1
|
Karan S, Opdensteinen P, Ma Y, De Oliveira JFA, Steinmetz NF. A replicon-based COVID-19 vaccine candidate delivered by tobacco mosaic virus-like particles. Vaccine 2025; 53:127063. [PMID: 40168732 DOI: 10.1016/j.vaccine.2025.127063] [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: 08/21/2024] [Revised: 03/12/2025] [Accepted: 03/20/2025] [Indexed: 04/03/2025]
Abstract
The COVID-19 pandemic highlights the opportunity for mRNA vaccines and their nanotechnology carriers to make an impact as a countermeasure to infectious disease. As alternative to the synthetic lipid nanoparticles or mammalian viruses, we developed a tobacco mosaic virus (TMV)-based mRNA vaccine delivery platform. Specifically, purified coat protein from TMV was used to package a self-amplifying Nodamura replicon expressing the receptor binding domain (RBD) from the Omicron strain of SARS-CoV-2. The replicon construct contains the origin of assembly sequence from the tobacco mosaic virus (TMV) for encapsulation and mRNA stabilization. The nanoparticle vaccine was obtained through in vitro assembly using purified TMV coat proteins and in vitro transcribed mRNA cassettes. Cell assays confirmed delivery of self-amplifying mRNA vaccine, amplification of the transgene and expression of the target protein, RBD, in mammalian cells. Immunization of mice yielded RBD-specific IgG antibodies that demonstrated neutralization of SARS-CoV-2 using an in vitro neutralization assay. The TMV platform nanotechnology does not require ultralow freezers for storage or distribution; and the in vitro assembly method provide 'plug-and-play' to adapt the vaccine formulation rapidly as new strains or diseases emerge. Finally, opportunity exists to produce and self-assemble the vaccine candidate in plants through molecular farming techniques, which may allow production in the region-for the region and could make a contribution to less resourced areas of the world.
Collapse
MESH Headings
- Tobacco Mosaic Virus/genetics
- Animals
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/genetics
- Mice
- Replicon
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- COVID-19/prevention & control
- COVID-19/immunology
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Humans
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Female
- Capsid Proteins/immunology
- Capsid Proteins/genetics
- Immunoglobulin G/immunology
- Immunoglobulin G/blood
- Mice, Inbred BALB C
- Nanoparticles
Collapse
Affiliation(s)
- Sweta Karan
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA 92093, USA; Shu and K.C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA; Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Patrick Opdensteinen
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA 92093, USA; Shu and K.C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA; Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Yifeng Ma
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA 92093, USA; Shu and K.C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA; Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Jessica Fernanda Affonso De Oliveira
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA 92093, USA; Shu and K.C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA; Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Nicole F Steinmetz
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA 92093, USA; Shu and K.C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA; Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States; Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States; Department of Radiology, University of California, San Diego, La Jolla, CA, United States; Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, United States; Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States; Center for Engineering in Cancer, Institute of Engineering Medicine, University of California, San Diego, La Jolla, CA, United States.
| |
Collapse
|
2
|
Bourguignon L, Hétu-Arbour R, Charpentier T, Bolduc M, Leclerc D, Heinonen KM, Lamarre A. Systemic administration of a viral nanoparticle neoadjuvant prevents lung metastasis development through emergency myelopoiesis. Oncoimmunology 2024; 13:2429846. [PMID: 39552216 PMCID: PMC11581170 DOI: 10.1080/2162402x.2024.2429846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024] Open
Abstract
Cancer presents a significant public health concern, particularly in the context of metastatic disease. Surgical removal of primary tumors, while essential, can inadvertently heighten the risk of metastasis. Thus, there is a critical need for innovative neoadjuvant therapies capable of curtailing metastatic progression before or immediately following tumor resection. Addressing this imperative, the papaya mosaic virus nanoparticle (PapMV) has demonstrated potent immunostimulatory capabilities against both viruses and tumors, effectively hindering their proliferation. Our study reveals that PapMV exerts a protective effect against lung metastasis when administered systemically prior to tumor implantation or during the early stages of metastasis in various mouse models of cancer. This anti-tumor effect is initiated by the recruitment of myeloid cells in the lungs. These cells adopt a pro-inflammatory profile, secreting cytokines such as IFN-α, thus fostering a tumor microenvironment inhospitable to tumor progression. Crucially, this protective mechanism hinges on the presence of macrophages before treatment. TLR7 and IFN-I signaling pathways also play pivotal roles in this process. Furthermore, our findings demonstrate that PapMV triggers the activation of the bone marrow emergency response, which accounts for the influx of myeloid cells into the lungs. This study unveils a novel aspect of PapMV's functionality. By bolstering the immune system, PapMV confers robust protection against metastasis at an early stage of disease progression. This discovery holds promise for therapeutic intervention, particularly as a preemptive measure prior to or just after surgical intervention.
Collapse
Affiliation(s)
- Léa Bourguignon
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, QC, Canada
| | - Roxann Hétu-Arbour
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, QC, Canada
| | - Tania Charpentier
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, QC, Canada
| | - Marilène Bolduc
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Center, Laval University, Quebec City, QC, Canada
| | - Denis Leclerc
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Center, Laval University, Quebec City, QC, Canada
| | - Krista M. Heinonen
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, QC, Canada
| | - Alain Lamarre
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, QC, Canada
| |
Collapse
|
3
|
Karan S, Affonso De Oliveira JF, Moreno-Gonzalez MA, Steinmetz NF. A Self-Amplifying Human Papillomavirus 16 Vaccine Candidate Delivered by Tobacco Mosaic Virus-Like Particles. ACS APPLIED BIO MATERIALS 2024; 7:7675-7683. [PMID: 39512153 PMCID: PMC11648571 DOI: 10.1021/acsabm.4c01239] [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] [Indexed: 11/15/2024]
Abstract
Virus-like particles (VLPs) are naturally occurring delivery platforms with potential for mRNA vaccines that can be used as an alternative to lipid nanoparticles. Here we describe a self-amplifying mRNA vaccine based on tobacco mosaic virus (TMV) expressing a mutated E7 protein from human papillomavirus 16 (HPV16). E7 is an early gene that plays a central role in viral replication and the oncogenic transformation of host cells, but nononcogenic mutant E7 proteins can suppress this activity. Immunization studies involving the delivery of self-amplifying mutant E7 mRNA packaged with TMV coat proteins confirmed the elicitation of E7-specific IgG antibodies. Additional in vitro splenocyte proliferation and cytokine profiling assays indicated the activation of humoral and cellular immune responses. We conclude that TMV particles are suitable for the delivery of mRNA vaccines and can preserve their integrity and functionality in vitro and in vivo.
