1
|
Huda MN, Nurunnabi M. Potential Application of Exosomes in Vaccine Development and Delivery. Pharm Res 2022; 39:2635-2671. [PMID: 35028802 PMCID: PMC8757927 DOI: 10.1007/s11095-021-03143-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023]
Abstract
Exosomes are cell-derived components composed of proteins, lipid, genetic information, cytokines, and growth factors. They play a vital role in immune modulation, cell-cell communication, and response to inflammation. Immune modulation has downstream effects on the regeneration of damaged tissue, promoting survival and repair of damaged resident cells, and promoting the tumor microenvironment via growth factors, antigens, and signaling molecules. On top of carrying biological messengers like mRNAs, miRNAs, fragmented DNA, disease antigens, and proteins, exosomes modulate internal cell environments that promote downstream cell signaling pathways to facilitate different disease progression and induce anti-tumoral effects. In this review, we have summarized how vaccines modulate our immune response in the context of cancer and infectious diseases and the potential of exosomes as vaccine delivery vehicles. Both pre-clinical and clinical studies show that exosomes play a decisive role in processes like angiogenesis, prognosis, tumor growth metastasis, stromal cell activation, intercellular communication, maintaining cellular and systematic homeostasis, and antigen-specific T- and B cell responses. This critical review summarizes the advancement of exosome based vaccine development and delivery, and this comprehensive review can be used as a valuable reference for the broader delivery science community.
Collapse
Affiliation(s)
- Md Nurul Huda
- Department of Pharmaceutical Sciences, University of Texas at El Paso School of Pharmacy, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Enviromental Science and Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, University of Texas at El Paso School of Pharmacy, 1101 N. Campbell St, El Paso, TX, 79902, USA.
- Enviromental Science and Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA.
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA.
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, USA.
| |
Collapse
|
2
|
Jarai BM, Stillman Z, Fromen CA. Hydrogel nanoparticle degradation influences the activation and survival of primary macrophages. J Mater Chem B 2021; 9:7246-7257. [PMID: 34226910 PMCID: PMC8446340 DOI: 10.1039/d1tb00982f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The effect of nanoparticle (NP) internalization on cell fate has emerged as an important consideration for nanomedicine design, as macrophages and other phagocytes are the primary clearance mechanisms of administered NP formulations. Pro-survival signaling is thought to be concurrent with phagocytosis and recent work has shown increased macrophage survival following lysosomal processing of internalized NPs. These observations have opened the door to explorations of NP physiochemical properties aimed at tuning the NP-driven macrophage survival at the lysosomal synapse. Here, we report that NP-induced macrophage survival and activation is strongly dependent on NP degradation rate using a series of thiol-containing poly(ethylene glycol) diacrylate-based NPs of equivalent size and zeta potential. Rapidly degrading, high thiol-containing NPs allowed for dramatic enhancement of cell longevity that was concurrent with macrophage stimulation after 2 weeks in ex vivo culture. While equivalent NP internalization resulted in suppressed caspase activity across the NP series, macrophage activation was correlated with increasing thiol content, leading to increased lysosomal activity and a robust pro-survival phenotype. Our results provide insight on tuning NP physiochemical properties as design handles for maximizing ex vivo macrophage longevity, which has implications for improving macrophage-based immune assays, biomanufacturing, and cell therapies.
Collapse
Affiliation(s)
- Bader M Jarai
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy St., Newark, DE 19716, USA.
| | - Zachary Stillman
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy St., Newark, DE 19716, USA.
| | - Catherine A Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy St., Newark, DE 19716, USA.
| |
Collapse
|
3
|
Recent advances in nano/microparticle-based oral vaccines. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2021; 51:425-438. [PMID: 34150345 PMCID: PMC8196935 DOI: 10.1007/s40005-021-00537-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/07/2021] [Indexed: 12/19/2022]
Abstract
Background Vaccines are often recognized as one of the most cost-effective public health interventions in controlling infectious diseases. Most pathogens infiltrate the body from mucosal sites, primarily from the oral and pulmonary region and reach the systemic circulation where disease manifestation starts. Traditional needle-based vaccines are usually not capable of inducing immunity at the mucosal sites where pathogen infiltrates start, but induces systemic immunity. In contrast to needle-based vaccines, mucosally administered vaccines induce immunity at both the mucosal sites and systemically. The oral route of immunization is the most convenient way to administer the vaccines. However, due to the complicated and hostile gastrointestinal structure and environment, vaccines need to overcome major hurdles while retaining their stability and immunogenicity. Area covered This review will briefly discuss different barriers to oral vaccine development. It gives a brief overview of different types of nano/microparticle-based oral vaccines and discusses how physicochemical characteristics of the particles influence overall immunity after oral immunization. Expert opinion Formulation strategies using novel lipid and polymer-based nano/microparticle platforms retain stability and antigenicity of vaccines against the harsh gastrointestinal condition. The physicochemical properties of particles can be uniquely tailored to prolong the release of antigens, and attached ligands (M-cells and APC-ligands) can precisely target uptake by immune cells. These represent viable strategies for efficient delivery of oral vaccines.
Collapse
|
4
|
Jarai BM, Stillman Z, Bomb K, Kloxin AM, Fromen CA. Biomaterials-Based Opportunities to Engineer the Pulmonary Host Immune Response in COVID-19. ACS Biomater Sci Eng 2021; 7:1742-1764. [PMID: 33356134 PMCID: PMC7784663 DOI: 10.1021/acsbiomaterials.0c01287] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/25/2020] [Indexed: 02/08/2023]
Abstract
The COVID-19 pandemic caused by the global spread of the SARS-CoV-2 virus has led to a staggering number of deaths worldwide and significantly increased burden on healthcare as nations scramble to find mitigation strategies. While significant progress has been made in COVID-19 diagnostics and therapeutics, effective prevention and treatment options remain scarce. Because of the potential for the SARS-CoV-2 infections to cause systemic inflammation and multiple organ failure, it is imperative for the scientific community to evaluate therapeutic options aimed at modulating the causative host immune responses to prevent subsequent systemic complications. Harnessing decades of expertise in the use of natural and synthetic materials for biomedical applications, the biomaterials community has the potential to play an especially instrumental role in developing new strategies or repurposing existing tools to prevent or treat complications resulting from the COVID-19 pathology. Leveraging microparticle- and nanoparticle-based technology, especially in pulmonary delivery, biomaterials have demonstrated the ability to effectively modulate inflammation and may be well-suited for resolving SARS-CoV-2-induced effects. Here, we provide an overview of the SARS-CoV-2 virus infection and highlight current understanding of the host's pulmonary immune response and its contributions to disease severity and systemic inflammation. Comparing to frontline COVID-19 therapeutic options, we highlight the most significant untapped opportunities in immune engineering of the host response using biomaterials and particle technology, which have the potential to improve outcomes for COVID-19 patients, and identify areas needed for future investigations. We hope that this work will prompt preclinical and clinical investigations of promising biomaterials-based treatments to introduce new options for COVID-19 patients.
Collapse
Affiliation(s)
- Bader M. Jarai
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| | - Zachary Stillman
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| | - Kartik Bomb
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| | - April M. Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716
| | - Catherine A. Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| |
Collapse
|
5
|
Thakur N, Thakur S, Chatterjee S, Das J, Sil PC. Nanoparticles as Smart Carriers for Enhanced Cancer Immunotherapy. Front Chem 2020; 8:597806. [PMID: 33409265 PMCID: PMC7779678 DOI: 10.3389/fchem.2020.597806] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy for the treatment of many forms of cancer by stimulating body's own immune system. This therapy not only eradicates tumor cells by inducing strong anti-tumor immune response but also prevent their recurrence. The clinical cancer immunotherapy faces some insurmountable challenges including high immune-mediated toxicity, lack of effective and targeted delivery of cancer antigens to immune cells and off-target side effects. However, nanotechnology offers some solutions to overcome those limitations, and thus can potentiate the efficacy of immunotherapy. This review focuses on the advancement of nanoparticle-mediated delivery of immunostimulating agents for efficient cancer immunotherapy. Here we have outlined the use of the immunostimulatory nanoparticles as a smart carrier for effective delivery of cancer antigens and adjuvants, type of interactions between nanoparticles and the antigen/adjuvant as well as the factors controlling the interaction between nanoparticles and the receptors on antigen presenting cells. Besides, the role of nanoparticles in targeting/activating immune cells and modulating the immunosuppressive tumor microenvironment has also been discussed extensively. Finally, we have summarized some theranostic applications of the immunomodulatory nanomaterials in treating cancers based on the earlier published reports.
