1
|
Kane G, Lusi C, Brassil M, Atukorale P. Engineering approaches for innate immune-mediated tumor microenvironment remodeling. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 21:100406. [PMID: 38213392 PMCID: PMC10777078 DOI: 10.1016/j.iotech.2023.100406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Cancer immunotherapy offers transformative promise particularly for the treatment of lethal cancers, since a correctly trained immune system can comprehensively orchestrate tumor clearance with no need for continued therapeutic intervention. Historically, the majority of immunotherapies have been T cell-focused and have included immune checkpoint inhibitors, chimeric antigen receptor T cells, and T-cell vaccines. Unfortunately T-cell-focused therapies have failed to achieve optimal efficacy in most solid tumors largely because of a highly immunosuppressed 'cold' or immune-excluded tumor microenvironment (TME). Recently, a rapidly growing treatment paradigm has emerged that focuses on activation of tumor-resident innate antigen-presenting cells, such as dendritic cells and macrophages, which can drive a proinflammatory immune response to remodel the TME from 'cold' or immune-excluded to 'hot'. Early strategies for TME remodeling centered on free cytokines and agonists, but these approaches have faced significant hurdles in both delivery and efficacy. Systemic toxicity from off-target inflammation is a paramount concern in these therapies. To address this critical gap, engineering approaches have provided the opportunity to add 'built-in' capabilities to cytokines, agonists, and other therapeutic agents to mediate improved delivery and efficacy. Such capabilities have included protective encapsulation to shield them from degradation, targeting to direct them with high specificity to tumors, and co-delivery strategies to harness synergistic proinflammatory pathways. Here, we review innate immune-mediated TME remodeling engineering approaches that focus on cytokines, innate immune agonists, immunogenic viruses, and cell-based methods, highlighting emerging preclinical approaches and strategies that are either being tested in clinical trials or already Food and Drug Administration approved.
Collapse
Affiliation(s)
- G.I. Kane
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - C.F. Lusi
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - M.L. Brassil
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - P.U. Atukorale
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, USA
| |
Collapse
|
2
|
Karan S, Jung E, Boone C, Steinmetz NF. Synergistic combination therapy using cowpea mosaic virus intratumoral immunotherapy and Lag-3 checkpoint blockade. Cancer Immunol Immunother 2024; 73:51. [PMID: 38349406 PMCID: PMC10864561 DOI: 10.1007/s00262-024-03636-2] [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: 11/30/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024]
Abstract
Immune checkpoint therapy (ICT) for cancer can yield dramatic clinical responses; however, these may only be observed in a minority of patients. These responses can be further limited by subsequent disease recurrence and resistance. Combination immunotherapy strategies are being developed to overcome these limitations. We have previously reported enhanced efficacy of combined intratumoral cowpea mosaic virus immunotherapy (CPMV IIT) and ICT approaches. Lymphocyte-activation gene-3 (LAG-3) is a next-generation inhibitory immune checkpoint with broad expression across multiple immune cell subsets. Its expression increases on activated T cells and contributes to T cell exhaustion. We observed heightened efficacy of a combined CPMV IIT and anti-LAG-3 treatment in a mouse model of melanoma. Further, LAG-3 expression was found to be increased within the TME following intratumoral CPMV administration. The integration of CPMV IIT with LAG-3 inhibition holds significant potential to improve treatment outcomes by concurrently inducing a comprehensive anti-tumor immune response, enhancing local immune activation, and mitigating T cell exhaustion.
Collapse
Affiliation(s)
- Sweta Karan
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, USA
| | - Eunkyeong Jung
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, USA
| | - Christine Boone
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA.
| | - Nicole F Steinmetz
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, USA.
- Department of Radiology, University of California, San Diego, La Jolla, CA, 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, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, USA.
- Center for Engineering in Cancer, Institute of Engineering Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
3
|
Chen YL, Bao CJ, Duan JL, Xie Y, Lu WL. Overcoming biological barriers by virus-like drug particles for drug delivery. Adv Drug Deliv Rev 2023; 203:115134. [PMID: 37926218 DOI: 10.1016/j.addr.2023.115134] [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: 09/09/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Virus-like particles (VLPs) have natural structural antigens similar to those found in viruses, making them valuable in vaccine immunization. Furthermore, VLPs have demonstrated significant potential in drug delivery, and emerged as promising vectors for transporting chemical drug, genetic drug, peptide/protein, and even nanoparticle drug. With virus-like permeability and strong retention, they can effectively target specific organs, tissues or cells, facilitating efficient intracellular drug release. Further modifications allow VLPs to transfer across various physiological barriers, thus acting the purpose of efficient drug delivery and accurate therapy. This article provides an overview of VLPs, covering their structural classifications, deliverable drugs, potential physiological barriers in drug delivery, strategies for overcoming these barriers, and future prospects.
Collapse
Affiliation(s)
- Yu-Ling Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Chun-Jie Bao
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jia-Lun Duan
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Xie
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Wan-Liang Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
4
|
Azizi M, Shahgolzari M, Fathi-Karkan S, Ghasemi M, Samadian H. Multifunctional plant virus nanoparticles: An emerging strategy for therapy of cancer. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1872. [PMID: 36450366 DOI: 10.1002/wnan.1872] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 12/05/2022]
Abstract
Cancer therapy requires sophisticated treatment strategies to obtain the highest success. Nanotechnology is enabling, revolutionizing, and multidisciplinary concepts to improve conventional cancer treatment modalities. Nanomaterials have a central role in this scenario, explaining why various nanomaterials are currently being developed for cancer therapy. Viral nanoparticles (VNPs) have shown promising performance in cancer therapy due to their unique features. VNPs possess morphological homogeneity, ease of functionalization, biocompatibility, biodegradability, water solubility, and high absorption efficiency that are beneficial for cancer therapy applications. In the current review paper, we highlight state-of-the-art properties and potentials of plant viruses, strategies for multifunctional plant VNPs formulations, potential applications and challenges in VNPs-based cancer therapy, and finally practical solutions to bring potential cancer therapy one step closer to real applications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Mehdi Azizi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Dental Implants Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehdi Shahgolzari
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sonia Fathi-Karkan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Maryam Ghasemi
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hadi Samadian
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
5
|
Han H, Xing L, Chen BT, Liu Y, Zhou TJ, Wang Y, Zhang LF, Li L, Cho CS, Jiang HL. Progress on the pathological tissue microenvironment barrier-modulated nanomedicine. Adv Drug Deliv Rev 2023; 200:115051. [PMID: 37549848 DOI: 10.1016/j.addr.2023.115051] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/21/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Imbalance in the tissue microenvironment is the main obstacle to drug delivery and distribution in the human body. Before penetrating the pathological tissue microenvironment to the target site, therapeutic agents are usually accompanied by three consumption steps: the first step is tissue physical barriers for prevention of their penetration, the second step is inactivation of them by biological molecules, and the third step is a cytoprotective mechanism for preventing them from functioning on specific subcellular organelles. However, recent studies in drug-hindering mainly focus on normal physiological rather than pathological microenvironment, and the repair of damaged physiological barriers is also rarely discussed. Actually, both the modulation of pathological barriers and the repair of damaged physiological barriers are essential in the disease treatment and the homeostasis maintenance. In this review, we present an overview describing the latest advances in the generality of these pathological barriers and barrier-modulated nanomedicine. Overall, this review holds considerable significance for guiding the design of nanomedicine to increase drug efficacy in the future.
Collapse
Affiliation(s)
- Han Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Bi-Te Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling-Feng Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
6
|
Nikitin N, Vasiliev Y, Kovalenko A, Ryabchevskaya E, Kondakova O, Evtushenko E, Karpova O. Plant Viruses as Adjuvants for Next-Generation Vaccines and Immunotherapy. Vaccines (Basel) 2023; 11:1372. [PMID: 37631940 PMCID: PMC10458565 DOI: 10.3390/vaccines11081372] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023] Open
Abstract
Vaccines are the cornerstone of infectious disease control and prevention. The outbreak of SARS-CoV-2 has confirmed the urgent need for a new approach to the design of novel vaccines. Plant viruses and their derivatives are being used increasingly for the development of new medical and biotechnological applications, and this is reflected in a number of preclinical and clinical studies. Plant viruses have a unique combination of features (biosafety, low reactogenicity, inexpensiveness and ease of production, etc.), which determine their potential. This review presents the latest data on the use of plant viruses with different types of symmetry as vaccine components and adjuvants in cancer immunotherapy. The discussion concludes that the most promising approaches might be those that use structurally modified plant viruses (spherical particles) obtained from the Tobacco mosaic virus. These particles combine high adsorption properties (as a carrier) with strong immunogenicity, as has been confirmed using various antigens in animal models. According to current research, it is evident that plant viruses have great potential for application in the development of vaccines and in cancer immunotherapy.