Collapse
Affiliation(s)
- Sweta Karan
- Aiiso Yufeng Li Family Department of Chemical and Nanoengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Center for Nano Immuno Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jessica Fernanda Affonso De Oliveira
- Aiiso Yufeng Li Family Department of Chemical and Nanoengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Center for Nano Immuno Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Miguel A Moreno-Gonzalez
- Aiiso Yufeng Li Family Department of Chemical and Nanoengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Center for Nano Immuno Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Nicole F Steinmetz
- Aiiso Yufeng Li Family Department of Chemical and Nanoengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Center for Nano Immuno Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Radiology, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Institute for Materials Discovery and Design, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Center for Engineering in Cancer, Institute of Engineering Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
4
|
Yang K, Zeng Y, Wu X, Li J, Guo J. Strategies for developing self-assembled nanoparticle vaccines against SARS-CoV-2 infection. Front Immunol 2024; 15:1392898. [PMID: 39351240 PMCID: PMC11440195 DOI: 10.3389/fimmu.2024.1392898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/21/2024] [Indexed: 10/04/2024] Open
Abstract
In the recent history of the SARS-CoV-2 outbreak, vaccines have been a crucial public health tool, playing a significant role in effectively preventing infections. However, improving the efficacy while minimizing side effects remains a major challenge. In recent years, there has been growing interest in nanoparticle-based delivery systems aimed at improving antigen delivery efficiency and immunogenicity. Among these, self-assembled nanoparticles with varying sizes, shapes, and surface properties have garnered considerable attention. This paper reviews the latest advancements in the design and development of SARS-CoV-2 vaccines utilizing self-assembled materials, highlighting their advantages in delivering viral immunogens. In addition, we briefly discuss strategies for designing a broad-spectrum universal vaccine, which provides insights and ideas for dealing with possible future infectious sarbecoviruses.
Collapse
Affiliation(s)
- Kaiwen Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Youqin Zeng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Xinyu Wu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Jia Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Jinlin Guo
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| |
Collapse
|
5
|
Karan S, Durán-Meza AL, Chapman A, Tanimoto C, Chan SK, Knobler CM, Gelbart WM, Steinmetz NF. In Vivo Delivery of Spherical and Cylindrical In Vitro Reconstituted Virus-like Particles Containing the Same Self-Amplifying mRNA. Mol Pharm 2024; 21:2727-2739. [PMID: 38709860 PMCID: PMC11250921 DOI: 10.1021/acs.molpharmaceut.3c01105] [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] [Indexed: 05/08/2024]
Abstract
The dramatic effectiveness of recent mRNA (mRNA)-based COVID vaccines delivered in lipid nanoparticles has highlighted the promise of mRNA therapeutics in general. In this report, we extend our earlier work on self-amplifying mRNAs delivered in spherical in vitro reconstituted virus-like particles (VLPs), and on drug delivery using cylindrical virus particles. In particular, we carry out separate in vitro assemblies of a self-amplifying mRNA gene in two different virus-like particles: one spherical, formed with the capsid protein of cowpea chlorotic mottle virus (CCMV), and the other cylindrical, formed from the capsid protein of tobacco mosaic virus (TMV). The mRNA gene is rendered self-amplifying by genetically fusing it to the RNA-dependent RNA polymerase (RdRp) of Nodamura virus, and the relative efficacies of cell uptake and downstream protein expression resulting from their CCMV- and TMV-packaged forms are compared directly. This comparison is carried out by their transfections into cells in culture: expressions of two self-amplifying genes, enhanced yellow fluorescent protein (EYFP) and Renilla luciferase (Luc), packaged alternately in CCMV and TMV VLPs, are quantified by fluorescence and chemiluminescence levels, respectively, and relative numbers of the delivered mRNAs are measured by quantitative real-time PCR. The cellular uptake of both forms of these VLPs is further confirmed by confocal microscopy of transfected cells. Finally, VLP-mediated delivery of the self-amplifying-mRNA in mice following footpad injection is shown by in vivo fluorescence imaging to result in robust expression of EYFP in the draining lymph nodes, suggesting the potential of these plant virus-like particles as a promising mRNA gene and vaccine delivery modality. These results establish that both CCMV and TMV VLPs can deliver their in vitro packaged mRNA genes to immune cells and that their self-amplifying forms significantly enhance in situ expression. Choice of one VLP (CCMV or TMV) over the other will depend on which geometry of nucleocapsid is self-assembled more efficiently for a given length and sequence of RNA, and suggests that these plant VLP gene delivery systems will prove useful in a wide variety of medical applications, both preventive and therapeutic.
Collapse
Affiliation(s)
- Sweta Karan
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Ana Luisa Durán-Meza
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Abigail Chapman
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Cheylene Tanimoto
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Soo Khim Chan
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
| | - Charles M Knobler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - William M Gelbart
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- UCLA Molecular Biology Institute, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Institute for Materials Discovery and Design, University of California San Diego, La Jolla, California 92093, United States
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Radiology, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
- Center for Engineering in Cancer, Institute for Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
6
|
Liu C, Yu Y, Fang L, Wang J, Sun C, Li H, Zhuang J, Sun C. Plant-derived nanoparticles and plant virus nanoparticles: Bioactivity, health management, and delivery potential. Crit Rev Food Sci Nutr 2023; 64:8875-8891. [PMID: 37128778 DOI: 10.1080/10408398.2023.2204375] [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: 05/03/2023]
Abstract
Natural plants have acquired an increasing attention in biomedical research. Recent studies have revealed that plant-derived nanoparticles (PDNPs), which are nano-sized membrane vesicles released by plants, are one of the important material bases for the health promotion of natural plants. A great deal of research in this field has focused on nanoparticles derived from fresh vegetables and fruits. Generally, PDNPs contain lipids, proteins, nucleic acids, and other active small molecules and exhibit unique biological regulatory activity and editability. Specifically, they have emerged as important mediators of intercellular communication, and thus, are potentially suitable for therapeutic purposes. In this review, PDNPs were extensively explored; by evaluating them systematically starting from the origin and isolation, toward their characteristics, including morphological compositions, biological functions, and delivery potentials, as well as distinguishing them from plant-derived exosomes and highlighting the limitations of the current research. Meanwhile, we elucidated the variations in PDNPs infected by pathogenic microorganisms and emphasized on the biological functions and characteristics of plant virus nanoparticles. After clarifying these problems, it is beneficial to further research on PDNPs in the future and develop their clinical application value.
Collapse
Affiliation(s)
- Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Yang Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liguang Fang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jia Wang
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Chunjie Sun
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| |
Collapse
|
7
|
Shahgolzari M, Dianat-Moghadam H, Fiering S. Multifunctional plant virus nanoparticles in the next generation of cancer immunotherapies. Semin Cancer Biol 2022; 86:1076-1085. [PMID: 34375725 PMCID: PMC8821734 DOI: 10.1016/j.semcancer.2021.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 06/27/2021] [Accepted: 07/28/2021] [Indexed: 02/07/2023]
Abstract
Plant virus nanoparticles (PVNPs) have inherent immune stimulatory ability, and have been investigated as immune adjuvants to stimulate an anti-tumor immune response. The combination of immune stimulation, nanoparticle structure and the ability to deliver other therapeutic molecules provides a flexible platform for cancer immunotherapy. Researching multifunctional PVNPs and their modification will generate novel reagents for cancer immunotherapy. Here we review the properties of PVNPs, and their potential for clinical utilization to activate anti-tumor innate and lymphoid immune responses. PVNPs have potential utility for cancer immunotherapy as vaccine adjuvant, and delivery systems for other reagents as mono immunotherapy or combined with other immunotherapies. This review outlines the potential and challenges in developing PVNPs as cancer immunotherapy reagents.