Collapse
Affiliation(s)
- Neelam Thakur
- Himalayan Centre for Excellence in Nanotechnology, Shoolini University, Solan, India
- School of Advanced Chemical Sciences, Faculty of Basic Sciences, Shoolini University, Solan, India
| | - Saloni Thakur
- Himalayan Centre for Excellence in Nanotechnology, Shoolini University, Solan, India
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, India
| | | | - Joydeep Das
- Himalayan Centre for Excellence in Nanotechnology, Shoolini University, Solan, India
- School of Advanced Chemical Sciences, Faculty of Basic Sciences, Shoolini University, Solan, India
| | - Parames C. Sil
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
6
|
Nasirmoghadas P, Mousakhani A, Behzad F, Beheshtkhoo N, Hassanzadeh A, Nikoo M, Mehrabi M, Kouhbanani MAJ. Nanoparticles in cancer immunotherapies: An innovative strategy. Biotechnol Prog 2020; 37:e3070. [PMID: 32829506 DOI: 10.1002/btpr.3070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/14/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022]
Abstract
Cancer has been one of the most significant causes of mortality, worldwide. Cancer immunotherapy has recently emerged as a competent, cancer-fighting clinical strategy. Nevertheless, due to the difficulty of such treatments, costs, and off-target adverse effects, the implementation of cancer immunotherapy described by the antigen-presenting cell (APC) vaccine and chimeric antigen receptor T cell therapy ex vivo in large clinical trials have been limited. Nowadays, the nanoparticles theranostic system as a promising target-based modality provides new opportunities to improve cancer immunotherapy difficulties and reduce their adverse effects. Meanwhile, the appropriate engineering of nanoparticles taking into consideration nanoparticle characteristics, such as, size, shape, and surface features, as well as the use of these physicochemical properties for suitable biological interactions, provides new possibilities for the application of nanoparticles in cancer immunotherapy. In this review article, we focus on the latest state-of-the-art nanoparticle-based antigen/adjuvant delivery vehicle strategies to professional APCs and engineering specific T lymphocyte required for improving the efficiency of tumor-specific immunotherapy.
Collapse
Affiliation(s)
- Pourya Nasirmoghadas
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Akbar Mousakhani
- Department of Plant Sciences, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Farahnaz Behzad
- Research Institute for Fundamental Sciences (RIFS), University of Tabriz, Tabriz, Iran
| | - Nasrin Beheshtkhoo
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hassanzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Helal Iran Pharmaceutical and Clinical Complex, Tehran, Iran
| | - Mohsen Mehrabi
- Department of Medical Nanotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Amin Jadidi Kouhbanani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Hashemi V, Farhadi S, Ghasemi Chaleshtari M, Seashore-Ludlow B, Masjedi A, Hojjat-Farsangi M, Namdar A, Ajjoolabady A, Mohammadi H, Ghalamfarsa G, Jadidi-Niaragh F. Nanomedicine for improvement of dendritic cell-based cancer immunotherapy. Int Immunopharmacol 2020; 83:106446. [PMID: 32244048 DOI: 10.1016/j.intimp.2020.106446] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 02/08/2023]
Abstract
Dendritic cell (DC)-based cancer immunotherapy has shown impressive outcomes, including the development of the first FDA-approved anti-cancer vaccine. However, the clinical application of DC-based cancer immunotherapy is associated with various challenges. Promising novel tools for the administration of cancer vaccines has emerged from recent developments in nanoscale biomaterials. One current strategy to enhance targeted drug delivery, while minimizing drug-related toxicities, is the use of nanoparticles (NPs). These can be utilized for antigen delivery into DCs, which have been shown to provide potent T cell-stimulating effects. Therefore, NP delivery represents one promising approach for creating an effective and stable immune response without toxic side effects. The current review surveys cancer immunotherapy with particular attention toward NP-based delivery methods that target DCs.
Collapse
Affiliation(s)
- Vida Hashemi
- Department of Basic Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shohreh Farhadi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Brinton Seashore-Ludlow
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Ali Masjedi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; The Persian Gulf Marine Biotechnology Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Afshin Namdar
- Department of Oncology, Cross Cancer Institute, The University of Alberta, Edmonton, Alberta, Canada
| | - Amir Ajjoolabady
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Bhargava A, Srivastava RK, Mishra DK, Tiwari RR, Sharma RS, Mishra PK. Dendritic cell engineering for selective targeting of female reproductive tract cancers. Indian J Med Res 2019; 148:S50-S63. [PMID: 30964081 PMCID: PMC6469378 DOI: 10.4103/ijmr.ijmr_224_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Female reproductive tract cancers (FRCs) are considered as one of the most frequently occurring malignancies and a foremost cause of death among women. The late-stage diagnosis and limited clinical effectiveness of currently available mainstay therapies, primarily due to the developed drug resistance properties of tumour cells, further increase disease severity. In the past decade, dendritic cell (DC)-based immunotherapy has shown remarkable success and appeared as a feasible therapeutic alternative to treat several malignancies, including FRCs. Importantly, the clinical efficacy of this therapy is shown to be restricted by the established immunosuppressive tumour microenvironment. However, combining nanoengineered approaches can significantly assist DCs to overcome this tumour-induced immune tolerance. The prolonged release of nanoencapsulated tumour antigens helps improve the ability of DC-based therapeutics to selectively target and remove residual tumour cells. Incorporation of surface ligands and co-adjuvants may further aid DC targeting (in vivo) to overcome the issues associated with the short DC lifespan, immunosuppression and imprecise uptake. We herein briefly discuss the necessity and progress of DC-based therapeutics in FRCs. The review also sheds lights on the future challenges to design and develop clinically effective nanoparticles-DC combinations that can induce efficient anti-tumour immune responses and prolong patients’ survival.
Collapse
Affiliation(s)
- Arpit Bhargava
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | | | - Dinesh Kumar Mishra
- School of Pharmacy & Technology Management, Narsee Monjee Institute of Management & Studies, Shirpur, India
| | - Rajnarayan R Tiwari
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Radhey Shyam Sharma
- Division of Reproductive Biology, Maternal & Child Health, Indian Council of Medical Research, New Delhi, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| |
Collapse
|
9
|
Qi X, Lu Q, Hu J, Xiong S. Spontaneous C-cleavage of a truncated intein as fusion tag to produce tag-free VP1 inclusion body nanoparticle vaccine against CVB3-induced viral myocarditis by the oral route. Microb Cell Fact 2019; 18:66. [PMID: 30947747 PMCID: PMC6449988 DOI: 10.1186/s12934-019-1115-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/28/2019] [Indexed: 12/14/2022] Open
Abstract
Background Oral vaccine is highly desired for infectious disease which is caused by pathogens infection through the mucosal surface. The design of suitable vaccine delivery system is ongoing for the antigen protection from the harsh gastric environment and target to the Peyer’s patches to induce sufficient mucosal immune responses. Among various potential delivery systems, bacterial inclusion bodies have been widely used as delivery systems in the field of nanobiomedicine. However, a large number of heterologous complex proteins could be difficult to propagate in E. coli and fusion partners are often used to enhance target protein expression. As a safety concern the fusion protein need to be removed from the target protein to get tag-free protein, especially for the production of protein antigen in vaccinology. Until now, there is no report on how to remove fusion tag from inclusion body particles in vitro and in vivo. Coxsackievirus B3 (CVB3) is a leading causative agent of viral myocarditis and orally protein vaccine is high desired for CVB3-induced myocarditis. In this context, we explored a tag-free VP1 inclusion body nanoparticles production protocol though a truncated Ssp DnaX mini-intein spontaneous C-cleavage in vivo and also exploited the VP1 inclusion bodies as an oral protein nanoparticle vaccine to protect mice against CVB3-induced myocarditis. Results We successfully produced the tag-free VP1 inclusion body nanoparticle antigen of CVB3 and orally administrated to mice. The results showed that the tag-free VP1 inclusion body nanoparticles as an effective antigen delivery system targeting to the Peyer’s patches had the capacity to induce mucosal immunity as well as to efficiently protect mice from CVB3 induce myocarditis without any adjuvant. Then, we proposed the use of VP1 inclusion body nanoparticles as good candidate for oral vaccine to against CVB3-induced myocarditis. Conclusions Our tag-free inclusion body nanoparticles production procedure is easy and low cost and may have universal applicability to produce a variety of tag-free inclusion body nanoparticles for oral vaccine. Electronic supplementary material The online version of this article (10.1186/s12934-019-1115-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xingmei Qi
- The Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Qian Lu
- The Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - JingPing Hu
- The Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Sidong Xiong
- The Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
10
|
Liu X, Liu J, Liu D, Han Y, Xu H, Liu L, Leng X, Kong D. A cell-penetrating peptide-assisted nanovaccine promotes antigen cross-presentation and anti-tumor immune response. Biomater Sci 2019; 7:5516-5527. [DOI: 10.1039/c9bm01183h] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Exogenous antigens processed in the cytosol and subsequently cross-presented on major histocompatibility complex class I (MHC-I) molecules activate cytotoxic CD8+ lymphocytes (CTL), which are crucial in cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaoxuan Liu
- Tianjin Key Laboratory of Biomaterials
- Institute of Biomedical Engineering
- Peking Union Medical College & Chinese Academy of Medical Sciences
- Tianjin 300192
- China
| | - Jiale Liu
- Tianjin Key Laboratory of Biomaterials
- Institute of Biomedical Engineering
- Peking Union Medical College & Chinese Academy of Medical Sciences
- Tianjin 300192
- China
| | - Dan Liu
- Tianjin Key Laboratory of Biomaterials
- Institute of Biomedical Engineering
- Peking Union Medical College & Chinese Academy of Medical Sciences
- Tianjin 300192
- China
| | - Yanfeng Han
- School of Biomedical Sciences
- University of Queensland
- St Lucia QLD 4072
- Australia
| | - Haiyan Xu
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences
- School of Basic Medicine Peking Union Medical College
- Beijing100730
- China
| | - Lanxia Liu
- Tianjin Key Laboratory of Biomaterials
- Institute of Biomedical Engineering
- Peking Union Medical College & Chinese Academy of Medical Sciences
- Tianjin 300192
- China
| | - Xigang Leng
- Tianjin Key Laboratory of Biomaterials
- Institute of Biomedical Engineering
- Peking Union Medical College & Chinese Academy of Medical Sciences
- Tianjin 300192
- China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology
- Key Laboratory of Bioactive Materials of Ministry of Education
- College of Life Science
- Nankai University
- Tianjin 300071
| |
Collapse
|
11
|
AlMatar M, Makky EA, AlMandeal H, Eker E, Kayar B, Var I, Köksal F. Does the Development of Vaccines Advance Solutions for Tuberculosis? Curr Mol Pharmacol 2018; 12:83-104. [PMID: 30474542 DOI: 10.2174/1874467212666181126151948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/06/2018] [Accepted: 10/17/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Mycobacterium tuberculosis (Mtb) is considered as one of the most efficacious human pathogens. The global mortality rate of TB stands at approximately 2 million, while about 8 to 10 million active new cases are documented yearly. It is, therefore, a priority to develop vaccines that will prevent active TB. The vaccines currently used for the management of TB can only proffer a certain level of protection against meningitis, TB, and other forms of disseminated TB in children; however, their effectiveness against pulmonary TB varies and cannot provide life-long protective immunity. Based on these reasons, more efforts are channeled towards the development of new TB vaccines. During the development of TB vaccines, a major challenge has always been the lack of diversity in both the antigens contained in TB vaccines and the immune responses of the TB sufferers. Current efforts are channeled on widening both the range of antigens selection and the range of immune response elicited by the vaccines. The past two decades witnessed a significant progress in the development of TB vaccines; some of the discovered TB vaccines have recently even completed the third phase (phase III) of a clinical trial. OBJECTIVE The objectives of this article are to discuss the recent progress in the development of new vaccines against TB; to provide an insight on the mechanism of vaccine-mediated specific immune response stimulation, and to debate on the interaction between vaccines and global interventions to end TB.
