1
|
Han MM, Fan YK, Zhang Y, Dong ZQ. Advances in herbal polysaccharides-based nano-drug delivery systems for cancer immunotherapy. J Drug Target 2024; 32:311-324. [PMID: 38269853 DOI: 10.1080/1061186x.2024.2309661] [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: 10/13/2023] [Accepted: 01/20/2024] [Indexed: 01/26/2024]
Abstract
The boom in cancer immunotherapy has provided many patients with a better chance of survival, but opportunities often come with challenges. Single immunotherapy is not good enough to eradicate tumours, and often fails to achieve the desired therapeutic effect because of the low targeting of immunotherapy drugs, and causes more side effects. As a solution to this problem, researchers have developed several nano Drug Delivery Systems (NDDS) to deliver immunotherapeutic agents to achieve good therapeutic outcomes. However, traditional drug delivery systems (DDS) have disadvantages such as poor bioavailability, high cytotoxicity, and difficulty in synthesis, etc. Herbal Polysaccharides (HPS), derived from natural Chinese herbs, inherently possess low toxicity. Furthermore, the biocompatibility, biodegradability, hydrophilicity, ease of modification, and immunomodulatory activities of HPS offer unique advantages in substituting traditional DDS. This review initially addresses the current developments and challenges in immunotherapy. Subsequently, it focuses on the immunomodulatory mechanisms of HPS and their design as nanomedicines for targeted drug delivery in tumour immunotherapy. Our findings reveal that HPS-based nanomedicines exhibit significant potential in enhancing the efficacy of cancer immunotherapy, providing crucial theoretical foundations and practical guidelines for future clinical applications.
Collapse
Affiliation(s)
- Miao-Miao Han
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Yi-Kai Fan
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Yun Zhang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
- Joint Research Center for Chinese Medicinal Herbs, IMPLAD, ABRC & ACCL, Beijing, China
| | - Zheng-Qi Dong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
- Joint Research Center for Chinese Medicinal Herbs, IMPLAD, ABRC & ACCL, Beijing, China
| |
Collapse
|
2
|
Bhowmik D, Bhuyan A, Gunalan S, Kothandan G, Kumar D. In silico and immunoinformatics based multiepitope subunit vaccine design for protection against visceral leishmaniasis. J Biomol Struct Dyn 2024; 42:9731-9752. [PMID: 37655736 DOI: 10.1080/07391102.2023.2252901] [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: 02/12/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023]
Abstract
Visceral leishmaniasis (VL) is a vector-borne neglected tropical protozoan disease with high fatality and no certified vaccine. Conventional vaccine preparation is challenging and tedious. Here in this work, we created a global multiepitope subunit vaccination against VL utilizing innovative immunoinformatics technique based on the extensively conserved epitopic regions of the PrimPol protein of Leishmania donovani consisting of four subunits which were analyzed and studied, out of which DNA primase large subunit and DNA polymerase α subunit B were evaluated as antigens by Vaxijen 2.0. The multiepitope vaccine design includes a single adjuvant β-defensins, eight CTL epitopes, eight HTL epitopes, seven linear BCL epitopes and one discontinuous BCL epitope to induce innate, cellular and humoral immune responses against VL. The Expasy ProtParam tool characterized the physiochemical parameters of the vaccine. At the same time, SOLpro evaluated our vaccine constructs to be soluble upon expression. We also modeled the stable tertiary structure of our vaccine construct through Robetta modeling for molecular docking studies with toll-like receptor proteins through HADDOCK 2.4. Simulations based on molecular dynamics revealed an intact vaccine and TLR8 complex, supporting our vaccine design's immunogenicity. Also, the immune simulation of our vaccine by the C-ImmSim server demonstrated the potency of the multiepitope vaccine construct to induce proper immune response for host defense. Codon optimization and in silico cloning of our vaccine further assured high expression. The outcomes of our study on multiepitope vaccine design significantly produced a potential candidate against VL and can potentially eradicate the disease in the future after clinical investigations.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Deep Bhowmik
- Deparment of Microbiology, Assam University, Silchar, Assam, India
| | - Achyut Bhuyan
- Deparment of Microbiology, Assam University, Silchar, Assam, India
| | - Seshan Gunalan
- Biopolymer Modelling Laboratory, Centre of Advanced Study in Crystallography and Biophysics, Guindy Campus, University of Madras, Chennai, India
| | - Gugan Kothandan
- Biopolymer Modelling Laboratory, Centre of Advanced Study in Crystallography and Biophysics, Guindy Campus, University of Madras, Chennai, India
| | - Diwakar Kumar
- Deparment of Microbiology, Assam University, Silchar, Assam, India
| |
Collapse
|
3
|
An W, Li T, Tian X, Fu X, Li C, Wang Z, Wang J, Wang X. Allergies to Allergens from Cats and Dogs: A Review and Update on Sources, Pathogenesis, and Strategies. Int J Mol Sci 2024; 25:10520. [PMID: 39408849 PMCID: PMC11476515 DOI: 10.3390/ijms251910520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Inhalation allergies caused by cats and dogs can lead to a range of discomforting symptoms, such as rhinitis and asthma, in humans. With the increasing popularity of and care provided to these companion animals, the allergens they produce pose a growing threat to susceptible patients' health. Allergens from cats and dogs have emerged as significant risk factors for triggering asthma and allergic rhinitis worldwide; however, there remains a lack of systematic measures aimed at assisting individuals in recognizing and preventing allergies caused by these animals. This review provides comprehensive insights into the classification of cat and dog allergens, along with their pathogenic mechanisms. This study also discusses implementation strategies for prevention and control measures, including physical methods, gene-editing technology, and immunological approaches, as well as potential strategies for enhancing allergen immunotherapy combined with immunoinformatics. Finally, it presents future prospects for the prevention and treatment of human allergies caused by cats and dogs. This review will improve knowledge regarding allergies to cats and dogs while providing insights into potential targets for the development of next-generation treatments.
Collapse
Affiliation(s)
- Wei An
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ting Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, No. 20, Dongda Street, Beijing 100071, China;
| | - Xinya Tian
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiaoxin Fu
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Chunxiao Li
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Zhenlong Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Jinquan Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiumin Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| |
Collapse
|
4
|
Sinani G, Sessevmez M, Şenel S. Applications of Chitosan in Prevention and Treatment Strategies of Infectious Diseases. Pharmaceutics 2024; 16:1201. [PMID: 39339237 PMCID: PMC11434819 DOI: 10.3390/pharmaceutics16091201] [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: 08/12/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Chitosan is the most commonly investigated functional cationic biopolymer in a wide range of medical applications due to its promising properties such as biocompatibility, biodegradability, and bioadhesivity, as well as its numerous bioactive properties. Within the last three decades, chitosan and its derivatives have been investigated as biomaterials for drug and vaccine delivery systems, besides for their bioactive properties. Due to the functional groups in its structure, it is possible to tailor the delivery systems with desired properties. There has been a great interest in the application of chitosan-based systems also for the prevention and treatment of infectious diseases, specifically due to their antimicrobial, antiviral, and immunostimulatory effects. In this review, recent applications of chitosan in the prevention and treatment of infectious diseases are reviewed, and possibilities and limitations with regards to technical and regulatory aspects are discussed. Finally, the future perspectives on utilization of chitosan as a biomaterial are discussed.
Collapse
Affiliation(s)
- Genada Sinani
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Altinbas University, 34147 Istanbul, Türkiye;
| | - Melike Sessevmez
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, 34116 Istanbul, Türkiye;
| | - Sevda Şenel
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe Univesity, 06100 Ankara, Türkiye
| |
Collapse
|
5
|
Li Y, Wang C, Lv H, Li J, Zhang X, Zhang S, Shen Q, Wu Q, Liu Y, Peng R, Liu Z. Manganese-Modified Aluminum Adjuvant Enhances both Humoral and Cellular Immune Responses. Adv Healthc Mater 2024:e2401675. [PMID: 39177146 DOI: 10.1002/adhm.202401675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/11/2024] [Indexed: 08/24/2024]
Abstract
Aluminum adjuvants remain the most commonly used vaccine adjuvants. Being rather effective in triggering humoral immunity, however, aluminum adjuvants usually show limited abilities in activating cellular immunities. Herein, by adding manganese ions during the preparation of aluminum adjuvant, a manganese-modified aluminum (Mn-Al) adjuvant is obtained, which can effectively stimulate both humoral and cellular immune responses. Such Mn-Al adjuvant can enhance antigen adsorption and promote antigen internalization by dendritic cells (DCs). Subsequently, the released Mn2+ can activate the cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes pathway to further promote DC activation. When combines with the model antigen ovalbumin (OVA), the Mn-Al-adjuvantes vaccine can induce high levels of antigen-specific antibody titers and high proportions of antigen-specific cytotoxic T cells in vivo. Moreover, the Mn-Al-adjuvanted vaccine elicited stronger antigen-specific humoral and cellular immune responses than high-dose of the aluminum-based adjuvant. Additionally, immunization of mice with OVA in the presence of the Mn-Al adjuvant significantly inhibited the growth of B16-OVA tumors. Furthermore, when formulated with human papillomavirus antigens, Mn-Al-adjuvanted vaccines show better in vivo vaccination performance than aluminum-adjuvanted vaccines. Therefore, the manganese-modified aluminum adjuvant may thus become a new vaccine adjuvant with the potential to replace conventional aluminum adjuvants.
Collapse
Affiliation(s)
- Yaxin Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Chenya Wang
- InnoBM Pharmaceuticals Co., Ltd., Suzhou, Jiangsu, 215123, China
| | - Haoyuan Lv
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Jingting Li
- Jiangsu Recbio Technology Co., Ltd., Taizhou, Jiangsu, 225300, China
| | - Xupei Zhang
- Jiangsu Recbio Technology Co., Ltd., Taizhou, Jiangsu, 225300, China
| | - Shiyuan Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Qing Shen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Qianqian Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yong Liu
- Jiangsu Recbio Technology Co., Ltd., Taizhou, Jiangsu, 225300, China
| | - Rui Peng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
6
|
Coelho CH, Marquez S, Nguemwo Tentokam BC, Berhe AD, Miura K, Rao VN, Long CA, Doumbo OK, Sagara I, Healy S, Kleinstein SH, Duffy PE. Antibody gene features associated with binding and functional activity in malaria vaccine-derived human mAbs. NPJ Vaccines 2024; 9:144. [PMID: 39127706 DOI: 10.1038/s41541-024-00929-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
The impact of adjuvants on malaria vaccine-induced antibody repertoire is poorly understood. Here, we characterize the impact of two adjuvants, Alhydrogel® and AS01, on antibody clonotype diversity, binding and function, post malaria vaccination. We expressed 132 recombinant anti-Pfs230D1 human monoclonal antibodies (mAbs) from participants immunized with malaria transmission-blocking vaccine Pfs230D1, formulated with either Alhydrogel® or AS01. Anti-Pfs230D1 mAbs generated by Alhydrogel® formulation showed higher binding frequency to Pfs230D1 compared to AS01 formulation, although the frequency of functional mAbs was similar between adjuvant groups. Overall, the AS01 formulation induced anti-Pfs230D1 functional antibodies from a broader array of germline sequences versus the Alhydrogel® formulation. All mAbs using IGHV1-69 gene from the Alhydrogel® cohort bound to recombinant Pfs230D1, but did not block parasite transmission to mosquitoes, similar to the IGHV1-69 mAbs isolated from the AS01 cohort. These findings may help inform vaccine design and adjuvant selection for immunization with Plasmodium antigens.
Collapse
Affiliation(s)
- Camila H Coelho
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- C-VARPP- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Immunology Precision Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Susanna Marquez
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Bergeline C Nguemwo Tentokam
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anne D Berhe
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - Vishal N Rao
- C-VARPP- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - Ogobara K Doumbo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Issaka Sagara
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Sara Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Steven H Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, 06511, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
7
|
Quispe-Salcedo A, Yamazaki T, Ohshima H. Effects of Synthetic Toll-Like Receptor 9 Ligand Molecules on Pulpal Immunomodulatory Response and Repair after Injuries. Biomolecules 2024; 14:931. [PMID: 39199319 PMCID: PMC11353191 DOI: 10.3390/biom14080931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Synthetic oligodeoxynucleotides (ODNs) containing unmethylated cytosine-phosphate-guanine (CpG) motifs (CpG-ODNs) are ligand molecules for Toll-like receptor 9 (TLR9), which is expressed by odontoblasts in vitro and dental pulp cells. This study determined the effects of CpG-ODNs on pulpal immunomodulatory response and repair following injury. Briefly, the upper right first molars of three-week-old mice were extracted, immersed in Type A (D35) or B (K3) CpG-ODN solutions (0.1 or 0.8 mM) for 30 min, and then replanted. Pulpal healing and immunomodulatory activity were assessed by hematoxylin-eosin and AZAN staining, as well as immunohistochemistry. One week following the operation, inflammatory reactions occurred in all of the experimental groups; however, re-revascularization and newly formed hard tissue deposition were observed in the pulp chamber of all groups at week 2. A positive trend in the expression of immune cell markers was observed toward the CpG-ODN groups at 0.1 mM. Our data suggest that synthetic CpG-ODN solutions at low concentrations may evoke a long-lasting macrophage-TLR9-mediated pro-inflammatory, rather than anti-inflammatory, response in the dental pulp to modulate the repair process and hard tissue formation. Further studies are needed to determine the effects of current immunomodulatory agents in vitro and in vivo and develop treatment strategies for dental tissue regeneration.