Collapse
Affiliation(s)
- Nikolai Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | | | - Angelina Kovalenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - Ekaterina Ryabchevskaya
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - Olga Kondakova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - Ekaterina Evtushenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - Olga Karpova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| |
Collapse
|
7
|
Shahgolzari M, Venkataraman S, Osano A, Akpa PA, Hefferon K. Plant Virus Nanoparticles Combat Cancer. Vaccines (Basel) 2023; 11:1278. [PMID: 37631846 PMCID: PMC10459942 DOI: 10.3390/vaccines11081278] [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: 05/19/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/27/2023] Open
Abstract
Plant virus nanoparticles (PVNPs) have garnered considerable interest as a promising nanotechnology approach to combat cancer. Owing to their biocompatibility, stability, and adjustable surface functionality, PVNPs hold tremendous potential for both therapeutic and imaging applications. The versatility of PVNPs is evident from their ability to be tailored to transport a range of therapeutic agents, including chemotherapy drugs, siRNA, and immunomodulators, thereby facilitating targeted delivery to the tumor microenvironment (TME). Furthermore, PVNPs may be customized with targeting ligands to selectively bind to cancer cell receptors, reducing off-target effects. Additionally, PVNPs possess immunogenic properties and can be engineered to exhibit tumor-associated antigens, thereby stimulating anti-tumor immune responses. In conclusion, the potential of PVNPs as a versatile platform for fighting cancer is immense, and further research is required to fully explore their potential and translate them into clinical applications.
Collapse
Affiliation(s)
- Mehdi Shahgolzari
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran
| | - Srividhya Venkataraman
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Anne Osano
- Department of Natural Sciences, Bowie State University, Bowie, MD 20715, USA
| | - Paul Achile Akpa
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka 410001, Enugu State, Nigeria
| | - Kathleen Hefferon
- Department of Microbiology, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
8
|
Huang G, Liu L, Pan H, Cai L. Biomimetic Active Materials Guided Immunogenic Cell Death for Enhanced Cancer Immunotherapy. SMALL METHODS 2023; 7:e2201412. [PMID: 36572642 DOI: 10.1002/smtd.202201412] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/22/2022] [Indexed: 05/17/2023]
Abstract
Despite immunotherapy emerging as a vital approach to improve cancer treatment, the activation of efficient immune responses is still hampered by immunosuppression, especially due to the low tumor immunogenicity. Immunogenic cell death (ICD) is a promising strategy to reshape the tumor microenvironment (TME) for achieving high immunogenicity. Various stimuli are able to effectively initiate their specific ICD by utilizing the corresponding ICD-inducer. However, the ICD-guided antitumor immune effects are usually impaired by various biological barriers and TME-associated immune resistance. Biomimetic active materials are being extensively explored as guided agents for ICD due to their unique advantages. In this review, two major strategies are systematically introduced that have been employed to exploit biomimetic active materials guided ICD for cancer immunotherapy, mainly including naive organism-derived nanoagents and engineered bioactive platforms. This review outlines the recent advances in the field at biomimetic active materials guided physiotherapy, chemotherapy, and biotherapy for ICD induction. The advances and challenges of biomimetic active materials guided ICD for cancer immunotherapy applications are further discussed in future clinical practice. This review provides an overview of the advances of biomimetic active materials for targeting immunoregulation and treatment and can contribute to the future of advanced antitumor combination therapy.
Collapse
Affiliation(s)
- Guojun Huang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
9
|
Shah S, Famta P, Tiwari V, Kotha AK, Kashikar R, Chougule MB, Chung YH, Steinmetz NF, Uddin M, Singh SB, Srivastava S. Instigation of the epoch of nanovaccines in cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1870. [PMID: 36410742 PMCID: PMC10182210 DOI: 10.1002/wnan.1870] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/03/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022]
Abstract
Cancer is an unprecedented proliferation of cells leading to abnormalities in differentiation and maturation. Treatment of primary and metastatic cancer is challenging. In addition to surgery, chemotherapy and radiation therapies have been conventionally used; however, they suffer from severe toxicity and non-specificity. Immunotherapy, the science of programming the body's own defense system against cancer has gained tremendous attention in the last few decades. However, partial immunogenic stimulation, premature degradation and inability to activate dendritic and helper T cells has resulted in limited clinical success. The era of nanomedicine has brought about several breakthroughs in various pharmaceutical and biomedical fields. Hereby, we review and discuss the interplay of tumor microenvironment (TME) and the immunological cascade and how they can be employed to develop nanoparticle-based cancer vaccines and immunotherapies. Nanoparticles composed of lipids, polymers and inorganic materials contain useful properties suitable for vaccine development. Proteinaceous vaccines derived from mammalian viruses, bacteriophages and plant viruses also have unique advantages due to their immunomodulation capabilities. This review accounts for all such considerations. Additionally, we explore how attributes of nanotechnology can be utilized to develop successful nanomedicine-based vaccines for cancer therapy. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, INDIA
| | - Paras Famta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, INDIA
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering, & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, INDIA
| | - Arun K Kotha
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Rama Kashikar
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Mahavir Bhupal Chougule
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Young Hun Chung
- Departments of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nicole F. Steinmetz
- Departments of Bioengineering, NanoEngineering, Radiology, Moores Cancer Center, Center for Nano-ImmunoEngineering, Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mohammad Uddin
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, INDIA
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, INDIA
| |
Collapse
|
10
|
Li ZZ, He JY, Wu Q, Liu B, Bu LL. Recent advances in targeting myeloid-derived suppressor cells and their applications to radiotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:233-264. [PMID: 37438019 DOI: 10.1016/bs.ircmb.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of heterogenous immature myeloid cells with potent immune suppressive properties that not only constrain anti-tumor immune activation and functions, promote tumor progression, but also contribute to treatment resistance and tumor relapse. Targeting MDSCs may be a promising new cancer treatment method, but there is still a problem of low treatment efficiency. Combined application with radiotherapy may be a potential method to solve this problem. Drug delivery systems (DDSs) provide more efficient targeted drug delivery capability and can reduce the toxicity and side effects of drugs. Recent advance in DDSs targeting development, recruitment, differentiation, and elimination of MDSCs have shown promising effect in reversing immune inhibition and in overcoming radiotherapy resistance. In this review, we systematically summarized DDSs applied to target MDSCs for the first time, and classified and discussed it according to its different mechanisms of action. In addition, this paper also reviewed the biological characteristics of MDSCs and their role in the initiation, progression, and metastasis of cancer. Moreover, this review also summarizes the role of DDSs targeting MDSCs in radiosensitization. Finally, the future development of DDSs targeting MDSCs is also prospected.