Collapse
Affiliation(s)
- Manaf AlMatar
- Department of Biotechnology, Institute of Natural and Applied Sciences (Fen Bilimleri Enstitusu) Cukurova University, Adana, Turkey
| | - Essam A Makky
- Department of Biotechnology, Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang (UMP), Kuantan, Malaysia
| | - Husam AlMandeal
- Freiburg Universität, Moltkestraße 90, 76133 karlsruhe Augenklinik, Germany
| | - Emel Eker
- Department of Medical Microbiology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Begüm Kayar
- Department of Medical Microbiology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Işıl Var
- Department of Food Engineering, Agricultural Faculty, Cukurova University, Adana, Turkey
| | - Fatih Köksal
- Department of Medical Microbiology, Faculty of Medicine, Cukurova University, Adana, Turkey
| |
Collapse
|
12
|
Mohamed SH, Arafa AS, Mady WH, Fahmy HA, Omer LM, Morsi RE. Preparation and immunological evaluation of inactivated avian influenza virus vaccine encapsulated in chitosan nanoparticles. Biologicals 2018; 51:46-53. [DOI: 10.1016/j.biologicals.2017.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 10/18/2022] Open
|
13
|
Afroz S, Medhi H, Maity S, Minhas G, Battu S, Giddaluru J, Kumar K, Paik P, Khan N. Mesoporous ZnO nanocapsules for the induction of enhanced antigen-specific immunological responses. NANOSCALE 2017; 9:14641-14653. [PMID: 28936523 DOI: 10.1039/c7nr03697c] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The application of nanotechnology in vaccinology has fuelled rapid advancement towards the design and development of nanovaccines. Nanoparticles have been found to enhance vaccine efficacy through the spatiotemporal orchestration of antigen delivery to secondary lymphoid organs and antigen-presentation by Antigen Presenting Cells (APCs) synchronized with stimulation of innate and adaptive immune responses. Metal based nanoparticles (MNPs) have been extensively engineered for the generation of nanovaccines owing to their intrinsic adjuvant-like properties and immunomodulatory functions. Furthermore, mesoporous nanocapsules of late have attracted researchers due to their precise size and exclusive capacity to encapsulate a wide range of biomolecules and their sustained release at the targeted sites. Herein, we have designed a novel mesoporous ZnO nanocapsule (mZnO) having a size of ∼12 nm with an average pore diameter of 2.5 nm, using a surfactant-free sonochemical method and investigated its immunomodulatory properties by using Ova loaded mZnO nanocapsules [mZnO(Ova)] in a mice model. Our findings show that mZnO(Ova) administration steered the enhanced expansion of antigen-specific T-cells and induction of IFN-γ producing effector CD4+ and CD8+ T-cells. Also, antigen-specific IgG levels were enriched in both the serum and lymph nodes of mZnO(Ova) immunized mice. Further, we noticed a substantial increase in serum IgG2a or IgG2b levels and IFN-γ secretion in Ova restimulated splenocytes from mZnO(Ova) immunized mice, indicating that mZnO(Ova) skew Th1 type immune response. Overall, the uniqueness of mZnO nanocapsules in terms of the defined particle to pore numbers ratio (maximum of three cavities per particle) allows loading antigens efficiently. Given these features in combination with its immunomodulatory characteristics reinforces the idea that mZnO could be used as an effective antigen-adjuvant platform for the development of novel nano-based vaccines against multiple diseases.
Collapse
Affiliation(s)
- Sumbul Afroz
- School of Life Sciences, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad-500046, Telangana, India.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Grimaldi AM, Incoronato M, Salvatore M, Soricelli A. Nanoparticle-based strategies for cancer immunotherapy and immunodiagnostics. Nanomedicine (Lond) 2017; 12:2349-2365. [PMID: 28868980 DOI: 10.2217/nnm-2017-0208] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although recent successes in clinical trials are strengthening research focused on cancer immunology, the poor immunogenicity and off-target side effects of immunotherapeutics remain major challenges in translating these promising approaches to clinically feasible therapies in the treatment of a large range of tumors. Nanotechnology offers target-based approaches, which have shown significant improvements in the rapidly advancing field of cancer immunotherapy. Here, we first discuss the chemical and physical features of nanoparticulate systems that can be tuned to address the anticancer immune response, and then review recent, key examples of the exploited strategies, ranging from nanovaccines to NPs revising the tumor immunosuppressive microenvironment, up to immunotherapeutic multimodal NPs. Finally, the paper concludes by identifying the promising and outstanding challenges the field of emerging nanotechnologies is facing for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Andrea Soricelli
- IRCCS SDN, Via Gianturco 113, 80143, Naples, Italy.,Department of Motor Sciences & Healthiness, University of Naples Parthenope, via Medina 40, 80133, Naples, Italy
| |
Collapse
|
15
|
Ghinnagow R, Cruz LJ, Macho-Fernandez E, Faveeuw C, Trottein F. Enhancement of Adjuvant Functions of Natural Killer T Cells Using Nanovector Delivery Systems: Application in Anticancer Immune Therapy. Front Immunol 2017; 8:879. [PMID: 28798749 PMCID: PMC5529346 DOI: 10.3389/fimmu.2017.00879] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/10/2017] [Indexed: 11/13/2022] Open
Abstract
Type I natural killer T (NKT) cells have gained considerable interest in anticancer immune therapy over the last decade. This “innate-like” T lymphocyte subset has the unique ability to recognize foreign and self-derived glycolipid antigens in association with the CD1d molecule expressed by antigen-presenting cells. An important property of these cells is to bridge innate and acquired immune responses. The adjuvant function of NKT cells might be exploited in the clinics. In this review, we discuss the approaches currently being used to target NKT cells for cancer therapy. In particular, we highlight ongoing strategies utilizing NKT cell-based nanovaccines to optimize immune therapy.
Collapse
Affiliation(s)
- Reem Ghinnagow
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - Luis Javier Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Elodie Macho-Fernandez
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - Christelle Faveeuw
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - François Trottein
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| |
Collapse
|
16
|
Abstract
AbstractCancer immunotherapy based on tumor vaccine is very promising and intriguing for carcinoma treatment. Herein, antitumor nanovaccines consisting of self-assembled chitosan (CS) nanoparticles and two-component mucin1 (MUC1) glycopeptide antigens were reported. Two different kinds of polyanionic electrolyte [sodium tripolyphosphate (TPP) and γ-poly-L-glutamic acid (γ-PGA)] were combined with chitosan polymers to fabricate the diameter of nearly 400–500 nm CS nanoparticles by electrostatic interactions. The nanovaccines were constructed by physically mixing MUC1 glycopeptide antigens with CS nanoparticles, which reduced vaccine constructing complexity compared with traditional chemical total synthetic vaccines. Immunological studies revealed that the CS/γ-PGA nanoparticle could dramatically enhance the immunogenicity of peptide epitope and produce significantly high titers of IgG antibody which was even better than Freund’s adjuvant-containing vaccines.
