1
|
Heidarnejad F, Namvar A, Sadat SM, Pordanjani PM, Rezaei F, Namdari H, Arjmand S, Bolhassani A. In silico designing of novel epitope-based peptide vaccines against HIV-1. Biotechnol Lett 2024; 46:315-354. [PMID: 38403788 DOI: 10.1007/s10529-023-03464-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 11/16/2023] [Accepted: 12/21/2023] [Indexed: 02/27/2024]
Abstract
The HIV-1 virus has been regarded as a catastrophe for human well-being. The global incidence of HIV-1-infected individuals is increasing. Hence, development of effective immunostimulatory molecules has recently attracted an increasing attention in the field of vaccine design against HIV-1 infection. In this study, we explored the impacts of CD40L and IFN-γ as immunostimulatory adjuvants for our candidate HIV-1 Nef vaccine in human and mouse using immunoinformatics analyses. Overall, 18 IFN-γ-based vaccine constructs (9 constructs in human and 9 constructs in mouse), and 18 CD40L-based vaccine constructs (9 constructs in human and 9 constructs in mouse) were designed. To find immunogenic epitopes, important characteristics of each component (e.g., MHC-I and MHC-II binding, and peptide-MHC-I/MHC-II molecular docking) were determined. Then, the selected epitopes were applied to create multiepitope constructs. Finally, the physicochemical properties, linear and discontinuous B cell epitopes, and molecular interaction between the 3D structure of each construct and CD40, IFN-γ receptor or toll-like receptors (TLRs) were predicted. Our data showed that the full-length CD40L and IFN-γ linked to the N-terminal region of Nef were capable of inducing more effective immune response than multiepitope vaccine constructs. Moreover, molecular docking of the non-allergenic full-length- and epitope-based CD40L and IFN-γ constructs to their cognate receptors, CD40 and IFN-γ receptors, and TLRs 4 and 5 in mouse were more potent than in human. Generally, these findings suggest that the full forms of these adjuvants could be more efficient for improvement of HIV-1 Nef vaccine candidate compared to the designed multiepitope-based constructs.
Collapse
Affiliation(s)
| | - Ali Namvar
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Seyed Mehdi Sadat
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Rezaei
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Haideh Namdari
- Iranian Tissue Bank Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Arjmand
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
2
|
Sadeghi L, Bolhassani A, Mohit E, Baesi K, Aghasadeghi MR. Heterologous DNA Prime/Protein Boost Immunization Targeting Nef-Tat Fusion Antigen Induces Potent T-cell Activity and in vitro Anti-SCR HIV-1 Effects. Curr HIV Res 2024; 22:109-119. [PMID: 38712371 DOI: 10.2174/011570162x297602240430142231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Heterologous combinations in vaccine design are an effective approach to promote T cell activity and antiviral effects. The goal of this study was to compare the homologous and heterologous regimens targeting the Nef-Tat fusion antigen to develop a human immunodeficiency virus-1 (HIV-1) therapeutic vaccine candidate. METHODS At first, the DNA and protein constructs harboring HIV-1 Nef and the first exon of Tat as linked form (pcDNA-nef-tat and Nef-Tat protein) were prepared in large scale and high purity. The generation of the Nef-Tat protein was performed in the E. coli expression system using an IPTG inducer. Then, we evaluated and compared immune responses of homologous DNA prime/ DNA boost, homologous protein prime/ protein boost, and heterologous DNA prime/protein boost regimens in BALB/c mice. Finally, the ability of mice splenocytes to secret cytokines after exposure to single-cycle replicable (SCR) HIV-1 was compared between immunized and control groups in vitro. RESULTS The nef-tat gene was successfully subcloned in eukaryotic pcDNA3.1 (-) and prokaryotic pET-24a (+) expression vectors. The recombinant Nef-Tat protein was generated in the E. coli Rosetta strain under optimized conditions as a clear band of ~ 35 kDa detected on SDS-PAGE. Moreover, transfection of pcDNA-nef-tat into HEK-293T cells was successfully performed using Lipofectamine 2000, as confirmed by western blotting. The immunization studies showed that heterologous DNA prime/protein boost regimen could significantly elicit the highest levels of Ig- G2a, IFN-γ, and Granzyme B in mice as compared to homologous DNA/DNA and protein/protein regimens. Moreover, the secretion of IFN-γ was higher in DNA/protein regimens than in DNA/DNA and protein/protein regimens after exposure of mice splenocytes to SCR HIV-1 in vitro. CONCLUSION The chimeric HIV-1 Nef-Tat antigen was highly immunogenic, especially when applied in a heterologous prime/ boost regimen. This regimen could direct immune response toward cellular immunity (Th1 and CTL activity) and increase IFN-γ secretion after virus exposure.
Collapse
Affiliation(s)
- Leila Sadeghi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Elham Mohit
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kazem Baesi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
3
|
Van NH, Vy NT, Van Toi V, Dao AH, Lee BJ. Nanostructured lipid carriers and their potential applications for versatile drug delivery via oral administration. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100064] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
4
|
Espinar-Buitrago M, Muñoz-Fernández MA. New Approaches to Dendritic Cell-Based Therapeutic Vaccines Against HIV-1 Infection. Front Immunol 2022; 12:719664. [PMID: 35058917 PMCID: PMC8763680 DOI: 10.3389/fimmu.2021.719664] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
Due to the success of combined antiretroviral therapy (cART) in recent years, the pathological outcome of Human Immunodeficiency Virus type 1 (HIV-1) infection has improved substantially, achieving undetectable viral loads in most cases. Nevertheless, the presence of a viral reservoir formed by latently infected cells results in patients having to maintain treatment for life. In the absence of effective eradication strategies against HIV-1, research efforts are focused on obtaining a cure. One of these approaches is the creation of therapeutic vaccines. In this sense, the most promising one up to now is based on the establishing of the immunological synapse between dendritic cells (DCs) and T lymphocytes (TL). DCs are one of the first cells of the immune system to encounter HIV-1 by acting as antigen presenting cells, bringing about the interaction between innate and adaptive immune responses mediated by TL. Furthermore, TL are the end effector, and their response capacity is essential in the adaptive elimination of cells infected by pathogens. In this review, we summarize the knowledge of the interaction between DCs with TL, as well as the characterization of the specific T-cell response against HIV-1 infection. The use of nanotechnology in the design and improvement of vaccines based on DCs has been researched and presented here with a special emphasis.