Collapse
Affiliation(s)
- Zi-Zhan Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Jing-Yu He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Bing Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China.
| | - Lin-Lin Bu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
11
|
Ou J, Zhu M, Ju X, Xu D, Lu G, Li K, Jiang W, Wan C, Zhao Y, Han Y, Tian Y, Niu Z. One-Dimensional Rod-like Tobacco Mosaic Virus Promotes Macrophage Polarization for a Tumor-Suppressive Microenvironment. NANO LETTERS 2023; 23:2056-2064. [PMID: 36695738 DOI: 10.1021/acs.nanolett.2c03809] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The phenotype of tumor-associated macrophages plays an important role in their function of regulating the tumor immune microenvironment. The M1-phenotype macrophages display tumor-killing and immune activating functions. Here we show that the tobacco mosaic virus (TMV), a rod-like plant virus, can polarize macrophages to an M1 phenotype and shape a tumor-suppressive microenvironment. RAW 264.7 cells and bone marrow derived-macrophages (BMDMs) can recognize TMV via Toll-like receptor-4, and then the MAPK and NF-κB signaling pathways are activated, leading to the production of pro-inflammatory factors. Furthermore, the in vivo assessments on a subcutaneous co-injection tumor model show that the TMV-polarized BMDMs shape a tumor-suppressive microenvironment, resulting in remarkable delay of 4T1 tumor growth. Another in vivo assessment on an established tumor model indicates the high tumor-metastasis-inhibiting capacity of TMV-polarized BMDMs. This work suggests a role for this plant virus in macrophage-mediated therapeutic approaches and provides a strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- Jinzhao Ou
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Meng Zhu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Xiaoyan Ju
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Dandan Xu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Guojun Lu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Kejia Li
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Wei Jiang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Chenxiao Wan
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Yuexia Zhao
- Biochemical Engineering College, Beijing Union University, No. 97, North Fourth Ring East Road, Beijing 100023, P.R. China
| | - Yongping Han
- Biochemical Engineering College, Beijing Union University, No. 97, North Fourth Ring East Road, Beijing 100023, P.R. China
| | - Ye Tian
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Zhongwei Niu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| |
Collapse
|
12
|
Emerging Trends in Nano-Driven Immunotherapy for Treatment of Cancer. Vaccines (Basel) 2023; 11:vaccines11020458. [PMID: 36851335 PMCID: PMC9968063 DOI: 10.3390/vaccines11020458] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 02/18/2023] Open
Abstract
Despite advancements in the development of anticancer medications and therapies, cancer still has the greatest fatality rate due to a dismal prognosis. Traditional cancer therapies include chemotherapy, radiotherapy, and targeted therapy. The conventional treatments have a number of shortcomings, such as a lack of selectivity, non-specific cytotoxicity, suboptimal drug delivery to tumour locations, and multi-drug resistance, which results in a less potent/ineffective therapeutic outcome. Cancer immunotherapy is an emerging and promising strategy to elicit a pronounced immune response against cancer. Immunotherapy stimulates the immune system with cancer-specific antigens or immune checkpoint inhibitors to overcome the immune suppressive tumour microenvironment and kill the cancer cells. However, delivery of the antigen or immune checkpoint inhibitors and activation of the immune response need to circumvent the issues pertaining to short lifetimes and effect times, as well as adverse effects associated with off-targeting, suboptimal, or hyperactivation of the immune system. Additional challenges posed by the tumour suppressive microenvironment are less tumour immunogenicity and the inhibition of effector T cells. The evolution of nanotechnology in recent years has paved the way for improving treatment efficacy by facilitating site-specific and sustained delivery of the therapeutic moiety to elicit a robust immune response. The amenability of nanoparticles towards surface functionalization and tuneable physicochemical properties, size, shape, and surfaces charge have been successfully harnessed for immunotherapy, as well as combination therapy, against cancer. In this review, we have summarized the recent advancements made in choosing different nanomaterial combinations and their modifications made to enable their interaction with different molecular and cellular targets for efficient immunotherapy. This review also highlights recent trends in immunotherapy strategies to be used independently, as well as in combination, for the destruction of cancer cells, as well as prevent metastasis and recurrence.
Collapse
|
13
|
Jung E, Mao C, Bhatia M, Koellhoffer EC, Fiering SN, Steinmetz NF. Inactivated Cowpea Mosaic Virus for In Situ Vaccination: Differential Efficacy of Formalin vs UV-Inactivated Formulations. Mol Pharm 2023; 20:500-507. [PMID: 36399598 PMCID: PMC9812890 DOI: 10.1021/acs.molpharmaceut.2c00744] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cowpea mosaic virus (CPMV) has been developed as a promising nanoplatform technology for cancer immunotherapy; when applied as in situ vaccine, CPMV exhibits potent, systemic, and durable efficacy. While CPMV is not infectious to mammals, it is infectious to legumes; therefore, agronomic safety needs to be addressed to broaden the translational application of CPMV. RNA-containing formulations are preferred over RNA-free virus-like particles because the RNA and protein, each, contribute to CPMV's potent antitumor efficacy. We have previously optimized inactivation methods to develop CPMV that contains RNA but is not infectious to plants. We established that inactivated CPMV has reduced efficacy compared to untreated, native CPMV. However, a systematic comparison between native CPMV and different inactivated forms of CPMV was not done. Therefore, in this study, we directly compared the therapeutic efficacies and mechanisms of immune activation of CPMV, ultraviolet- (UV-), and formalin (Form)-inactivated CPMV to explain the differential efficacies. In a B16F10 melanoma mouse tumor model, Form-CPMV suppressed the tumor growth with prolonged survival (there were no statistical differences comparing CPMV and Form-CPMV). In comparison, UV-CPMV inhibited tumor growth significantly but not as well as Form-CPMV or CPMV. The reduced therapeutic efficacy of UV-CPMV is explained by the degree of cross-linking and aggregated state of the RNA, which renders it inaccessible for sensing by Toll-like receptor (TLR) 7/8 to activate immune responses. The mechanistic studies showed that the highly aggregated state of UV-CPMV inhibited TLR7 signaling more so than for the Form-CPMV formulation, reducing the secretion of interleukin-6 (IL-6) and interferon-α (IFN-α), cytokines associated with TLR7 signaling. These findings support the translational development of Form-CPMV as a noninfectious immunotherapeutic agent.
Collapse
Affiliation(s)
| | | | - Misha Bhatia
- Department of Nanoengineering, University of, California San Diego, La Jolla, California 92093, United, States
| | - Edward C. Koellhoffer
- Radiology, University of California San Diego, La Jolla, California 92093, United States
| | - Steven N. Fiering
- Department of Microbiology and, Immunology and Dartmouth Cancer Center, Dartmouth, Geisel School of Medicine, Hanover, New Hampshire 03755, United States
| | - Nicole F. Steinmetz
- Department of Nanoengineering, Radiology, Bioengineering, Moores Cancer Center, Center for Nano-Immuno Engineering, and Institute for Materials, Design and Discovery, University of California San Diego, La, Jolla, California 92093, United States
| |
Collapse
|
14
|
Mao C, Beiss V, Ho GW, Fields J, Steinmetz NF, Fiering S. In situ vaccination with cowpea mosaic virus elicits systemic antitumor immunity and potentiates immune checkpoint blockade. J Immunother Cancer 2022; 10:e005834. [PMID: 36460333 PMCID: PMC9723958 DOI: 10.1136/jitc-2022-005834] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND In situ vaccination (ISV) is a cancer immunotherapy strategy in which immunostimulatory reagents are introduced directly into a tumor to stimulate antitumor immunity both against the treated tumor and systemically against untreated tumors. Recently, we showed that cowpea mosaic virus (CPMV) is a potent multi-toll-like receptor (TLR) agonist with potent efficacy for treating tumors in mice and dogs by ISV. However, ISV with CPMV alone does not uniformly treat all mouse tumor models tested, however this can be overcome through strategic combinations. More insight is needed to delineate potency and mechanism of systemic antitumor immunity and abscopal effect. METHOD We investigated the systemic efficacy (abscopal effect) of CPMV ISV with a two-tumor mouse model using murine tumor lines B16F10, 4T1, CT26 and MC38. Flow cytometry identified changes in cell populations responsible for systemic efficacy of CPMV. Transgenic knockout mice and depleting antibodies validated the role of relevant candidate cell populations and cytokines. We evaluated these findings and engineered a multicomponent combination therapy to specifically target the candidate cell population and investigated its systemic efficacy, acquired resistance and immunological memory in mouse models. RESULTS ISV with CPMV induces systemic antitumor T-cell-mediated immunity that inhibits growth of untreated tumors and requires conventional type-1 dendritic cells (cDC1s). Furthermore, using multiple tumor mouse models resistant to anti-programmed death 1 (PD-1) therapy, we tested the hypothesis that CPMV along with local activation of antigen-presenting cells with agonistic anti-CD40 can synergize and strengthen antitumor efficacy. Indeed, this combination ISV strategy induces an influx of CD8+ T cells, triggers regression in both treated local and untreated distant tumors and potentiates tumor responses to anti-PD-1 therapy. Moreover, serial ISV overcomes resistance to anti-PD-1 therapy and establishes tumor-specific immunological memory. CONCLUSIONS These findings provide new insights into in situ TLR activation and cDC1 recruitment as effective strategies to overcome resistance to immunotherapy in treated and untreated tumors.