Collapse
Affiliation(s)
- Angela Quispe-Salcedo
- Division of Anatomy and Cell Biology of the Hard Tissue, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| | - Tomohiko Yamazaki
- Research Center for Macromolecules and Biomaterials, National Institute of Material Sciences (NIMS), Tsukuba 305-0047, Japan;
| | - Hayato Ohshima
- Division of Anatomy and Cell Biology of the Hard Tissue, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| |
Collapse
|
8
|
Sadeghi L, Bolhassani A, Mohit E, Baesi K, Aghasadeghi MR, Milani A, Agi E. Engineered ClearColi™-derived outer membrane vesicles as functional carriers for development of HIV-1 therapeutic vaccine candidate. Microb Pathog 2024; 193:106749. [PMID: 38879140 DOI: 10.1016/j.micpath.2024.106749] [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: 02/27/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 07/24/2024]
Abstract
Bacteria-derived outer membrane vesicles (OMVs) can be engineered to incorporate foreign antigens. This study explored the potential of ClearColi™-derived OMVs as a natural adjuvant and a carrier (recombinant OMVs or rOMVs) for development of an innovative therapeutic vaccine candidate harboring HIV-1 Nef and Nef-Tat antigens. Herein, the rOMVs containing CytolysinA (ClyA)-Nef and ClyA-Nef-Tat fusion proteins were isolated from ClearColi™ strain. The presence of Nef and Nef-Tat proteins on their surface (rOMVNef and rOMVNef-Tat) was confirmed by western blotting after proteinase K treatment. Immune responses induced by Nef and Nef-Tat proteins emulsified with Montanide® ISA720 or mixed with OMVs, and also rOMVNef and rOMVNef-Tat were investigated in BALB/c mice. Additionally, the potency of splenocytes exposed to single-cycle replicable (SCR) HIV-1 virions was assessed for the secretion of cytokines in vitro. Our findings showed that the rOMVs as an antigen carrier (rOMVNef and rOMVNef-Tat) induced higher levels of IgG2a, IFN-γ and granzyme B compared to OMVs as an adjuvant (Nef + OMV and Nef-Tat + OMV), and also Montanide® ISA720 (Nef + Montanide and Nef-Tat + Montanide). Moreover, IFN-γ level in splenocytes isolated from mice immunized with rOMVNef-Tat was higher than other regimens after exposure to SCR virions. Generally, ClearColi™-derived rOMVs can serve as potent carriers for developing effective vaccines against HIV-1 infection.
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
| | | | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Elnaz Agi
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| |
Collapse
|
9
|
Wang R, Huang H, Yu C, Li X, Wang Y, Xie L. Current status and future directions for the development of human papillomavirus vaccines. Front Immunol 2024; 15:1362770. [PMID: 38983849 PMCID: PMC11231394 DOI: 10.3389/fimmu.2024.1362770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 06/11/2024] [Indexed: 07/11/2024] Open
Abstract
The development of human papillomavirus (HPV) vaccines has made substantive progress, as represented by the approval of five prophylactic vaccines since 2006. Generally, the deployment of prophylactic HPV vaccines is effective in preventing newly acquired infections and incidences of HPV-related malignancies. However, there is still a long way to go regarding the prevention of all HPV infections and the eradication of established HPV infections, as well as the subsequent progression to cancer. Optimizing prophylactic HPV vaccines by incorporating L1 proteins from more HPV subtypes, exploring adjuvants that reinforce cellular immune responses to eradicate HPV-infected cells, and developing therapeutic HPV vaccines used either alone or in combination with other cancer therapeutic modalities might bring about a new era getting closer to the vision to get rid of HPV infection and related diseases. Herein, we summarize strategies for the development of HPV vaccines, both prophylactic and therapeutic, with an emphasis on the selection of antigens and adjuvants, as well as implications for vaccine efficacy based on preclinical studies and clinical trials. Additionally, we outline current cutting-edge insights on formulation strategies, dosing schedules, and age expansion among HPV vaccine recipients, which might play important roles in addressing barriers to vaccine uptake, such as vaccine hesitancy and vaccine availability.
Collapse
Affiliation(s)
- Rui Wang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Hongpeng Huang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Chulin Yu
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Xuefeng Li
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Yang Wang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
- Cell Culture Engineering Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
10
|
Ahmed M, Kurungottu P, Swetha K, Atla S, Ashok N, Nagamalleswari E, Bonam SR, Sahu BD, Kurapati R. Role of NLRP3 inflammasome in nanoparticle adjuvant-mediated immune response. Biomater Sci 2024. [PMID: 38867716 DOI: 10.1039/d4bm00439f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is pivotal in orchestrating the immune response induced by nanoparticle adjuvants. Understanding the intricate mechanisms underlying the activation of NLRP3 inflammasome by these adjuvants is crucial for deciphering their immunomodulatory properties. This review explores the involvement of the NLRP3 inflammasome in mediating immune responses triggered by nanoparticle adjuvants. It delves into the signaling pathways and cellular mechanisms involved in NLRP3 activation, highlighting its significance in modulating the efficacy and safety of nanoparticle-based adjuvants. A comprehensive grasp of the interplay between NLRP3 inflammasome and nanoparticle adjuvants holds promise for optimizing vaccine design and advancing immunotherapeutic strategies.
Collapse
Affiliation(s)
- Momitul Ahmed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Pavithra Kurungottu
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - K Swetha
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Sandeep Atla
- Texas A&M Drug Discovery Center, Department of Chemistry, Texas A&M University, College Station, Texas 77843, USA
| | - Nivethitha Ashok
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Easa Nagamalleswari
- MTCC and Gene Bank, CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| |
Collapse
|
11
|
Ahmad S, Demneh FM, Rehman B, Almanaa TN, Akhtar N, Pazoki-Toroudi H, Shojaeian A, Ghatrehsamani M, Sanami S. In silico design of a novel multi-epitope vaccine against HCV infection through immunoinformatics approaches. Int J Biol Macromol 2024; 267:131517. [PMID: 38621559 DOI: 10.1016/j.ijbiomac.2024.131517] [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: 11/11/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
Infection with the hepatitis C virus (HCV) is one of the causes of liver cancer, which is the world's sixth most prevalent and third most lethal cancer. The current treatments do not prevent reinfection; because they are expensive, their usage is limited to developed nations. Therefore, a prophylactic vaccine is essential to control this virus. Hence, in this study, an immunoinformatics method was applied to design a multi-epitope vaccine against HCV. The best B- and T-cell epitopes from conserved regions of the E2 protein of seven HCV genotypes were joined with the appropriate linkers to design a multi-epitope vaccine. In addition, cholera enterotoxin subunit B (CtxB) was included as an adjuvant in the vaccine construct. This study is the first to present this epitopes-adjuvant combination. The vaccine had acceptable physicochemical characteristics. The vaccine's 3D structure was predicted and validated. The vaccine's binding stability with Toll-like receptor 2 (TLR2) and TLR4 was confirmed using molecular docking and molecular dynamics (MD) simulation. The immune simulation revealed the vaccine's efficacy by increasing the population of B and T cells in response to vaccination. In silico expression in Escherichia coli (E. coli) was also successful.
Collapse
Affiliation(s)
- Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar 25000, Pakistan; Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, P.O. Box 36, Lebanon; Department of Natural Sciences, Lebanese American University, Beirut, P.O. Box 36, Lebanon
| | - Fatemeh Mobini Demneh
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Bushra Rehman
- Institute of Biotechnology and Microbiology, Bacha khan University, Charsadda, Pakistan
| | - Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nahid Akhtar
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, India
| | - Hamidreza Pazoki-Toroudi
- Department of Physiology & Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Shojaeian
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahdi Ghatrehsamani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Samira Sanami
- Abnormal Uterine Bleeding Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
12
|
Lv X, Martin J, Hoover H, Joshi B, Wilkens M, Ullisch DA, Leibold T, Juchum JS, Revadkar S, Kalinovska B, Keith J, Truby A, Liu G, Sun E, Haserick J, DeGnore J, Conolly J, Hill AV, Baldoni J, Kensil C, Levey D, Spencer AJ, Gorr G, Findeis M, Tanne A. Chemical and biological characterization of vaccine adjuvant QS-21 produced via plant cell culture. iScience 2024; 27:109006. [PMID: 38361610 PMCID: PMC10867646 DOI: 10.1016/j.isci.2024.109006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/07/2023] [Accepted: 01/19/2024] [Indexed: 02/17/2024] Open
Abstract
Many vaccines, including those using recombinant antigen subunits, rely on adjuvant(s) to enhance the efficacy of the host immune responses. Among the few adjuvants clinically approved, QS-21, a saponin-based immunomodulatory molecule isolated from the tree bark of Quillaja saponaria (QS) is used in complex formulations in approved effective vaccines. High demand of the QS raw material as well as manufacturing scalability limitation has been barriers here. We report for the first-time successful plant cell culture production of QS-21 having structural, chemical, and biologic, properties similar to the bark extracted product. These data ensure QS-21 and related saponins are broadly available and accessible to drug developers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - John S. Juchum
- Phyton Biotech LLC, 1503 Cliveden Avenue, Delta, BC V3M 6P7, Canada
| | | | | | | | - Adam Truby
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | | | | | | | - Adrian V.S. Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | | | - Alexandra J. Spencer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Hunter Medical Research Institute, School of Biomedical Sciences and Pharmacy, College of Health, Medicine & Wellbeing; Immune Health Program, New Lambton Heights, NSW, Australia
| | | | | | | |
Collapse
|
13
|
Lin YJ, Zimmermann J, Schülke S. Novel adjuvants in allergen-specific immunotherapy: where do we stand? Front Immunol 2024; 15:1348305. [PMID: 38464539 PMCID: PMC10920236 DOI: 10.3389/fimmu.2024.1348305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Type I hypersensitivity, or so-called type I allergy, is caused by Th2-mediated immune responses directed against otherwise harmless environmental antigens. Currently, allergen-specific immunotherapy (AIT) is the only disease-modifying treatment with the potential to re-establish clinical tolerance towards the corresponding allergen(s). However, conventional AIT has certain drawbacks, including long treatment durations, the risk of inducing allergic side effects, and the fact that allergens by themselves have a rather low immunogenicity. To improve AIT, adjuvants can be a powerful tool not only to increase the immunogenicity of co-applied allergens but also to induce the desired immune activation, such as promoting allergen-specific Th1- or regulatory responses. This review summarizes the knowledge on adjuvants currently approved for use in human AIT: aluminum hydroxide, calcium phosphate, microcrystalline tyrosine, and MPLA, as well as novel adjuvants that have been studied in recent years: oil-in-water emulsions, virus-like particles, viral components, carbohydrate-based adjuvants (QS-21, glucans, and mannan) and TLR-ligands (flagellin and CpG-ODN). The investigated adjuvants show distinct properties, such as prolonging allergen release at the injection site, inducing allergen-specific IgG production while also reducing IgE levels, as well as promoting differentiation and activation of different immune cells. In the future, better understanding of the immunological mechanisms underlying the effects of these adjuvants in clinical settings may help us to improve AIT.