Collapse
Affiliation(s)
- Marisierra Espinar-Buitrago
- Section Head Immunology, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Ma Angeles Muñoz-Fernández
- Section Head Immunology, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Spanish Human Immunodeficiency Virus- Hospital Gregorio Marañón (HIV-HGM) BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
5
|
Palameta S, Manrique-Rincón AJ, Toscaro JM, Semionatto IF, Fonseca MC, Rosa RS, Ruas LP, Oliveira PS, Bajgelman MC. Boosting antitumor response with PSMA-targeted immunomodulatory VLPs, harboring costimulatory TNFSF ligands and GM-CSF cytokine. Mol Ther Oncolytics 2022; 24:650-662. [PMID: 35284623 PMCID: PMC8898762 DOI: 10.1016/j.omto.2022.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/11/2022] [Indexed: 11/29/2022] Open
Abstract
Therapeutic strategies based on immunomodulation have improved cancer therapy. Most approaches target co-stimulatory pathways or the inhibition of immunosuppressive mechanisms, to enhance immune response and overcome the immune tolerance of tumors. Here, we propose a novel platform to deliver targeted immunomodulatory signaling, enhancing antitumor response. The platform is based on virus-like particles derived from lentiviral capsids. These particles may be engineered to harbor multifunctional ligands on the surface that drive tropism to the tumor site and deliver immunomodulatory signaling, boosting the antitumor response. We generated virus-like particles harboring a PSMA-ligand, TNFSF co-stimulatory ligands 4-1BBL or OX40L, and a membrane-anchored GM-CSF cytokine. The virus-like particles are driven to PSMA-expressing tumors and deliver immunomodulatory signaling from the TNFSF surface ligands and the anchored GM-CSF, inducing T cell proliferation, inhibition of regulatory T cells, and potentiating elimination of tumor cells. The PSMA-targeted particles harboring immunomodulators enhanced antitumor activity in immunocompetent challenged mice and may be explored as a potential tool for cancer immunotherapy.
Collapse
|
6
|
Kaur A, Kanwar R, Kaushik D, Sakala IG, Honda-Okubo Y, Petrovsky N, Salunke DB, Mehta SK. Combined delivery of TLR2 and TLR7 agonists by Nanostructured lipid carriers induces potent vaccine adjuvant activity in mice. Int J Pharm 2021; 613:121378. [PMID: 34915144 DOI: 10.1016/j.ijpharm.2021.121378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/12/2021] [Accepted: 12/09/2021] [Indexed: 10/19/2022]
Abstract
Toll-like receptor (TLR) agonists are promising adjuvants and the combination of TLR agonists enhance immune responses by providing synergistic immune activity via triggering different signalling pathways. However, systematic cytotoxicity due to the immediate release of such immune potentiators from the site of injection hampers its clinical performance. Nanostructured lipid carriers (NLCs) offer a possibility to incorporate multiple TLR agonists with high encapsulation efficiency and slow drug release. Herein, we synthesized NLCs from didodecyldimethylammonium bromide (D12DAB) and oleic acid and used these to co-encapsulate a Pam2CS derivative (T-2, TLR2 agonist) with an imidazoquinoline derivative (T-7, TLR7 agonist) as a combination vaccine adjuvant. Hydrodynamic diameter and zeta potential of the prepared NLCs were found to be in the range of 200-500 nm and 23-27 mV, respectively. Spherical shape and size of prepared NLCs were also assessed through Field Emission Scanning Electron Microscopy (FE-SEM) and Transmission Electron Microscopy (TEM) analysis. In-vitro release studies of T-7 demonstrated sustained release and the addition of lipopeptide T-2 augmented encapsulation efficiency (from 84 to 92.9%) with a slight trigger in the release percentage. All NLC formulations were screened in TLR2/1, TLR2/6, TLR7 and TLR8 reporter cell lines and loaded NLC formulation showed high TLR2 and TLR7 agonistic activity. Adjuvant potency was evaluated through intramuscular immunization of female C57BL/6 mice with recombinant hepatitis B surface antigen and influenza hemagglutinin protein. T-2 and T-7 loaded NLCs induced good protective efficacy in mice challenged with a lethal dose of influenza virus.
Collapse
Affiliation(s)
- Arshpreet Kaur
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India
| | - Rohini Kanwar
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; Mehr Chand Mahajan DAV College for Women, Chandigarh, India
| | - Deepender Kaushik
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India
| | - Isaac G Sakala
- Vaxine Pty Ltd, Warradale, Australia; College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Yoshikazu Honda-Okubo
- Vaxine Pty Ltd, Warradale, Australia; College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Warradale, Australia; College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Deepak B Salunke
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India.
| | - Surinder K Mehta
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India.
| |
Collapse
|
7
|
Development, formulation, and cellular mechanism of a lipophilic copper chelator for the treatment of Wilson's disease. Int J Pharm 2021; 609:121193. [PMID: 34673167 DOI: 10.1016/j.ijpharm.2021.121193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022]
Abstract
Copper homeostasis is finely regulated in human to avoid any detrimental impact of free intracellular copper ions. Upon copper accumulation, biliary excretion is triggered in liver thanks to trafficking of the ATP7B copper transporter to bile canaliculi. However, in Wilson's disease this protein is mutated leading to copper accumulation. Current therapy uses Cu chelators acting extracellularly and requiring a life-long treatment with side effects. Herein, a new Cu(I) pro-chelator was encapsulated in long-term stable nanostructured lipid carriers. Cellular assays revealed that the pro-chelator protects hepatocytes against Cu-induced cell death. Besides, the cellular stresses induced by moderate copper concentrations, including protein unfolding, are counteracted by the pro-chelator. These data showed the pro-chelator efficiency to deliver intracellularly an active chelator that copes with copper stress and surpasses current and under development chelators. Although its biological activity is more mitigated, the pro-chelator nanolipid formulation led to promising results. This innovative approach is of outmost importance in the quest of better treatments for Wilson's disease.
