1
|
Chege H, Githigia S, Gathumbi J, Chege N, Ojuok R, Odaba J, Mwalimu S, Oboge H, Steinaa L, Nene V, Lacasta A. An Improved Theileria parva Sporozoite Seroneutralization Assay for the Identification of East Coast Fever Immune Correlates. Antibodies (Basel) 2024; 13:100. [PMID: 39727483 DOI: 10.3390/antib13040100] [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: 09/04/2024] [Revised: 11/04/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Immune correlates of protection are ideal tools to predict treatment or vaccine efficacy. However, the accuracy of the immune correlate and the capability to robustly predict the outcome of a vaccine candidate are determined by the performance of the in vitro immunoassay used. Several Theileria parva sporozoite seroneutralization assays have previously been used to assess antibody functional activities; however, a common limitation has been the need for fresh material, target cells and sporozoites, and operator-to-operator bias. An improved assay represents a positive step toward overcoming challenges associated with variability and it might provide a more reliable means of establishing an immune correlate with protection after sub-unit vaccine administration. METHODS Herein, we describe key improvements, among them, (1) the use of frozen parasites and target cells to avoid batch-to-batch variations and (2) the development of a new assay read-out based on the detection of infected cells through flow cytometry, instead of the use of Giemsa staining and microscopic evaluation, in order to improve the reproducibility of the results. RESULTS The improved seroneutralization assay is not only able to detect the individual neutralizing capacity of antibodies; it also detects the additive effect of antibody combinations. CONCLUSIONS This effect is described for the first time in Theileria parva and is of great interest for new antigen discovery and/or the epitope discovery of already known antigens like p67, opening a new avenue for the identification of ECF immune correlates of protection and the in vitro down-selection of new Theileria parva vaccine candidates, thereby contributing to reducing the use of animals in challenge experiments.
Collapse
Affiliation(s)
- Hannah Chege
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
- Department of Veterinary Pathology, Microbiology and Parasitology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - Samuel Githigia
- Department of Veterinary Pathology, Microbiology and Parasitology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - James Gathumbi
- Department of Veterinary Pathology, Microbiology and Parasitology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - Naomi Chege
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Rose Ojuok
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Josiah Odaba
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Stephen Mwalimu
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Harriet Oboge
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
- Department of Veterinary Pathology, Microbiology and Parasitology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - Lucilla Steinaa
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Vishvanath Nene
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Anna Lacasta
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| |
Collapse
|
2
|
Mozafari A, Rahmani M, Yasini Nasab Y, Shahsavandi S, Jafari M, Salmanian AH. The heterologous expression of novel recombinant protein composed of HN and F moieties of Newcastle disease virus and immunogenicity evaluation in mouse model. IRANIAN JOURNAL OF MICROBIOLOGY 2024; 16:655-665. [PMID: 39534294 PMCID: PMC11551657 DOI: 10.18502/ijm.v16i5.16801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Background and Objectives The rapid spread of Newcastle disease (ND), driven by extensive commercial exchange in the poultry industry, necessitates urgent preventive measures. Although effective vaccines against the Newcastle disease virus (NDV) have been used since 1940, recent outbreaks and the limitations of current vaccines highlight the need for improved solutions. Advances in synthetic biology, reverse vaccinology, molecular biology, and recombinant DNA technology over the past 20 years have led to the development of recombinant vaccines, which offer enhanced protection and broader immunogenic coverage against NDV. This study aimed to express the immunogenic domains of Hemagglutinin Neuraminidase (HN) and Fusion (F) glycoproteins, linked to the heat-labile enterotoxin B subunit (LTB) bio-adjuvant, to develop an effective and reliable recombinant vaccine for NDV. Materials and Methods In this study, the L(HN)2F protein, composed of the LTB bio-adjuvant and the immunogenic regions of the doubled Hemagglutinin Neuraminidase (HN-HN) and Fusion (F) epitope, was expressed in Escherichia coli. Subcutaneous injection was used to evaluate the humoral immune response in mice and the result was compared with B1 vaccine. Results The induction of strong humoral immune responses proved the strong immunoreactivity of the recombinant protein. Conclusion The IgG elicited by the recombinant proteins was comparable to that of the commercial B1 vaccine against NDV, indicating its potential as a viable candidate for further development and evaluation as a recombinant vaccine against NDV.