Collapse
Affiliation(s)
- Yen-Ju Lin
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Stefan Schülke
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
- Section Research Allergology (ALG 5), Division of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
14
|
Huete-Carrasco J, Lynch RI, Ward RW, Lavelle EC. Rational design of polymer-based particulate vaccine adjuvants. Eur J Immunol 2024; 54:e2350512. [PMID: 37994660 DOI: 10.1002/eji.202350512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 11/24/2023]
Abstract
Vaccination is considered one of the major milestones in modern medicine, facilitating the control and eradication of life-threatening infectious diseases. Vaccine adjuvants are a key component of many vaccines, serving to steer antigen-specific immune responses and increase their magnitude. Despite major advances in the field of adjuvant research over recent decades, our understanding of their mechanism of action remains incomplete. This hinders our capacity to further improve these adjuvant technologies, so addressing how adjuvants induce and control the induction of innate and adaptive immunity is a priority. Investigating how adjuvant physicochemical properties, such as size and charge, exert immunomodulatory effects can provide valuable insights and serve as the foundation for the rational design of vaccine adjuvants. Most clinically applied adjuvants are particulate in nature and polymeric particulate adjuvants present advantages due to stability, biocompatibility profiles, and flexibility in terms of formulation. These properties can impact on antigen release kinetics and biodistribution, cellular uptake and targeting, and drainage to the lymphatics, consequently dictating the induction of innate, cellular, and humoral adaptive immunity. A current focus is to apply rational design principles to the development of adjuvants capable of eliciting robust cellular immune responses including CD8+ cytotoxic T-cell and Th1-biased CD4+ T-cell responses, which are required for vaccines against intracellular pathogens and cancer. This review highlights recent advances in our understanding of how particulate adjuvants, especially polymer-based particulates, modulate immune responses and how this can be used as a guide for improved adjuvant design.
Collapse
Affiliation(s)
- Jorge Huete-Carrasco
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Roisin I Lynch
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN) & Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin, Ireland
| | - Ross W Ward
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN) & Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
15
|
Bjerkhaug AU, Ramalingham S, Mboizi R, Le Doare K, Klingenberg C. The immunogenicity and safety of Group B Streptococcal maternal vaccines: A systematic review. Vaccine 2024; 42:84-98. [PMID: 38072754 DOI: 10.1016/j.vaccine.2023.11.056] [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: 10/24/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 01/01/2024]
Abstract
PURPOSE To systematically review immunogenicity and safety data of maternal group B streptococcal (GBS) vaccines in published clinical trials until July 2023. METHODS EMBASE, MEDLINE, Cochrane Library and clinicaltrial.gov. databases were searched for clinical studies that reported immunogenicity and/or safety of GBS vaccine in non-pregnant adults, pregnant women and infants between 1st of January 1996 to 31st of July 2023. Pairs of reviewers independently selected, data extracted, and assessed the risk of bias of the studies. Discrepancies were resolved by consensus. (PROSPERO CRD42020185213). RESULTS We retrieved 1472 records from the literature search; 20 studies and 6 sub-studies were included, involving 4440 non-pregnant participants and 1325 pregnant women with their newborns. There was a significantly higher IgG Geometric Mean Concentration (GMC) and IgG placental transfer ratios in vaccinated compared to placebo groups, with peak response 4-8 weeks after vaccination. Placental transfer ratio varied from 0.4 to 1.4 across five studies. The different clinical trials used different assays that limited direct comparison. There were no significant differences in the risk of serious adverse events (adjusted OR 0.73; 95 % CI 0.49-1.07), serious adverse events leading to withdrawal (adjusted OR 0.44; 95 % CI 0.13-1.51), and systemic illness or fever (adjusted OR 1.05; 95 % CI 0.26-4.19) between the vaccine and placebo groups. CONCLUSIONS The published clinical trials show significant IgG GMC response in subjects receiving the conjugated capsular polysaccharide and surface subunit protein vaccines compared to placebo. In current clinical trials of experimental GBS maternal vaccines, there have been no observed serious adverse events of special interest directly linked to vaccination.
Collapse
Affiliation(s)
- Aline U Bjerkhaug
- Paediatric Research Group, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway; Department of Paediatrics and Adolescence Medicine, University Hospital of North Norway, Tromsø, Norway.
| | - Shouwmika Ramalingham
- Paediatric Research Group, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Robert Mboizi
- Makerere University Johns Hopkins University (MU-JHU) Research Collaboration (MUJHU CARE LTD), Kampala, Uganda
| | - Kirsty Le Doare
- Makerere University Johns Hopkins University (MU-JHU) Research Collaboration (MUJHU CARE LTD), Kampala, Uganda; Centre for Neonatal and Paediatric Infection, Maternal and Neonatal Vaccine Immunology Research Group, St Georgés University of London, United Kingdom
| | - Claus Klingenberg
- Paediatric Research Group, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway; Department of Paediatrics and Adolescence Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
16
|
He S, Liu S. Zwitterionic materials for nucleic acid delivery and therapeutic applications. J Control Release 2024; 365:919-935. [PMID: 38103789 DOI: 10.1016/j.jconrel.2023.12.017] [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: 11/28/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Nucleic acid therapeutics have demonstrated substantial potential in combating various diseases. However, challenges persist, particularly in the delivery of multifunctional nucleic acids. To address this issue, numerous gene delivery vectors have been developed to fully unlock the potential of gene therapy. The advancement of innovative materials with exceptional delivery properties is critical to propel the clinical translation of nucleic acid drugs. Cationic vector materials have received extensive attention, while zwitterionic materials remain relatively underappreciated in delivery. In this review, we outline a diverse range of zwitterionic material nucleic acid carriers, predominantly encompassing zwitterionic lipids, polymers and peptides. Their respective chemical structures, synthesis approaches, properties, advantages, and therapeutic applications are summarized and discussed. Furthermore, we highlight the challenges and future opportunities associated with the development of zwitterionic vector materials. This review will aid to understand the zwitterionic materials in aiding gene delivery, contributing to the continual progress of nucleic acid therapeutics.
Collapse
Affiliation(s)
- Shun He
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shuai Liu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
17
|
Parmaksız S, Pekcan M, Özkul A, Türkmen E, Rivero-Arredondo V, Ontiveros-Padilla L, Forbes N, Perrie Y, López-Macías C, Şenel S. In vivo evaluation of new adjuvant systems based on combination of Salmonella Typhi porins with particulate systems: Liposomes versus polymeric particles. Int J Pharm 2023; 648:123568. [PMID: 37925042 DOI: 10.1016/j.ijpharm.2023.123568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/06/2023]
Abstract
Subunit vaccines that have weak immunogenic activity require adjuvant systems for enhancedcellular and long-acting humoral immune responses. Both lipid-based and polymeric-based particulate adjuvants have been widely investigated to induce the desired immune responses against the subunit vaccines. The adjuvant efficacy of these particulate adjuvants depends upon their physicochemical properties such as particle size, surface charge, shape and their composition. Previously, we showed in vitro effect of adjuvant systems based on combination of chitosan and Salmonella Typhi porins in microparticle or nanoparticle form, which were spherical with positive surface charge. In the present study, we have further developed an adjuvant system based on combination of porins with liposomes (cationic and neutral) and investigated the adjuvant effect of both the liposomal and polymeric systems in BALB/c mice using a model antigen, ovalbumin. Humoral immune responses were determined following priming and booster dose at 15-day intervals. In overall, IgM and IgG levels were induced in the presence of both the liposomal and polymeric adjuvant systems indicating the positive impact of combination with porins. The highest IgM levels were obtained on Day 8, and liposomal adjuvant systems were found to elicit significantly higher IgM levels compared to polymeric systems. IgG levels were increased significantly after booster, particularly more profound with the micro-sized polymeric system when compared to cationic liposomal system with nano-size. Our results demonstrated that the developed particulate systems are promising both as an adjuvant and delivery system, providing enhanced immune responses against subunit antigens, and have the potential for long-term protection.
Collapse
Affiliation(s)
- Selin Parmaksız
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06100 Ankara, Turkey
| | - Mert Pekcan
- Ankara University, Faculty of Veterinary Medicine, Department of Biochemistry, 06110 Ankara, Turkey
| | - Aykut Özkul
- Ankara University, Faculty of Veterinary Medicine, Department of Virology, Ankara University, 06110 Ankara, Turkey
| | - Ece Türkmen
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06100 Ankara, Turkey
| | - Vanessa Rivero-Arredondo
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Luis Ontiveros-Padilla
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Neil Forbes
- University of Strathclyde, Strathclyde Institute of Pharmacy and Biomedical Sciences, Glasgow, United Kingdom
| | - Yvonne Perrie
- University of Strathclyde, Strathclyde Institute of Pharmacy and Biomedical Sciences, Glasgow, United Kingdom
| | - Constantino López-Macías
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Sevda Şenel
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06100 Ankara, Turkey.
| |
Collapse
|
18
|
Vakili B, Karami-Darehnaranji M, Mirzaei E, Hosseini F, Nezafat N. Graphene oxide as novel vaccine adjuvant. Int Immunopharmacol 2023; 125:111062. [PMID: 37866317 DOI: 10.1016/j.intimp.2023.111062] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/30/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
To improve antigen immunogenicity and promote long-lasting immunity, vaccine formulations have been appropriately supplemented with adjuvants. Graphene has been found to enhance the presentation of antigens to CD8+ T cells, as well as stimulating innate immune responses and inflammatory factors. Its properties, such as large surface area, water stability, and high aspect ratio, make it a suitable candidate for delivering biological substances. Graphene-based nanomaterials have recently attracted significant attention as a new type of vaccine adjuvants due to their potential role in the activation of immune responses. Due to the limited functionality of some approved human adjuvants for use, the development of new all-purpose adjuvants is urgently required. Research on the immunological and biomedical use of graphene oxide (GO) indicates that these nanocarriers possess excellent physicochemical properties, acceptable biocompatibility, and a high capacity for drug loading. Graphene-based nanocarriers also could improve the function of some immune cells such as dendritic cells and macrophages through specific signaling pathways. However, GO injection can lead to significant oxidative stress and inflammation. Various surface functionalization protocols have been employed to reduce possible adverse effects of GO, such as aggregation of GO in biological liquids and induce cell death. Furthermore, these modifications enhance the properties of functionalized-GO's qualities, making it an excellent carrier and adjuvant. Shedding light on different physicochemical and structural properties of GO and its derivatives has led to their application in various therapeutic and drug delivery fields. In this review, we have endeavored to elaborate on different aspects of GO.
Collapse
Affiliation(s)
- Bahareh Vakili
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboubeh Karami-Darehnaranji
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Esmaeil Mirzaei
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farnaz Hosseini
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Computational Vaccine and Drug Design Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
19
|
Li X, Chen Y, Wang R, Lu E, Luo K, Sha X. Enhancement of cancer immunotherapy using CRT valgus tumor cell membranes coated bacterial whole peptidoglycan combined with radiotherapy. Int J Pharm 2023; 646:123430. [PMID: 37742823 DOI: 10.1016/j.ijpharm.2023.123430] [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: 07/09/2023] [Revised: 08/23/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023]
Abstract
Immunotherapy has achieved some success in preclinical and clinical studies, but the immunosuppressive tumor microenvironment (TME) leads to a low response rate of this therapy. In this paper, we describe a calreticulin (CRT) valgus CT-26 tumor cell membranes-coated bacterial whole peptidoglycan (WPG) from P. aeruginosa (CPW/SR) with a high rate of the STING agonist loading. In the construct, WPG from P. aeruginosa (P.WPG) was used as a carrier with the immunoadjuvant function while synergistically promoting the maturation of dendritic cells (DCs) through the delivery of the STING agonist SR-717. CRT valgus tumor cell membranes were identified and internalized by DCs via CRT on the surface. In addition, this construct was able to reverse the immunosuppressive TME in vivo and achieve synergies with radiotherapy by creating a personalized tumor vaccine, therefore achieving more resultful antitumor efficacy. In conclusion, CPW/SR constructed in this paper provides a new approach for achieving efficient cancer immunotherapy and combination therapy.