Collapse
|
8
|
Shah S, Chougule MB, Kotha AK, Kashikar R, Godugu C, Raghuvanshi RS, Singh SB, Srivastava S. Nanomedicine based approaches for combating viral infections. J Control Release 2021; 338:80-104. [PMID: 34375690 PMCID: PMC8526416 DOI: 10.1016/j.jconrel.2021.08.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022]
Abstract
Millions of people die each year from viral infections across the globe. There is an urgent need to overcome the existing gap and pitfalls of the current antiviral therapy which include increased dose and dosing frequency, bioavailability challenges, non-specificity, incidences of resistance and so on. These stumbling blocks could be effectively managed by the advent of nanomedicine. Current review emphasizes over an enhanced understanding of how different lipid, polymer and elemental based nanoformulations could be potentially and precisely used to bridle the said drawbacks in antiviral therapy. The dawn of nanotechnology meeting vaccine delivery, role of RNAi therapeutics in antiviral treatment regimen, various regulatory concerns towards clinical translation of nanomedicine along with current trends and implications including unexplored research avenues for advancing the current drug delivery have been discussed in detail.
Collapse
Affiliation(s)
- Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mahavir Bhupal Chougule
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, MS, USA; Department Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Arun K Kotha
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, MS, USA; Department Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Rama Kashikar
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, MS, USA; Department Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Chandraiah Godugu
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Rajeev Singh Raghuvanshi
- Indian Pharmacopoeia Commission, Ministry of Health & Family Welfare, Government of India, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
9
|
Gao X, Gong J, Cai Y, Wang J, Wen J, Peng L, Ji H, Jiang S, Guo D. Chitosan modified squalene nanostructured lipid carriers as a promising adjuvant for freeze-dried ovalbumin vaccine. Int J Biol Macromol 2021; 188:855-862. [PMID: 34411614 DOI: 10.1016/j.ijbiomac.2021.08.074] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/30/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023]
Abstract
As immune adjuvants assisting vaccines, nanoparticle delivery systems have been widely exploited. Squalene, the major ingredient of approved adjuvant MF59, has great potential in activating immune responses. In the current study, model antigen ovalbumin (OVA) was encapsulated into squalene-based nanostructured lipid carriers (NLCs), and the chitosan, a cationic polysaccharide, was used for modifying nanoparticles to develop a functionalized and cationic nanoparticle delivery system (OVA-csNLCs). Firstly, the optimal formulation of csNLCs was successfully screened out, and had hydrodynamic diameter of 235.80 ± 5.99 nm and zeta potential of 34.90 ± 6.95 mV. Then, the generated OVA-csNLCs had no significant difference in hydrodynamic diameter and exhibited lower zeta potential of 19.03 ± 0.31 mV and high encapsulation efficiency of 83.4%. Sucrose (10%, w/w) was selected as optimal lyoprotectant, exhibiting good stability of OVA-csNLCs in the form of freeze-dried powder. More importantly, the OVA-csNLCs effectively promoted OVA antigen uptake by macrophage, significantly enhanced the level of OVA-specific IgG, and induced a Th2-based immune response in vivo. Furthermore, mice immunization experiment demonstrated that OVA-csNLCs had well biocompatibility and facilitated spleen lymphocytes proliferation. Above findings indicate that chitosan modified squalene nanostructured lipid carriers show promise as antigen delivery system and an open adjuvant platform.
Collapse
Affiliation(s)
- Xiuge Gao
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Jiahao Gong
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Ying Cai
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Jiacai Wang
- Shandong Vocational Animal Science and Veterinary College, 88 Shengli East Street, Weifang 261061, China
| | - Jia Wen
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Lin Peng
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Hui Ji
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Shanxiang Jiang
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Dawei Guo
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China.
| |
Collapse
|
10
|
Dey AK, Nougarède A, Clément F, Fournier C, Jouvin-Marche E, Escudé M, Jary D, Navarro FP, Marche PN. Tuning the Immunostimulation Properties of Cationic Lipid Nanocarriers for Nucleic Acid Delivery. Front Immunol 2021; 12:722411. [PMID: 34497612 PMCID: PMC8419413 DOI: 10.3389/fimmu.2021.722411] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/04/2021] [Indexed: 11/25/2022] Open
Abstract
Nonviral systems, such as lipid nanoparticles, have emerged as reliable methods to enable nucleic acid intracellular delivery. The use of cationic lipids in various formulations of lipid nanoparticles enables the formation of complexes with nucleic acid cargo and facilitates their uptake by target cells. However, due to their small size and highly charged nature, these nanocarrier systems can interact in vivo with antigen-presenting cells (APCs), such as dendritic cells (DCs) and macrophages. As this might prove to be a safety concern for developing therapies based on lipid nanocarriers, we sought to understand how they could affect the physiology of APCs. In the present study, we investigate the cellular and metabolic response of primary macrophages or DCs exposed to the neutral or cationic variant of the same lipid nanoparticle formulation. We demonstrate that macrophages are the cells affected most significantly and that the cationic nanocarrier has a substantial impact on their physiology, depending on the positive surface charge. Our study provides a first model explaining the impact of charged lipid materials on immune cells and demonstrates that the primary adverse effects observed can be prevented by fine-tuning the load of nucleic acid cargo. Finally, we bring rationale to calibrate the nucleic acid load of cationic lipid nanocarriers depending on whether immunostimulation is desirable with the intended therapeutic application, for instance, gene delivery or messenger RNA vaccines.