Collapse
Affiliation(s)
- Chenkai Mao
- Microbiology and Immunology, Dartmouth College Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Veronique Beiss
- Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Gregory W Ho
- Microbiology and Immunology, Dartmouth College Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Jennifer Fields
- Microbiology and Immunology, Dartmouth College Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Nicole F Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Steven Fiering
- Microbiology and Immunology, Dartmouth College Geisel School of Medicine, Lebanon, New Hampshire, USA
- Geisel School of Medicine at Dartmouth, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire, USA
| |
Collapse
|
15
|
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: 8.5] [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
|
16
|
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: 2.0] [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
|
17
|
Liao Z, Huang J, Lo PC, Lovell JF, Jin H, Yang K. Self-adjuvanting cancer nanovaccines. J Nanobiotechnology 2022; 20:345. [PMID: 35883176 PMCID: PMC9316869 DOI: 10.1186/s12951-022-01545-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/04/2022] [Indexed: 11/12/2022] Open
Abstract
Nanovaccines, a new generation of vaccines that use nanoparticles as carriers and/or adjuvants, have been widely used in the prevention and treatment of various diseases, including cancer. Nanovaccines have sparked considerable interest in cancer therapy due to a variety of advantages, including improved access to lymph nodes (LN), optimal packing and presentation of antigens, and induction of a persistent anti-tumor immune response. As a delivery system for cancer vaccines, various types of nanoparticles have been designed to facilitate the delivery of antigens and adjuvants to lymphoid organs and antigen-presenting cells (APCs). Particularly, some types of nanoparticles are able to confer an immune-enhancing capability and can themselves be utilized for adjuvant-like effect for vaccines, suggesting a direction for a better use of nanomaterials and the optimization of cancer vaccines. However, this role of nanoparticles in vaccines has not been well studied. To further elucidate the role of self-adjuvanting nanovaccines in cancer therapy, we review the mechanisms of antitumor vaccine adjuvants with respect to nanovaccines with self-adjuvanting properties, including enhancing cross-presentation, targeting signaling pathways, biomimicking of the natural invasion process of pathogens, and further unknown mechanisms. We surveyed self-adjuvanting cancer nanovaccines in clinical research and discussed their advantages and challenges. In this review, we classified self-adjuvanting cancer nanovaccines according to the underlying immunomodulatory mechanism, which may provide mechanistic insights into the design of nanovaccines in the future.
Collapse
Affiliation(s)
- Zhiyun Liao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Honglin Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
18
|
Microbial-Derived Toll-like Receptor Agonism in Cancer Treatment and Progression. Cancers (Basel) 2022; 14:cancers14122923. [PMID: 35740589 PMCID: PMC9221178 DOI: 10.3390/cancers14122923] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/02/2022] [Accepted: 06/13/2022] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Toll like receptors (TLRs) are a group of transmembrane receptors belonging to the class of pattern recognition receptors (PRR), which are involved in recognition of pathogen associated molecular patterns (PAMPs), inducing immune response. During the past decade, a number of preclinical and clinical breakthroughs in the field of TLR agonists has immerged in cancer research and some of these agents have performed exceptionally well in clinical trials. Based on evidence from scientific studies, we draw attention to several microbial based TLR agonists and discuss their relevance in various cancer and explore various microbial based TLR agonists for developing effective immunotherapeutic strategies against cancer. Abstract Toll-like receptors (TLRs) are typical transmembrane proteins, which are essential pattern recognition receptors in mediating the effects of innate immunity. TLRs recognize structurally conserved molecules derived from microbes and damage-associated molecular pattern molecules that play an important role in inflammation. Since the first discovery of the Toll receptor by the team of J. Hoffmann in 1996, in Drosophila melanogaster, numerous TLRs have been identified across a wide range of invertebrate and vertebrate species. TLR stimulation leads to NF-κB activation and the subsequent production of pro-inflammatory cytokines and chemokines, growth factors and anti-apoptotic proteins. The expression of TLRs has also been observed in many tumors, and their stimulation results in tumor progression or regression, depending on the TLR and tumor type. The anti-tumoral effects can result from the activation of anti-tumoral immune responses and/or the direct induction of tumor cell death. The pro-tumoral effects may be due to inducing tumor cell survival and proliferation or by acting on suppressive or inflammatory immune cells in the tumor microenvironment. The aim of this review is to draw attention to the effects of TLR stimulation in cancer, the activation of various TLRs by microbes in different types of tumors, and, finally, the role of TLRs in anti-cancer immunity and tumor rejection.
Collapse
|
19
|
Kondakova OA, Evtushenko EA, Baranov OA, Nikitin NA, Karpova OV. Structurally Modified Plant Viruses and Bacteriophages with Helical Structure. Properties and Applications. BIOCHEMISTRY (MOSCOW) 2022; 87:548-558. [PMID: 35790410 PMCID: PMC9201271 DOI: 10.1134/s0006297922060062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Structurally modified virus particles can be obtained from the rod-shaped or filamentous virions of plant viruses and bacteriophages by thermal or chemical treatment. They have recently attracted attention of the researchers as promising biogenic platforms for the development of new biotechnologies. This review presents data on preparation, structure, and properties of the structurally modified virus particles. In addition, their biosafety for animals is considered, as well as the areas of application of such particles in biomedicine. A separate section is devoted to one of the most relevant and promising areas for the use of structurally modified plant viruses – design of vaccine candidates based on them.
Collapse
Affiliation(s)
- Olga A Kondakova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | | | - Oleg A Baranov
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Nikolai A Nikitin
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Olga V Karpova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| |
Collapse
|
20
|
Feng C, Li Y, Ferdows BE, Patel DN, Ouyang J, Tang Z, Kong N, Chen E, Tao W. Emerging vaccine nanotechnology: From defense against infection to sniping cancer. Acta Pharm Sin B 2022; 12:2206-2223. [PMID: 35013704 PMCID: PMC8730377 DOI: 10.1016/j.apsb.2021.12.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/24/2021] [Accepted: 11/16/2021] [Indexed: 02/07/2023] Open
Abstract
Looking retrospectively at the development of humanity, vaccination is an unprecedented medical landmark that saves lives by harnessing the human immune system. During the ongoing coronavirus disease 2019 (COVID-19) pandemic, vaccination is still the most effective defense modality. The successful clinical application of the lipid nanoparticle-based Pfizer/BioNTech and Moderna mRNA COVID-19 vaccines highlights promising future of nanotechnology in vaccine development. Compared with conventional vaccines, nanovaccines are supposed to have advantages in lymph node accumulation, antigen assembly, and antigen presentation; they also have, unique pathogen biomimicry properties because of well-organized combination of multiple immune factors. Beyond infectious diseases, vaccine nanotechnology also exhibits considerable potential for cancer treatment. The ultimate goal of cancer vaccines is to fully mobilize the potency of the immune system as a living therapeutic to recognize tumor antigens and eliminate tumor cells, and nanotechnologies have the requisite properties to realize this goal. In this review, we summarize the recent advances in vaccine nanotechnology from infectious disease prevention to cancer immunotherapy and highlight the different types of materials, mechanisms, administration methods, as well as future perspectives.
Collapse
Affiliation(s)
- Chan Feng
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Bijan Emiliano Ferdows
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dylan Neal Patel
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jiang Ouyang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zhongmin Tang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Enguo Chen
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Corresponding authors. Fax: +001 857 307 2337 (Wei Tao).