Collapse
Affiliation(s)
- Xinhong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yiting Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Rui Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Enhao Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Kuankuan Luo
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xianyi Sha
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; The Institutes of Integrative Medicine of Fudan University, Shanghai, China.
| |
Collapse
|
20
|
Ko MK, Kim HW, Park SH, Park JH, Kim SM, Lee MJ. The role of zinc sulfate in enhancing cellular and humoral immune responses to foot-and-mouth disease vaccine. Virus Res 2023; 335:199189. [PMID: 37536380 PMCID: PMC10432855 DOI: 10.1016/j.virusres.2023.199189] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Foot-and-mouth disease (FMD) is a rapidly propagating infectious disease of cloven-hoofed animals, especially cattle and pigs, affecting the productivity and profitability of the livestock industry. Presently, FMD is controlled and prevented using vaccines; however, conventional FMD vaccines have several disadvantages, including short vaccine efficacy, low antibody titers, and safety issues in pigs, indicating the need for further studies. Here, we evaluated the efficacy of a novel bivalent vaccine containing zinc sulfate as an immunostimulant and FMD type O and A antigens (O PA2 and A YC, respectively) against FMD virus in mice and pigs. Zinc sulfate induced cellular immunity in murine peritoneal exudate cells (PECs) and porcine peripheral blood mononuclear cells (PBMCs) by increasing IFNγ secretion. Additionally, FMD vaccine containing O PA2 and A YC antigens and zinc sulfate induced early, mid-, and long-term immune responses in mice and pigs, and enhanced cellular and humoral immunity by regulating the expression of pathogen recognition receptors (PRRs), transcription factors, co-stimulatory molecules, and cytokines in porcine PBMCs from vaccinated pigs. Overall, these results indicated that the novel immunostimulant zinc sulfate induced potent cellular and humoral immune responses by stimulating antigen-presenting cells (APCs) and T and B cells, and enhanced long-term immunity by promoting the expression of co-stimulatory molecules. These outcomes suggest that zinc sulfate could be used as a novel vaccine immunostimulant for difficult-to-control viral diseases, such as African swine fever (ASF) or COVID-19.
Collapse
Affiliation(s)
- Mi-Kyeong Ko
- Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea
| | - Hyeong Won Kim
- Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea
| | - So Hui Park
- Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea
| | - Jong-Hyeon Park
- Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea
| | - Su-Mi Kim
- Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea
| | - Min Ja Lee
- Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea.
| |
Collapse
|
21
|
Díaz-Dinamarca DA, Salazar ML, Escobar DF, Castillo BN, Valdebenito B, Díaz P, Manubens A, Salazar F, Troncoso MF, Lavandero S, Díaz J, Becker MI, Vásquez AE. Surface immunogenic protein from Streptococcus agalactiae and Fissurella latimarginata hemocyanin are TLR4 ligands and activate MyD88- and TRIF dependent signaling pathways. Front Immunol 2023; 14:1186188. [PMID: 37790926 PMCID: PMC10544979 DOI: 10.3389/fimmu.2023.1186188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/29/2023] [Indexed: 10/05/2023] Open
Abstract
The development of vaccine adjuvants is of interest for the management of chronic diseases, cancer, and future pandemics. Therefore, the role of Toll-like receptors (TLRs) in the effects of vaccine adjuvants has been investigated. TLR4 ligand-based adjuvants are the most frequently used adjuvants for human vaccines. Among TLR family members, TLR4 has unique dual signaling capabilities due to the recruitment of two adapter proteins, myeloid differentiation marker 88 (MyD88) and interferon-β adapter inducer containing the toll-interleukin-1 receptor (TIR) domain (TRIF). MyD88-mediated signaling triggers a proinflammatory innate immune response, while TRIF-mediated signaling leads to an adaptive immune response. Most studies have used lipopolysaccharide-based ligands as TLR4 ligand-based adjuvants; however, although protein-based ligands have been proven advantageous as adjuvants, their mechanisms of action, including their ability to undergo structural modifications to achieve optimal immunogenicity, have been explored less thoroughly. In this work, we characterized the effects of two protein-based adjuvants (PBAs) on TLR4 signaling via the recruitment of MyD88 and TRIF. As models of TLR4-PBAs, we used hemocyanin from Fissurella latimarginata (FLH) and a recombinant surface immunogenic protein (rSIP) from Streptococcus agalactiae. We determined that rSIP and FLH are partial TLR4 agonists, and depending on the protein agonist used, TLR4 has a unique bias toward the TRIF or MyD88 pathway. Furthermore, when characterizing gene products with MyD88 and TRIF pathway-dependent expression, differences in TLR4-associated signaling were observed. rSIP and FLH require MyD88 and TRIF to activate nuclear factor kappa beta (NF-κB) and interferon regulatory factor (IRF). However, rSIP and FLH have a specific pattern of interleukin 6 (IL-6) and interferon gamma-induced protein 10 (IP-10) secretion associated with MyD88 and TRIF recruitment. Functionally, rSIP and FLH promote antigen cross-presentation in a manner dependent on TLR4, MyD88 and TRIF signaling. However, FLH activates a specific TRIF-dependent signaling pathway associated with cytokine expression and a pathway dependent on MyD88 and TRIF recruitment for antigen cross-presentation. Finally, this work supports the use of these TLR4-PBAs as clinically useful vaccine adjuvants that selectively activate TRIF- and MyD88-dependent signaling to drive safe innate immune responses and vigorous Th1 adaptive immune responses.
Collapse
Affiliation(s)
- Diego A. Díaz-Dinamarca
- Sección de Biotecnología, Subdepartamento, Innovación, Desarrollo, Transferencia Tecnológica (I+D+T) y Evaluación de Tecnologías Sanitarias (ETESA), Instituto de Salud Pública, Santiago, Chile
- Laboratorio de Inmunología, Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
- Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Michelle L. Salazar
- Laboratorio de Inmunología, Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - Daniel F. Escobar
- Sección de Biotecnología, Subdepartamento, Innovación, Desarrollo, Transferencia Tecnológica (I+D+T) y Evaluación de Tecnologías Sanitarias (ETESA), Instituto de Salud Pública, Santiago, Chile
| | - Byron N. Castillo
- Laboratorio de Inmunología, Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - Bastián Valdebenito
- Sección de Biotecnología, Subdepartamento, Innovación, Desarrollo, Transferencia Tecnológica (I+D+T) y Evaluación de Tecnologías Sanitarias (ETESA), Instituto de Salud Pública, Santiago, Chile
| | - Pablo Díaz
- Sección de Biotecnología, Subdepartamento, Innovación, Desarrollo, Transferencia Tecnológica (I+D+T) y Evaluación de Tecnologías Sanitarias (ETESA), Instituto de Salud Pública, Santiago, Chile
| | | | - Fabián Salazar
- Laboratorio de Inmunología, Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
- Investigación y Desarrollo, BIOSONDA S.A., Santiago, Chile
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Mayarling F. Troncoso
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Químicas y Farmacéuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Químicas y Farmacéuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Janepsy Díaz
- Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago, Chile
| | - María Inés Becker
- Laboratorio de Inmunología, Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
- Investigación y Desarrollo, BIOSONDA S.A., Santiago, Chile
| | - Abel E. Vásquez
- Sección de Biotecnología, Subdepartamento, Innovación, Desarrollo, Transferencia Tecnológica (I+D+T) y Evaluación de Tecnologías Sanitarias (ETESA), Instituto de Salud Pública, Santiago, Chile
- Facultad de Ciencias de la Salud, Escuela de Medicina, Universidad del Alba, Santiago, Chile
| |
Collapse
|
22
|
Lee W, Kim YJ, Lee SJ, Ahn DG, Kim SJ. Current Status of Epidemiology, Diagnosis, Therapeutics, and Vaccines for the Re-Emerging Human Monkeypox Virus. J Microbiol Biotechnol 2023; 33:981-991. [PMID: 37519276 PMCID: PMC10468680 DOI: 10.4014/jmb.2306.06033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/01/2023]
Abstract
Monkeypox (Mpox) virus, a member of the Poxviridae family, causes a severe illness similar to smallpox, which is characterized by symptoms such as high fever, rash, and pustules. Human-to-human transmission cases have been reported but remained low since the first recorded case of human infection occurred in the Congo in 1970. Recently, Mpox has re-emerged, leading to an alarming surge in infections worldwide since 2022, originating in the United Kingdom. Consequently, the World Health Organization (WHO) officially declared the '2022-23 Mpox outbreak'. Currently, no specific therapy or vaccine is available for Mpox. Therefore, patients infected with Mpox are treated using conventional therapies developed for smallpox. However, the vaccines developed for smallpox have demonstrated only partial efficacy against Mpox, allowing viral transmission among humans. In this review, we discuss the current epidemiology of the ongoing Mpox outbreak and provide an update on the progress made in diagnosis, treatment, and development of vaccines for Mpox.
Collapse
Affiliation(s)
- Wooseong Lee
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Yu-Jin Kim
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Su Jin Lee
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Dae-Gyun Ahn
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Seong-Jun Kim
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| |
Collapse
|
23
|
Buonaguro L, Tagliamonte M. Peptide-based vaccine for cancer therapies. Front Immunol 2023; 14:1210044. [PMID: 37654484 PMCID: PMC10467431 DOI: 10.3389/fimmu.2023.1210044] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/31/2023] [Indexed: 09/02/2023] Open
Abstract
Different strategies based on peptides are available for cancer treatment, in particular to counter-act the progression of tumor growth and disease relapse. In the last decade, in the context of therapeutic strategies against cancer, peptide-based vaccines have been evaluated in different tumor models. The peptides selected for cancer vaccine development can be classified in two main type: tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs), which are captured, internalized, processed and presented by antigen-presenting cells (APCs) to cell-mediated immunity. Peptides loaded onto MHC class I are recognized by a specific TCR of CD8+ T cells, which are activated to exert their cytotoxic activity against tumor cells presenting the same peptide-MHC-I complex. This process is defined as active immunotherapy as the host's immune system is either de novo activated or restimulated to mount an effective, tumor-specific immune reaction that may ultimately lead to tu-mor regression. However, while the preclinical data have frequently shown encouraging results, therapeutic cancer vaccines clinical trials, including those based on peptides have not provided satisfactory data to date. The limited efficacy of peptide-based cancer vaccines is the consequence of several factors, including the identification of specific target tumor antigens, the limited immunogenicity of peptides and the highly immunosuppressive tumor microenvironment (TME). An effective cancer vaccine can be developed only by addressing all such different aspects. The present review describes the state of the art for each of such factors.
Collapse
Affiliation(s)
| | - Maria Tagliamonte
- Innovative Immunological Models Unit, Istituto Nazionale Tumori - IRCCS - “Fond G. Pascale”, Naples, Italy
| |
Collapse
|
24
|
Coelho CH, Marquez S, Tentokam BCN, Berhe AD, Miura K, Long CA, Sagara I, Healy S, Kleinstein SH, Duffy PE. Antibody gene features associated with binding and functional activity in vaccine-derived human mAbs targeting malaria parasites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551554. [PMID: 37781572 PMCID: PMC10541113 DOI: 10.1101/2023.08.01.551554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Adjuvants have been essential to malaria vaccine development, but their impact on the vaccine-induced antibody repertoire is poorly understood. Here, we used cDNA sequences from antigen-specific single memory B cells to express 132 recombinant human anti-Pfs230 monoclonal antibodies (mAbs). Alhydrogel®-induced mAbs demonstrated higher binding to Pfs230D1, although functional activity was similar between adjuvants. All Alhydrogel® mAbs using IGHV1-69 gene bound to recombinant Pfs230D1, but none blocked parasite transmission to mosquitoes; similarly, no AS01 mAb using IGHV1-69 blocked transmission. Functional mAbs from both Alhydrogel® and AS01 vaccines used IGHV3-21 and IGHV3-30 genes. Antibodies with the longest CDR3 sequences were associated with binding but not functional activity. This study assesses adjuvant effects on antibody clonotype diversity during malaria vaccination.
Collapse
Affiliation(s)
- Camila H. Coelho
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY
| | - Susanna Marquez
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Bergeline C. Nguemwo Tentokam
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anne D. Berhe
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector and Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland, USA
| | - Carole A. Long
- Laboratory of Malaria and Vector and Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland, USA
| | - Issaka Sagara
- Malaria Research and Training Center, University of Sciences, Techniques, and Technology, Bamako, Mali
| | - Sara Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven H. Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
25
|
Hendricksen AT, Ezzatpour S, Pulukuri AJ, Ryan AT, Flanagan TJ, Frantz W, Buchholz DW, Ortega V, Monreal IA, Sahler JM, Nielsen AE, Aguilar HC, Mancini RJ. Thermophobic Trehalose Glycopolymers as Smart C-Type Lectin Receptor Vaccine Adjuvants. Adv Healthc Mater 2023; 12:e2202918. [PMID: 37002787 PMCID: PMC11212414 DOI: 10.1002/adhm.202202918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/06/2023] [Indexed: 04/04/2023]
Abstract
Herein, this work reports the first synthetic vaccine adjuvants that attenuate potency in response to small, 1-2 °C changes in temperature about their lower critical solution temperature (LCST). Adjuvant additives significantly increase vaccine efficacy. However, adjuvants also cause inflammatory side effects, such as pyrexia, which currently limits their use. To address this, a thermophobic vaccine adjuvant engineered to attenuate potency at temperatures correlating to pyrexia is created. Thermophobic adjuvants are synthesized by combining a rationally designed trehalose glycolipid vaccine adjuvant with thermoresponsive poly-N-isoporpylacrylamide (NIPAM) via reversible addition fragmentation chain transfer (RAFT) polymerization. The resulting thermophobic adjuvants exhibit LCSTs near 37 °C, and self-assembled into nanoparticles with temperature-dependent sizes (90-270 nm). Thermophobic adjuvants activate HEK-mMINCLE and other innate immune cell lines as well as primary mouse bone marrow derived dendritic cells (BMDCs) and bone marrow derived macrophages (BMDMs). Inflammatory cytokine production is attenuated under conditions mimicking pyrexia (above the LCST) relative to homeostasis (37 °C) or below the LCST. This thermophobic behavior correlated with decreased adjuvant Rg is observed by DLS, as well as glycolipid-NIPAM shielding interactions are observed by NOESY-NMR. In vivo, thermophobic adjuvants enhance efficacy of a whole inactivated influenza A/California/04/2009 virus vaccine, by increasing neutralizing antibody titers and CD4+ /44+ /62L+ lung and lymph node central memory T cells, as well as providing better protection from morbidity after viral challenge relative to unadjuvanted control vaccine. Together, these results demonstrate the first adjuvants with potency regulated by temperature. This work envisions that with further investigation, this approach can enhance vaccine efficacy while maintaining safety.