Collapse
|
17
|
Yang Z, Xu M, Jia Z, Zhang Y, Wang L, Zhang H, Wang J, Song M, Zhao Y, Wu Z, Zhao L, Yin Z, Hong Z. A novel antigen delivery system induces strong humoral and CTL immune responses. Biomaterials 2017; 134:51-63. [DOI: 10.1016/j.biomaterials.2017.04.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/17/2017] [Accepted: 04/19/2017] [Indexed: 12/17/2022]
|
18
|
Noble J, Zimmerman A, Fromen CA. Potent Immune Stimulation from Nanoparticle Carriers Relies on the Interplay of Adjuvant Surface Density and Adjuvant Mass Distribution. ACS Biomater Sci Eng 2017; 3:560-571. [DOI: 10.1021/acsbiomaterials.6b00756] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jeffery Noble
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Anthony Zimmerman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Catherine A. Fromen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
19
|
Goodwin ZI, Pascual DW. Brucellosis vaccines for livestock. Vet Immunol Immunopathol 2016; 181:51-58. [DOI: 10.1016/j.vetimm.2016.03.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 03/11/2016] [Accepted: 03/14/2016] [Indexed: 01/18/2023]
|
20
|
Nair M, Jayant RD, Kaushik A, Sagar V. Getting into the brain: Potential of nanotechnology in the management of NeuroAIDS. Adv Drug Deliv Rev 2016; 103:202-217. [PMID: 26944096 PMCID: PMC4935582 DOI: 10.1016/j.addr.2016.02.008] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 12/18/2022]
Abstract
In spite of significant advances in antiretroviral (ARV) therapy, the elimination of human immunodeficiency virus (HIV) reservoirs from the periphery and the central nervous system (CNS) remains a formidable task. The incapability of ARV to go across the blood-brain barrier (BBB) after systemic administration makes the brain one of the dominant HIV reservoirs. Thus, screening, monitoring, and elimination of HIV reservoirs from the brain remain a clinically daunting and key task. The practice and investigation of nanomedicine possesses potentials for therapeutics against neuroAIDS. This review highlights the advancements in nanoscience and nanotechnology to design and develop specific size therapeutic cargo for efficient navigation across BBB so as to recognize and eradicate HIV brain reservoirs. Different navigation and drug release strategies, their biocompatibility and efficacy with related challenges and future prospects are also discussed. This review would be an excellent platform to understand nano-enable multidisciplinary research to formulate efficient nanomedicine for the management of neuroAIDS.
Collapse
Key Words
- Anti-retroviral (ARV) therapy
- Blood–brain barrier (BBB)
- Bradykinin (PubChem CID: 439,201)
- CNS drug delivery
- Enfuvirtide (PubChem CID: 16,130,199), Lamivudine & Zidovudine (PubChem CID: 160,352)
- Ferrous oxide or iron (II) oxide (PubChem CID: 14,945)
- Foscarnet sodium (PubChem CID: 44,561)
- HIV monitoring
- HIV-1
- Magnetic nanoparticle
- Mannitol (PubChem CID: 6251)
- Nanotechnology
- Neopterin (PubChem CID: 4455)
- NeuroAIDS
- Pluronic-P85 (PubChem CID: 24,751)
- Saquinavir mesylate (PubChem CID: 60,934)
- Tenofovir disoproxil fumarate (PubChem CID: 6,398,764)
Collapse
Affiliation(s)
- Madhavan Nair
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Rahul Dev Jayant
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Ajeet Kaushik
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Vidya Sagar
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
21
|
Development of nanostructures in the diagnosis of drug hypersensitivity reactions. Curr Opin Allergy Clin Immunol 2016; 16:300-7. [DOI: 10.1097/aci.0000000000000282] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
22
|
Torres-Sangiao E, Holban AM, Gestal MC. Advanced Nanobiomaterials: Vaccines, Diagnosis and Treatment of Infectious Diseases. Molecules 2016; 21:molecules21070867. [PMID: 27376260 PMCID: PMC6273484 DOI: 10.3390/molecules21070867] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/21/2016] [Accepted: 06/25/2016] [Indexed: 11/16/2022] Open
Abstract
The use of nanoparticles has contributed to many advances due to their important properties such as, size, shape or biocompatibility. The use of nanotechnology in medicine has great potential, especially in medical microbiology. Promising data show the possibility of shaping immune responses and fighting severe infections using synthetic materials. Different studies have suggested that the addition of synthetic nanoparticles in vaccines and immunotherapy will have a great impact on public health. On the other hand, antibiotic resistance is one of the major concerns worldwide; a recent report of the World Health Organization (WHO) states that antibiotic resistance could cause 300 million deaths by 2050. Nanomedicine offers an innovative tool for combating the high rates of resistance that we are fighting nowadays, by the development of both alternative therapeutic and prophylaxis approaches and also novel diagnosis methods. Early detection of infectious diseases is the key to a successful treatment and the new developed applications based on nanotechnology offer an increased sensibility and efficiency of the diagnosis. The aim of this review is to reveal and discuss the main advances made on the science of nanomaterials for the prevention, diagnosis and treatment of infectious diseases. Highlighting innovative approaches utilized to: (i) increasing the efficiency of vaccines; (ii) obtaining shuttle systems that require lower antibiotic concentrations; (iii) developing coating devices that inhibit microbial colonization and biofilm formation.
Collapse
Affiliation(s)
- Eva Torres-Sangiao
- Department of Microbiology and Parasitology, University Santiago de Compostela, Galicia 15782, Spain.
| | - Alina Maria Holban
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest 060101, Romania.
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Bucharest 060042, Romania.
| | - Monica Cartelle Gestal
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens (UGA), GA 30602, USA.
| |
Collapse
|
23
|
Jones DS, Rowe CG, Chen B, Reiter K, Rausch KM, Narum DL, Wu Y, Duffy PE. A Method for Producing Protein Nanoparticles with Applications in Vaccines. PLoS One 2016; 11:e0138761. [PMID: 26950441 PMCID: PMC4780713 DOI: 10.1371/journal.pone.0138761] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 09/03/2015] [Indexed: 11/18/2022] Open
Abstract
A practical method is described for synthesizing conjugated protein nanoparticles using thioether (thiol-maleimide) cross-linking chemistry. This method fills the need for a reliable and reproducible synthesis of protein conjugate vaccines for preclinical studies, which can be adapted to produce comparable material for clinical studies. The described method appears to be generally applicable to the production of nanoparticles from a variety of soluble proteins having different structural features. Examples presented include single-component particles of the malarial antigens AMA1, CSP and Pfs25, and two component particles comprised of those antigens covalently cross-linked with the immunogenic carrier protein EPA (a detoxified form of exotoxin A from Pseudomonas aeruginosa). The average molar masses (Mw) of particles in the different preparations ranged from 487 kDa to 3,420 kDa, with hydrodynamic radii (Rh) ranging from 12.1 nm to 38.3 nm. The antigenic properties and secondary structures of the proteins within the particles appear to be largely intact, with no significant changes seen in their far UV circular dichroism spectra, or in their ability to bind conformation-dependent monoclonal antibodies. Mice vaccinated with mixed particles of Pfs25 or CSP and EPA generated significantly greater antigen-specific antibody levels compared with mice vaccinated with the respective unmodified monomeric antigens, validating the potential of antigen-EPA nanoparticles as vaccines.
Collapse
Affiliation(s)
- David S. Jones
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
- * E-mail:
| | - Christopher G. Rowe
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - Beth Chen
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - Karine Reiter
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - Kelly M. Rausch
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - David L. Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - Yimin Wu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| |
Collapse
|
24
|
Abstract
During the last decades significant progress has been made in the field of cancer immunotherapy. However, cancer vaccines have not been successful in clinical trials due to poor immunogenicity of antigen, limitations of safety associated with traditional systemic delivery as well as the complex regulation of the immune system in tumor microenvironment. In recent years, nanotechnology-based delivery systems have attracted great interest in the field of immunotherapy since they provide new opportunities to fight the cancer. In particular, for delivery of cancer vaccines, multifunctional nanoparticles present many advantages such as targeted delivery to immune cells, co-delivery of therapeutic agents, reduced adverse outcomes, blocked immune checkpoint molecules, and amplify immune activation via the use of stimuli-responsive or immunostimulatory materials. In this review article, we highlight recent progress and future promise of multifunctional nanoparticles that have been applied to enhance the efficiency of cancer vaccines.