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Corresponding authors. Fax: +001 857 307 2337 (Wei Tao).
| |
Collapse
|
21
|
McNulty MJ, Schwartz A, Delzio J, Karuppanan K, Jacobson A, Hart O, Dandekar A, Giritch A, Nandi S, Gleba Y, McDonald KA. Affinity Sedimentation and Magnetic Separation With Plant-Made Immunosorbent Nanoparticles for Therapeutic Protein Purification. Front Bioeng Biotechnol 2022; 10:865481. [PMID: 35573255 PMCID: PMC9092175 DOI: 10.3389/fbioe.2022.865481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/04/2022] [Indexed: 11/20/2022] Open
Abstract
The virus-based immunosorbent nanoparticle is a nascent technology being developed to serve as a simple and efficacious agent in biosensing and therapeutic antibody purification. There has been particular emphasis on the use of plant virions as immunosorbent nanoparticle chassis for their diverse morphologies and accessible, high yield manufacturing via plant cultivation. To date, studies in this area have focused on proof-of-concept immunosorbent functionality in biosensing and purification contexts. Here we consolidate a previously reported pro-vector system into a single Agrobacterium tumefaciens vector to investigate and expand the utility of virus-based immunosorbent nanoparticle technology for therapeutic protein purification. We demonstrate the use of this technology for Fc-fusion protein purification, characterize key nanomaterial properties including binding capacity, stability, reusability, and particle integrity, and present an optimized processing scheme with reduced complexity and increased purity. Furthermore, we present a coupling of virus-based immunosorbent nanoparticles with magnetic particles as a strategy to overcome limitations of the immunosorbent nanoparticle sedimentation-based affinity capture methodology. We report magnetic separation results which exceed the binding capacity reported for current industry standards by an order of magnitude.
Collapse
Affiliation(s)
- Matthew J. McNulty
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
| | | | - Jesse Delzio
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
| | - Kalimuthu Karuppanan
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
| | - Aaron Jacobson
- Department of Plant Sciences, University of California, Davis, Davis, CA, United States
| | - Olivia Hart
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
| | - Abhaya Dandekar
- Department of Plant Sciences, University of California, Davis, Davis, CA, United States
| | | | - Somen Nandi
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
- Global HealthShare® Initiative, University of California, Davis, Davis, CA, United States
| | | | - Karen A. McDonald
- Department of Chemical Engineering, University of California, Davis, Davis, CA, United States
- Global HealthShare® Initiative, University of California, Davis, Davis, CA, United States
- *Correspondence: Karen A. McDonald,
| |
Collapse
|
22
|
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
|
23
|
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: 0.5] [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
|
24
|
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: 8.5] [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
|
25
|
Chen QW, Qiao JY, Liu XH, Zhang C, Zhang XZ. Customized materials-assisted microorganisms in tumor therapeutics. Chem Soc Rev 2021; 50:12576-12615. [PMID: 34605834 DOI: 10.1039/d0cs01571g] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Microorganisms have been extensively applied as active biotherapeutic agents or drug delivery vehicles for antitumor treatment because of their unparalleled bio-functionalities. Taking advantage of the living attributes of microorganisms, a new avenue has been opened in anticancer research. The integration of customized functional materials with living microorganisms has demonstrated unprecedented potential in solving existing questions and even conferring microorganisms with updated antitumor abilities and has also provided an innovative train of thought for enhancing the efficacy of microorganism-based tumor therapy. In this review, we have summarized the emerging development of customized materials-assisted microorganisms (MAMO) (including bacteria, viruses, fungi, microalgae, as well as their components) in tumor therapeutics with an emphasis on the rational utilization of chosen microorganisms and tailored materials, the ingenious design of biohybrid systems, and the efficacious antitumor mechanisms. The future perspectives and challenges in this field are also discussed.
Collapse
Affiliation(s)
- Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Ji-Yan Qiao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Xin-Hua Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Cheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| |
Collapse
|
26
|
Gorbet MJ, Singh A, Mao C, Fiering S, Ranjan A. Using nanoparticles for in situ vaccination against cancer: mechanisms and immunotherapy benefits. Int J Hyperthermia 2021; 37:18-33. [PMID: 33426995 DOI: 10.1080/02656736.2020.1802519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy to treat cancer is now an established clinical approach. Immunotherapy can be applied systemically, as done with checkpoint blockade antibodies, but it can also be injected directly into identified tumors, in a strategy of in situ vaccination (ISV). ISV is designed to stimulate a strong local antitumor immune response involving both innate and adaptive immune cells, and through this generate a systemic antitumor immune response against metastatic tumors. A variety of ISVs have been utilized to generate an immunostimulatory tumor microenvironment (TME). These include attenuated microorganisms, recombinant proteins, small molecules, physical disruptors of TME (alternating magnetic and focused ultrasound heating, photothermal therapy, and radiotherapy), and more recently nanoparticles (NPs). NPs are attractive and unique since they can load multiple drugs or other reagents to influence immune and cancer cell functions in the TME, affording a unique opportunity to stimulate antitumor immunity. Here, we describe the NP-ISV therapeutic mechanisms, review chemically synthesized NPs (i.e., liposomes, polymeric, chitosan-based, inorganic NPs, etc.), biologically derived NPs (virus and bacteria-based NPs), and energy-activated NP-ISVs in the context of their use as local ISV. Data suggests that NP-ISVs can enhance outcomes of immunotherapeutic regimens including those utilizing tumor hyperthermia and checkpoint blockade therapies.
Collapse
Affiliation(s)
| | - Akansha Singh
- College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Chenkai Mao
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center at Dartmouth and Dartmouth Hitchcock, Lebanon, NH, USA
| | - Ashish Ranjan
- College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| |
Collapse
|
27
|
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: 15.0] [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
|
28
|
Rahimian N, Miraei HR, Amiri A, Ebrahimi MS, Nahand JS, Tarrahimofrad H, Hamblin MR, Khan H, Mirzaei H. Plant-based vaccines and cancer therapy: Where are we now and where are we going? Pharmacol Res 2021; 169:105655. [PMID: 34004270 DOI: 10.1016/j.phrs.2021.105655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 10/21/2022]
Abstract
Therapeutic vaccines are an effective approach in cancer therapy for treating the disease at later stages. The Food and Drug Administration (FDA) recently approved the first therapeutic cancer vaccine, and further studies are ongoing in clinical trials. These are expected to result in the future development of vaccines with relatively improved efficacy. Several vaccination approaches are being studied in pre-clinical and clinical trials, including the generation of anti-cancer vaccines by plant expression systems.This approach has advantages, such as high safety and low costs, especially for the synthesis of recombinant proteins. Nevertheless, the development of anti-cancer vaccines in plants is faced with some technical obstacles.Herein, we summarize some vaccines that have been used in cancer therapy, with an emphasis on plant-based vaccines.
Collapse
Affiliation(s)
- Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Hamid Reza Miraei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashahd, Iran
| | | | - Javid Sadri Nahand
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Tarrahimofrad
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 20282028, South Africa
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
29
|
Viana IMDO, Roussel S, Defrêne J, Lima EM, Barabé F, Bertrand N. Innate and adaptive immune responses toward nanomedicines. Acta Pharm Sin B 2021; 11:852-870. [PMID: 33747756 PMCID: PMC7955583 DOI: 10.1016/j.apsb.2021.02.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/04/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022] Open
Abstract
Since the commercialization of the first liposomes used for drug delivery, Doxil/Caelyx® and Myocet®, tremendous progress has been made in understanding interactions between nanomedicines and biological systems. Fundamental work at the interface of engineering and medicine has allowed nanomedicines to deliver therapeutic small molecules and nucleic acids more efficiently. While nanomedicines are used in oncology for immunotherapy or to deliver combinations of cytotoxics, the clinical successes of gene silencing approaches like patisiran lipid complexes (Onpattro®) have paved the way for a variety of therapies beyond cancer. In parallel, the global severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has highlighted the potential of mRNA vaccines to develop immunization strategies at unprecedented speed. To rationally design therapeutic and vaccines, chemists, materials scientists, and drug delivery experts need to better understand how nanotechnologies interact with the immune system. This review presents a comprehensive overview of the innate and adaptative immune systems and emphasizes the intricate mechanisms through which nanomedicines interact with these biological functions.