Collapse
Affiliation(s)
- Aaron T. Hendricksen
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - Shahrzad Ezzatpour
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Anunay J. Pulukuri
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - Austin T. Ryan
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - Tatum J. Flanagan
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - William Frantz
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - David W. Buchholz
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Victoria Ortega
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Isaac A. Monreal
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Julie M. Sahler
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Amy E. Nielsen
- Astante Therapeutics Inc.120 N Pine Street, Suite 270ASpokaneWA99202USA
| | - Hector C. Aguilar
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Rock J. Mancini
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
- Department of Chemistry and BiochemistryMiami University651 E. High StreetOxfordOH45056USA
| |
Collapse
|
26
|
Parizi LF, Githaka NW, Logullo C, Zhou J, Onuma M, Termignoni C, da Silva Vaz I. Universal Tick Vaccines: Candidates and Remaining Challenges. Animals (Basel) 2023; 13:2031. [PMID: 37370541 DOI: 10.3390/ani13122031] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/29/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Recent advancements in molecular biology, particularly regarding massively parallel sequencing technologies, have enabled scientists to gain more insight into the physiology of ticks. While there has been progress in identifying tick proteins and the pathways they are involved in, the specificities of tick-host interaction at the molecular level are not yet fully understood. Indeed, the development of effective commercial tick vaccines has been slower than expected. While omics studies have pointed to some potential vaccine immunogens, selecting suitable antigens for a multi-antigenic vaccine is very complex due to the participation of redundant molecules in biological pathways. The expansion of ticks and their pathogens into new territories and exposure to new hosts makes it necessary to evaluate vaccine efficacy in unusual and non-domestic host species. This situation makes ticks and tick-borne diseases an increasing threat to animal and human health globally, demanding an urgent availability of vaccines against multiple tick species and their pathogens. This review discusses the challenges and advancements in the search for universal tick vaccines, including promising new antigen candidates, and indicates future directions in this crucial research field.
Collapse
Affiliation(s)
- Luís Fernando Parizi
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | | | - Carlos Logullo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Jinlin Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Misao Onuma
- Department of Infectious Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Carlos Termignoni
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre 90040-060, Brazil
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
- Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre 91540-000, Brazil
| |
Collapse
|
27
|
Singh R, Sharma R, Varshney R, Mal G, Ghosh M, Singh B. Evaluation of immunological adjuvant activities of saponin rich fraction from the fruits of Asparagus adscendens Roxb. with less adverse reactions. Drug Chem Toxicol 2023; 46:557-565. [PMID: 35484852 DOI: 10.1080/01480545.2022.2067170] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The hemolytic activity, in vitro as well as in vivo toxicity, and immunomodulatory potential of saponins-rich fraction of Asparagus adscendens Roxb. fruit (AA-SRF) have been assessed in this study in order to explore AA-SRF as an alternative safer adjuvant to standard Quil-A saponin. The AA-SRF showed lower hemolytic activity (HD50 = 301.01 ± 1.63 µg/ml) than Quil-A (HD50 = 17.15 ± 2.12 µg/ml). The sulforhodamine B assay also revealed that AA-SRF was less toxic to VERO cells (IC50≥200 ± 4.32 µg/ml) than Quil-A (IC50 = 60 ± 2.78 µg/ml). The AA-SRF did not lead to mortality in mice up to 1.6 mg and was much safer than Quil-A for in vivo use. Conversely, mice were subcutaneously immunized with OVA 100 μg alone or along with Alum (200 μg) or Quil-A (10 μg) or AA-SRF (50 μg/100 μg/200 μg) on days 0 and 14. The AA-SRF at 100 μg dose best supported the LPS/Con A primed splenocyte proliferation activity, elevated the serum OVA-specific total IgG antibody, IL-12, CD4 titer and upsurged CD3/CD19 expression in spleen as well as lymph node sections which in turn advocated its adjuvant potential. Thus, AA-SRF can be further studied for use as a safe alternative adjuvant in vaccines.
Collapse
Affiliation(s)
- Rahul Singh
- Animal Experimental Pathology, Central Ayurvedic Research Institute for Drug Development, CCRAS, Ministry of Ayush, Kolkata, India
| | - Rinku Sharma
- Disease Investigation Laboratory, ICAR-Indian Veterinary Research Institute, Regional Station, Palampur, India
| | - Rajat Varshney
- Department of Veterinary Microbiology, FVAS, IAS, RGSC, Banaras Hindu University, Mirzapur, India
| | - Gorakh Mal
- Disease Investigation Laboratory, ICAR-Indian Veterinary Research Institute, Regional Station, Palampur, India
| | - Mayukh Ghosh
- Department of Veterinary Physiology and Biochemistry, FVAS, IAS, RGSC, Banaras Hindu University, Mirzapur, India
| | - Birbal Singh
- Disease Investigation Laboratory, ICAR-Indian Veterinary Research Institute, Regional Station, Palampur, India
| |
Collapse
|
28
|
Villarraza J, Fuselli A, Gugliotta A, Garay E, Rodríguez MC, Fontana D, Antuña S, Gastaldi V, Battagliotti JM, Tardivo MB, Alvarez D, Castro E, Cassataro J, Ceaglio N, Prieto C. A COVID-19 vaccine candidate based on SARS-CoV-2 spike protein and immune-stimulating complexes. Appl Microbiol Biotechnol 2023; 107:3429-3441. [PMID: 37093307 PMCID: PMC10124706 DOI: 10.1007/s00253-023-12520-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/21/2023] [Accepted: 04/04/2023] [Indexed: 04/25/2023]
Abstract
Spike protein from SARS-CoV-2, the etiologic agent of the COVID-19 pandemic disease, constitutes a structural protein that proved to be the main responsible for neutralizing antibody production. Thus, its sequence is highly considered for the design of candidate vaccines. Animal cell culture represents the best option for the production of subunit vaccines based on recombinant proteins since they introduce post-translational modifications that are important to mimic the natural antigenic epitopes. Particularly, the human cell line HEK293T has been explored and used for the production of biotherapeutics since the products derived from them present human-like post-translational modifications that are important for the protein's activity and immunogenicity. The aim of this study was to produce and characterize a potential vaccine for COVID-19 based on the spike ectodomain (S-ED) of SARS-CoV-2 and two different adjuvants: aluminum hydroxide (AH) and immune-stimulating complexes (ISCOMs). The S-ED was produced in sHEK293T cells using a 1-L stirred tank bioreactor operated in perfusion mode and purified. S-ED characterization revealed the expected size and morphology. High N-glycan content was confirmed. S-ED-specific binding with the hACE2 (human angiotensin-converting enzyme 2) receptor was verified. The immunogenicity of S-ED was evaluated using AH and ISCOMs. Both formulations demonstrated the presence of anti-RBD antibodies in the plasma of immunized mice, being significantly higher for the latter adjuvant. Also, higher levels of IFN-γ and IL-4 were detected after the ex vivo immune stimulation of spleen-derived MNCs from ISCOMs immunized mice. Further analysis confirmed that S-ED/ISCOMs elicit neutralizing antibodies against SARS-CoV-2. KEY POINTS: Trimeric SARS-CoV-2 S-ED was produced in stable recombinant sHEK cells in serum-free medium. A novel S-ED vaccine formulation induced potent humoral and cellular immunity. S-ED formulated with ISCOMs adjuvant elicited a highly neutralizing antibody titer.
Collapse
Affiliation(s)
- Javier Villarraza
- UNL, CONICET, FBCB, Centro Biotecnológico del Litoral, Santa Fe, Pcia., Santa Fe, Argentina
| | - Antonela Fuselli
- UNL, CONICET, FBCB, Centro Biotecnológico del Litoral, Santa Fe, Pcia., Santa Fe, Argentina
| | - Agustina Gugliotta
- UNL, CONICET, FBCB, Centro Biotecnológico del Litoral, Santa Fe, Pcia., Santa Fe, Argentina.
| | - Ernesto Garay
- UNL, CONICET, FBCB, Centro Biotecnológico del Litoral, Santa Fe, Pcia., Santa Fe, Argentina
| | | | - Diego Fontana
- Biotecnofe S.A. PTLC, Santa Fe, Pcia., Santa Fe, Argentina
- UNL, FBCB, Centro Biotecnológico del Litoral, Santa Fe, Pcia., Santa Fe, Argentina
| | | | - Victoria Gastaldi
- UNL, CONICET, FBCB, Centro Biotecnológico del Litoral, Santa Fe, Pcia., Santa Fe, Argentina
- Biotecnofe S.A. PTLC, Santa Fe, Pcia., Santa Fe, Argentina
| | | | | | - Diego Alvarez
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Eliana Castro
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Juliana Cassataro
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Natalia Ceaglio
- UNL, CONICET, FBCB, Centro Biotecnológico del Litoral, Santa Fe, Pcia., Santa Fe, Argentina
| | - Claudio Prieto
- Biotecnofe S.A. PTLC, Santa Fe, Pcia., Santa Fe, Argentina
- UNL, FBCB, Centro Biotecnológico del Litoral, Santa Fe, Pcia., Santa Fe, Argentina
- Cellargen Biotech SRL, Santa Fe, Pcia., Santa Fe, Argentina
| |
Collapse
|
29
|
Bonam SR, Hu H. Next-Generation Vaccines Against COVID-19 Variants: Beyond the Spike Protein. ZOONOSES (BURLINGTON, MASS.) 2023; 3:10.15212/zoonoses-2023-0003. [PMID: 38031548 PMCID: PMC10686570 DOI: 10.15212/zoonoses-2023-0003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Vaccines are among the most effective medical countermeasures against infectious diseases. The current Coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spurred the scientific strategies to fight against the disease. Since 2020, a great number of vaccines based on different platforms have been in development in response to the pandemic, among which mRNA, adenoviral vector, and subunit vaccines have been clinically approved for use in humans. These first-generation COVID-19 vaccines largely target the viral spike (S) protein and aim for eliciting potent neutralizing antibodies. With the emergence of SARS-CoV-2 variants, especially the highly transmissible Omicron strains, the S-based vaccine strategies have been faced constant challenges due to strong immune escape by the variants. The coronavirus nucleocapsid (N) is one of the viral proteins that induces strong T-cell immunity and is more conserved across different SARS-CoV-2 variants. Inclusion of N in the development of COVID-19 vaccines has been reported. Here, we briefly reviewed and discussed COVID-19 disease, current S-based vaccine strategies, and focused on the immunobiology of N protein in SARS-CoV-2 host immunity, as well as the next-generation vaccine strategies involving N protein, to combat current and emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA 77555
| | - Haitao Hu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA 77555
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA 77555
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA 77555
| |
Collapse
|
30
|
Zhao D, Chen X, Wang L, Zhang J, Zhao Z, Yue N, Zhu Y, Fei W, Li X, Tan L, He W. Bidirectional and persistent immunomodulation of Astragalus polysaccharide as an adjuvant of influenza and recombinant SARS-CoV-2 vaccine. Int J Biol Macromol 2023; 234:123635. [PMID: 36801224 PMCID: PMC9932796 DOI: 10.1016/j.ijbiomac.2023.123635] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023]
Abstract
Respiratory viral infections, such as coronavirus disease of 2019 (COVID-19) and influenza, cause significant morbidity and mortality and have become a worldwide public health concern with tremendous economic and societal burdens. Vaccination is a major strategy for preventing infections. However, some new vaccines have an unmet need for impairing responses in certain individuals, especially COVID-19 vaccines, despite ongoing vaccine and adjuvant research. Here, we evaluated the effectiveness of Astragalus polysaccharide (APS), a bioactive polysaccharide extracted from the traditional Chinese herb Astragalus membranaceus as an immune adjuvant to regulate the efficacy of influenza split vaccine (ISV) and recombinant severe acute respiratory syndrome (SARS)-Cov-2 vaccine in mice. Our data indicated that APS as an adjuvant can facilitate the induction of high levels of hemagglutination inhibition (HAI) titer and specific antibody immunoglobulin G (IgG) and confer protection against the lethal challenge of influenza A viruses, including increased survival and amelioration of weight loss in mice immunized with the ISV. RNA sequencing (RNA-seq) analysis revealed that the NF-κB and Fc gamma R-mediated phagocytosis signaling pathways are essential for the immune response of mice immunized with the recombinant SARS-Cov-2 vaccine (RSV). Another important finding was that bidirectional immunomodulation of APS on cellular and humoral immunity was observed, and APS-adjuvant-induced antibodies persisted at a high level for at least 20 weeks. These findings suggest that APS is a potent adjuvant for influenza and COVID-19 vaccines, and has the advantages of bidirectional immunoregulation and persistent immunity.