Collapse
Affiliation(s)
- Arindam K. Dey
- Univ. Grenoble Alpes, St Martin d’Hères, France
- Institute for Advanced Biosciences, Research Center INSERM U1209, CNRS UMR5309, La Tronche, France
| | - Adrien Nougarède
- Univ. Grenoble Alpes, St Martin d’Hères, France
- CEA, LETI, Division for Biology and Healthcare Technologies, Microfluidic Systems and Bioengineering Lab, Grenoble, France
| | - Flora Clément
- Univ. Grenoble Alpes, St Martin d’Hères, France
- Institute for Advanced Biosciences, Research Center INSERM U1209, CNRS UMR5309, La Tronche, France
- Univ. Grenoble Alpes, CEA, INSERM, IRIG, Biomics, Grenoble, France
| | - Carole Fournier
- Univ. Grenoble Alpes, St Martin d’Hères, France
- Institute for Advanced Biosciences, Research Center INSERM U1209, CNRS UMR5309, La Tronche, France
| | - Evelyne Jouvin-Marche
- Univ. Grenoble Alpes, St Martin d’Hères, France
- Institute for Advanced Biosciences, Research Center INSERM U1209, CNRS UMR5309, La Tronche, France
| | - Marie Escudé
- Univ. Grenoble Alpes, St Martin d’Hères, France
- CEA, LETI, Division for Biology and Healthcare Technologies, Microfluidic Systems and Bioengineering Lab, Grenoble, France
| | - Dorothée Jary
- Univ. Grenoble Alpes, St Martin d’Hères, France
- CEA, LETI, Division for Biology and Healthcare Technologies, Microfluidic Systems and Bioengineering Lab, Grenoble, France
| | - Fabrice P. Navarro
- Univ. Grenoble Alpes, St Martin d’Hères, France
- CEA, LETI, Division for Biology and Healthcare Technologies, Microfluidic Systems and Bioengineering Lab, Grenoble, France
| | - Patrice N. Marche
- Univ. Grenoble Alpes, St Martin d’Hères, France
- Institute for Advanced Biosciences, Research Center INSERM U1209, CNRS UMR5309, La Tronche, France
| |
Collapse
|
11
|
Larijani MS, Pouriayevali MH, Sadat SM, Ramezani A. Production of Recombinant HIV-1 p24-Nef Protein in Two Forms as Potential Candidate Vaccines in Three Vehicles. Curr Drug Deliv 2021; 17:387-395. [PMID: 32183667 DOI: 10.2174/1567201817666200317121728] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 01/11/2020] [Accepted: 02/13/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND Different approaches have been investigated to develop a preventive or therapeutic vaccine, although none of them has been fully practical. Therapeutic vaccines against HIV-1 have been studied with the aim of eliminating the virus from reservoir cells with or without HAART (Highly Active Antiretroviral Therapy). Fusion proteins with the most immunogenic features among conserved regions can facilitate this achievement in such a variable virus. To achieve the most immunogenic and also conserved regions, bioinformatics tools are widely used to predict antigens' features before applying them. OBJECTIVE This study aimed at the in vitro evaluation of p24 -Nef fusion protein based on the previous in silico design to achieve a potential therapeutic subunit vaccine against HIV-1. METHODS The truncated form of p24-Nef using AAY flexible linker and the full protein were expressed and evaluated in the prokaryotic system and confirmed by western blotting. We also used pcDNA3.1 to transfect Lenti-X 293T cells. Moreover, lentiviral vectors were applied to produce recombinant virions harboring the genes of interest and cell transduction. RESULTS Both fusion proteins in a truncated and a full form were expressed and confirmed by Anti Nef polyclonal antibody in western blotting. Recombinant virions were generated and transduced Lenti-X 293T cells confirming by immunofluorescence microscope and p24 ELISA assay kit. Transduced cells were analyzed by SDS-PAGE and western blotting, which resulted in approved protein expression. CONCLUSION Fusion protein of p24 and Nef is well expressed in eukaryotic cell lines according to its pre-evaluated features by bioinformatics tools.
Collapse
Affiliation(s)
- Mona Sadat Larijani
- Department of Hepatitis, AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Hassan Pouriayevali
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Ref Lab), Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Mehdi Sadat
- Department of Hepatitis, AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Amitis Ramezani
- Department of Clinical Research, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
12
|
Jazayeri SD, Lim HX, Shameli K, Yeap SK, Poh CL. Nano and Microparticles as Potential Oral Vaccine Carriers and Adjuvants Against Infectious Diseases. Front Pharmacol 2021; 12:682286. [PMID: 34149426 PMCID: PMC8206556 DOI: 10.3389/fphar.2021.682286] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Mucosal surfaces are the first site of infection for most infectious diseases and oral vaccination can provide protection as the first line of defense. Unlike systemic administration, oral immunization can stimulate cellular and humoral immune responses at both systemic and mucosal levels to induce broad-spectrum and long-lasting immunity. Therefore, to design a successful vaccine, it is essential to stimulate the mucosal as well as systemic immune responses. Successful oral vaccines need to overcome the harsh gastrointestinal environment such as the extremely low pH, proteolytic enzymes, bile salts as well as low permeability and the low immunogenicity of vaccines. In recent years, several delivery systems and adjuvants have been developed for improving oral vaccine delivery and immunogenicity. Formulation of vaccines with nanoparticles and microparticles have been shown to improve antigen stability, availability and adjuvanticity as well as immunostimulatory capacity, target delivery and specific release. This review discusses how nanoparticles (NPs) and microparticles (MPs) as oral carriers with adjuvant characteristics can be beneficial in oral vaccine development.