Collapse
|
30
|
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: 16] [Impact Index Per Article: 5.3] [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
|
31
|
Chariou PL, Beiss V, Ma Y, Steinmetz NF. In situ vaccine application of inactivated CPMV nanoparticles for cancer immunotherapy. MATERIALS ADVANCES 2021; 2:1644-1656. [PMID: 34368764 PMCID: PMC8323807 DOI: 10.1039/d0ma00752h] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/26/2021] [Indexed: 05/24/2023]
Abstract
Cowpea mosaic virus (CPMV) is currently in the development pipeline for multiple biomedical applications, including cancer immunotherapy. In particular the application of CPMV as in situ vaccine has shown promise; here the plant viral nanoparticle is used as an adjuvant and is injected directly into a tumor to reverse immunosuppression and prime systemic anti-tumor immunity. Efficacy of this CPMV-based cancer immunotherapy has been demonstrated in multiple tumor mouse models and canine cancer patients. However, while CPMV is non-infectious to mammals, it is infectious to legumes and therefore, from a safety perspective, it is desired to develop non-infectious CPMV formulations. Non-infectious virus-like particles of CPMV devoid of nucleic acids have been produced; nevertheless, efficacy of such empty CPMV nanoparticles does not match efficacy of nucleic acid-laden CPMV. The multivalent capsid activates the innate immune system through pathogen pattern recognition receptors (PRRs) such as toll-like receptors (TLRs); the RNA cargo provides additional signaling through TLR-7/8, which boosts the efficacy of this adjuvant. Therefore, in this study, we set out to develop RNA-laden, but non-infectious CPMV. We report inactivation of CPMV using UV light and chemical inactivation using β-propiolactone (βPL) or formalin. 7.5 J cm-2 UV, 50 mM βPL or 1 mM formalin was determined to be sufficient to inactivate CPMV and prevented plant infection. We compared the immunogenicity of native CPMV and inactivated CPMV formulations in vitro and in vivo using RAW-Blue™ reporter cells and a murine syngeneic, orthotropic melanoma model (using B16F10 cells and C57BL6 mice). While the in vitro assay indicated activation of the RAW-Blue™ reporter cells by formaldehyde and UV-inactivated CPMV at levels comparable to native CPMV; βPL-inactivated CPMV appeared to have diminished activity. Tumor mouse model experiments indicate potent efficacy of the chemically-inactivated CPMV (UV-treated CPMV was not tested) leading to tumor regression and increased survival; efficacy was somewhat reduced when compared to CPMV, however these samples outperformed the empty CPMV nanoparticles. These results will facilitate the translational development of safe and potent CPMV-based cancer immunotherapies.
Collapse
Affiliation(s)
- Paul L. Chariou
- Department of Bioengineering, University of California-San DiegoLa JollaCA 92039USA
| | - Veronique Beiss
- Department of NanoEngineering, University of California-San DiegoLa JollaCA 92039USA
| | - Yifeng Ma
- Department of NanoEngineering, University of California-San DiegoLa JollaCA 92039USA
| | - Nicole F. Steinmetz
- Department of Bioengineering, University of California-San DiegoLa JollaCA 92039USA
- Department of NanoEngineering, University of California-San DiegoLa JollaCA 92039USA
- Department of Radiology, University of California-San DiegoLa JollaCA 92039USA
- Moores Cancer Center, University of California-San DiegoLa JollaCA 92039USA
- Center for Nano-ImmunoEngineering, University of California-San DiegoLa JollaCA 92039USA
- Institute for Materials Discovery and Design, University of California-San DiegoLa JollaCA 92039USA
| |
Collapse
|
32
|
Xu J, Wang C. Cell-derived vesicles for delivery of cancer immunotherapy. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2020.00031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In recent years, cancer immunotherapy has received unprecedented attention due to the clinical achievements. The applications of biomedical engineering and materials science to cancer immunotherapy have solved the challenges caused by immunotherapy to a certain extent. Among them, cell-derived vesicles are natural biomaterials chosen as carriers or immune-engineering in view of their many unique advantages. This review will briefly introduce the recent applications of cell-derived vesicles for cancer immunotherapy.
Collapse
Affiliation(s)
- Jialu Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| |
Collapse
|
33
|
Abstract
In recent years, cancer immunotherapy has received unprecedented attention due to the clinical achievements. The applications of biomedical engineering and materials science to cancer immunotherapy have solved the challenges caused by immunotherapy to a certain extent. Among them, cell-derived vesicles are natural biomaterials chosen as carriers or immune-engineering in view of their many unique advantages. This review will briefly introduce the recent applications of cell-derived vesicles for cancer immunotherapy.
Collapse
Affiliation(s)
- Jialu Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| |
Collapse
|
34
|
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: 1.0] [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
|
35
|
Shukla S, Wang C, Beiss V, Cai H, Washington T, Murray AA, Gong X, Zhao Z, Masarapu H, Zlotnick A, Fiering S, Steinmetz NF. The unique potency of Cowpea mosaic virus (CPMV) in situ cancer vaccine. Biomater Sci 2020; 8:5489-5503. [PMID: 32914796 PMCID: PMC8086234 DOI: 10.1039/d0bm01219j] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immunosuppressive tumor microenvironment enables cancer to resist immunotherapies. We have established that intratumoral administration of plant-derived Cowpea mosaic virus (CPMV) nanoparticles as an in situ vaccine overcomes the local immunosuppression and stimulates a potent anti-tumor response in several mouse cancer models and canine patients. CPMV does not infect mammalian cells but acts as a danger signal that leads to the recruitment and activation of innate and subsequently, adaptive immune cells. In the present study we addressed whether other icosahedral viruses or virus-like particles (VLPs) of plant, bacteriophage and mammalian origin can be similarly employed as intratumoral immunotherapy. Our results indicate that CPMV in situ vaccine outperforms Cowpea chlorotic mottle virus (CCMV), Physalis mosaic virus (PhMV), Sesbania mosaic virus (SeMV), bacteriophage Qβ VLPs, or Hepatitis B virus capsids (HBVc). Furthermore, ex vivo and in vitro assays reveal unique features of CPMV that makes it an inherently stronger immune stimulant.
Collapse
Affiliation(s)
- Sourabh Shukla
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA.
| | - Chao Wang
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA.
| | - Veronique Beiss
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA.
| | - Hui Cai
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA.
| | - Torus Washington
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA.
| | - Abner A Murray
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Xingjian Gong
- Department of Bioengineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zhongchao Zhao
- Molecular and Cellular Biochemistry Department, Indiana University Bloomington, IN 47405, USA
| | - Hema Masarapu
- Department of Virology, Sri Venkateswara University, Tirupati 517502, India
| | - Adam Zlotnick
- Molecular and Cellular Biochemistry Department, Indiana University Bloomington, IN 47405, USA
| | - Steven Fiering
- Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA. and Department of Bioengineering, University of California-San Diego, La Jolla, CA 92039, USA and Department of Radiology, University of California-San Diego, La Jolla, CA 92039, USA and Moores Cancer Center, University of California-San Diego, La Jolla, CA 92039, USA and Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
| |
Collapse
|
36
|
Shahgolzari M, Pazhouhandeh M, Milani M, Yari Khosroushahi A, Fiering S. Plant viral nanoparticles for packaging and in vivo delivery of bioactive cargos. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1629. [PMID: 32249552 DOI: 10.1002/wnan.1629] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/14/2020] [Accepted: 02/21/2020] [Indexed: 01/15/2023]
Abstract
Nanoparticles have unique capabilities and considerable promise for many different biological uses. One capability is delivering bioactive cargos to specific cells, tissues, or organisms. Depending on the task, there are multiple variables to consider including nanoparticle selection, targeting strategies, and incorporating cargo so it can be delivered in a biologically active form. One nanoparticle option, genetically controlled plant viral nanoparticles (PVNPs), is highly uniform within a given virus but quite variable between viruses with a broad range of useful properties. PVNPs are flexible and versatile tools for incorporating and delivering a wide range of small or large molecule cargos. Furthermore, PVNPs can be modified to create nanostructures that can solve problems in medical, environmental, and basic research. This review discusses the currently available techniques for delivering bioactive cargos with PVNPs and potential cargos that can be delivered with these strategies. This article is categorized under: Implantable Materials and Surgical Technologies > Nanomaterials and Implants Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Mehdi Shahgolzari
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maghsoud Pazhouhandeh
- Biotechnology Department, Agricultural Faculty, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Morteza Milani
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - 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
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth and Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| |
Collapse
|
37
|
Abdou P, Wang Z, Chen Q, Chan A, Zhou DR, Gunadhi V, Gu Z. Advances in engineering local drug delivery systems for cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1632. [PMID: 32255276 PMCID: PMC7725287 DOI: 10.1002/wnan.1632] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy aims to leverage the immune system to suppress the growth of tumors and to inhibit metastasis. The recent promising clinical outcomes associated with cancer immunotherapy have prompted research and development efforts towards enhancing the efficacy of immune checkpoint blockade, cancer vaccines, cytokine therapy, and adoptive T cell therapy. Advancements in biomaterials, nanomedicine, and micro-/nano-technology have facilitated the development of enhanced local delivery systems for cancer immunotherapy, which can enhance treatment efficacy while minimizing toxicity. Furthermore, locally administered cancer therapies that combine immunotherapy with chemotherapy, radiotherapy, or phototherapy have the potential to achieve synergistic antitumor effects. Herein, the latest studies on local delivery systems for cancer immunotherapy are surveyed, with an emphasis on the therapeutic benefits associated with the design of biomaterials and nanomedicines. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Peter Abdou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Road, Suzhou, 215123, Jiangsu, PR China
| | - Amanda Chan
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Daojia R. Zhou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Vivienne Gunadhi
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
38
|
Chung YH, Cai H, Steinmetz NF. Viral nanoparticles for drug delivery, imaging, immunotherapy, and theranostic applications. Adv Drug Deliv Rev 2020; 156:214-235. [PMID: 32603813 PMCID: PMC7320870 DOI: 10.1016/j.addr.2020.06.024] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 02/06/2023]
Abstract
Viral nanoparticles (VNPs) encompass a diverse array of naturally occurring nanomaterials derived from plant viruses, bacteriophages, and mammalian viruses. The application and development of VNPs and their genome-free versions, the virus-like particles (VLPs), for nanomedicine is a rapidly growing. VLPs can encapsulate a wide range of active ingredients as well as be genetically or chemically conjugated to targeting ligands to achieve tissue specificity. VLPs are manufactured through scalable fermentation or molecular farming, and the materials are biocompatible and biodegradable. These properties have led to a wide range of applications, including cancer therapies, immunotherapies, vaccines, antimicrobial therapies, cardiovascular therapies, gene therapies, as well as imaging and theranostics. The use of VLPs as drug delivery agents is evolving, and sufficient research must continuously be undertaken to translate these therapies to the clinic. This review highlights some of the novel research efforts currently underway in the VNP drug delivery field in achieving this greater goal.