Collapse
Affiliation(s)
- Danping Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiuhong Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Linyuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| | - Jianjun Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Zhongpeng Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| | - Na Yue
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yingli Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wenting Fei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyu Li
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lingyun Tan
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Wei He
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
31
|
Hussain H, Ganesh A, Milane L, Amiji M. Lessons learned from the SARS-CoV-2 pandemic; from nucleic acid nanomedicines, to clinical trials, herd immunity, and the vaccination divide. Expert Opin Drug Deliv 2023; 20:489-506. [PMID: 36890642 DOI: 10.1080/17425247.2023.2189697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
INTRODUCTION In November 2019, the idea of a zoonotic virus crossing over to human transmission in a seafood market in Wuhan, China, and then soaring across the globe to claim over 6.3 million lives and persisting to date, seemed more like wild science fiction than a future reality. As the SARS-CoV-2 pandemic continues, it is important to hallmark the imprints the pandemic has made on science. AREAS COVERED This review covers the biology of SARS-CoV-2, vaccine formulations and trials, the concept of 'herd resistance,' and the vaccination divide. EXPERT OPINION The SARS-CoV-2 pandemic has changed the landscape of medicine. The rapid approval of SARS-CoV-2 vaccines has changed the culture of drug development and clinical approvals. This change is already leading to more accelerated trials. The RNA vaccines have opened the market for nucleic acid therapies and the applications are limitless - from cancer to influenza. A phenomenon that has occurred is that the low efficacy of current vaccines and the rapid mutation rate of the virus is preventing herd immunity from being attained. Instead, herd resistance is being acquired. Even with future, more effective vaccines, anti-vaccination attitudes will continue to challenge the quest for SARS-CoV-2 herd immunity.
Collapse
Affiliation(s)
| | - Aishwarya Ganesh
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Lara Milane
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| |
Collapse
|
32
|
Swetha K, Kotla NG, Tunki L, Jayaraj A, Bhargava SK, Hu H, Bonam SR, Kurapati R. Recent Advances in the Lipid Nanoparticle-Mediated Delivery of mRNA Vaccines. Vaccines (Basel) 2023; 11:658. [PMID: 36992242 PMCID: PMC10059764 DOI: 10.3390/vaccines11030658] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/10/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023] Open
Abstract
Lipid nanoparticles (LNPs) have recently emerged as one of the most advanced technologies for the highly efficient in vivo delivery of exogenous mRNA, particularly for COVID-19 vaccine delivery. LNPs comprise four different lipids: ionizable lipids, helper or neutral lipids, cholesterol, and lipids attached to polyethylene glycol (PEG). In this review, we present recent the advances and insights for the design of LNPs, as well as their composition and properties, with a subsequent discussion on the development of COVID-19 vaccines. In particular, as ionizable lipids are the most critical drivers for complexing the mRNA and in vivo delivery, the role of ionizable lipids in mRNA vaccines is discussed in detail. Furthermore, the use of LNPs as effective delivery vehicles for vaccination, genome editing, and protein replacement therapy is explained. Finally, expert opinion on LNPs for mRNA vaccines is discussed, which may address future challenges in developing mRNA vaccines using highly efficient LNPs based on a novel set of ionizable lipids. Developing highly efficient mRNA delivery systems for vaccines with improved safety against some severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants remains difficult.
Collapse
Affiliation(s)
- K. Swetha
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India
| | - Niranjan G. Kotla
- Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India
| | - Lakshmi Tunki
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Centre for Advanced Materials and Industrial Chemistry (CAMIC), School of Science, RMIT University, Melbourne, VIC 3001, Australia
| | - Arya Jayaraj
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India
| | - Suresh K. Bhargava
- Centre for Advanced Materials and Industrial Chemistry (CAMIC), School of Science, RMIT University, Melbourne, VIC 3001, Australia
| | - Haitao Hu
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India
| |
Collapse
|
33
|
Yi Y, Yu M, Li W, Zhu D, Mei L, Ou M. Vaccine-like nanomedicine for cancer immunotherapy. J Control Release 2023; 355:760-778. [PMID: 36822241 DOI: 10.1016/j.jconrel.2023.02.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023]
Abstract
The successful clinical application of immune checkpoint blockade (ICB) and chimeric antigen receptor T cells (CAR-T) therapeutics has attracted extensive attention to immunotherapy, however, their drawbacks such as limited specificity, persistence and toxicity haven't met the high expectations on efficient cancer treatments. Therapeutic cancer vaccines which instruct the immune system to capture tumor specific antigens, generate long-term immune memory and specifically eliminate cancer cells gradually become the most promising strategies to eradicate tumor. However, the disadvantages of some existing vaccines such as weak immunogenicity and in vivo instability have restricted their development. Nanotechnology has been recently incorporated into vaccine fabrication and exhibited promising results for cancer immunotherapy. Nanoparticles promote the stability of vaccines, as well as enhance antigen recognition and presentation owing to their nanometer size which promotes internalization of antigens by phagocytic cells. The surface modification with targeting units further permits the delivery of vaccines to specific cells. Meanwhile, nanocarriers with adjuvant effect can improve the efficacy of vaccines. In addition to classic vaccines composed of antigens and adjuvants, the nanoparticle-mediated chemotherapy, radiotherapy and certain other therapeutics could induce the release of tumor antigens in situ, which therefore effectively simulate antitumor immune responses. Such vaccine-like nanomedicine not only kills primary tumors, but also prevents tumor recurrence and helps eliminate metastatic tumors. Herein, we introduce recent developments in nanoparticle-based delivery systems for antigen delivery and in situ antitumor vaccination. We will also discuss the remaining opportunities and challenges of nanovaccine in clinical translation towards cancer treatment.
Collapse
Affiliation(s)
- Yunfei Yi
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China; School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Meitong Ou
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
34
|
Verma SK, Mahajan P, Singh NK, Gupta A, Aggarwal R, Rappuoli R, Johri AK. New-age vaccine adjuvants, their development, and future perspective. Front Immunol 2023; 14:1043109. [PMID: 36911719 PMCID: PMC9998920 DOI: 10.3389/fimmu.2023.1043109] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/26/2023] [Indexed: 02/26/2023] Open
Abstract
In the present scenario, immunization is of utmost importance as it keeps us safe and protects us from infectious agents. Despite the great success in the field of vaccinology, there is a need to not only develop safe and ideal vaccines to fight deadly infections but also improve the quality of existing vaccines in terms of partial or inconsistent protection. Generally, subunit vaccines are known to be safe in nature, but they are mostly found to be incapable of generating the optimum immune response. Hence, there is a great possibility of improving the potential of a vaccine in formulation with novel adjuvants, which can effectively impart superior immunity. The vaccine(s) in formulation with novel adjuvants may also be helpful in fighting pathogens of high antigenic diversity. However, due to the limitations of safety and toxicity, very few human-compatible adjuvants have been approved. In this review, we mainly focus on the need for new and improved vaccines; the definition of and the need for adjuvants; the characteristics and mechanisms of human-compatible adjuvants; the current status of vaccine adjuvants, mucosal vaccine adjuvants, and adjuvants in clinical development; and future directions.
Collapse
Affiliation(s)
| | - Pooja Mahajan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Nikhlesh K. Singh
- Integrative Biosciences Center, Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI, United States
| | - Ankit Gupta
- Microbiology Division, Defence Research and Development Establishment, Gwalior, India
| | - Rupesh Aggarwal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | - Atul Kumar Johri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
35
|
Astragalus Saponins, Astragaloside VII and Newly Synthesized Derivatives, Induce Dendritic Cell Maturation and T Cell Activation. Vaccines (Basel) 2023; 11:vaccines11030495. [PMID: 36992079 DOI: 10.3390/vaccines11030495] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/04/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Astragaloside VII (AST VII), a triterpenic saponin isolated from Astragalus species, shows promise as a vaccine adjuvant, as it supported a balanced Th1/Th2 immune response in previous in vivo studies. However, the underlying mechanisms of its adjuvant activity have not been defined. Here, we investigated the impact of AST VII and its newly synthesized semi-synthetic analogs on human whole blood cells, as well as on mouse bone marrow-derived dendritic cells (BMDCs). Cells were stimulated with AST VII and its derivatives in the presence or absence of LPS or PMA/ionomycin and the secretion of cytokines and the expression of activation markers were analyzed using ELISA and flow cytometry, respectively. AST VII and its analogs increased the production of IL-1β in PMA/ionomycin-stimulated human whole blood cells. In LPS-treated mouse BMDCs, AST VII increased the production of IL-1β and IL-12, and the expression of MHC II, CD86, and CD80. In mixed leukocyte reaction, AST VII and derivatives increased the expression of the activation marker CD44 on mouse CD4+ and CD8+ T cells. In conclusion, AST VII and its derivatives strengthen pro-inflammatory responses and support dendritic cell maturation and T cell activation in vitro. Our results provide insights into the mechanisms of the adjuvant activities of AST VII and its analogs, which will be instrumental to improve their utility as a vaccine adjuvant.
Collapse
|
36
|
THEME: "Vaccines and Vaccine Adjuvants/Immunomodulators for Infectious Diseases". Vaccines (Basel) 2023; 11:vaccines11020383. [PMID: 36851261 PMCID: PMC9965514 DOI: 10.3390/vaccines11020383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 01/12/2023] [Indexed: 02/11/2023] Open
Abstract
The discovery of vaccines has enabled the successful prevention of many deadly infectious diseases, decreased the overall mortality rate, and improved life expectancy worldwide [...].
