing Vocational College of Transportation, Chongqing, 400715, China
| | - Huilin Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Channakeshava Sokke Umeshappa
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
44
|
Liu D, Liu J, Ma B, Deng B, Leng X, Kong D, Liu L. A simple self-adjuvanting biomimetic nanovaccine self-assembled with the conjugate of phospholipids and nucleotides can induce a strong cancer immunotherapeutic effect. Biomater Sci 2021; 9:84-92. [PMID: 33016303 DOI: 10.1039/d0bm01333a] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Biomimetic nanoparticles have potential applications in many fields due to their favorable properties. Here, we developed a self-adjuvanting biomimetic anti-tumor nanovaccine, which was self-assembled with an amphiphilic conjugate synthesized with the phospholipids of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and hydrophilic Toll-like receptor (TLR9) agonist CpG ODN. The nanovaccine could not only provide effective initial antigen stimulation and sustained long-term antigen supply with a controlled release, but also induce antigen cross-presentation via the MHC-I pathway initiating CD8+ T-cell responses. Moreover, the dense nucleotide shell around the nanovaccine could promote antigen endocytosis via various receptor-mediated pathways into dendritic cells. CpG ODN interacted with TLR9 triggering the cytokine secretion of TNF-α and IL-10, which further boosted the anti-tumor humoral and cellular immune responses, which led to a significant tumor suppressive effect and remarkable survival prolongation. So, this nanovaccine self-assembled with phospholipid-nucleotide amphiphiles can serve as a safe, simple and efficient approach for anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Dan Liu
- The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin 300192, China.
| | | | | | | | | | | | | |
Collapse
|
45
|
Gao S, Li Y, Xiao D, Zhou M, Cai X, Lin Y. Tetrahedral Framework Nucleic Acids Induce Immune Tolerance and Prevent the Onset of Type 1 Diabetes. NANO LETTERS 2021; 21:4437-4446. [PMID: 33955221 DOI: 10.1021/acs.nanolett.1c01131] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A failure in immune tolerance leads to autoimmune destruction of insulin-producing β-cells, leading to type 1 diabetes (T1D). Inhibiting autoreactive T cells and inducing regulatory T cells (Tregs) to re-establish immune tolerance are promising approaches to prevent the onset of T1D. Here, we investigated the ability of tetrahedral framework nucleic acids (tFNAs) to induce immune tolerance and prevent T1D in nonobese diabetic (NOD) mice. In prediabetic NOD mice, tFNAs treatment led to maintenance of normoglycemia and reduced incidence of diabetes. Moreover, the tFNAs (250 nM) treatment preserved the mass and function of β-cells, increased the frequency of Tregs, and suppressed autoreactive T cells, leading to immune tolerance. Collectively, our results demonstrate that tFNAs treatment aids glycemic control, provides β-cell protection, and prevents the onset of T1D in NOD mice by immunomodulation. These results highlight the potential of tFNAs for the prevention of autoimmune T1D.
Collapse
Affiliation(s)
- Shaojingya Gao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yanjing Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mi Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
46
|
Khorsandi K, Hosseinzadeh R, Sadat Esfahani H, Keyvani-Ghamsari S, Ur Rahman S. Nanomaterials as drug delivery systems with antibacterial properties: current trends and future priorities. Expert Rev Anti Infect Ther 2021; 19:1299-1323. [PMID: 33755503 DOI: 10.1080/14787210.2021.1908125] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction:Despite extensive advances in the production and synthesis of antibiotics, infectious diseases are one of the main problems of the 21st century due to multidrug-resistant (MDR) distributing in organisms. Therefore, researchers in nanotechnology have focused on new strategies to formulate and synthesis the different types of nanoparticles (NPs) with antimicrobial properties.Areas covered:The present review focuses on nanoparticles which are divided into two groups, organic (micelles, liposomes, polymer-based and lipid-based NPs) and inorganic (metals and metal oxides). NPs can penetrate the cell wall then destroy permeability of cell membrane, the structure and function of cell macromolecules by producing of reactive oxygen species (ROS) and eventually kill the bacteria. Moreover, their characteristics and mechanism in various bacteria especially MDR bacteria and finally their biocompatibility and the factors affecting their activity have been discussed.Expert opinion:Nanotechnology has led to higher drug absorption, targeted drug delivery and fewer side effects. NPs can overcome MDR through affecting several targets in the bacteria cell and synergistically increase the effectiveness of current antibiotics. Moreover, organic NPs with regard to their biodegradability and biocompatibility characteristics can be suitable agents for medical applications. However, they are less stable in environment in comparison to inorganic NPs.