Collapse
Affiliation(s)
| | - Hui Xuan Lim
- Centre for Virus and Vaccine Research, Subang Jaya, Malaysia
| | - Kamyar Shameli
- Malaysia-Japan International Institute of Technology, Universiti Teknologi Malaysia, Kuala Lumpur, Malaysia
| | - Swee Keong Yeap
- Department of Marine Biotechnology, China-Asean College of Marine Sciences, Xiamen University Malaysia, Sepang, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, Subang Jaya, Malaysia
| |
Collapse
|
13
|
Khojasteh NF, Fekri M, Shabani SH, Milani A, Baesi K, Bolhassani A. Evaluation of HIV-1 Regulatory and Structural Proteins as Antigen Candidate in Mice and Humans. Curr HIV Res 2021; 19:225-237. [PMID: 33243125 DOI: 10.2174/1570162x18999201125212131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The diagnosis of HIV infection is important among different groups. Moreover, combination antiretroviral therapy is used to treat HIV-1, but it cannot eradicate the infection. Thus, the development of therapeutic vaccines, along with antiretroviral therapy, is recommended. This study evaluates the values of four HIV proteins as antigen candidates in therapeutic vaccine design as well as a possible diagnostic marker for HIV infection in humans. METHODS In this study, the HIV-1 Tat and Rev regulatory proteins and structural Gp120 and p24 proteins were generated in E. coli expression system. Their immunogenicity was evaluated in BALB/ c mice using homologous and heterologous prime/boost strategies. Moreover, the detection of anti- HIV IgG antibodies against these recombinant proteins was assessed in untreated (Naïve/ HIV-infected), treated, and drug-resistant patients compared to the healthy (control) group as a possible diagnostic marker for HIV infection. RESULTS In humans, our results showed that among HIV-1 proteins, anti-Gp120 antibody was not detected in treated individuals compared to the healthy (control) group. The levels of anti-Gp120 antibody were significantly different between the treated group and Naïve as well as drug-resistant subjects. Moreover, the level of anti-p24 antibody was significantly lower in the treated group than the Naive group. In mice, the results of immunization indicated that the Rev antigen could significantly induce IgG2a, IgG2b, and IFN-γ secretion aimed at Th1 response as well as Granzyme B generation as CTL activity in comparison with other antigens. Furthermore, the heterologous DNA prime/ protein boost regimen was more potent than the homologous regimen for stimulation of cellular immunity. CONCLUSION Briefly, the levels of both anti-Gp120 and anti-p24 antibodies can be considered for the diagnosis of the HIV-infected individuals in different groups compared to the healthy group. Moreover, among four recombinant proteins, Rev elicited Th1 cellular immunity and CTL activity in mice as an antigen candidate in therapeutic vaccine development.
Collapse
Affiliation(s)
| | - Mehrshad Fekri
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Kazem Baesi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
14
|
Duarte JL, Filippo LDD, Araujo VHS, Oliveira AEMDFM, de Araújo JTC, Silva FBDR, Pinto MC, Chorilli M. Nanotechnology as a tool for detection and treatment of arbovirus infections. Acta Trop 2021; 216:105848. [PMID: 33524384 DOI: 10.1016/j.actatropica.2021.105848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/21/2021] [Accepted: 01/23/2021] [Indexed: 12/19/2022]
Abstract
Arboviruses are medically important viruses that cause high rates of infection all over the world. In addition, the severity of the symptoms and the inadequate diagnostic methods represent a challenge far beyond eradicating the vector. The lack of specific treatments for arbovirus infections reflects the imminent need for new research for safe and efficient medicines to treat these infections. Nanotechnology is an innovative approach currently used as a platform for developing new treatments, thus improving the biopharmaceutical properties of drugs. It can also be applied to the development of diagnostic devices, improving their detection capacity. The purpose of this paper is to review recent research on the use of nanotechnology for developing new treatments and detection devices for arbovirus infections. Interestingly, it was found that only a few studies report on the use of nanotechnology to treat arbovirus infections and that most of these reports focus on the fabrication of diagnostic tools. Also, some papers report on the use of nanotechnology for the development of vaccines, which in association with mosquito eradication programs could effectively reduce the high rates of infections by these viruses.
Collapse
Affiliation(s)
- Jonatas Lobato Duarte
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Leonardo Delello Di Filippo
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Victor Hugo Sousa Araujo
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Anna Eliza Maciel de Faria Mota Oliveira
- Federal University of Amapá - UNIFAP, Department of Health and biological sciences, Rodovia Juscelino Kubitschek, Km 02, Jardim Marco Zero, Macapá-AP, 68903-361, Brazil
| | - Jennifer Thayanne Cavalcante de Araújo
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Flávia Benini da Rocha Silva
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Mara Cristina Pinto
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Marlus Chorilli
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil.