Collapse
Affiliation(s)
- Mehdi Shahgolzari
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States.
| |
Collapse
|
8
|
Multifunctional Plant Virus Nanoparticles for Targeting Breast Cancer Tumors. Vaccines (Basel) 2022; 10:vaccines10091431. [PMID: 36146510 PMCID: PMC9502313 DOI: 10.3390/vaccines10091431] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Breast cancer treatment using plant-virus-based nanoparticles (PVNPs) has achieved considerable success in preclinical studies. PVNP-based breast cancer therapies include non-targeted and targeted nanoplatforms for delivery of anticancer therapeutic chemo and immune agents and cancer vaccines for activation of local and systemic antitumor immunity. Interestingly, PVNP platforms combined with other tumor immunotherapeutic options and other modalities of oncotherapy can improve tumor efficacy treatment. These applications can be achieved by encapsulation of a wide range of active ingredients and conjugating ligands for targeting immune and tumor cells. This review presents the current breast cancer treatments based on PVNP platforms.
Collapse
|
9
|
Shan X, Gong X, Li J, Wen J, Li Y, Zhang Z. Current approaches of nanomedicines in the market and various stage of clinical translation. Acta Pharm Sin B 2022; 12:3028-3048. [PMID: 35865096 PMCID: PMC9293719 DOI: 10.1016/j.apsb.2022.02.025] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/16/2021] [Accepted: 02/21/2022] [Indexed: 12/11/2022] Open
Abstract
Compared with traditional drug therapy, nanomedicines exhibit intriguing biological features to increase therapeutic efficiency, reduce toxicity and achieve targeting delivery. This review provides a snapshot of nanomedicines that have been currently launched or in the clinical trials, which manifests a diversified trend in carrier types, applied indications and mechanisms of action. From the perspective of indications, this article presents an overview of the applications of nanomedicines involving the prevention, diagnosis and treatment of various diseases, which include cancer, infections, blood disorders, cardiovascular diseases, immuno-associated diseases and nervous system diseases, etc. Moreover, the review provides some considerations and perspectives in the research and development of nanomedicines to facilitate their translations in clinic.
Collapse
Affiliation(s)
- Xiaoting Shan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Gong
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Jingyuan Wen
- School of Pharmacy, University of Auckland, Auckland 1142, New Zealand
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
10
|
Shahgolzari M, Fiering S. Emerging Potential of Plant Virus Nanoparticles (PVNPs) in Anticancer Immunotherapies. JOURNAL OF CANCER IMMUNOLOGY 2022; 4:22-29. [PMID: 35600219 PMCID: PMC9121906 DOI: 10.33696/cancerimmunol.4.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer immunotherapies using plant virus nanoparticles (PVNPs) have achieved considerable success in preclinical studies. PVNP based nanoplatforms can be endogenous immune adjuvants and act as nanocarriers that stabilize and deliver cancer antigens and exogenous immune adjuvants. Although they do not infect mammalian cells, PVNPs are viruses and they are variably recognized by pathogen pattern recognition receptors (PRR), activate innate immune cells including antigen-presenting cells (APCs), and increase the expression of costimulatory molecules. Novel immunotherapy strategies use them as in situ vaccines (ISV) that can effectively inhibit tumor growth after intratumoral administration and generate expanded systemic antitumor immunity. PVNPs combined with other tumor immunotherapeutic options and other modalities of oncotherapy can improve both local and systemic anti-tumor immune responses. While not yet in clinical trials in humans, there is accelerating interest and research of the potential of PVNPs for ISV immune therapy for cancer. Thus, antitumor efficacy of PVNPs by themselves, or loaded with soluble toll-like receptor (TLR) agonists and/or cancer antigens, will likely enter human trials over the next few years and potentially contribute to next-generation antitumor immune-based therapies.
Collapse
Affiliation(s)
- Mehdi Shahgolzari
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Steven Fiering
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH, United States
- Norris Cotton Cancer Center, Dartmouth Geisel School of Medicine and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| |
Collapse
|
11
|
Boone CE, Wang L, Gautam A, Newton IG, Steinmetz NF. Combining nanomedicine and immune checkpoint therapy for cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1739. [PMID: 34296535 PMCID: PMC8906799 DOI: 10.1002/wnan.1739] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/07/2021] [Accepted: 06/24/2021] [Indexed: 01/03/2023]
Abstract
Cancer immunotherapy has emerged as a pillar of the cancer therapy armamentarium. Immune checkpoint therapy (ICT) is a mainstay of modern immunotherapy. Although ICT monotherapy has demonstrated remarkable clinical efficacy in some patients, the majority do not respond to treatment. In addition, many patients eventually develop resistance to ICT, disease recurrence, and toxicity from off-target effects. Combination therapy is a keystone strategy to overcome the limitations of monotherapy. With the integration of ICT and any therapy that induces tumor cell lysis and release of tumor-associated antigens (TAAs), ICT is expected to strengthen the coordinated innate and adaptive immune responses to TAA release and promote systemic, cellular antitumor immunity. Nanomedicine is well poised to facilitate combination ICT. Nanoparticles with delivery and/or immunomodulation capacities have been successfully combined with ICT in preclinical applications. Delivery nanoparticles protect and control the targeted release of their cargo. Inherently immunomodulatory nanoparticles can facilitate immunogenic cell death, modification of the tumor microenvironment, immune cell mimicry and modulation, and/or in situ vaccination. Nanoparticles are frequently multifunctional, combining multiple treatment strategies into a single platform with ICT. Nanomedicine and ICT combinations have great potential to yield novel, powerful treatments for patients with cancer. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
| | - Lu Wang
- Department of Bioengineering, University of California, San Diego, La Jolla CA 92039, USA
| | - Aayushma Gautam
- Department of Bioengineering, University of California, San Diego, La Jolla CA 92039, USA
| | - Isabel G. Newton
- Department of Radiology, University of California, San Diego, La Jolla CA 92039, USA,Veterans Administration San Diego Healthcare System, 3350 La Jolla Village Drive San Diego, CA 92161
| | | |
Collapse
|
12
|
Possamaï D, Hanafi LA, Bellemare-Pelletier A, Hamelin K, Thébault P, Hébert MJ, Gagnon É, Leclerc D, Lapointe R. MHC class I antigen cross-presentation mediated by PapMV nanoparticles in human antigen-presenting cells is dependent on autophagy. PLoS One 2022; 16:e0261987. [PMID: 34972158 PMCID: PMC8719699 DOI: 10.1371/journal.pone.0261987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 12/14/2021] [Indexed: 12/23/2022] Open
Abstract
Nanoparticles made of the coat protein of papaya mosaic virus (PapMV) and a single-strand RNA were previously shown to be an efficient antigen presentation system for the trigger of cellular immunity. Engineering of PapMV nano with a cytotoxic T lymphocyte epitope was previously shown activating specific T lymphocytes through a proteasome-independent major histocompatibility complex class I (MHC-I) cross-presentation. In this study, we provide new insights into the mechanism of the MHC-I cross-presentation mediated by PapMV nanoparticles. We demonstrate that PapMV nanoparticles do not require the transporter associated with antigen presentation (TAP), but rather depend on lysosome acidification and cathepsin S protease activity for presentation of the T cell epitope. We have also linked the induction of autophagy with this vacuolar MHC-I cross-presentation process. Interestingly, autophagy is induced in antigen-presenting cells after PapMV nanoparticles exposure and inhibition of autophagy reduce MHC-I cross-presentation. This study demonstrates that autophagy is associated with TAP- and proteasome-independent MHC-I cross-presentation. A deeper understanding of the autophagy-dependent MHC-I cross-presentation will be useful in designing vaccination platforms that aim to trigger an efficient cytotoxic T lymphocyte response.