Collapse
Affiliation(s)
- Tayebeh Saleh
- a Department of Nanobiotechnology , Faculty of Biological Sciences, Tarbiat Modares University , Tehran , Iran
| | - Seyed Abbas Shojaosadati
- b Biotechnology Group, Faculty of Chemical Engineering, Tarbiat Modares University , Tehran , Iran
| |
Collapse
|
25
|
Harde H, Siddhapura K, Agrawal AK, Jain S. Development of dual toxoid-loaded layersomes for complete immunostimulatory response following peroral administration. Nanomedicine (Lond) 2016; 10:1077-91. [PMID: 25929566 DOI: 10.2217/nnm.14.177] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
AIM Present study reports the development of divalent vaccine with enhanced protection, permeation and presentation following peroral immunization. MATERIALS & METHODS Layersomes were prepared by layer-by-layer tuning of polyelectrolytes on liposomes template. The developed system was evaluated for in vitro stability of antigen and layersomes, cell-based assays and immunization experiments in mice. RESULTS Layersomes exhibited enhanced stability in simulated biological fluids, still preserving the integrity, biological activity and conformational stability of toxoids. Layersomes also exhibited complete and protective (>0.1 IU/ml) immunostimulatory response include serum IgG titer, mucosal sIgA titer and cytokines (IL-2 and IFN-γ) levels following peroral administration. CONCLUSION The positive findings of proposed strategy are expected to contribute significantly in the field of stable liposomes technology and peroral immunization.
Collapse
Affiliation(s)
- Harshad Harde
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, SAS Nagar, Punjab 160 062, India
| | | | | | | |
Collapse
|
26
|
Silva AL, Soema PC, Slütter B, Ossendorp F, Jiskoot W. PLGA particulate delivery systems for subunit vaccines: Linking particle properties to immunogenicity. Hum Vaccin Immunother 2016; 12:1056-69. [PMID: 26752261 PMCID: PMC4962933 DOI: 10.1080/21645515.2015.1117714] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Among the emerging subunit vaccines are recombinant protein- and synthetic peptide-based vaccine formulations. However, proteins and peptides have a low intrinsic immunogenicity. A common strategy to overcome this is to co-deliver (an) antigen(s) with (an) immune modulator(s) by co-encapsulating them in a particulate delivery system, such as poly(lactic-co-glycolic acid) (PLGA) particles. Particulate PLGA formulations offer many advantages for antigen delivery as they are biocompatible and biodegradable; can protect the antigens from degradation and clearance; allow for co-encapsulation of antigens and immune modulators; can be targeted to antigen presenting cells; and their particulate nature can increase uptake and cross-presentation by mimicking the size and shape of an invading pathogen. In this review we discuss the pros and cons of using PLGA particulate formulations for subunit vaccine delivery and provide an overview of formulation parameters that influence their adjuvanticity and the ensuing immune response.
Collapse
Affiliation(s)
- A L Silva
- a Division of Drug Delivery Technology , Leiden Academic Center for Drug Research, Leiden University , Leiden , The Netherlands
| | - P C Soema
- b Intravacc (Institute for Translational Vaccinology) , Bilthoven , The Netherlands
| | - B Slütter
- a Division of Drug Delivery Technology , Leiden Academic Center for Drug Research, Leiden University , Leiden , The Netherlands.,c Cluster BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University , Leiden , The Netherlands
| | - F Ossendorp
- d Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , Leiden , The Netherlands
| | - W Jiskoot
- a Division of Drug Delivery Technology , Leiden Academic Center for Drug Research, Leiden University , Leiden , The Netherlands
| |
Collapse
|
27
|
Gupta A, Bahal R, Gupta M, Glazer PM, Saltzman WM. Nanotechnology for delivery of peptide nucleic acids (PNAs). J Control Release 2016; 240:302-311. [PMID: 26776051 DOI: 10.1016/j.jconrel.2016.01.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 12/10/2015] [Accepted: 01/04/2016] [Indexed: 12/22/2022]
Abstract
Over the past three decades, peptide nucleic acids have been employed in numerous chemical and biological applications. Peptide nucleic acids possess enormous potential because of their superior biophysical properties, compared to other oligonucleotide chemistries. However, for therapeutic applications, intracellular delivery of peptide nucleic acids remains a challenge. In this review, we summarize the progress that has been made in delivering peptide nucleic acids to intracellular targets. In addition, we emphasize recent nanoparticle-based strategies for efficient delivery of conventional and chemically-modified peptides nucleic acids.
Collapse
Affiliation(s)
- Anisha Gupta
- Department of Therapeutic Radiology, Yale University, New Haven, CT, USA
| | - Raman Bahal
- Department of Therapeutic Radiology, Yale University, New Haven, CT, USA
| | - Meera Gupta
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Department of Chemical Engineering, Indian Institute of Technology-Delhi, New Delhi, India
| | - Peter M Glazer
- Department of Therapeutic Radiology, Yale University, New Haven, CT, USA; Department of Genetics, Yale University, New Haven, CT, USA.
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
28
|
Vacas-Córdoba E, Climent N, De La Mata FJ, Plana M, Gómez R, Pion M, García F, Muñoz-Fernández MÁ. Dendrimers as nonviral vectors in dendritic cell-based immunotherapies against human immunodeficiency virus: steps toward their clinical evaluation. Nanomedicine (Lond) 2015; 9:2683-702. [PMID: 25529571 DOI: 10.2217/nnm.14.172] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Although the antiretroviral therapy has led to a long-term control of HIV-1, it does not cure the disease. Therefore, several strategies are being explored to develop an effective HIV vaccine, such as the use of dendritic cells (DCs). DC-based immunotherapies bear different limitations, but one of the most critical point is the antigen loading into DCs. Nanotechnology offers new tools to overcome these constraints. Dendrimers have been proposed as carriers for targeted delivery of HIV antigens in DCs. These nanosystems can release the antigens in a controlled manner leading to a more potent specific immune response. This review focuses on the first steps for clinical development of dendrimers to assess their safety and potential use in DC-based immunotherapies against HIV.
Collapse
Affiliation(s)
- Enrique Vacas-Córdoba
- Laboratorio InmunoBiología Molecular, Sección Inmunologia, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria del Gregorio Marañón, C/Dr. Esquerdo 46, 28007, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Seth A, Oh DB, Lim YT. Nanomaterials for enhanced immunity as an innovative paradigm in nanomedicine. Nanomedicine (Lond) 2015; 10:959-75. [PMID: 25867860 DOI: 10.2217/nnm.14.200] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Since the advent of nanoparticle technology, novel and versatile properties of nanomaterials have been introduced, which has constantly expanded their applications in therapeutics. Introduction of nanomaterials for immunomodulation has opened up new avenues with tremendous potential. Interesting properties of nanoparticles, such as adjuvanticity, capability to enhance cross-presentation, polyvalent presentation, siRNA delivery for silencing of immunesuppressive gene, targeting and imaging of immune cells have been known to have immense utility in vaccination and immunotherapy. A thorough understanding of the merits associated with nanomaterials is crucial for designing of modular and versatile nanovaccines, for improved immune response. With the emerging prerequisites of vaccination, nanomaterial-based immune stimulation, seems to be capable of taking the field of immunization to a next higher level.
Collapse
Affiliation(s)
- Anushree Seth
- Graduate School of Analytical Science & Technology, Chungnam National University, Daejeon 305-764, South Korea
| | | | | |
Collapse
|
30
|
Epsilon-caprolactone modified polyethylenimine for highly efficient antigen delivery and chemical exchange saturation transfer functional MR imaging. Biomaterials 2015; 56:219-28. [DOI: 10.1016/j.biomaterials.2015.03.049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/25/2015] [Accepted: 03/27/2015] [Indexed: 11/21/2022]
|
31
|
Pretreatment with Huperzine A-Loaded Poly(lactide-co-glycolide) Nanoparticles Protects against Lethal Effects of Soman-Induced in Mice. ACTA ACUST UNITED AC 2015. [DOI: 10.4028/www.scientific.net/kem.645-646.1374] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Huperzine A (HupA), an alkaloid isolated from theChinese club moss, is a reversible inhibitor of cholinesterases which cross the blood-brain barrier and show high specificity for acetylcholinesterase (AChE). However, HupA induces unwanted side effects in an effective dose against nerve agent poisoning. In the present study, HupA–loaded poly (lactide-co-glycolide) nanoparticles (HupA-PLGA-NP) were prepared using the O/W emulsion solvent evaporation method. The results of SEM demonstrated that HupA-PLGA-NP had an spherical shape and a smooth surface without pores. It’s mean diameter and PDI were 208.5±3.6nm and 0.09±0.01 respectively. The Zeta potential was-35.3±1.8mV and the drug loading was 2.86±0.6%.In vitrodrug release studies showed that HupA-PLGA-NP had a sustained-release behavior in phosphate buffer solution, The accumulated amount of HupA was about 72.1% at 48h with a low burst release within 30min. The LD50values of HupA and HupA-PLGA-NP were 1.40 and 4.85mg/kg respectively, showing that the toxicity of HupA was reduced by 3.5 times. We evaluated the protective efficacy for different doses of HupA or HupA-PLGA-NP against 1.0×LD95(143.0μg/kg) soman toxicity. The results confirmed that HupA (0.3~0.5mg/kg) or HupA-PLGA-NP (0.5~1.5mg/kg) could ensure animals survive. However, about 10% of the animals injected with HupA (0.8mg/kg) died, while no animals died when injected with HupA-PLGA-NP (1.5mg/kg). Aim to 100% survival rate, the effective protective time (12h) of HupA-PLGA-NP (0.5mg/kg,iv) against 1.0×LD95soman toxicity in mice was significantly prolonged compared with that of HupA (4h). The study of AChE activity showed that whole-blood and supernatant of brain diluted by 80-fold and 10-fold respectively were optimum in this study. AChE inhibition after administration of HupA and HupA-PLGA-NP (0.5mg/kg,iv) was recorded and analyzed, The peak values of AChE inhibition in whole-blood and brain by HupA-PLGA-NP (17.6% and 21.8%) were lower than those by HupA (33.7% and 31.9%) and AChE inhibition time by HupA-PLGA-NP was longer than that by HupA. These data confirmed that HupA-PLGA-NP had less toxic and more longer time than HupA against 1.0×LD95soman poisoning and warrant further development as a potent medical countermeasure against chemical warfare nerve agents (CWNAs) poisoning.