Collapse
Affiliation(s)
- Young Hun Chung
- Department of Bioengineering, University of California-San Diego, La Jolla, CA 92093, United States
| | - Hui Cai
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92093, United States
| | - Nicole F Steinmetz
- Department of Bioengineering, University of California-San Diego, La Jolla, CA 92093, United States; Department of NanoEngineering, 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.
| |
Collapse
|
39
|
Tong Q, Qiu N, Ji J, Ye L, Zhai G. Research Progress in Bioinspired Drug Delivery Systems. Expert Opin Drug Deliv 2020; 17:1269-1288. [PMID: 32543953 DOI: 10.1080/17425247.2020.1783235] [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: 12/21/2022]
Abstract
INTRODUCTION To tackle challenges associated with traditional drug carriers, investigators have explored cells, cellular membrane, and macromolecular components including proteins and exosomes for the fabrication of delivery vehicles, owing to their excellent biocompatibility, lower toxicity, lower immunogenicity and similarities with the host. Biomacromolecule- and biomimetic nanoparticle (NP)-based drug/gene carriers are drawing immense attention, and biomimetic drug delivery systems (BDDSs) have been conceived and constructed. AREAS COVERED This review focuses on BDDS based on mammalian cells, including blood cells, cancer cells, adult stem cells, endogenous proteins, pathogens and extracellular vesicles (EVs). EXPERT OPINION Compared with traditional drug delivery systems (DDSs), BDDSs are based on biological nanocarriers, exhibiting superior biocompatibility, fewer side effects, natural targeting, and diverse modifications. In addition to directly employing natural biomaterials such as cells, proteins, pathogens and EVs as carriers, BDDSs offer these advantages by mimicking the structure of natural nanocarriers through bioengineering technologies. Furthermore, BDDSs demonstrate fewer limitations and irregularities than natural materials and can overcome several shortcomings associated with natural carriers. Although research remains ongoing to resolve these limitations, it is anticipated that BDDSs possess the potential to overcome challenges associated with traditional DDS, with a promising future in the treatment of human diseases.
Collapse
Affiliation(s)
- Qirong Tong
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Na Qiu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| |
Collapse
|
40
|
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: 40] [Impact Index Per Article: 10.0] [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
|
41
|
Zhou J, Kroll AV, Holay M, Fang RH, Zhang L. Biomimetic Nanotechnology toward Personalized Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901255. [PMID: 31206841 PMCID: PMC6918015 DOI: 10.1002/adma.201901255] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/07/2019] [Indexed: 04/14/2023]
Abstract
While traditional approaches for disease management in the era of modern medicine have saved countless lives and enhanced patient well-being, it is clear that there is significant room to improve upon the current status quo. For infectious diseases, the steady rise of antibiotic resistance has resulted in super pathogens that do not respond to most approved drugs. In the field of cancer treatment, the idea of a cure-all silver bullet has long been abandoned. As a result of the challenges facing current treatment and prevention paradigms in the clinic, there is an increasing push for personalized therapeutics, where plans for medical care are established on a patient-by-patient basis. Along these lines, vaccines, both against bacteria and tumors, are a clinical modality that could benefit significantly from personalization. Effective vaccination strategies could help to address many challenging disease conditions, but current vaccines are limited by factors such as a lack of potency and antigenic breadth. Recently, researchers have turned toward the use of biomimetic nanotechnology as a means of addressing these hurdles. Recent progress in the development of biomimetic nanovaccines for antibacterial and anticancer applications is discussed, with an emphasis on their potential for personalized medicine.
Collapse
Affiliation(s)
- Jiarong Zhou
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ashley V Kroll
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Maya Holay
- 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
|
42
|
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: 117] [Impact Index Per Article: 29.3] [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
|
43
|
Shukla S, Wang C, Beiss V, Steinmetz NF. Antibody Response against Cowpea Mosaic Viral Nanoparticles Improves In Situ Vaccine Efficacy in Ovarian Cancer. ACS NANO 2020; 14:2994-3003. [PMID: 32133838 PMCID: PMC8085886 DOI: 10.1021/acsnano.9b07865] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Cancer immunotherapies are designed to facilitate recognition and elimination of transformed cells by the immune system. We have established the immunotherapeutic efficacy of the plant virus cowpea mosaic virus (CPMV) as an in situ vaccine in several syngeneic tumor mouse models as well as in companion dogs with metastatic melanoma. Intratumoral injection of CPMV modulates the local tumor microenvironment to relieve immunosuppression and potentiate antitumor immunity. The viral nucleocapsid that drives this antitumor immunity, however, also is a potent immunogen itself, and thus immune response in the form of anti-CPMV antibodies is expected during the treatment based on repeat administrations. Moreover, being part of the food chain, pre-existing antibodies to plant viruses may be prevalent. The presence of such pre-existing anti-CPMV immunity could potentially impact immunotherapeutic efficacy of the in situ vaccine and could have translational implications. To address such concerns, this study evaluated the efficacy of CPMV in situ vaccine in the presence of pre-existing antibodies in a syngeneic mouse model of ovarian cancer. Our results indicate that prior exposure to CPMV had no negative impact on the efficacy of CPMV in situ vaccine. Strikingly, an improved efficacy of CPMV in situ vaccine was observed. This study therefore presents an important milestone in the translational development of plant viral-based in situ vaccines and alleviates concerns about the presence of anti-CPMV antibodies, which are developed during the course of treatment but have no impact on immunotherapeutic efficacy.
Collapse
|
44
|
Chariou PL, Ortega-Rivera OA, Steinmetz NF. Nanocarriers for the Delivery of Medical, Veterinary, and Agricultural Active Ingredients. ACS NANO 2020; 14:2678-2701. [PMID: 32125825 PMCID: PMC8085836 DOI: 10.1021/acsnano.0c00173] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Nanocarrier-based delivery systems can be used to increase the safety and efficacy of active ingredients in medical, veterinary, or agricultural applications, particularly when such ingredients are unstable, sparingly soluble, or cause off-target effects. In this review, we highlight the diversity of nanocarrier materials and their key advantages compared to free active ingredients. We discuss current trends based on peer-reviewed research articles, patent applications, clinical trials, and the nanocarrier formulations already approved by regulatory bodies. Although most nanocarriers have been engineered to combat cancer, the number of formulations developed for other purposes is growing rapidly, especially those for the treatment of infectious diseases and parasites affecting humans, livestock, and companion animals. The regulation and prohibition of many pesticides have also fueled research to develop targeted pesticide delivery systems based on nanocarriers, which maximize efficacy while minimizing the environmental impact of agrochemicals.