Collapse
Affiliation(s)
- David Possamaï
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montréal, Québec, Canada
| | - Laïla-Aïcha Hanafi
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montréal, Québec, Canada
| | - Angélique Bellemare-Pelletier
- Institut de Recherche en Immunologie et Cancérologie, Montréal, Québec, Canada
- Département de Microbiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Katia Hamelin
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montréal, Québec, Canada
| | - Paméla Thébault
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montréal, Québec, Canada
| | - Marie-Josée Hébert
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montréal, Québec, Canada
| | - Étienne Gagnon
- Institut de Recherche en Immunologie et Cancérologie, Montréal, Québec, Canada
- Département de Microbiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Denis Leclerc
- Centre de recherche en infectiologie, Centre hospitalier universitaire de Québec, Québec, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université Laval, Québec, Québec, Canada
| | - Réjean Lapointe
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montréal, Québec, Canada
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
13
|
Cui SN, Tan HY, Fan GC. Immunopathological Roles of Neutrophils in Virus Infection and COVID-19. Shock 2021; 56:345-351. [PMID: 33534399 PMCID: PMC8354486 DOI: 10.1097/shk.0000000000001740] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/14/2020] [Accepted: 01/20/2021] [Indexed: 01/08/2023]
Abstract
ABSTRACT Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been spread around the world and is currently affecting global public health. Clinical evidence indicates that the elevated number of peripheral neutrophils and higher ratio of neutrophils-to-lymphocytes are correlated with severe outcomes in COVID-19 patients, suggesting the possible immunopathological role of neutrophils during SARS-CoV-2 infection. As an abundant innate immune cell type, neutrophils are well known for their contributions to antimicrobial defense. However, their dysfunction is also associated with different inflammatory signatures during the pathogenesis of infection. Herein, in this mini-review, we summarize the recent progress on the potential role of neutrophils during COVID-19-associated inflammatory responses. In particular, we highlight the interactions between neutrophils and viruses as well as the relationship of neutrophils with cytokine storm and thrombosis in COVID-19 patients. Lastly, we discuss the importance of neutrophils as potential therapeutic targets for COVID-19.
Collapse
Affiliation(s)
- Shu-Nan Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Anesthesiology, Peking University Cancer Hospital and Institute, Beijing, China
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Hong-Yu Tan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Anesthesiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
14
|
Varshney D, Qiu SY, Graf TP, McHugh KJ. Employing Drug Delivery Strategies to Overcome Challenges Using TLR7/8 Agonists for Cancer Immunotherapy. AAPS JOURNAL 2021; 23:90. [PMID: 34181117 DOI: 10.1208/s12248-021-00620-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022]
Abstract
Toll-like receptors (TLRs) are a potential target for cancer immunotherapy due to their role in the activation of the innate immune system. More specifically, TLR7 and TLR8, two structurally similar pattern recognition receptors that trigger interferon and cytokine responses, have proven to be therapeutically relevant targets for cancer in numerous preclinical and clinical studies. When triggered by an agonist, such as imiquimod or resiquimod, the TLR7/8 activation pathway induces cellular and humoral immune responses that can kill cancer cells with high specificity. Unfortunately, TLR7/8 agonists also present a number of issues that must be overcome prior to broad clinical implementation, such as poor drug solubility and systemic toxic effects. To overcome the key limitations of TLR7/8 agonists as a cancer therapy, biomaterial-based drug delivery systems have been developed. These delivery devices are highly diverse in their design and include systems that can be directly administered to the tumor, passively accumulated in relevant cancerous and lymph tissues, triggered by environmental stimuli, or actively targeted to specific physiological areas and cellular populations. In addition to improved delivery systems, recent studies have also demonstrated the potential benefits of TLR7/8 agonist co-delivery with other types of therapies, particularly checkpoint inhibitors, cancer vaccines, and chemotherapeutics, which can yield impressive anti-cancer effects. In this review, we discuss recent advances in the development of TLR7/8 agonist delivery systems and provide perspective on promising future directions.
Collapse
Affiliation(s)
- Dhruv Varshney
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, USA
| | - Sherry Yue Qiu
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA
| | - Tyler P Graf
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA.
| |
Collapse
|
15
|
Mao C, Beiss V, Fields J, Steinmetz NF, Fiering S. Cowpea mosaic virus stimulates antitumor immunity through recognition by multiple MYD88-dependent toll-like receptors. Biomaterials 2021; 275:120914. [PMID: 34126409 DOI: 10.1016/j.biomaterials.2021.120914] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/10/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
Cowpea mosaic virus (CPMV), a non-enveloped plant virus, and empty CPMV (eCPMV), a virus-like particle (VLP) composed of CPMV capsid without nucleic acids, are potent in situ cancer vaccines when administered intratumorally (I.T.). However, it is unclear how immune cells recognize these nanoparticles and why they are immunogenic, which was investigated in this study. CPMV generated stronger selective induction of cytokines and chemokines in naïve mouse splenocytes and exhibited more potent anti-tumor efficacy than eCPMV. MyD88 is required for both CPMV- and eCPMV-elicited immune responses. Screening with human embryonic kidney (HEK)-293 cell toll-like receptor (TLR) reporter assays along with experiments in corresponding TLR-/- mice indicated CPMV and eCPMV capsids are recognized by MyD88-dependent TLR2 and TLR4. CPMV, but not eCPMV, is additionally recognized by TLR7. Secretion of type I interferons (IFNs), which requires the interaction between TLR7 and encapsulated single-stranded RNAs (ssRNAs), is critical to CPMV's better efficacy. The same recognition mechanisms are also functional in human peripheral blood mononuclear cells (PBMCs). Overall, these findings link CPMV immunotherapy efficacy with molecular recognition, provide rationale for how to develop more potent viral particles, accentuate the value of multi-TLR agonists as in situ cancer vaccines, and highlight the functional importance of type I IFNs for in situ vaccination.