Collapse
|
37
|
Zong C, Wu Q, Shao T, Dong Z, Liu Q. Exploiting the anaerobic fermentation of alfalfa as a renewable source of squalene. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:221-232. [PMID: 35857393 DOI: 10.1002/jsfa.12134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/12/2022] [Accepted: 07/20/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The use of alfalfa is a promising response to the increasing demand for squalene. Ensiling could enhance the squalene content of fresh alfalfa and silage. To investigate and exploit the anaerobic fermentation of forage as a new squalene source, alfalfa was ensiled without (CON) or with molasses (ML) and sunflower seed oil (SSL) for 10, 40, and 70 days. RESULTS Naturally ensiled alfalfa was of poor quality but had up to 1.93 times higher squalene content (P < 0.001) than fresh alfalfa. The squalene-producing bacteria were found to be cocci lactic acid bacteria (LAB). Adding ML and SSL decreased squalene content (P = 0.002 and P < 0.001) by 6.89% and 11.6%, respectively. Multiple linear regression models and correlation analysis indicated that squalene synthase was the key enzyme for squalene synthesis. The addition of ML and SSL altered the structure of LAB communities, mainly decreasing the relative abundance of cocci LAB, which was responsible for squalene synthesis, and changing the fermentation products (lactic acid, propionic acid, and ammonia-N) influencing the squalene-related enzymes, thereby decreasing squalene production. Compared with squalene production from the reference bacteria (Pediococcus acidilactici Ch-2, Rhodopseudomonas palustris, Bacillus subtilis, engineered Escherichia coli), alfalfa silage had the potential to be a new squalene source. CONCLUSION Natural ensiled alfalfa was a promising source for squalene, and ensiling was a potential pathway to obtain novel high-yield squalene bacteria. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Cheng Zong
- Institute of Ensiling and Processing of Grass, College of Agro-grassland Science, Nanjing Agricultural University, Nanjing, China
| | - Qifeng Wu
- Institute of Ensiling and Processing of Grass, College of Agro-grassland Science, Nanjing Agricultural University, Nanjing, China
| | - Tao Shao
- Institute of Ensiling and Processing of Grass, College of Agro-grassland Science, Nanjing Agricultural University, Nanjing, China
| | - Zhihao Dong
- Institute of Ensiling and Processing of Grass, College of Agro-grassland Science, Nanjing Agricultural University, Nanjing, China
| | - Qinhua Liu
- Institute of Ensiling and Processing of Grass, College of Agro-grassland Science, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
38
|
Bonam SR, Platenburg PP, Bayry J. Retrospective analysis on the immunopotentiating mechanism of an emulsion-based vaccine adjuvant on human antigen presenting cells. Front Immunol 2023; 13:1086752. [PMID: 36700217 PMCID: PMC9868768 DOI: 10.3389/fimmu.2022.1086752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
We retrospectively analyzed the immunopotentiating mechanism of an oil-in-water (O/W) emulsion-based vaccine adjuvant LiteVax™ Adjuvant (LVA) that contains CMS (Maltose 4'-monosulphate 1,2,3,6,2',3',6'-heptadecanoic acid ester), squalane, Tween 80 in phosphate buffered saline. Despite being effective in animal models, the immunological mechanisms by which LVA exerts adjuvant function are not known. As dendritic cells (DC) are key for initiating and propagating the immune response, we have investigated the effect of LVA and of its components on the DC function. We show that CMS but not LVA significantly enhances the expression of DC activation-associated markers, cytokine secretion, and CD4+ T cell responses. On the other hand, CMS ZERO [non-sulphated sucrose fatty acid esters (ZERO)], used as a control, had no such activity. Our data identified the unique nature of CMS in LVA, and propose that LVA acts as a delivery system, and CMS acts as an immunostimulatory agent.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche desCordeliers, Sorbonne Université, Université de Paris, Paris, France
| | | | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche desCordeliers, Sorbonne Université, Université de Paris, Paris, France,Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad, India,*Correspondence: Jagadeesh Bayry, ;
| |
Collapse
|
39
|
Ko KH, Cha SB, Lee SH, Bae HS, Ham CS, Lee MG, Kim DH, Han SH. A novel defined TLR3 agonist as an effective vaccine adjuvant. Front Immunol 2023; 14:1075291. [PMID: 36761735 PMCID: PMC9902914 DOI: 10.3389/fimmu.2023.1075291] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/06/2023] [Indexed: 01/25/2023] Open
Abstract
Synthetic double-stranded RNA analogs recognized by Toll-like receptor 3 (TLR3) are an attractive adjuvant candidate for vaccines, especially against intracellular pathogens or tumors, because of their ability to enhance T cell and antibody responses. Although poly(I:C) is a representative dsRNA with potent adjuvanticity, its clinical application has been limited due to heterogeneous molecular size, inconsistent activity, poor stability, and toxicity. To overcome these limitations, we developed a novel dsRNA-based TLR3 agonist named NexaVant (NVT) by using PCR-coupled bidirectional in vitro transcription. Agarose gel electrophoresis and reverse phase-HPLC analysis demonstrated that NVT is a single 275-kDa homogeneous molecule. NVT appears to be stable since its appearance, concentration, and molecular size were unaffected under 6 months of accelerated storage conditions. Moreover, preclinical evaluation of toxicity under good laboratory practices showed that NVT is a safe substance without any signs of serious toxicity. NVT stimulated TLR3 and increased the expression of viral nucleic acid sensors TLR3, MDA-5, and RIG-1. When intramuscularly injected into C57BL/6 mice, ovalbumin (OVA) plus NVT highly increased the migration of dendritic cells (DCs), macrophages, and neutrophils into inguinal lymph node (iLN) compared with OVA alone. In addition, NVT substantially induced the phenotypic markers of DC maturation and activation including MHC-II, CD40, CD80, and CD86 together with IFN-β production. Furthermore, NVT exhibited an appropriate adjuvanticity because it elevated OVA-specific IgG, in particular, higher levels of IgG2c (Th1-type) but lower IgG1 (Th2-type). Concomitantly, NVT increased the levels of Th1-type T cells such as IFN-γ+CD4+ and IFN-γ+CD8+ cells in response to OVA stimulation. Collectively, we suggest that NVT with appropriate safety and effectiveness is a novel and promising adjuvant for vaccines, especially those requiring T cell mediated immunity such as viral and cancer vaccines.
Collapse
Affiliation(s)
- Kwang Hyun Ko
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea.,Interdisciplinary Program in Genetic Engineering, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seung Bin Cha
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Seung-Hwan Lee
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Hyun Shik Bae
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Chul Soo Ham
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Min-Gyu Lee
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Dong-Ho Kim
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Seung Hyun Han
- Interdisciplinary Program in Genetic Engineering, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea.,Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
40
|
Tong Y, Lu G, Wang Z, Hao S, Zhang G, Sun H. Tubeimuside I improves the efficacy of a therapeutic Fusobacterium nucleatum dendritic cell-based vaccine against colorectal cancer. Front Immunol 2023; 14:1154818. [PMID: 37207216 PMCID: PMC10189021 DOI: 10.3389/fimmu.2023.1154818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/24/2023] [Indexed: 05/21/2023] Open
Abstract
Introduction Fusobacterium nucleatum (F. nucleatum) infection has been confirmed to be associated with the development, chemoresistance, and immune evasion of colorectal cancer (CRC). The complex relationship between the microorganism, host cells, and the immune system throughout all stages of CRC progression, which makes the development of new therapeutic methods difficult. Methods We developed a new dendritic cell (DC) vaccine to investigate the antitumor efficacy of CRC immunotherapy strategies. By mediating a specific mode of interaction between the bacteria, tumor, and host, we found a new plant-derived adjuvant, tubeimuside I (TBI), which simultaneously improved the DC vaccine efficacy and inhibited the F. nucleatum infection. Encapsulating TBI in a nanoemulsion greatly improved the drug efficacy and reduced the drug dosage and administration times. Results The nanoemulsion encapsulated TBI DC vaccine exhibited an excellent antibacterial and antitumor effect and improved the survival rate of CRC mice by inhibiting tumor development and progression. Discussion In this study, we provide a effective strategy for developing a DC-based vaccine against CRC and underlies the importance of further understanding the mechanism of CRC processes caused by F. nucleatum.
Collapse
Affiliation(s)
- Yanan Tong
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, China
| | - Guoxiu Lu
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, China
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
| | - Zhiguo Wang
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, China
| | - Shanhu Hao
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, China
| | - Guoxu Zhang
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, China
- *Correspondence: Guoxu Zhang, ; Hongwu Sun,
| | - Hongwu Sun
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
- *Correspondence: Guoxu Zhang, ; Hongwu Sun,
| |
Collapse
|
41
|
Lozano JM, Muller S. Monkeypox: potential vaccine development strategies. Trends Pharmacol Sci 2023; 44:15-19. [PMID: 36563658 PMCID: PMC9730257 DOI: 10.1016/j.tips.2022.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/29/2022] [Accepted: 10/31/2022] [Indexed: 12/13/2022]
Abstract
A multicountry outbreak of monkeypox has gained global attention. Basic research including structural and immunological investigation on monkeypox virus (MPXV) is central to design effective solutions of treatment with antivirals and appropriate vaccines. We summarize some information about this virus and its re-emergence and the current vaccines that are proposed to limit its spread and present some possible avenues for developing new vaccines.
Collapse
Affiliation(s)
- José Manuel Lozano
- Universidad Nacional de Colombia-Sede Bogotá, Departamento de Farmacia, Mimetismo molecular de los Agentes infecciosos, Bogotá, DC, Colombia.
| | - Sylviane Muller
- Centre National de la Recherche scientifique-Université de Strasbourg, Biotechnology and Cell Signalling Unit, Neuroimmunology and Peptide Therapeutics Team, Strasbourg Drug Discovery and Development Institute, Strasbourg, France; University of Strasbourg Institute for Advanced Study, Strasbourg, France; Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
42
|
Nitin S, Srinivasa R. B, Monica MS, Thyago H. C. Incursions by severe acute respiratory syndrome coronavirus-2 on the host anti-viral immunity during mild, moderate, and severe coronavirus disease 2019 disease. EXPLORATION OF MEDICINE 2022. [DOI: 10.37349/ei.2022.00084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection in the human host can lead to various clinical manifestations, from symptomless carriers to mild to moderate to severe/critical illness. Therefore, the clinical classification of SARS-CoV-2 disease, based on severity, is a reliable way to predict disease states in SARS-CoV-2 infection. Recent studies on genomics, transcriptomics, epigenomics, and immunogenomics, along with spatial analysis of immune cells have delineated and defined the categorization of these disease groups using these high throughout technologies. These technologies hold the promise of providing not only a detailed but a holistic view of SARS-CoV-2-led pathogenesis. The main genomic, cellular, and immunologic features of each disease category, and what separates them spatially and molecularly are discussed in this brief review to provide a foundational spatial understanding of SARS-CoV-2 immunopathogenesis.
Collapse
Affiliation(s)
- Saksena Nitin
- Institute for Health and Sport, Victoria University, Footscray Campus, Melbourne VIC. 3011, Australia; Aegros Therapeutics Pty Ltd, Macquarie Park, Sydney 2019, Australia
| | - Bonam Srinivasa R.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Miranda-Saksena Monica
- Westmead Institute of Medical Research (WIMR), Herpes Virus Laboratory, Sydney 2145, Australia
| | - Cardoso Thyago H.
- OMICS Centre of Excellence, G42 Healthcare, Mazdar City, Abu Dhabi 3079, United Arab Emirates
| |
Collapse
|
43
|
Kim H, Yu J, Bai D, Nahm MH, Wang P. Potentiating pneumococcal glycoconjugate vaccine PCV13 with saponin adjuvant VSA-1. Front Immunol 2022; 13:1079047. [PMID: 36578488 PMCID: PMC9790987 DOI: 10.3389/fimmu.2022.1079047] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
VSA-1 is a semisynthetic saponin adjuvant prepared from naturally occurring Momordica saponin and capable of stimulating antigen-specific humoral and cellular immune responses. Its immunostimulating activity in enhancing the immune responses induced by the clinical glycoconjugate pneumococcal vaccine PCV13 is compared with QS-21 in female BALB/c mice. Both VSA-1 and QS-21 boosted IgG and opsonic antibodies titers against seven selected serotypes, including serotypes 3, 14, and 19A that are involved in most PCV13 breakthroughs. Since VSA-1 is much more accessible and of lower toxicity than QS-21, it can be a practical saponin immunostimulant to be included in a new glycoconjugate pneumococcal vaccine formulation.
Collapse
Affiliation(s)
- Hyunjung Kim
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jigui Yu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Di Bai
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Moon H. Nahm
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States,World Health Organization (WHO) Pneumococcal Serology Reference Laboratory, University of Alabama at Birmingham, Birmingham, AL, United States,*Correspondence: Moon H. Nahm, ; Pengfei Wang,
| | - Pengfei Wang
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, United States,*Correspondence: Moon H. Nahm, ; Pengfei Wang,
| |
Collapse
|
44
|
Wang K, Zhang T, Liu M, Wang D, Zhu H, Wang Z, Yu F, Liu Y, Zhao W. Synthesis and immunological evaluation of Mincle ligands-based antitumor vaccines. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.108065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
45
|
Campos DMDO, Silva MKD, Barbosa ED, Leow CY, Fulco UL, Oliveira JIN. Exploiting reverse vaccinology approach for the design of a multiepitope subunit vaccine against the major SARS-CoV-2 variants. Comput Biol Chem 2022; 101:107754. [PMID: 36037724 PMCID: PMC9385604 DOI: 10.1016/j.compbiolchem.2022.107754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 11/03/2022]
Abstract
The current COVID-19 pandemic, an infectious disease caused by the novel coronavirus (SARS-CoV-2), poses a threat to global health because of its high rate of spread and death. Currently, vaccination is the most effective method to prevent the spread of this disease. In the present study, we developed a novel multiepitope vaccine against SARS-CoV-2 containing Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (BA.1) variants. To this end, we performed a robust immunoinformatics approach based on multiple epitopes of the four structural proteins of SARS-CoV-2 (S, M, N, and E) from 475 SARS-CoV-2 genomes sequenced from the regions with the highest number of registered cases, namely the United States, India, Brazil, France, Germany, and the United Kingdom. To investigate the best immunogenic epitopes for linear B cells, cytotoxic T lymphocytes (CTL), and helper T lymphocytes (HTL), we evaluated antigenicity, allergenicity, conservation, immunogenicity, toxicity, human population coverage, IFN-inducing, post-translational modifications, and physicochemical properties. The tertiary structure of a vaccine prototype was predicted, refined, and validated. Through docking experiments, we evaluated its molecular coupling to the key immune receptor Toll-Like Receptor 3 (TLR3). To improve the quality of docking calculations, quantum mechanics/molecular mechanics calculations (QM/MM) were used, with the QM part of the simulations performed using the density functional theory formalism (DFT). Cloning and codon optimization were performed for the successful expression of the vaccine in E. coli. Finally, we investigated the immunogenic properties and immune response of our SARS-CoV-2 multiepitope vaccine. The results of the simulations show that administering our prototype three times significantly increases the antibody response and decreases the amount of antigens. The proposed vaccine candidate should therefore be tested in clinical trials for its efficacy in neutralizing SARS-CoV-2.