Collapse
|
32
|
Adak AK, Li BY, Lin CC. Advances in multifunctional glycosylated nanomaterials: preparation and applications in glycoscience. Carbohydr Res 2015; 405:2-12. [DOI: 10.1016/j.carres.2014.07.026] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 01/13/2023]
|
33
|
Zhang H, Jiang Y, Zhao SG, Jiang LQ, Meng Y, Liu P, Kim MO, Li S. Selective neuronal targeting, protection and signaling network analysis via dopamine-mediated mesoporous silica nanoparticles. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00038f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Mesoporous silica nanoparticle (MSN)-mediated glutathione (GSH) delivery for targeted protection of dopaminergic neuronal cells.
Collapse
Affiliation(s)
- Hailong Zhang
- Laboratory of Clinical Chemistry & Drug Innovation
- The Second Hospital of Shandong University
- Jinan
- PR China
| | - Yuhua Jiang
- Cancer Centre
- The Second Hospital of Shandong University
- Jinan
- PR China
| | - Sheng-gang Zhao
- Department of Cardiology
- The Second Hospital of Jiaxing
- Jiaxing
- PR China
| | - Li-qin Jiang
- Department of Cardiology
- The Second Hospital of Jiaxing
- Jiaxing
- PR China
| | - Yan Meng
- Department of Urology
- The Second Hospital of Shandong University
- Jinan
- PR China
| | - Peng Liu
- Department of Chemistry
- University of Toronto
- Toronto
- Canada
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 plus)
- College of Natural Sciences
- Gyeongsang National University
- Jinju
- Republic of Korea
| | - Shupeng Li
- Laboratory of Clinical Chemistry & Drug Innovation
- The Second Hospital of Shandong University
- Jinan
- PR China
- Cancer Centre
| |
Collapse
|
34
|
Saluja SS, Hanlon DJ, Sharp FA, Hong E, Khalil D, Robinson E, Tigelaar R, Fahmy TM, Edelson RL. Targeting human dendritic cells via DEC-205 using PLGA nanoparticles leads to enhanced cross-presentation of a melanoma-associated antigen. Int J Nanomedicine 2014; 9:5231-46. [PMID: 25419128 PMCID: PMC4235494 DOI: 10.2147/ijn.s66639] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Targeting antigen to dendritic cells (DCs) is a powerful and novel strategy for vaccination. Priming or loading DCs with antigen controls whether subsequent immunity will develop and hence whether effective vaccination can be achieved. The goal of our present work was to increase the potency of DC-based antitumor vaccines by overcoming inherent limitations associated with antigen stability and cross-presentation. Nanoparticles prepared from the biodegradable polymer poly(lactic-co-glycolic acid) have been extensively used in clinical settings for drug delivery and are currently the subject of intensive investigation as antigen delivery vehicles for vaccine applications. Here we describe a nanoparticulate delivery system with the ability to simultaneously carry a high density of protein-based antigen while displaying a DC targeting ligand on its surface. Utilizing a targeting motif specific for the DC-associated surface ligand DEC-205, we show that targeted nanoparticles encapsulating a MART-127–35 peptide are both internalized and cross-presented with significantly higher efficiency than isotype control-coated nanoparticles in human cells. In addition, the DEC-205-labeled nanoparticles rapidly escape from the DC endosomal compartment and do not colocalize with markers of early (EEA-1) or late endosome/lysosome (LAMP-1). This indicates that encapsulated antigens delivered by nanoparticles may have direct access to the class I cytoplasmic major histocompatibility complex loading machinery, overcoming the need for “classical” cross-presentation and facilitating heightened DC stimulation of anti-tumor CD8+ T-cells. These results indicate that this delivery system provides a flexible and versatile methodology to deliver melanoma-associated antigen to DCs, with both high efficiency and heightened potency.
Collapse
Affiliation(s)
- Sandeep S Saluja
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Douglas J Hanlon
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Fiona A Sharp
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Enping Hong
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - David Khalil
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Eve Robinson
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Robert Tigelaar
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Tarek M Fahmy
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA ; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Richard L Edelson
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
35
|
Sánchez-Rodríguez J, Vacas-Córdoba E, Gómez R, De La Mata FJ, Muñoz-Fernández MÁ. Nanotech-derived topical microbicides for HIV prevention: the road to clinical development. Antiviral Res 2014; 113:33-48. [PMID: 25446339 DOI: 10.1016/j.antiviral.2014.10.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/20/2014] [Accepted: 10/29/2014] [Indexed: 11/15/2022]
Abstract
More than three decades since its discovery, HIV infection remains one of the most aggressive epidemics worldwide, with more than 35 million people infected. In sub-Saharan Africa, heterosexual transmissions represent nearly 80% of new infections, with 50% of these occurring in women. In an effort to stop the dramatic spread of the HIV epidemic, new preventive treatments, such as microbicides, have been developed. Nanotechnology has revolutionized this field by designing and engineering novel highly effective nano-sized materials as microbicide candidates. This review illustrates the most recent advances in nanotech-derived HIV prevention strategies, as well as the main steps required to translate promising in vitro results into clinical trials.
Collapse
Affiliation(s)
- Javier Sánchez-Rodríguez
- Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Enrique Vacas-Córdoba
- Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Rafael Gómez
- Dendrimers for Biomedical Applications Group (BioInDen), University of Alcalá, Alcalá de Henares, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - F Javier De La Mata
- Dendrimers for Biomedical Applications Group (BioInDen), University of Alcalá, Alcalá de Henares, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Ma Ángeles Muñoz-Fernández
- Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
36
|
Harde H, Agrawal AK, Jain S. Development of stabilized glucomannosylated chitosan nanoparticles using tandem crosslinking method for oral vaccine delivery. Nanomedicine (Lond) 2014; 9:2511-29. [DOI: 10.2217/nnm.13.225] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aim: The aim of this study was to develop a novel platform technology, comprising of stable glucomannosylated chitosan nanoparticles, for oral immunization. Materials & methods: Chitosan nanoparticles were stabilized by tandem crosslinking using tripolyphosphate followed by glutaraldehyde. Process and formulation variables were optimized using a ‘Box–Behnken’ design. The in vitro and in vivo performances were established in RAW 264.7 and BALB/c mice, respectively. Results: The lyophilized formulation was exceptionally stable in simulated biological media and the enclosed antigen was conformationally stable. The mechanistic understanding of glucomannosylated chitosan nanoparticles in RAW 264.7 revealed transcellular uptake via both mannose and glucose transporter-mediated endocytosis. Glucomannan modification resulted in significantly higher systemic (serum IgG titer), mucosal (secretory IgA) and cell-mediated (IL-2 and IFN-γ) immune responses in comparison with nonmodified chitosan nanoparticles. Conclusion: The present strategy is expected to contribute some novel tools for the oral delivery of numerous biomacromolecules. Original submitted 8 August 2013; Revised submitted 15 December 2013
Collapse
Affiliation(s)
- Harshad Harde
- Center for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, SAS Nagar, Punjab, 160062, India
| | - Ashish Kumar Agrawal
- Center for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, SAS Nagar, Punjab, 160062, India
| | - Sanyog Jain
- Center for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, SAS Nagar, Punjab, 160062, India
| |
Collapse
|
37
|
Sehgal K, Ragheb R, Fahmy TM, Dhodapkar MV, Dhodapkar KM. Nanoparticle-mediated combinatorial targeting of multiple human dendritic cell (DC) subsets leads to enhanced T cell activation via IL-15-dependent DC crosstalk. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:2297-305. [PMID: 25080481 PMCID: PMC6195217 DOI: 10.4049/jimmunol.1400489] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Most vaccines depend on coadministration of Ags and adjuvants that activate APCs. Nanoparticles (NPs) have emerged as an attractive vehicle for synchronized delivery of Ags and adjuvants to APCs and can be targeted to specific cell types, such as dendritic cells (DCs), which are potent APCs. Which subset of human DCs should be targeted for optimal activation of T cell immunity, however, remains unknown. In this article, we describe a poly-lactic-coglycolic acid-based NP platform, wherein avidin-decorated NPs can be targeted to multiple human DC subsets via biotinylated Abs. Both BDCA3(+) and monocyte-derived DC-SIGN(+) NP-loaded DCs were equally effective at generating Ag-specific human T cells in culture, including against complex peptide mixtures from viral and tumor Ags across multiple MHC molecules. Ab-mediated targeting of NPs to distinct DC subsets led to enhanced T cell immunity. However, combination targeting to both DC-SIGN and BDCA3(+) DCs led to significantly greater activation of T cells compared with targeting either DC subset alone. Enhanced T cell activation following combination targeting depended on DC-mediated cytokine release and was IL-15 dependent. These data demonstrate that simultaneous targeting of multiple DC subsets may improve NP vaccines by engaging DC crosstalk and provides a novel approach to improving vaccines against pathogens and tumors.