Collapse
|
45
|
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: 17] [Impact Index Per Article: 4.3] [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
|
46
|
Santoni M, Zampieri R, Avesani L. Plant Virus Nanoparticles for Vaccine Applications. Curr Protein Pept Sci 2020; 21:344-356. [PMID: 32048964 DOI: 10.2174/1389203721666200212100255] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 09/16/2019] [Accepted: 10/19/2019] [Indexed: 12/29/2022]
Abstract
In the rapidly evolving field of nanotechnology, plant virus nanoparticles (pVNPs) are emerging as powerful tools in diverse applications ranging from biomedicine to materials science. The proteinaceous structure of plant viruses allows the capsid structure to be modified by genetic engineering and/or chemical conjugation with nanoscale precision. This means that pVNPs can be engineered to display peptides and proteins on their external surface, including immunodominant peptides derived from pathogens allowing pVNPs to be used for active immunization. In this context, pVNPs are safer than VNPs derived from mammalian viruses because there is no risk of infection or reversion to pathogenicity. Furthermore, pVNPs can be produced rapidly and inexpensively in natural host plants or heterologous production platforms. In this review, we discuss the use of pVNPs for the delivery of peptide antigens to the host immune in pre-clinical studies with the final aim of promoting systemic immunity against the corresponding pathogens. Furthermore, we described the versatility of plant viruses, with innate immunostimulatory properties, in providing a huge natural resource of carriers that can be used to develop the next generation of sustainable vaccines.
Collapse
Affiliation(s)
- Mattia Santoni
- Department of Biotechnology, University of Verona. Strada Le Grazie, 15. 37134 Verona, Italy
| | | | - Linda Avesani
- Department of Biotechnology, University of Verona. Strada Le Grazie, 15. 37134 Verona, Italy
- Diamante srl. Strada Le Grazie, 15. 37134 Verona, Italy
| |
Collapse
|
47
|
|
48
|
Bamogo PKA, Brugidou C, Sérémé D, Tiendrébéogo F, Djigma FW, Simpore J, Lacombe S. Virus-based pharmaceutical production in plants: an opportunity to reduce health problems in Africa. Virol J 2019; 16:167. [PMID: 31888686 PMCID: PMC6937724 DOI: 10.1186/s12985-019-1263-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Developing African countries face health problems that they struggle to solve. The major causes of this situation are high therapeutic and logistical costs. Plant-made therapeutics are easy to produce due to the lack of the safety considerations associated with traditional fermenter-based expression platforms, such as mammalian cells. Plant biosystems are easy to scale up and inexpensive, and they do not require refrigeration or a sophisticated medical infrastructure. These advantages provide an opportunity for plant-made pharmaceuticals to counteract diseases for which medicines were previously inaccessible to people in countries with few resources. MAIN BODY The techniques needed for plant-based therapeutic production are currently available. Viral expression vectors based on plant viruses have greatly enhanced plant-made therapeutic production and have been exploited to produce a variety of proteins of industrial, pharmaceutical and agribusiness interest. Some neglected tropical diseases occurring exclusively in the developing world have found solutions through plant bioreactor technology. Plant viral expression vectors have been reported in the production of therapeutics against these diseases occurring exclusively in the third world, and some virus-derived antigens produced in plants exhibit appropriate antigenicity and immunogenicity. However, all advances in the use of plants as bioreactors have been made by companies in Europe and America. The developing world is still far from acquiring this technology, although plant viral expression vectors may provide crucial help to overcome neglected diseases. CONCLUSION Today, interest in these tools is rising, and viral amplicons made in and for Africa are in progress. This review describes the biotechnological advances in the field of plant bioreactors, highlights factors restricting access to this technology by those who need it most and proposes a solution to overcome these limitations.
Collapse
Affiliation(s)
- Pingdwende Kader Aziz Bamogo
- Interactions Plantes Microorganismes et Environnement (IPME), IRD, CIRAD, Université Montpellier, 911 Avenue Agropolis BP64501, 34394, Montpellier Cedex 5, France
- Laboratoire de Virologie et de Biotechnologies Végétales, Institut de L'Environnement et de Recherches Agricoles (INERA)/LMI Patho-Bios, 01BP476, Ouagadougou 01, Burkina Faso
- Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), Ecole Doctorale Sciences et Technologie, Université Joseph Ki-Zerbo; Centre de Recherche Biomoléculaire Piétro Annigoni (CERBA), Ouagadougou 01, BP, 364, Burkina Faso
| | - Christophe Brugidou
- Interactions Plantes Microorganismes et Environnement (IPME), IRD, CIRAD, Université Montpellier, 911 Avenue Agropolis BP64501, 34394, Montpellier Cedex 5, France
- Laboratoire de Virologie et de Biotechnologies Végétales, Institut de L'Environnement et de Recherches Agricoles (INERA)/LMI Patho-Bios, 01BP476, Ouagadougou 01, Burkina Faso
| | - Drissa Sérémé
- Laboratoire de Virologie et de Biotechnologies Végétales, Institut de L'Environnement et de Recherches Agricoles (INERA)/LMI Patho-Bios, 01BP476, Ouagadougou 01, Burkina Faso
| | - Fidèle Tiendrébéogo
- Laboratoire de Virologie et de Biotechnologies Végétales, Institut de L'Environnement et de Recherches Agricoles (INERA)/LMI Patho-Bios, 01BP476, Ouagadougou 01, Burkina Faso
| | - Florencia Wendkuuni Djigma
- Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), Ecole Doctorale Sciences et Technologie, Université Joseph Ki-Zerbo; Centre de Recherche Biomoléculaire Piétro Annigoni (CERBA), Ouagadougou 01, BP, 364, Burkina Faso
| | - Jacques Simpore
- Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), Ecole Doctorale Sciences et Technologie, Université Joseph Ki-Zerbo; Centre de Recherche Biomoléculaire Piétro Annigoni (CERBA), Ouagadougou 01, BP, 364, Burkina Faso
| | - Séverine Lacombe
- Interactions Plantes Microorganismes et Environnement (IPME), IRD, CIRAD, Université Montpellier, 911 Avenue Agropolis BP64501, 34394, Montpellier Cedex 5, France.
- Laboratoire de Virologie et de Biotechnologies Végétales, Institut de L'Environnement et de Recherches Agricoles (INERA)/LMI Patho-Bios, 01BP476, Ouagadougou 01, Burkina Faso.
| |
Collapse
|
49
|
|
50
|
Li C, Wang J, Wang Y, Gao H, Wei G, Huang Y, Yu H, Gan Y, Wang Y, Mei L, Chen H, Hu H, Zhang Z, Jin Y. Recent progress in drug delivery. Acta Pharm Sin B 2019; 9:1145-1162. [PMID: 31867161 PMCID: PMC6900554 DOI: 10.1016/j.apsb.2019.08.003] [Citation(s) in RCA: 422] [Impact Index Per Article: 84.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/10/2019] [Accepted: 07/16/2019] [Indexed: 01/05/2023] Open
Abstract
Drug delivery systems (DDS) are defined as methods by which drugs are delivered to desired tissues, organs, cells and subcellular organs for drug release and absorption through a variety of drug carriers. Its usual purpose to improve the pharmacological activities of therapeutic drugs and to overcome problems such as limited solubility, drug aggregation, low bioavailability, poor biodistribution, lack of selectivity, or to reduce the side effects of therapeutic drugs. During 2015-2018, significant progress in the research on drug delivery systems has been achieved along with advances in related fields, such as pharmaceutical sciences, material sciences and biomedical sciences. This review provides a concise overview of current progress in this research area through its focus on the delivery strategies, construction techniques and specific examples. It is a valuable reference for pharmaceutical scientists who want to learn more about the design of drug delivery systems.
Collapse
Affiliation(s)
- Chong Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jiancheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yiguang Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Gang Wei
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Yongzhuo Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Haijun Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yong Gan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yongjun Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lin Mei
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Huabing Chen
- School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Haiyan Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhiping Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|