| |
Collapse
|
15
|
Sadat Larijani M, Ramezani A, Mashhadi Abolghasem Shirazi M, Bolhassani A, Pouriayevali MH, Shahbazi S, Sadat SM. Evaluation of transduced dendritic cells expressing HIV-1 p24-Nef antigens in HIV-specific cytotoxic T cells induction as a therapeutic candidate vaccine. Virus Res 2021; 298:198403. [PMID: 33775753 DOI: 10.1016/j.virusres.2021.198403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 10/21/2022]
Abstract
Various approaches have been investigated to prevent or eliminate HIV-1 since 1981. However, the virus has been affecting human population worldwide with no effective vaccine yet. The conserved regions among the viral genes are suitable targets in mutable viruses to induce the immune responses via an effective delivery platform. In this study, we aimed at evaluation of p24 and nef in two forms of full and truncated genes as two fusion antigenic forms according to our previous bioinformatics analysis. The designed antigens were then transferred through ex vivo generated dendritic cells and also proteins in BALB/c to assess and compare immunogenicity. p24 and Nef amino acid sequences were aligned, then, the most conserved regions were selected and two fusion forms as the truncated (p24:80-231aa-Nef:120-150aa) and the full from (p24-Nef) were cloned and expressed in prokaryotic and eukaryotic systems. Lentiviral vectors were applied to generate recombinant virions harboring the genes of interest to transduce generated murine dendritic cells. BALB/c mice received the recombinant DCs or recombinant proteins according to the defined schedule. IgG development was assessed to determine humoral immune activity and cellular immune responses were evaluated by IL-5 and IFN-y induction. Granzyme B secretion was also investigated to determine CTL activity in different immunized groups. The data showed high induction of cellular immune responses in dendritic cell immunization specifically in immunized mice with the truncated form of the p24 and Nef by high secretion of IFN-y and strong CTL activity. Moreover, protein/ DC prime-boost formulation led to stronger Th1 pathway and strong CTL activation in comparison with other formulations. The generated recombinant dendritic cells expressing p24-Nef induced humoral and cellular immunity in a Th1 pathway specifically with the in silico predicted truncated antigen which could be of high value as a dendritic cell therapeutic vaccine candidate against HIV-1.
Collapse
Affiliation(s)
- Mona Sadat Larijani
- Clinical Research Department, Pasteur Institute of Iran, Tehran, Iran; Hepatitis, AIDS and Blood borne diseases Department, Pasteur Institute of Iran, Tehran, Iran
| | - Amitis Ramezani
- Clinical Research Department, Pasteur Institute of Iran, Tehran, Iran.
| | | | - Azam Bolhassani
- Hepatitis, AIDS and Blood borne diseases Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Hassan Pouriayevali
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Reference Laboratory), Pasteur Institute of Iran, Tehran, Iran
| | - Sepideh Shahbazi
- Hepatitis, AIDS and Blood borne diseases Department, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Mehdi Sadat
- Hepatitis, AIDS and Blood borne diseases Department, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
16
|
D'haese S, Lacroix C, Garcia F, Plana M, Ruta S, Vanham G, Verrier B, Aerts JL. Off the beaten path: Novel mRNA-nanoformulations for therapeutic vaccination against HIV. J Control Release 2020; 330:1016-1033. [PMID: 33181204 DOI: 10.1016/j.jconrel.2020.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/16/2022]
Abstract
Over the last few years, immunotherapy for HIV in general and therapeutic vaccination in particular, has received a tremendous boost, both in preclinical research and in clinical applications. This interest is based on the evidence that the immune system plays a crucial role in controlling HIV infection, as shown for long-term non-progressors and elite controllers, and that immune responses can be manipulated towards targeting conserved epitopes. So far, the most successful approach has been vaccination with autologous dendritic cells (DCs) loaded ex vivo with antigens and activation signals. Although this approach offers much promise, it also comes with significant drawbacks such as the requirement of a specialized infrastructure and expertise, as well as major challenges for logistics and storage, making it extremely time consuming and costly. Therefore, methods are being developed to avoid the use of ex vivo generated, autologous DCs. One of these methods is based on mRNA for therapeutic vaccination. mRNA has proven to be a very promising vaccine platform, as the coding information for any desired protein, including antigens and activation signals, can be generated in a very short period of time, showing promise both as an off-the-shelf therapy and as a personalized approach. However, an important drawback of this approach is the short half-life of native mRNA, due to the presence of ambient RNases. In addition, proper immunization requires that the antigens are expressed, processed and presented at the right immunological site (e.g. the lymphoid tissues). An ambivalent aspect of mRNA as a vaccine is its capacity to induce type I interferons, which can have beneficial adjuvant effects, but also deleterious effects on mRNA stability and translation. Thus, proper formulation of the mRNA is crucially important. Many approaches for RNA formulation have already been tested, with mixed success. In this review we discuss the state-of-the-art and future trends for mRNA-nanoparticle formulations for HIV vaccination, both in the prophylactic and in the therapeutic setting.
Collapse
Affiliation(s)
- Sigrid D'haese
- Neuro-Aging & Viro-Immunotherapy (NAVI), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Céline Lacroix
- Institute for the Biology and Chemistry of Proteins (IBCP), Lyon, France
| | | | | | - Simona Ruta
- Carol Davila University of Medicine and Pharmacy, Stefan S. Nicolau Institute of Virology, Bucharest, Romania
| | - Guido Vanham
- Institute of Tropical Medicine and University of Antwerp, Antwerp, Belgium
| | - Bernard Verrier
- Institute for the Biology and Chemistry of Proteins (IBCP), Lyon, France
| | - Joeri L Aerts
- Neuro-Aging & Viro-Immunotherapy (NAVI), Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| |
Collapse
|
17
|
Bhardwaj P, Bhatia E, Sharma S, Ahamad N, Banerjee R. Advancements in prophylactic and therapeutic nanovaccines. Acta Biomater 2020; 108:1-21. [PMID: 32268235 PMCID: PMC7163188 DOI: 10.1016/j.actbio.2020.03.020] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
Vaccines activate suitable immune responses to fight against diseases but can possess limitations such as compromised efficacy and immunogenic responses, poor stability, and requirement of adherence to multiple doses. ‘Nanovaccines’ have been explored to elicit a strong immune response with the advantages of nano-sized range, high antigen loading, enhanced immunogenicity, controlled antigen presentation, more retention in lymph nodes and promote patient compliance by a lower frequency of dosing. Various types of nanoparticles with diverse pathogenic or foreign antigens can help to overcome immunotolerance and alleviate the need of booster doses as required with conventional vaccines. Nanovaccines have the potential to induce both cell-mediated and antibody-mediated immunity and can render long-lasting immunogenic memory. With such properties, nanovaccines have shown high potential for the prevention of infectious diseases such as acquired immunodeficiency syndrome (AIDS), malaria, tuberculosis, influenza, and cancer. Their therapeutic potential has also been explored in the treatment of cancer. The various kinds of nanomaterials used for vaccine development and their effects on immune system activation have been discussed with special relevance to their implications in various pathological conditions. Statement of Significance Interaction of nanoparticles with the immune system has opened multiple avenues to combat a variety of infectious and non-infectious pathological conditions. Limitations of conventional vaccines have paved the path for nanomedicine associated benefits with a hope of producing effective nanovaccines. This review highlights the role of different types of nanovaccines and the role of nanoparticles in modulating the immune response of vaccines. The applications of nanovaccines in infectious and non-infectious diseases like malaria, tuberculosis, AIDS, influenza, and cancers have been discussed. It will help the readers develop an understanding of mechanisms of immune activation by nanovaccines and design appropriate strategies for novel nanovaccines.