Collapse
Affiliation(s)
- Kartik Sehgal
- Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, CT 06510; Department of Medicine, Yale School of Medicine, Yale University, New Haven, CT 06510
| | - Ragy Ragheb
- Department of Biomedical Engineering, Yale School of Engineering, Yale University, New Haven, CT 06510
| | - Tarek M Fahmy
- Department of Biomedical Engineering, Yale School of Engineering, Yale University, New Haven, CT 06510; Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT 06510; and
| | - Madhav V Dhodapkar
- Department of Medicine, Yale School of Medicine, Yale University, New Haven, CT 06510; Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT 06510; and Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT 06510
| | - Kavita M Dhodapkar
- Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, CT 06510; Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT 06510
| |
Collapse
|
38
|
Deering RP, Kommareddy S, Ulmer JB, Brito LA, Geall AJ. Nucleic acid vaccines: prospects for non-viral delivery of mRNA vaccines. Expert Opin Drug Deliv 2014; 11:885-99. [PMID: 24665982 DOI: 10.1517/17425247.2014.901308] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Nucleic acid-based vaccines are being developed as a means to combine the positive attributes of both live-attenuated and subunit vaccines. Viral vectors and plasmid DNA vaccines have been extensively evaluated in human clinical trials and have been shown to be safe and immunogenic, although none have been licensed for human use. More recently, mRNA-based vaccine alternatives have emerged and might offer certain advantages over their DNA-based counterparts. AREAS COVERED This review describes the two main categories of mRNA vaccines: conventional non-amplifying and self-amplifying mRNA. It summarizes the initial clinical proof-of-concept studies and outlines the preclinical testing of the next wave of innovations for the technology. Finally, this review highlights the versatile functionality of the mRNA molecule and introduces opportunities for future improvements in vaccine design. EXPERT OPINION The prospects for mRNA vaccines are very promising. Like other types of nucleic acid vaccines, mRNA vaccines have the potential to combine the positive attributes of live attenuated vaccines while obviating many potential safety limitations. Although data from initial clinical trials appear encouraging, mRNA vaccines are far from a commercial product. These initial approaches have spurred innovations in vector design, non-viral delivery, large-scale production and purification of mRNA to quickly move the technology forward. Some improvements have already been tested in preclinical models for both prophylactic and therapeutic vaccine targets and have demonstrated their ability to elicit potent and broad immune responses, including functional antibodies, type 1 T helper cells-type T cell responses and cytotoxic T cells. Though the initial barriers for this nucleic acid vaccine approach seem to be overcome, in our opinion, the future and continued success of this approach lies in a more extensive evaluation of the many non-viral delivery systems described in the literature and gaining a better understanding of the mechanism of action to allow rational design of next generation technologies.
Collapse
Affiliation(s)
- Raquel P Deering
- Novartis Vaccines, Inc. , 350 Massachusetts Ave, Cambridge, MA 02139 , USA +1 617 871 3745 ;
| | | | | | | | | |
Collapse
|
39
|
Lefebvre DE, Pearce B, Fine JH, Chomyshyn E, Ross N, Halappanavar S, Tayabali AF, Curran I, Bondy GS. In vitro enhancement of mouse T helper 2 cell sensitization to ovalbumin allergen by carbon black nanoparticles. Toxicol Sci 2014; 138:322-32. [PMID: 24449417 DOI: 10.1093/toxsci/kfu010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Agglomerated carbon black nanoparticles (CBNPs) administered via respiratory or subcutaneous routes have been shown to promote allergic sensitization to coadministered ovalbumin (OVA) protein in rodents. In the present study, we aimed to model and elucidate the mechanism of this adjuvanticity using an in vitro assay based on T cell sensitization to ovalbumin₃₂₃₋₃₃₉ peptide (OVA(p)). CBNP base particles of 22 and 39 nm were characterized and termed CBNP22 and CBNP39 powders. Splenic leukocytes derived from transgenic DO11.10 mice were exposed to suspensions of media alone, concanavalin A mitogen, CBNP agglomerates smaller than 220 nm, OVA(p) alone, OVA(p) + anti-CD28 costimulant, OVA(p) + cyclosporin A immunosuppressant, or OVA(p) + CBNPs. Samples were analyzed at 72 h post-exposure. Proliferation rate, a marker of cellular mitosis, was assessed. Polymerase chain reaction arrays were used to assess genes involved in allergic response pathways. The mitogen control, costimulatory control, and immunosuppressive control chemicals modified the T helper cell proliferation rate. CBNP22 mildly reduced proliferation at 12 μg/ml, but CBNP39 did not. Gene expression analysis of cells treated with OVA(p) showed that coincubation with 12 μg/ml CBNP22 enhanced gene expression of interleukin-4 (IL-4), IL-10, and IL-13, all allergy-associated Th2 cytokines. Coincubation of OVA(p) with 12 μg/ml CBNP39 significantly enhanced IL-13 gene expression concurrent with downregulation of the Th1-associated transcription factor Stat4. IL-4 and IL-13 protein secretion reflected the mRNA trends. The changes were consistently higher in cells exposed to CBNP22 than CBNP39, suggesting that smaller particle size, higher surface area, and higher purity were associated with the direct adjuvant effect on Th2 cells in this genetically susceptible model of OVA allergy.
Collapse
Affiliation(s)
- David E Lefebvre
- Bureau of Chemical Safety, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Cherif MS, Shuaibu MN, Kodama Y, Kurosaki T, Helegbe GK, Kikuchi M, Ichinose A, Yanagi T, Sasaki H, Yui K, Tien NH, Karbwang J, Hirayama K. Nanoparticle formulation enhanced protective immunity provoked by PYGPI8p-transamidase related protein (PyTAM) DNA vaccine in Plasmodium yoelii malaria model. Vaccine 2014; 32:1998-2006. [PMID: 24440206 DOI: 10.1016/j.vaccine.2014.01.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 11/23/2013] [Accepted: 01/02/2014] [Indexed: 11/15/2022]
Abstract
We have previously reported the new formulation of polyethylimine (PEI) with gamma polyglutamic acid (γ-PGA) nanoparticle (NP) to have provided Plasmodium yoelii merozoite surface protein-1 (PyMSP-1) plasmid DNA vaccine with enhanced protective cellular and humoral immunity in the lethal mouse malaria model. PyGPI8p-transamidase-related protein (PyTAM) was selected as a possible candidate vaccine antigen by using DNA vaccination screening from 29 GPI anchor and signal sequence motif positive genes picked up using web-based bioinformatics tools; though the observed protection was not complete. Here, we observed augmented protective effect of PyTAM DNA vaccine by using PEI and γ-PGA complex as delivery system. NP-coated PyTAM plasmid DNA immunized mice showed a significant survival rate from lethal P. yoelii challenge infection compared with naked PyTAM plasmid or with NP-coated empty plasmid DNA group. Antigen-specific IgG1 and IgG2b subclass antibody levels, proportion of CD4 and CD8T cells producing IFN-γ in the splenocytes and IL-4, IFN-γ, IL-12 and TNF-α levels in the sera and in the supernatants from ex vivo splenocytes culture were all enhanced by the NP-coated PyTAM DNA vaccine. These data indicates that NP augments PyTAM protective immune response, and this enhancement was associated with increased DC activation and concomitant IL-12 production.