Collapse
Affiliation(s)
- Chenkai Mao
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH, 03755, United States
| | - Veronique Beiss
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Jennifer Fields
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH, 03755, United States; Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical System, Lebanon, NH, 03756, United States
| | - Nicole F Steinmetz
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, 92093, United States; Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, United States; Department of Radiology, University of California, San Diego, La Jolla, CA, 92093, United States; Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093, United States; Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, 92093, United States; Institute for Materials Design and Discovery, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH, 03755, United States; Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical System, Lebanon, NH, 03756, United States.
| |
Collapse
|
16
|
Nkanga CI, Steinmetz NF. The pharmacology of plant virus nanoparticles. Virology 2021; 556:39-61. [PMID: 33545555 PMCID: PMC7974633 DOI: 10.1016/j.virol.2021.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/14/2022]
Abstract
The application of nanoparticles for medical purposes has made enormous strides in providing new solutions to health problems. The observation that plant virus-based nanoparticles (VNPs) can be repurposed and engineered as smart bio-vehicles for targeted drug delivery and imaging has launched extensive research for improving the therapeutic and diagnostic management of various diseases. There is evidence that VNPs are promising high value nanocarriers with potential for translational development. This is mainly due to their unique features, encompassing structural uniformity, ease of manufacture and functionalization by means of expression, chemical biology and self-assembly. While the development pipeline is moving rapidly, with many reports focusing on engineering and manufacturing aspects to tailor the properties and efficacy of VNPs, fewer studies have focused on gaining insights into the nanotoxicity of this novel platform nanotechnology. Herein, we discuss the pharmacology of VNPs as a function of formulation and route of administration. VNPs are reviewed in the context of their application as therapeutic adjuvants or nanocarrier excipients to initiate, enhance, attenuate or impede the formulation's toxicity. The summary of the data however also underlines the need for meticulous VNP structure-nanotoxicity studies to improve our understanding of their in vivo fates and pharmacological profiles to pave the way for translation of VNP-based formulations into the clinical setting.
Collapse
Affiliation(s)
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA, 92039, United States; Department of Bioengineering, Department of Radiology, Center for NanoImmunoEngineering, Moores Cancer Center, Institute for Materials Discovery and Design, University of California-San Diego, La Jolla, CA, 92039, United States.
| |
Collapse
|
17
|
Shahgolzari M, Pazhouhandeh M, Milani M, Fiering S, Khosroushahi AY. Alfalfa mosaic virus nanoparticles-based in situ vaccination induces antitumor immune responses in breast cancer model. Nanomedicine (Lond) 2021; 16:97-107. [PMID: 33442986 DOI: 10.2217/nnm-2020-0311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Preclinical and clinical studies show that local and systemic antitumor efficacy is achievable by in situ vaccination (ISV) using plant virus nanoparticles in which immunostimulatory reagents are directly administered into the tumor rather than systemically. Aim: To investigate a minimally studied plant virus nanoparticle, alfalfa mosaic virus (AMV), for ISV treatment of 4T1, the very aggressive and metastatic murine triple-negative breast cancer model. Materials & methods: AMV nanoparticles were propagated and characterized. Their treatment impact on in vivo tumors were analyzed using determination of inherent immunogenicity, cytokine analysis, western blotting analysis and immunohistochemistry methodologies. Results: AMV used as an ISV significantly slowed down tumor progression and prolonged survival through immune mechanisms (p < 0.001). Conclusion: Mechanistic studies show that ISV with AMV increases costimulatory molecules, inflammatory cytokines and immune effector cell infiltration and downregulates immune-suppressive molecules.
Collapse
Affiliation(s)
- Mehdi Shahgolzari
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maghsoud Pazhouhandeh
- Biotechnology Dept. Agriculture Fac. Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Morteza Milani
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Steven Fiering
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03756, USA.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth & Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Ahmad Yari Khosroushahi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
Modulation of Antigen Display on PapMV Nanoparticles Influences Its Immunogenicity. Vaccines (Basel) 2021; 9:vaccines9010033. [PMID: 33435570 PMCID: PMC7829862 DOI: 10.3390/vaccines9010033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
Background: The papaya mosaic virus (PapMV) vaccine platform is a rod-shaped nanoparticle made of the recombinant PapMV coat protein (CP) self-assembled around a noncoding single-stranded RNA (ssRNA) template. The PapMV nanoparticle induces innate immunity through stimulation of the Toll-like receptors (TLR) 7 and 8. The display of the vaccine antigen at the surface of the nanoparticle, associated with the co-stimulation signal via TLR7/8, ensures a strong stimulation of the immune response, which is ideal for the development of candidate vaccines. In this study, we assess the impact of where the peptide antigen is fused, whether at the surface or at the extremities of the nanoparticles, on the immune response directed to that antigen. Methods: Two different peptides from influenza A virus were used as model antigens. The conserved M2e peptide, derived from the matrix protein 2 was chosen as the B-cell epitope, and a peptide derived from the nucleocapsid was chosen as the cytotoxic T lymphocytes (CTL) epitope. These peptides were coupled at two different positions on the PapMV CP, the N- (PapMV-N) or the C-terminus (PapMV-C), using the transpeptidase activity of Sortase A (SrtA). The immune responses, both humoral and CD8+ T-cell-mediated, directed to the peptide antigens in the two different fusion contexts were analyzed and compared. The impact of coupling density at the surface of the nanoparticle was also investigated. Conclusions: The results demonstrate that coupling of the peptide antigens at the N-terminus (PapMV-N) of the PapMV CP led to an enhanced immune response to the coupled peptide antigens as compared to coupling to the C-terminus. The difference between the two vaccine platforms is linked to the enhanced capacity of the PapMV-N vaccine platform to stimulate TLR7/8. We also demonstrated that the strength of the immune response increases with the density of coupling at the surface of the nanoparticles.
Collapse
|
19
|
A Randomized Controlled Study to Evaluate the Safety and Reactogenicity of a Novel rVLP-Based Plant Virus Nanoparticle Adjuvant Combined with Seasonal Trivalent Influenza Vaccine Following Single Immunization in Healthy Adults 18-50 Years of Age. Vaccines (Basel) 2020; 8:vaccines8030393. [PMID: 32698532 PMCID: PMC7564144 DOI: 10.3390/vaccines8030393] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/20/2020] [Accepted: 07/06/2020] [Indexed: 02/02/2023] Open
Abstract
Inactivated influenza vaccines efficacy is variable and often poor. We conducted a phase 1 trial (NCT02188810), to assess the safety and immunogenicity of a novel nanoparticle Toll-like receptor 7/8 agonist adjuvant (Papaya Mosaic Virus) at different dose levels combined with trivalent influenza vaccine in healthy persons 18–50 years of age. Hemagglutination-inhibition assays, antibody to Influenza A virus nucleoprotein and peripheral blood mononuclear cells for measurement of interferon-gamma ELISPOT response to influenza antigens, Granzyme B and IFNγ:IL-10 ratio were measured. The most common adverse events were transient mild to severe injection site pain and no safety signals were observed. A dose-related adjuvant effect was observed. Geometric mean hemagglutination-inhibition titers increased at day 28 in most groups and waned over time, but fold-antibody responses were poor in all groups. Cell mediated immunity results were consistent with humoral responses. The Papaya Mosaic Virus adjuvant in doses of 30 to 240 µg combined with reduced influenza antigen content was safe with no signals up to 3 years after vaccination. A dose-related adjuvant effect was observed and immunogenicity results suggest that efficacy study should be conducted in influenza antigen-naïve participants.