Collapse
Affiliation(s)
- Daniel Melo de Oliveira Campos
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, 59064-741, Natal/RN, Brazil.
| | - Maria Karolaynne da Silva
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, 59064-741, Natal/RN, Brazil.
| | - Emmanuel Duarte Barbosa
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, 59064-741, Natal/RN, Brazil.
| | | | - Umberto Laino Fulco
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, 59064-741, Natal/RN, Brazil.
| | - Jonas Ivan Nobre Oliveira
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, 59064-741, Natal/RN, Brazil.
| |
Collapse
|
46
|
Enhancing the Effect of Nucleic Acid Vaccines in the Treatment of HPV-Related Cancers: An Overview of Delivery Systems. Pathogens 2022; 11:pathogens11121444. [PMID: 36558778 PMCID: PMC9781236 DOI: 10.3390/pathogens11121444] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
Prophylactic vaccines against human papillomavirus (HPV) have proven efficacy in those who have not been infected by the virus. However, they do not benefit patients with established tumors. Therefore, the development of therapeutic options for HPV-related malignancies is critical. Third-generation vaccines based on nucleic acids are fast and simple approaches to eliciting adaptive immune responses. However, techniques to boost immunogenicity, reduce degradation, and facilitate their capture by immune cells are frequently required. One option to overcome this constraint is to employ delivery systems that allow selective antigen absorption and help modulate the immune response. This review aimed to discuss the influence of these different systems on the response generated by nucleic acid vaccines. The results indicate that delivery systems based on lipids, polymers, and microorganisms such as yeasts can be used to ensure the stability and transport of nucleic acid vaccines to their respective protein synthesis compartments. Thus, in view of the limitations of nucleic acid-based vaccines, it is important to consider the type of delivery system to be used-due to its impact on the immune response and desired final effect.
Collapse
|
47
|
Kim H, Bai D, Ghosh S, Franks ML, Wang X, Yan C, Liu Z, Zhang P, Michalek SM, Leavenworth JW, Wang P. Structure-Activity Relationship Study of Momordica Saponin II Derivatives as Vaccine Adjuvants. J Med Chem 2022; 65:14589-14598. [PMID: 36318612 PMCID: PMC10202417 DOI: 10.1021/acs.jmedchem.2c01087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
VSA-2 is a recently developed semisynthetic saponin immunostimulant. It is prepared by incorporating a terminal-functionalized side chain to the branched trisaccharide domain at the C3 position of Momordica saponin II (MS II) isolated from the seeds of perennial Momordica cochinchinensis Spreng. Direct comparison of VSA-2 and the clinically proven saponin adjuvant QS-21 shows that VSA-2 is comparable to QS-21 in enhancing humoral and cellular immune responses. Structure-activity relationship studies show that structural changes in the side chain have a significant impact on saponins' adjuvant activity. However, with the VSA-2 molecular framework intact, the new VSA-2 analogues with various substitution(s) at the terminal benzyl group of the side chain retain the ability of potentiating antigen-specific humoral and cellular responses.
Collapse
Affiliation(s)
- Hyunjung Kim
- Department of Chemistry, University of Alabama at Birmingham, 901 14th Street South, Birmingham, Alabama 35294, United States
| | - Di Bai
- Department of Chemistry, University of Alabama at Birmingham, 901 14th Street South, Birmingham, Alabama 35294, United States
| | - Sadashib Ghosh
- Department of Neurosurgery, University of Alabama at Birmingham, 901 14th Street South, Birmingham, Alabama 35294, United States
| | - Michael L Franks
- Department of Neurosurgery, University of Alabama at Birmingham, 901 14th Street South, Birmingham, Alabama 35294, United States
| | - Xifeng Wang
- Department of Chemistry, Central China Normal University, 152 Luoyu Road, Wuhan, Hubei 430079, P. R. China
| | - Cheng Yan
- Department of Chemistry, Central China Normal University, 152 Luoyu Road, Wuhan, Hubei 430079, P. R. China
| | - Zheng Liu
- Department of Chemistry, Central China Normal University, 152 Luoyu Road, Wuhan, Hubei 430079, P. R. China
| | - Ping Zhang
- Department of Pediatric Dentistry, University of Alabama at Birmingham, 901 14th Street South, Birmingham, Alabama 35294, United States
| | - Suzanne M Michalek
- Department of Microbiology, University of Alabama at Birmingham, 901 14th Street South, Birmingham, Alabama 35294, United States
| | - Jianmei W Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, 901 14th Street South, Birmingham, Alabama 35294, United States
- Department of Microbiology, University of Alabama at Birmingham, 901 14th Street South, Birmingham, Alabama 35294, United States
- The O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, 901 14th Street South, Birmingham, Alabama 35294, United States
| | - Pengfei Wang
- Department of Chemistry, University of Alabama at Birmingham, 901 14th Street South, Birmingham, Alabama 35294, United States
- The O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, 901 14th Street South, Birmingham, Alabama 35294, United States
| |
Collapse
|
48
|
Ye L, Zhang Y, Wu S, Wang Z, Liu F, Wang C, Hu M. Immunoprotection Efficacy of Con A-Purified Proteins against Haemonchus contortus in Goats. Vaccines (Basel) 2022; 10:vaccines10111891. [PMID: 36366399 PMCID: PMC9696691 DOI: 10.3390/vaccines10111891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Parasitic nematodes are important pathogens that infect animals, causing significant economic losses globally. Current repeated treatments have led to widespread anthelmintic resistance in nematode populations, so vaccine development offers an alternative control approach. However, only one effective vaccine (named Barbervax) has been developed to protect animals against one of the most pathogenic nematodes of ruminants—Haemonchus contortus (the barber’s pole worm). This vaccine contains a dominant component, Concanavalin A (Con A) purified H11 antigen, which has been shown to induce high levels (>85%) of immune protection in sheep breeds, but in goat breeds, the immunoprotection test of this native protein is still lacking. Here, we evaluated the protective efficacy of low-dose Con A-purified proteins for controlling the H. contortus infection in goats. Four-month-old Boer goats were equally divided into two vaccinated groups of 5 μg and 10 μg native proteins, and one adjuvant control. Each goat was immunized subcutaneously thrice and then challenged with 7000 infective third-stage larvae (L3s). The fecal egg count (FEC), degree of anemia, antibody levels of serum and abomasum mucosa, as well as worm burdens, were detected in experimental goats. Our results showed that both 5 μg and 10 μg vaccinated groups induced the effective protection in goats, reduced mean FEC by 71.8% and 68.6%, and mean worm burdens by 69.8% and 61.6%, respectively, compared to the adjuvant control. In addition, we detected that the serum antibody responses to the Con A-purified proteins were dominated by the IgG subtype, but the mucosal antibody responses were not detected. These data demonstrate Con A-purified proteins induced effective immunoprotection in goats, and underline their significance for controlling this widespread parasite.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Min Hu
- Correspondence: (C.W.); (M.H.)
| |
Collapse
|
49
|
Del Bino L, Østerlid KE, Wu DY, Nonne F, Romano MR, Codée J, Adamo R. Synthetic Glycans to Improve Current Glycoconjugate Vaccines and Fight Antimicrobial Resistance. Chem Rev 2022; 122:15672-15716. [PMID: 35608633 PMCID: PMC9614730 DOI: 10.1021/acs.chemrev.2c00021] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Antimicrobial resistance (AMR) is emerging as the next potential pandemic. Different microorganisms, including the bacteria Acinetobacter baumannii, Clostridioides difficile, Escherichia coli, Enterococcus faecium, Klebsiella pneumoniae, Neisseria gonorrhoeae, Pseudomonas aeruginosa, non-typhoidal Salmonella, and Staphylococcus aureus, and the fungus Candida auris, have been identified by the WHO and CDC as urgent or serious AMR threats. Others, such as group A and B Streptococci, are classified as concerning threats. Glycoconjugate vaccines have been demonstrated to be an efficacious and cost-effective measure to combat infections against Haemophilus influenzae, Neisseria meningitis, Streptococcus pneumoniae, and, more recently, Salmonella typhi. Recent times have seen enormous progress in methodologies for the assembly of complex glycans and glycoconjugates, with developments in synthetic, chemoenzymatic, and glycoengineering methodologies. This review analyzes the advancement of glycoconjugate vaccines based on synthetic carbohydrates to improve existing vaccines and identify novel candidates to combat AMR. Through this literature survey we built an overview of structure-immunogenicity relationships from available data and identify gaps and areas for further research to better exploit the peculiar role of carbohydrates as vaccine targets and create the next generation of synthetic carbohydrate-based vaccines.
Collapse
Affiliation(s)
| | - Kitt Emilie Østerlid
- Leiden
Institute of Chemistry, Leiden University, 2300 RA Leiden, The Netherlands
| | - Dung-Yeh Wu
- Leiden
Institute of Chemistry, Leiden University, 2300 RA Leiden, The Netherlands
| | | | | | - Jeroen Codée
- Leiden
Institute of Chemistry, Leiden University, 2300 RA Leiden, The Netherlands
| | | |
Collapse
|
50
|
Islam SI, Sanjida S, Ahmed SS, Almehmadi M, Allahyani M, Aljuaid A, Alsaiari AA, Halawi M. Core Proteomics and Immunoinformatic Approaches to Design a Multiepitope Reverse Vaccine Candidate against Chagas Disease. Vaccines (Basel) 2022; 10:vaccines10101669. [PMID: 36298534 PMCID: PMC9607777 DOI: 10.3390/vaccines10101669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/23/2022] [Accepted: 10/02/2022] [Indexed: 11/05/2022] Open
Abstract
Chagas disease is a tropical ailment indigenous to South America and caused by the protozoan parasite Trypanosoma cruzi, which has serious health consequences globally. Insect vectors transmit the parasite and, due to the lack of vaccine availability and limited treatment options, we implemented an integrated core proteomics analysis to design a reverse vaccine candidate based on immune epitopes for disease control. Firstly, T. cruzi core proteomics was used to identify immunodominant epitopes. Therefore, we designed the vaccine sequence to be non-allergic, antigenic, immunogenic, and to have better solubility. After predicting the tertiary structure, docking and molecular dynamics simulation (MDS) were performed with TLR4, MHC-I, and MHC-II receptors to discover the binding affinities. The final vaccine design demonstrated significant hydrogen bond interactions upon docking with TLR4, MHC-I, and MHC-II receptors. This indicated the efficacy of the vaccine candidate. A server-based immune simulation approach was generated to predict the efficacy. Significant structural compactness and binding stability were found based on MDS. Finally, by optimizing codons on Escherichia coli K12, a high GC content and CAI value were obtained, which were then incorporated into the cloning vector pET2+ (a). Thus, the developed vaccine sequence may be a viable therapy option for Chagas disease.
Collapse
Affiliation(s)
- Sk Injamamul Islam
- The International Graduate Program of Veterinary Science and Technology (VST), Department of Veterinary Microbiology, Faculty of Veterinary Science and Technology, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: or
| | - Saloa Sanjida
- Department of Environmental Science and Technology, Faculty of Applied Science and Technology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Sheikh Sunzid Ahmed
- Department of Botany, Faculty of Biological Sciences, University of Dhaka, Dhaka 1000, Bangladesh
| | - Mazen Almehmadi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Mamdouh Allahyani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Abdulelah Aljuaid
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Ahad Amer Alsaiari
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Mustafa Halawi
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Jazan 54943, Saudi Arabia
| |
Collapse
|