Collapse
Affiliation(s)
- Mahamoud Sama Cherif
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan; Global COE Program, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan; Institut National de Santé Publique, Université de Conakry, Guinea
| | - Mohammed Nasir Shuaibu
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan; Global COE Program, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | | | | | - Gideon Kofi Helegbe
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Mihoko Kikuchi
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Akitoyo Ichinose
- Electron Microscopy Central Laboratory, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Tetsuo Yanagi
- Animal Research Center for Tropical Medicine, Nagasaki, Japan
| | - Hitoshi Sasaki
- Department of Hospital Pharmacy, Nagasaki University, Japan; Global COE Program, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Katsuyuki Yui
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Nguyen Huy Tien
- Department of Product Development, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Juntra Karbwang
- Department of Product Development, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan; Global COE Program, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.
| |
Collapse
|
41
|
Rancan F, Blume-Peytavi U, Vogt A. Utilization of biodegradable polymeric materials as delivery agents in dermatology. Clin Cosmet Investig Dermatol 2014; 7:23-34. [PMID: 24470766 PMCID: PMC3891488 DOI: 10.2147/ccid.s39559] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Biodegradable polymeric materials are ideal carrier systems for biomedical applications. Features like controlled and sustained delivery, improved drug pharmacokinetics, reduced side effects and safe degradation make the use of these materials very attractive in a lot of medical fields, with dermatology included. A number of studies have shown that particle-based formulations can improve the skin penetration of topically applied drugs. However, for a successful translation of these promising results into a clinical application, a more rational approach is needed to take into account the different properties of diseased skin and the fate of these polymeric materials after topical application. In fact, each pathological skin condition poses different challenges and the way diseased skin interacts with polymeric carriers might be markedly different to that of healthy skin. In most inflammatory skin conditions, the skin's barrier is impaired and the local immune system is activated. A better understanding of such mechanisms has the potential to improve the efficacy of carrier-based dermatotherapy. Such knowledge would allow the informed choice of the type of polymeric carrier depending on the skin condition to be treated, the type of drug to be loaded, and the desired release kinetics. Furthermore, a better control of polymer degradation and release properties in accordance with the skin environment would improve the safety and the selectivity of drug release. This review aims at summarizing the current knowledge on how polymeric delivery systems interact with healthy and diseased skin, giving an overview of the challenges that different pathological skin conditions pose to the development of safer and more specific dermatotherapies.
Collapse
Affiliation(s)
- Fiorenza Rancan
- Clinical Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Germany
| | - Ulrike Blume-Peytavi
- Clinical Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Germany
| | - Annika Vogt
- Clinical Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Germany
| |
Collapse
|
42
|
Sylvester A, Sivaraman B, Deb P, Ramamurthi A. Nanoparticles for localized delivery of hyaluronan oligomers towards regenerative repair of elastic matrix. Acta Biomater 2013; 9:9292-302. [PMID: 23917150 DOI: 10.1016/j.actbio.2013.07.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/17/2013] [Accepted: 07/24/2013] [Indexed: 10/26/2022]
Abstract
Abdominal aortic aneurysms (AAAs) are rupture-prone progressive dilations of the infrarenal aorta due to a loss of elastic matrix that lead to weakening of the aortic wall. Therapies to coax biomimetic regenerative repair of the elastic matrix by resident, diseased vascular cells may thus be useful to slow, arrest or regress AAA growth. Hyaluronan oligomers (HA-o) have been shown to induce elastic matrix synthesis by healthy and aneurysmal rat aortic smooth muscle cells (SMCs) in vitro but only via exogenous dosing, which potentially has side-effects and limitations to in vivo delivery towards therapy. In this paper, we describe the development of HA-o loaded poly(lactide-co-glycolide) nanoparticles (NPs) for targeted, controlled and sustained delivery of HA-o towards the elastogenic induction of aneurysmal rat aortic SMCs. These NPs were able to deliver HA-o over an extended period (>30 days) at previously determined elastogenic doses (0.2-20 μg ml(-1)). HA-o released from the NPs led to dose-dependent increases in elastic matrix synthesis, and the recruitment and activity of lysyl oxidase, the enzyme which cross-links elastin precursor molecules into mature fibers/matrix. Therefore, we were able to successfully develop a nanoparticle-based system for controlled and sustained HA-o delivery for the in vitro elastogenic induction of aneurysmal rat aortic smooth muscle cells.
Collapse
|
43
|
Ungaro F, Conte C, Quaglia F, Tornesello ML, Buonaguro FM, Buonaguro L. VLPs and particle strategies for cancer vaccines. Expert Rev Vaccines 2013; 12:1173-1193. [PMID: 24124878 DOI: 10.1586/14760584.2013.836909] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Effective delivery of tumor antigens to APCs is one of the key steps for eliciting a strong and durable immune response to tumors. Several cancer vaccines have been evaluated in clinical trials, based on soluble peptides, but results have not been fully satisfactory. To improve immunogenicity particles provide a valid strategy to display and/or incorporate epitopes which can be efficiently targeted to APCs for effective induction of adaptive immunity. In the present review, we report some leading technologies for developing particulate vaccines employed in cancer immunotherapy, highlighting the key parameters for a rational design to elicit both humoral and cellular responses.
Collapse
Affiliation(s)
- Francesca Ungaro
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131, Napoli, Italy
| | | | | | | | | | | |
Collapse
|
44
|
Geall AJ, Mandl CW, Ulmer JB. RNA: the new revolution in nucleic acid vaccines. Semin Immunol 2013; 25:152-9. [PMID: 23735226 DOI: 10.1016/j.smim.2013.05.001] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 04/29/2013] [Accepted: 05/03/2013] [Indexed: 01/02/2023]
Abstract
Nucleic acid vaccines have the potential to address issues of safety and effectiveness sometimes associated with vaccines based on live attenuated viruses and recombinant viral vectors. In addition, methods to manufacture nucleic acid vaccines are suitable as generic platforms and for rapid response, both of which will be very important for addressing newly emerging pathogens in a timely fashion. Plasmid DNA is the more widely studied form of nucleic acid vaccine and proof of principle in humans has been demonstrated, although no licensed human products have yet emerged. The RNA vaccine approach, based on mRNA and engineered RNA replicons derived from certain RNA viruses, is gaining increased attention and several vaccines are under investigation for infectious diseases, cancer and allergy. Human clinical trials are underway and the prospects for success are bright.
Collapse
Affiliation(s)
- Andrew J Geall
- Novartis Vaccines & Diagnostics, Cambridge, MA 02139, USA.
| | | | | |
Collapse
|
45
|
Knuschke T, Sokolova V, Rotan O, Wadwa M, Tenbusch M, Hansen W, Staeheli P, Epple M, Buer J, Westendorf AM. Immunization with Biodegradable Nanoparticles Efficiently Induces Cellular Immunity and Protects against Influenza Virus Infection. THE JOURNAL OF IMMUNOLOGY 2013; 190:6221-9. [DOI: 10.4049/jimmunol.1202654] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
46
|
Silva A, Rosalia R, Sazak A, Carstens M, Ossendorp F, Oostendorp J, Jiskoot W. Optimization of encapsulation of a synthetic long peptide in PLGA nanoparticles: Low-burst release is crucial for efficient CD8+ T cell activation. Eur J Pharm Biopharm 2013. [DOI: 10.1016/j.ejpb.2012.11.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
47
|
Handké N, Ficheux D, Rollet M, Delair T, Mabrouk K, Bertin D, Gigmes D, Verrier B, Trimaille T. Lysine-tagged peptide coupling onto polylactide nanoparticles coated with activated ester-based amphiphilic copolymer: A route to highly peptide-functionalized biodegradable carriers. Colloids Surf B Biointerfaces 2013; 103:298-303. [DOI: 10.1016/j.colsurfb.2012.10.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 10/19/2012] [Accepted: 10/22/2012] [Indexed: 11/25/2022]
|
48
|
García-Arévalo C, Bermejo-Martín JF, Rico L, Iglesias V, Martín L, Rodríguez-Cabello JC, Arias FJ. Immunomodulatory Nanoparticles from Elastin-Like Recombinamers: Single-Molecules for Tuberculosis Vaccine Development. Mol Pharm 2013; 10:586-97. [DOI: 10.1021/mp300325v] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Carmen García-Arévalo
- Bioforge Group, University of Valladolid, CIBER-BBN, Paseo de Belén
11, 47011 Valladolid, Spain
| | - Jesús F. Bermejo-Martín
- Infection and Immunity Medical Research Unit (IMI), Microbiology
Department, Hospital Clínico Universitario-IECSCYL, Ramón y Cajal 3, 47005 Valladolid, Spain
| | - Lucia Rico
- Infection and Immunity Medical Research Unit (IMI), Microbiology
Department, Hospital Clínico Universitario-IECSCYL, Ramón y Cajal 3, 47005 Valladolid, Spain
| | - Verónica Iglesias
- Infection and Immunity Medical Research Unit (IMI), Microbiology
Department, Hospital Clínico Universitario-IECSCYL, Ramón y Cajal 3, 47005 Valladolid, Spain
| | - Laura Martín
- Bioforge Group, University of Valladolid, CIBER-BBN, Paseo de Belén
11, 47011 Valladolid, Spain
| | | | - F. Javier Arias
- Bioforge Group, University of Valladolid, CIBER-BBN, Paseo de Belén
11, 47011 Valladolid, Spain
| |
Collapse
|
49
|
Meraz IM, Segura-Ibarra V, Leonard F, Gonzalez J, Ally S, Godin B, Serda RE. Biological Microniches Characterizing Pathological Lesions. Nanomedicine (Lond) 2013. [DOI: 10.1016/b978-0-08-098338-7.00006-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
50
|
Bhargava A, Mishra D, Banerjee S, Mishra PK. Engineered dendritic cells for gastrointestinal tumor immunotherapy: opportunities in translational research. J Drug Target 2012; 21:126-36. [PMID: 23061479 DOI: 10.3109/1061186x.2012.731069] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|