Collapse
|
18
|
Larijani MS, Ramezani A, Sadat SM. Updated Studies on the Development of HIV Therapeutic Vaccine. Curr HIV Res 2020; 17:75-84. [PMID: 31210114 DOI: 10.2174/1570162x17666190618160608] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND Among the various types of pharmaceuticals, vaccines have a special place. However, in the case of HIV, nearly after 40 years of its discovery, an effective vaccine still is not available. The reason lies in several facts mainly the variability and smartness of HIV as well as the complexity of the interaction between HIV and immune responses. A robust, effective, and longterm immunity is undoubtedly what a successful preventive vaccine should induce in order to prevent the infection of HIV. Failure of human trials to this end has led to the idea of developing therapeutic vaccines with the purpose of curing already infected patients by boosting their immune responses against the virus. Nevertheless, the exceptional ability of the virus to escape the immune system based on the genetically diverse envelope and variable protein products have made it difficult to achieve an efficient therapeutic vaccine. OBJECTIVE We aimed at studying and comparing different approaches to HIV therapeutic vaccines. METHODS In this review, we summarized the human trials undergoing on HIV therapeutic vaccination which are registered in the U.S. clinical trial database (clinicaltrials.gov). These attempts are divided into different tables, according to the type of formulation and application in order to classify and compare their results. RESULT/CONCLUSION Among several methods applied in studied clinical trials which are mainly divided into DNA, Protein, Peptide, Viral vectors, and Dendritic cell-based vaccines, protein vaccine strategy is based on Tat protein-induced anti-Tat Abs in 79% HIV patients. However, the studies need to be continued to achieve a durable efficient immune response against HIV-1.
Collapse
Affiliation(s)
- Mona Sadat Larijani
- Hepatitis, AIDS, and Bloodborne Diseases Department, Pasteur Institute of Iran, Tehran, Iran
| | - Amitis Ramezani
- Hepatitis, AIDS, and Bloodborne Diseases Department, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Mehdi Sadat
- Hepatitis, AIDS, and Bloodborne Diseases Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
19
|
|
20
|
Larijani MS, Sadat SM, Bolhassani A, Pouriayevali MH, Bahramali G, Ramezani A. In Silico Design and Immunologic Evaluation of HIV-1 p24-Nef Fusion Protein to Approach a Therapeutic Vaccine Candidate. Curr HIV Res 2019; 16:322-337. [PMID: 30605062 PMCID: PMC6446525 DOI: 10.2174/1570162x17666190102151717] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 12/04/2018] [Accepted: 12/27/2018] [Indexed: 01/24/2023]
Abstract
Background: Acquired immune deficiency syndrome (HIV/AIDS) has been a major glob-al health concern for over 38 years. No safe and effective preventive or therapeutic vaccine has been developed although many products have been investigated. Computational methods have facilitated vaccine developments in recent decades. Among HIV-1 proteins, p24 and Nef are two suitable targets to provoke the cellular immune response. However, the fusion form of these two proteins has not been analyzed in silico yet. Objective: This study aimed at the evaluation of possible fusion forms of p24 and Nef in order to achieve a potential therapeutic subunit vaccine against HIV-1. Method: In this study, various computational approaches have been applied to predict the most effec-tive fusion form of p24-Nef including CTL (Cytotoxic T lymphocytes) response, immunogenicity, conservation and population coverage. Moreover, binding to MHC (Major histocompatibility com-plex) molecules was assessed in both human and BALB/c. Results: After analyzing six possible fusion protein forms using AAY linker, we came up with the most practical form of p24 from 80 to 231 and Nef from 120 to 150 regions (according to their refer-ence sequence of HXB2 strain) using an AAY linker, based on their peptides affinity to MHC mole-cules which are located in a conserved region among different virus clades. The selected fusion protein contains seventeen MHC I antigenic epitopes, among them KRWIILGLN, YKRWIILGL, DIAG-TTSTL and FPDWQNYTP are fully conserved between the virus clades. Furthermore, analyzed class I CTL epitopes showed greater affinity binding to HLA-B 57*01, HLA-B*51:01 and HLA-B 27*02 molecules. The population coverage with the rate of >70% coverage in the Persian population supports this truncated form as an appropriate candidate against HIV-I virus. Conclusion: The predicted fusion protein, p24-AAY-Nef in a truncated form with a high rate of T cell epitopes and high conservancy rate among different clades, provides a helpful model for developing a therapeutic vaccine candidate against HIV-1.