Collapse
Affiliation(s)
- Khatereh Khorsandi
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Reza Hosseinzadeh
- Department of Medical Laser, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Homa Sadat Esfahani
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | | | - Saeed Ur Rahman
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan.,Department of Oral Biology, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| |
Collapse
|
47
|
Gu P, Zhang Y, Cai G, Liu Z, Hu Y, Liu J, Wang D. Administration Routes of Polyethylenimine-Coated PLGA Nanoparticles Encapsulating Angelica Sinensis Polysaccharide Vaccine Delivery System Affect Immune Responses. Mol Pharm 2021; 18:2274-2284. [PMID: 33926191 DOI: 10.1021/acs.molpharmaceut.1c00090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nanoparticle vaccine delivery systems have been emerging strategies for inducing potent immune responses to prevent and treat infectious diseases and cancers. The properties of nanoparticle vaccine delivery systems, such as nanoparticle size, surface charge, and antigen release kinetics, have been extensively studied and proven to effectively influence the efficacy of vaccine responses. However, a few types of research have focused on the influence of administration routes of nanoparticle vaccines on immune responses. Herein, to investigate how the administration routes affect the immune responses of nanoparticles vaccines, we developed a nanoparticles system (NPs), in which the ovalbumin (OVA) and Angelica sinensis polysaccharide (ASP) were incorporated into poly(lactic-co-glycolic acid) (PLGA) nanoparticles and the polyethylenimine (PEI) was coated on the surface of nanoparticles. The NPs vaccine was intramuscularly and subcutaneously injected (im and sc) into mice, and the immune responses induced by these two delivery routes were compared. The results showed that both im and sc administration of NPs vaccines elicited strong antigen-specific IgG, IgG1, and IgG2a antibody responses, with no significant difference. In contrast, NP vaccines with sc administration significantly enhanced immune responses, such as enhancing the recruitment and activation of dendritic cells (DCs) in lymph nodes (LNs), promoting the antigen transport into draining lymph nodes, increasing the secretion of cytokines, improving the ratio of CD4+T cells to CD8+ T cells, activating cytotoxic T lymphocyte response, and inducing a strong cellular immune response. These results may provide a new insight onto the development of vaccine delivery systems.
Collapse
Affiliation(s)
- Pengfei Gu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Yue Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Gaofeng Cai
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Yuanliang Hu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Jiaguo Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| |
Collapse
|
48
|
Guo Z, Kubiatowicz LJ, Fang RH, Zhang L. Nanotoxoids: Biomimetic Nanoparticle Vaccines against Infections. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Zhongyuan Guo
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| | - Luke J. Kubiatowicz
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| | - Ronnie H. Fang
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| |
Collapse
|
49
|
Zhang G, Fu X, Sun H, Zhang P, Zhai S, Hao J, Cui J, Hu M. Poly(ethylene glycol)-Mediated Assembly of Vaccine Particles to Improve Stability and Immunogenicity. ACS APPLIED MATERIALS & INTERFACES 2021; 13:13978-13989. [PMID: 33749241 DOI: 10.1021/acsami.1c00706] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
We report the one-step assembly of vaccine particles by encapsulating ovalbumin (OVA) and cytosine-phosphate-guanine oligodeoxynucleotides (CpG) into poly(ethylene glycol) (PEG)-mediated zeolitic imidazolate framework-8 nanoparticles (OVA-CpG@ZIF-8 NPs), where PEG improves the stability and dispersity of ZIF-8 NPs and the NPs protect the encapsulated OVA and CpG to circumvent the cold chain issue. Compared with free OVA and OVA-encapsulated ZIF-8 (OVA@ZIF-8) NPs, OVA-CpG@ZIF-8 NPs can enhance antigen uptake, cross-presentation, dendritic cell (DC) maturation, production of specific antibody and cytokines, and CD4+ T and CD8+ T cell activation. More importantly, the vaccine particles retain their bioactivity against enzymatic degradation, elevated temperatures, and long-term storage at ambient temperature. The study highlights the importance of PEG-mediated ZIF-8 NPs as a vaccine delivery system for the promising application of effective and cold chain-independent vaccination against diseases.