Collapse
|
20
|
Steinmetz NF, Lim S, Sainsbury F. Protein cages and virus-like particles: from fundamental insight to biomimetic therapeutics. Biomater Sci 2020; 8:2771-2777. [PMID: 32352101 PMCID: PMC8085892 DOI: 10.1039/d0bm00159g] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Protein cages (viral and non-viral) found in nature have evolved for a variety of purposes and are found in all kingdoms of life. The main functions of these nanoscale compartments are the protection and delivery of nucleic acids e.g. virus capsids, or the enrichment and sequestration of metabolons e.g. bacterial microcompartments. This review focuses on recent developments of protein cages for use in immunotherapy and therapeutic delivery. In doing so, we highlight the unique ways in which protein cages have informed on fundamental principles governing bio-nano interactions. With the enormous existing design space among naturally occurring protein cages, there is still much to learn from studying them as biomimetic particles.
Collapse
Affiliation(s)
- Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, CA 92093, USA and Department of Bioengineering, University of California, San Diego, CA 92093, USA and Department of Radiology, University of California, San Diego, CA 92093, USA and Moores Cancer Center, University of California, San Diego, CA 92093, USA and Center for Nano-ImmunoEngineering, University of California, San Diego, CA 92093, USA
| | - Sierin Lim
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore and NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University, Singapore 637457, Singapore
| | - Frank Sainsbury
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia. and Synthetic Biology Future Science Platform, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Brisbane, QLD 4001, Australia
| |
Collapse
|
21
|
Shields CW, Wang LLW, Evans MA, Mitragotri S. Materials for Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901633. [PMID: 31250498 DOI: 10.1002/adma.201901633] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/17/2019] [Indexed: 05/20/2023]
Abstract
Breakthroughs in materials engineering have accelerated the progress of immunotherapy in preclinical studies. The interplay of chemistry and materials has resulted in improved loading, targeting, and release of immunomodulatory agents. An overview of the materials that are used to enable or improve the success of immunotherapies in preclinical studies is presented, from immunosuppressive to proinflammatory strategies, with particular emphasis on technologies poised for clinical translation. The materials are organized based on their characteristic length scale, whereby the enabling feature of each technology is organized by the structure of that material. For example, the mechanisms by which i) nanoscale materials can improve targeting and infiltration of immunomodulatory payloads into tissues and cells, ii) microscale materials can facilitate cell-mediated transport and serve as artificial antigen-presenting cells, and iii) macroscale materials can form the basis of artificial microenvironments to promote cell infiltration and reprogramming are discussed. As a step toward establishing a set of design rules for future immunotherapies, materials that intrinsically activate or suppress the immune system are reviewed. Finally, a brief outlook on the trajectory of these systems and how they may be improved to address unsolved challenges in cancer, infectious diseases, and autoimmunity is presented.
Collapse
Affiliation(s)
- C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael A Evans
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
22
|
Albakri MM, Veliz FA, Fiering SN, Steinmetz NF, Sieg SF. Endosomal toll-like receptors play a key role in activation of primary human monocytes by cowpea mosaic virus. Immunology 2020; 159:183-192. [PMID: 31630392 PMCID: PMC6954739 DOI: 10.1111/imm.13135] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/20/2019] [Accepted: 10/14/2019] [Indexed: 12/30/2022] Open
Abstract
The plant virus, cowpea mosaic virus (CPMV), has demonstrated a remarkable capacity to induce anti-tumour immune responses following direct administration into solid tumours. The molecular pathways that account for these effects and the capacity of CPMV to activate human cells are not well defined. Here, we examine the ability of CPMV particles to activate human monocytes, dendritic cells (DCs) and macrophages. Monocytes in peripheral blood mononuclear cell cultures and purified CD14+ monocytes were readily activated by CPMV in vitro, leading to induction of HLA-DR, CD86, PD-L1, IL-15R and CXCL10 expression. Monocytes released chemokines, CXCL10, MIP-1α and MIP-1β into cell culture supernatants after incubation with CPMV. DC subsets (pDC and mDC) and monocyte-derived macrophages also demonstrated evidence of activation after incubation with CPMV. Inhibitors of spleen tyrosine kinase (SYK), endocytosis or endocytic acidification impaired the capacity of CPMV to activate monocytes. Furthermore, CPMV activation of monocytes was partially blocked by a TLR7/8 antagonist. These data demonstrate that CPMV activates human monocytes in a manner dependent on SYK signalling, endosomal acidification and with an important contribution from TLR7/8 recognition.
Collapse
Affiliation(s)
- Marwah M. Albakri
- Department of PathologySchool of MedicineCase Western Reserve UniversityClevelandOHUSA
- Department of Medical Laboratory TechnologyCollege of Applied Medical SciencesTaibah UniversityMedinaSaudi Arabia
| | - Frank A. Veliz
- Department of Biomedical EngineeringSchool of MedicineCase Western Reserve UniversityClevelandOHUSA
| | - Steven N. Fiering
- Department of Microbiology and ImmunologyGeisel School of Medicine at DartmouthNorris Cotton Cancer CenterLebanonNHUSA
| | - Nicole F. Steinmetz
- Department of NanoEngineeringUniversity of California San DiegoLa JollaCAUSA
- Department of RadiologyUniversity of California San DiegoLa JollaCAUSA
- Department of BioengineeringUniversity of California San DiegoLa JollaCAUSA
- Moores Cancer CenterUniversity of California San DiegoLa JollaCAUSA
| | - Scott F. Sieg
- Division of Infectious Diseases and HIV MedicineSchool of MedicineCase Western Reserve UniversityClevelandOHUSA
| |
Collapse
|
23
|
Nuñez-Rivera A, Fournier PGJ, Arellano DL, Rodriguez-Hernandez AG, Vazquez-Duhalt R, Cadena-Nava RD. Brome mosaic virus-like particles as siRNA nanocarriers for biomedical purposes. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2020; 11:372-382. [PMID: 32175217 PMCID: PMC7059527 DOI: 10.3762/bjnano.11.28] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/07/2020] [Indexed: 05/06/2023]
Abstract
There is an increasing interest in the use of plant viruses as vehicles for anti-cancer therapy. In particular, the plant virus brome mosaic virus (BMV) and cowpea chlorotic mottle virus (CCMV) are novel potential nanocarriers for different therapies in nanomedicine. In this work, BMV and CCMV were loaded with a fluorophore and assayed on breast tumor cells. The viruses BMV and CCMV were internalized into breast tumor cells. Both viruses, BMV and CCMV, did not show cytotoxic effects on tumor cells in vitro. However, only BMV did not activate macrophages in vitro. This suggests that BMV is less immunogenic and may be a potential carrier for therapy delivery in tumor cells. Furthermore, BMV virus-like particles (VLPs) were efficiently loaded with small interfering RNA (siRNA) without packaging signal. The gene silencing was demonstrated by VLPs loaded with siGFP and tested on breast tumor cells that constitutively express the green fluorescent protein (GPF). After VLP-siGFP treatment, GFP expression was efficiently inhibited corroborating the cargo release inside tumor cells and the gene silencing. In addition, BMV VLP carring siAkt1 inhibited the tumor growth in mice. These results show the attractive potential of plant virus VLPs to deliver molecular therapy to tumor cells with low immunogenic response.