Collapse
Affiliation(s)
- Mona Sadat Larijani
- Hepatitis, AIDS and Bloodborne Diseases Department, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Mehdi Sadat
- Hepatitis, AIDS and Bloodborne Diseases Department, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Hepatitis, AIDS and Bloodborne Diseases Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Hassan Pouriayevali
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Ref Lab), Pasteur Institute of Iran (IPI) Tehran, Iran
| | - Golnaz Bahramali
- Hepatitis, AIDS and Bloodborne Diseases Department, Pasteur Institute of Iran, Tehran, Iran
| | - Amitis Ramezani
- Hepatitis, AIDS and Bloodborne Diseases Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
21
|
Tohidi F, Sadat SM, Bolhassani A, Yaghobi R, Larijani MS. Induction of a Robust Humoral Response using HIV-1 VLPMPER-V3 as a Novel Candidate Vaccine in BALB/c Mice. Curr HIV Res 2019; 17:33-41. [DOI: 10.2174/1570162x17666190306124218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 01/10/2023]
Abstract
Background:
Several approaches have not been successful to suppress HIV (Human immunodeficiency
virus) infection among infected individuals or to prevent it yet. In order to expand
strong HIV specific humoral and cellular responses, Virus-like particles (VLPs) as potential vaccines
show significant increase in neutralizing antibodies secretion, T-cell count and also secretion
of cytokines.
Objective:
This study aimed at immunological evaluation of VLPs harboring high copy of MPERV3
in BALB/c mice.
Methods:
Female BALB/c mice were immunized with homologous and heterologous primeboosting
regimens of HIV-1 VLPMPER-V3. Their immune responses were evaluated for humoral responses
(Total IgG and IgG isotyping) and cellular responses (IFN-γ, IL-5 secretion, in vitro CTL
assay and T cell proliferation) and compared in immunized mice.
Results:
The data showed robust induction of humoral response in mice groups which received different
regimens of VLP. Furthermore, analysis of cytokine profile indicated that the highest IL-5 secretion
was related to VLP+M50 group and confirmed the dominance of Th2 immunity in this
group.
Conclusion:
This study showed that VLP MPER-V3 as a potential vaccine candidate has the potency as
an effective prophylactic vaccine and this finding guarantees further investigations to achieve a
promising HIV-1 vaccine candidate.
Collapse
Affiliation(s)
- Fatemeh Tohidi
- Department of Hepatitis, AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Mehdi Sadat
- Department of Hepatitis, AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis, AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Ramin Yaghobi
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mona Sadat Larijani
- Department of Hepatitis, AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
22
|
Development and validation of a novel UPLC-ELSD method for the assessment of lipid composition of nanomedicine formulation. Int J Pharm 2019; 566:11-23. [PMID: 31112794 DOI: 10.1016/j.ijpharm.2019.05.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 01/12/2023]
Abstract
Lipid nanocarriers incorporating glycerides, polyethylene glycol (PEG)-stearates and phospholipids have attracted great attention for in vivo diagnostic, in vivo imaging, activated or non-activated targeted drug delivery. For quality control purposes, the development of appropriate methods for the quantification of their lipid components is needed. In the present study, we developed an analytical method for lipid quantification in formulated nanoparticles. PEG-stearates and glycerides were analyzed in a single run by RP-UPLC-ELSD using a two-step gradient elution program, while the analysis of phospholipids was accomplished by HILIC-UPLC-ELSD after isolation using an SPE silica column. Using both isolated compounds and commercial lipid standards, calibration curves were produced using second-order polynomials to attain the quantitative evaluation of each lipid excipient. Relative standard deviation of all analytes was between 0.9% and 5.3% for intra-day precision and recovery ranged from 83.5% to 112.2%. The presented method was successfully implemented to study the manufacturing process and stability of the formulated lipid excipients during long-term storage and accelerated conditions. The formulation lipid yield was determined and found equal to 82.5%.
Collapse
|
23
|
Rai M, Jamil B. Nanoformulations: A Valuable Tool in the Therapy of Viral Diseases Attacking Humans and Animals. Nanotheranostics 2019. [PMCID: PMC7121811 DOI: 10.1007/978-3-030-29768-8_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Various viruses can be considered as one of the most frequent causes of human diseases, from mild illnesses to really serious sicknesses that end fatally. Numerous viruses are also pathogenic to animals and plants, and many of them, mutating, become pathogenic also to humans. Several cases of affecting humans by originally animal viruses have been confirmed. Viral infections cause significant morbidity and mortality in humans, the increase of which is caused by general immunosuppression of the world population, changes in climate, and overall globalization. In spite of the fact that the pharmaceutical industry pays great attention to human viral infections, many of clinically used antivirals demonstrate also increased toxicity against human cells, limited bioavailability, and thus, not entirely suitable therapeutic profile. In addition, due to resistance, a combination of antivirals is needed for life-threatening infections. Thus, the development of new antiviral agents is of great importance for the control of virus spread. On the other hand, the discovery and development of structurally new antivirals represent risks. Therefore, another strategy is being developed, namely the reformulation of existing antivirals into nanoformulations and investigation of various metal and metalloid nanoparticles with respect to their diagnostic, prophylactic, and therapeutic antiviral applications. This chapter is focused on nanoscale materials/formulations with the potential to be used for the treatment or inhibition of the spread of viral diseases caused by human immunodeficiency virus, influenza A viruses (subtypes H3N2 and H1N1), avian influenza and swine influenza viruses, respiratory syncytial virus, herpes simplex virus, hepatitis B and C viruses, Ebola and Marburg viruses, Newcastle disease virus, dengue and Zika viruses, and pseudorabies virus. Effective antiviral long-lasting and target-selective nanoformulations developed for oral, intravenous, intramuscular, intranasal, intrarectal, intravaginal, and intradermal applications are discussed. Benefits of nanoparticle-based vaccination formulations with the potential to secure cross protection against divergent viruses are outlined as well.
Collapse
Affiliation(s)
- Mahendra Rai
- Department of Biotechnology, Nanobiotechnology Laboratory, Amravati, Maharashtra, India, Department of Chemistry, Federal University of Piauí, Teresina, Piauí Brazil
| | - Bushra Jamil
- Department of DMLS, University of Lahore, Islamabad, Pakistan
| |
Collapse
|