Collapse
Affiliation(s)
- Guiqiang Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Xiao Fu
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Haifeng Sun
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Peiyu Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Shumei Zhai
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jingcheng Hao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong 250012, China
| | - Ming Hu
- School of Physics and Electronic Science, East China Normal University, Shanghai 200241, China
| |
Collapse
|
50
|
Yuan J, Zhang Y, Zhang Y, Mo Y, Zhang Q. Effects of metal nanoparticles on tight junction-associated proteins via HIF-1α/miR-29b/MMPs pathway in human epidermal keratinocytes. Part Fibre Toxicol 2021; 18:13. [PMID: 33740985 PMCID: PMC7980342 DOI: 10.1186/s12989-021-00405-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
Background The increasing use of metal nanoparticles in industry and biomedicine raises the risk for unintentional exposure. The ability of metal nanoparticles to penetrate the skin ranges from stopping at the stratum corneum to passing below the dermis and entering the systemic circulation. Despite the potential health risks associated with skin exposure to metal nanoparticles, the mechanisms underlying the toxicity of metal nanoparticles on skin keratinocytes remain unclear. In this study, we proposed that exposure of human epidermal keratinocytes (HaCaT) to metal nanoparticles, such as nickel nanoparticles, dysregulates tight-junction associated proteins by interacting with the HIF-1α/miR-29b/MMPs axis. Methods We performed dose-response and time-response studies in HaCaT cells to observe the effects of Nano-Ni or Nano-TiO2 on the expression and activity of MMP-2 and MMP-9, and on the expression of tight junction-associated proteins, TIMP-1, TIMP-2, miR-29b, and HIF-1α. In the dose-response studies, cells were exposed to 0, 10, or 20 μg/mL of Nano-Ni or Nano-TiO2 for 24 h. In the time-response studies, cells were exposed to 20 μg/mL of Nano-Ni for 12, 24, 48, or 72 h. After treatment, cells were collected to either assess the expression of mRNAs and miR-29b by real-time PCR or to determine the expression of tight junction-associated proteins and HIF-1α nuclear accumulation by Western blot and/or immunofluorescent staining; the conditioned media were collected to evaluate the MMP-2 and MMP-9 activities by gelatin zymography assay. To further investigate the mechanisms underlying Nano-Ni-induced dysregulation of tight junction-associated proteins, we employed a HIF-1α inhibitor, CAY10585, to perturb HIF-1α accumulation in one experiment, and transfected a miR-29b-3p mimic into the HaCaT cells before Nano-Ni exposure in another experiment. Cells and conditioned media were collected, and the expression and activities of MMPs and the expression of tight junction-associated proteins were determined as described above. Results Exposure of HaCaT cells to Nano-Ni resulted in a dose-dependent increase in the expression of MMP-2, MMP-9, TIMP-1, and TIMP-2 and the activities of MMP-2 and MMP-9. However, exposure of cells to Nano-TiO2 did not cause these effects. Nano-Ni caused a dose-dependent decrease in the expression of miR-29b and tight junction-associated proteins, such as ZO-1, occludin, and claudin-1, while Nano-TiO2 did not. Nano-Ni also caused a dose-dependent increase in HIF-1α nuclear accumulation. The time-response studies showed that Nano-Ni caused significantly increased expressions of MMP-2 at 24 h, MMP-9 at 12, 24, and 48 h, TIMP-1 from 24 to 72 h, and TIMP-2 from 12 to 72 h post-exposure. The expression of miR-29b and tight junction-associated proteins such as ZO-1, occludin, and claudin-1 decreased as early as 12 h post-exposure, and their levels declined gradually over time. Pretreatment of cells with a HIF-1α inhibitor, CAY10585, abolished Nano-Ni-induced miR-29b down-regulation and MMP-2/9 up-regulation. Introduction of a miR-29b-3p mimic into HaCaT cells by transfection before Nano-Ni exposure ameliorated Nano-Ni-induced increased expression and activity of MMP-2 and MMP-9 and restored Nano-Ni-induced down-regulation of tight junction-associated proteins. Conclusion Our study herein demonstrated that exposure of human epidermal keratinocytes to Nano-Ni caused increased HIF-1α nuclear accumulation and increased transcription and activity of MMP-2 and MMP-9 and down-regulation of miR-29b and tight junction-associated proteins. Nano-Ni-induced miR-29b down-regulation was through Nano-Ni-induced HIF-1α nuclear accumulation. Restoration of miR-29b level by miR-29b-3p mimic transfection abolished Nano-Ni-induced MMP-2 and MMP-9 activation and down-regulation of tight junction-associated proteins. In summary, our results demonstrated that Nano-Ni-induced dysregulation of tight junction-associated proteins in skin keratinocytes was via HIF-1α/miR-29b/MMPs pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-021-00405-2.
Collapse
Affiliation(s)
- Jiali Yuan
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
| | - Yue Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
| | - Yuanbao Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
| | - Yiqun Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
| | - Qunwei Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA.
| |
Collapse
|