Collapse
Affiliation(s)
- Alfredo Nuñez-Rivera
- Centro de Nanociencias y Nanotecnología - Universidad Nacional Autónoma de México (UNAM) – Ensenada, Baja California, México
- Centro de Investigación Científica y de Educación Superior de Ensenada, Baja California, (CICESE), Ensenada, Baja California, México
| | - Pierrick G J Fournier
- Centro de Investigación Científica y de Educación Superior de Ensenada, Baja California, (CICESE), Ensenada, Baja California, México
| | - Danna L Arellano
- Centro de Investigación Científica y de Educación Superior de Ensenada, Baja California, (CICESE), Ensenada, Baja California, México
| | - Ana G Rodriguez-Hernandez
- Centro de Nanociencias y Nanotecnología - Universidad Nacional Autónoma de México (UNAM) – Ensenada, Baja California, México
| | - Rafael Vazquez-Duhalt
- Centro de Nanociencias y Nanotecnología - Universidad Nacional Autónoma de México (UNAM) – Ensenada, Baja California, México
| | - Ruben D Cadena-Nava
- Centro de Nanociencias y Nanotecnología - Universidad Nacional Autónoma de México (UNAM) – Ensenada, Baja California, México
| |
Collapse
|
24
|
Gestal MC, Johnson HM, Harvill ET. Immunomodulation as a Novel Strategy for Prevention and Treatment of Bordetella spp. Infections. Front Immunol 2019; 10:2869. [PMID: 31921136 PMCID: PMC6923730 DOI: 10.3389/fimmu.2019.02869] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022] Open
Abstract
Well-adapted pathogens have evolved to survive the many challenges of a robust immune response. Defending against all host antimicrobials simultaneously would be exceedingly difficult, if not impossible, so many co-evolved organisms utilize immunomodulatory tools to subvert, distract, and/or evade the host immune response. Bordetella spp. present many examples of the diversity of immunomodulators and an exceptional experimental system in which to study them. Recent advances in this experimental system suggest strategies for interventions that tweak immunity to disrupt bacterial immunomodulation, engaging more effective host immunity to better prevent and treat infections. Here we review advances in the understanding of respiratory pathogens, with special focus on Bordetella spp., and prospects for the use of immune-stimulatory interventions in the prevention and treatment of infection.
Collapse
Affiliation(s)
- Monica C Gestal
- Department of Infectious Diseases, College of Veterinary Sciences, University of Georgia, Athens, GA, United States
| | - Hannah M Johnson
- Department of Infectious Diseases, College of Veterinary Sciences, University of Georgia, Athens, GA, United States
| | - Eric T Harvill
- Department of Infectious Diseases, College of Veterinary Sciences, University of Georgia, Athens, GA, United States
| |
Collapse
|
25
|
Wang C, Beiss V, Steinmetz NF. Cowpea Mosaic Virus Nanoparticles and Empty Virus-Like Particles Show Distinct but Overlapping Immunostimulatory Properties. J Virol 2019; 93:e00129-19. [PMID: 31375592 PMCID: PMC6803287 DOI: 10.1128/jvi.00129-19] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 07/16/2019] [Indexed: 12/30/2022] Open
Abstract
Cowpea mosaic virus (CPMV) is a plant virus that has been developed for multiple biomedical and nanotechnology applications, including immunotherapy. Two key platforms are available: virus nanoparticles (VNPs) based on the complete CMPV virion, including the genomic RNA, and virus-like nanoparticles (VLPs) based on the empty CPMV (eCPMV) virion. It is unclear whether these platforms differ in terms of immunotherapeutic potential. We therefore compared their physicochemical properties and immunomodulatory activities following in situ vaccination of an aggressive ovarian tumor mouse model (ID8-Defb29/Vegf-A). In physicochemical terms, CPMV and eCPMV were very similar, and both significantly increased the survival of tumor-bearing mice and showed promising antitumor efficacy. However, they demonstrated distinct yet overlapping immunostimulatory effects due to the presence of virus RNA in wild-type particles, indicating their suitability for different immunotherapeutic strategies. Specifically, we found that the formulations had similar effects on most secreted cytokines and immune cells, but the RNA-containing CPMV particles were uniquely able to boost populations of potent antigen-presenting cells, such as tumor-infiltrating neutrophils and activated dendritic cells. Our results will facilitate the development of CPMV and eCPMV as immunotherapeutic vaccine platforms with tailored responses.IMPORTANCE The engagement of antiviral effector responses caused by viral infection is essential when using viruses or virus-like particles (VLPs) as an immunotherapeutic agent. Here, we compare the chemophysical and immunostimulatory properties of wild-type cowpea mosaic virus (CPMV) (RNA containing) and eCPMV (RNA-free VLPs) produced from two expression systems (agrobacterium-based plant expression system and baculovirus-insect cell expression). CPMV and eCPMV could each be developed as novel adjuvants to overcome immunosuppression and thus promote tumor regression in ovarian cancer (and other tumor types). To our knowledge, this is the first study to define the immunotherapeutic differences between CPMV and eCPMV, which is essential for the further development of biomedical applications for plant viruses and the selection of rational combinations of immunomodulatory reagents.
Collapse
Affiliation(s)
- Chao Wang
- Department of NanoEngineering, University of California, San Diego, La Jolla, California, USA
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Veronique Beiss
- Department of NanoEngineering, University of California, San Diego, La Jolla, California, USA
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, California, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, California, USA
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
26
|
Laliberté-Gagné MÈ, Bolduc M, Thérien A, Garneau C, Casault P, Savard P, Estaquier J, Leclerc D. Increased Immunogenicity of Full-Length Protein Antigens through Sortase-Mediated Coupling on the PapMV Vaccine Platform. Vaccines (Basel) 2019; 7:vaccines7020049. [PMID: 31212772 PMCID: PMC6630801 DOI: 10.3390/vaccines7020049] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/06/2019] [Accepted: 06/08/2019] [Indexed: 01/16/2023] Open
Abstract
Background: Flexuous rod-shape nanoparticles—made of the coat protein of papaya mosaic virus (PapMV)—provide a promising vaccine platform for the presentation of viral antigens to immune cells. The PapMV nanoparticles can be combined with viral antigens or covalently linked to them. The coupling to PapMV was shown to improve the immune response triggered against peptide antigens (<39 amino acids) but it remains to be tested if large proteins can be coupled to this platform and if the coupling will lead to an immune response improvement. Methods: Two full-length recombinant viral proteins, the influenza nucleoprotein (NP) and the simian immunodeficiency virus group-specific protein antigen (GAG) were coupled to PapMV nanoparticles using sortase A. Mice were immunized with the nanoparticles coupled to the antigens and the immune response directed to the antigens were analyzed by ELISA and ELISPOT. Results: We showed the feasibility of coupling two different full-length proteins (GAG and NP) to the nanoparticle. We also showed that the coupling to PapMV nanoparticles improved significantly the humoral and the cytotoxic T lymphocyte (CTL) immune response to the antigens. Conclusion: This proof of concept demonstrates the versatility and the efficacy of the PapMV vaccine platform in the design of vaccines against viral diseases.
Collapse
Affiliation(s)
- Marie-Ève Laliberté-Gagné
- Department of Microbiology, Infectiology and Immunology, faculty of Medicine, Laval University, Quebec City, QC G1V 4G2, Canada.
| | - Marilène Bolduc
- Department of Microbiology, Infectiology and Immunology, faculty of Medicine, Laval University, Quebec City, QC G1V 4G2, Canada.
| | - Ariane Thérien
- Department of Microbiology, Infectiology and Immunology, faculty of Medicine, Laval University, Quebec City, QC G1V 4G2, Canada.
| | - Caroline Garneau
- Department of Microbiology, Infectiology and Immunology, faculty of Medicine, Laval University, Quebec City, QC G1V 4G2, Canada.
| | - Philippe Casault
- Department of Microbiology, Infectiology and Immunology, faculty of Medicine, Laval University, Quebec City, QC G1V 4G2, Canada.
| | - Pierre Savard
- Department of Neurosciences, faculty of Medicine, Laval University, Quebec City, QC G1V 4G2, Canada.
| | - Jérome Estaquier
- Department of Microbiology, Infectiology and Immunology, faculty of Medicine, Laval University, Quebec City, QC G1V 4G2, Canada.
| | - Denis Leclerc
- Department of Microbiology, Infectiology and Immunology, faculty of Medicine, Laval University, Quebec City, QC G1V 4G2, Canada.
| |
Collapse
|
27
|
Stegelmeier AA, van Vloten JP, Mould RC, Klafuric EM, Minott JA, Wootton SK, Bridle BW, Karimi K. Myeloid Cells during Viral Infections and Inflammation. Viruses 2019; 11:E168. [PMID: 30791481 PMCID: PMC6410039 DOI: 10.3390/v11020168] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/15/2019] [Accepted: 02/16/2019] [Indexed: 12/11/2022] Open
Abstract
Myeloid cells represent a diverse range of innate leukocytes that are crucial for mounting successful immune responses against viruses. These cells are responsible for detecting pathogen-associated molecular patterns, thereby initiating a signaling cascade that results in the production of cytokines such as interferons to mitigate infections. The aim of this review is to outline recent advances in our knowledge of the roles that neutrophils and inflammatory monocytes play in initiating and coordinating host responses against viral infections. A focus is placed on myeloid cell development, trafficking and antiviral mechanisms. Although known for promoting inflammation, there is a growing body of literature which demonstrates that myeloid cells can also play critical regulatory or immunosuppressive roles, especially following the elimination of viruses. Additionally, the ability of myeloid cells to control other innate and adaptive leukocytes during viral infections situates these cells as key, yet under-appreciated mediators of pathogenic inflammation that can sometimes trigger cytokine storms. The information presented here should assist researchers in integrating myeloid cell biology into the design of novel and more effective virus-targeted therapies.
Collapse
Affiliation(s)
- Ashley A Stegelmeier
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Jacob P van Vloten
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Robert C Mould
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Elaine M Klafuric
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Jessica A Minott
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Sarah K Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
28
|
Abstract
Cancer immunotherapy approaches have emerged as novel treatment regimens against cancer. A particularly interesting avenue is the concept of in situ vaccination, where immunostimulatory agents are introduced into an identified tumor to overcome local immunosuppression and, if successful, mount systemic antitumor immunity. We had previously shown that nanoparticles from cowpea mosaic virus (CPMV) are highly potent in inducing long-lasting antitumor immunity when used as an in situ vaccine in various tumor mouse models. Here we asked whether the nanoparticles from tobacco mosaic virus (TMV) could also be applied as an in situ vaccine and, if so, whether efficacy or mechanism of immune-activation would be affected by the nanoparticle size (300 × 18 nm native TMV vs 50 × 18 nm short TMV nanorods), shape (nanorods vs spherical TMV, termed SNP), or state of assembly (assembled TMV rod vs free coat protein, CP). Our studies indicate that CPMV, but less so TMV, elicits potent antitumor immunity after intratumoral treatment of dermal melanoma (B16F10 using C57BL/6 mice). TMV and TMVshort slowed tumor growth and increased survival time, however, at significantly lower potency compared to that of CPMV. There were no apparent differences between TMV, TMVshort, or the SNP indicating that the aspect ratio does not necessarily play a role in plant viral in situ vaccines. The free CPs did not elicit an antitumor response or immunostimulation, which may indicate that a multivalent assembly is required to trigger an innate immune recognition and activation. Differential potency of CPMV vs TMV can be explained with differences in immune-activation: data indicate that CPMV stimulates an antitumor response through recruitment of monocytes into the tumor microenvironment (TME), establishing signaling through the IFN-γ pathway, which also leads to recruitment of tumor-infiltrated neutrophils (TINs) and natural killer (NK) cells. Furthermore, the priming of the innate immune system also mounts an adaptive response with CD4+ and CD8+ T cell recruitment and establishment of effector memory cells. While the TMV treatment also lead to the recruitment of innate immune cells as well as T cells (although to a lesser degree), key differences were noted in cyto/chemokine profiling with TMV inducing a potent immune response early on characterized by strong pro-inflammatory cytokines, primarily IL-6. Together, data indicate that some plant viral nanotechnology platforms are more suitable for application as in situ vaccines than others; understanding the intricate differences and underlying mechanism of immune-activation may set the stage for clinical development of these technologies.
Collapse
Affiliation(s)
| | - Chao Wang
- Department of Biomedical Engineering
| | - Steven Fiering
- Department of Microbiology and Immunology
- Norris Cotton Cancer Center, Dartmouth University, Lebanon, New Hampshire 03756, United States
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering
- Department of Radiology
- Department of Materials Science and Engineering
- Macromolecular Science and Engineering
- Division of General Medical Sciences-Oncology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| |
Collapse
|
29
|
Hefferon KL. Repurposing Plant Virus Nanoparticles. Vaccines (Basel) 2018; 6:vaccines6010011. [PMID: 29443902 PMCID: PMC5874652 DOI: 10.3390/vaccines6010011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 01/26/2018] [Accepted: 02/02/2018] [Indexed: 12/21/2022] Open
Abstract
Plants have been explored for many years as inexpensive and versatile platforms for the generation of vaccines and other biopharmaceuticals. Plant viruses have also been engineered to either express subunit vaccines or act as epitope presentation systems. Both icosahedral and helical, filamentous-shaped plant viruses have been used for these purposes. More recently, plant viruses have been utilized as nanoparticles to transport drugs and active molecules into cancer cells. The following review describes the use of both icosahedral and helical plant viruses in a variety of new functions against cancer. The review illustrates the breadth of variation among different plant virus nanoparticles and how this impacts the immune response.
Collapse
|