Collapse
Affiliation(s)
- Atena Mozafari
- Department of Agricultural Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Mehregan Rahmani
- Department of Agricultural Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Yasaman Yasini Nasab
- Department of Agricultural Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Shahla Shahsavandi
- Razi Vaccine & Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Mahyat Jafari
- Department of Agricultural Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Ali Hatef Salmanian
- Department of Agricultural Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
3
|
Stump WH, Klingenberg HJ, Ott AC, Gonzales DM, Burns JM. Design and Evaluation of Chimeric Plasmodium falciparum Circumsporozoite Protein-Based Malaria Vaccines. Vaccines (Basel) 2024; 12:351. [PMID: 38675734 PMCID: PMC11053680 DOI: 10.3390/vaccines12040351] [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: 02/21/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Efficacy data on two malaria vaccines, RTS,S and R21, targeting Plasmodium falciparum circumsporozoite protein (PfCSP), are encouraging. Efficacy may be improved by induction of additional antibodies to neutralizing epitopes outside of the central immunodominant repeat domain of PfCSP. We designed four rPfCSP-based vaccines in an effort to improve the diversity of the antibody response. We also evaluated P. falciparum merozoite surface protein 8 (PfMSP8) as a malaria-specific carrier protein as an alternative to hepatitis B surface antigen. We measured the magnitude, specificity, subclass, avidity, durability, and efficacy of vaccine-induced antibodies in outbred CD1 mice. In comparison to N-terminal- or C-terminal-focused constructs, immunization with near full-length vaccines, rPfCSP (#1) or the chimeric rPfCSP/8 (#2), markedly increased the breadth of B cell epitopes recognized covering the N-terminal domain, junctional region, and central repeat. Both rPfCSP (#1) and rPfCSP/8 (#2) also elicited a high proportion of antibodies to conformation-dependent epitopes in the C-terminus of PfCSP. Fusion of PfCSP to PfMSP8 shifted the specificity of the T cell response away from PfCSP toward PfMSP8 epitopes. Challenge studies with transgenic Plasmodium yoelii sporozoites expressing PfCSP demonstrated high and consistent sterile protection following rPfCSP/8 (#2) immunization. Of note, antibodies to conformational C-terminal epitopes were not required for protection. These results indicate that inclusion of the N-terminal domain of PfCSP can drive responses to protective, repeat, and non-repeat B cell epitopes and that PfMSP8 is an effective carrier for induction of high-titer, durable anti-PfCSP antibodies.
Collapse
Affiliation(s)
| | | | | | | | - James M. Burns
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA; (W.H.S.); (H.J.K.); (A.C.O.); (D.M.G.)
| |
Collapse
|
4
|
Pourhashem Z, Nourani L, Sani JJ, Yousefi H, Pirahmadi S, Sabouri M, Raz A, Djadid ND, Zakeri S, Mehrizi AA. Evaluation of a new fusion antigen, cd loop and HAP2-GCS1 domain (cd-HAP) of Plasmodium falciparum Generative Cell Specific 1 antigen formulated with various adjuvants, as a transmission blocking vaccine. Malar J 2023; 22:374. [PMID: 38071314 PMCID: PMC10710725 DOI: 10.1186/s12936-023-04798-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Malaria is a major global health challenge, and for the elimination and eradication of this disease, transmission-blocking vaccines (TBVs) are a priority. Plasmodium falciparum Generative Cell Specific 1 (PfGCS1), a promising TBV candidate, is essential for gamete fertilization. The HAP2-GCS1 domain of this antigen as well as its cd loop could induce antibodies that partially inhibit transmission of P. falciparum. METHODS In the current study, a new synthetic fusion antigen containing cd loop and HAP2-GCS1 domain (cd-HAP) of PfGCS1 was evaluated as a transmission blocking vaccine candidate. Initially, the profile of naturally acquired IgG antibodies to the cd-HAP antigen was analysed in Iranian individuals infected with P. falciparum, to confirm that this new fusion protein has the appropriate structure containing common epitopes with the native form of PfGCS1. Then, the immunogenicity of cd-HAP was evaluated in BALB/c mice, using different adjuvant systems such as CpG, MPL, QS-21, and a combination of them (CMQ). Furthermore, the blocking efficacy of polyclonal antibodies induced against these formulations was also assessed by oocyst intensity and infection prevalence in the Standard Membrane Feeding Assay (SMFA). RESULTS The naturally acquired antibodies (dominantly IgG1 and IgG3 subclasses) induced in P. falciparum-infected individuals could recognize the cd-HAP antigen which implies that the new fusion protein has a proper conformation that mimics the native structure of PfGCS1. Concerning the immunogenicity of cd-HAP antigen, the highest IgG levels and titers, by a Th1-type immune profile, and elevated antibody avidity were induced in mice immunized with the cd-HAP antigen formulated with a combination of adjuvants (P < 0.0001). Additionally, cytokine profiling of the immunized mice displayed that a high level of IFN-γ response, a Th1-type immune response, was produced by splenocytes from immunized mice that received cd-HAP antigen in combination with CMQ adjuvants (P < 0.0001). This formulation of cd-HAP antigen with CMQ adjuvants could reduce oocyst intensity and infection prevalence by 82%, evidenced by the SMFA and hold significant implications for future malaria vaccine development. CONCLUSION Altogether, the results showed that cd-HAP antigen formulated with a combination of the adjuvants (CMQ), could be a promising formulation to develop a PfGCS1-based transmission-blocking vaccine.
Collapse
Affiliation(s)
- Zeinab Pourhashem
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Leila Nourani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Jafar J Sani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Hemn Yousefi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Mobina Sabouri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Abbasali Raz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Navid Dinparast Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
5
|
Muslimov A, Tereshchenko V, Shevyrev D, Rogova A, Lepik K, Reshetnikov V, Ivanov R. The Dual Role of the Innate Immune System in the Effectiveness of mRNA Therapeutics. Int J Mol Sci 2023; 24:14820. [PMID: 37834268 PMCID: PMC10573212 DOI: 10.3390/ijms241914820] [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: 07/13/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Advances in molecular biology have revolutionized the use of messenger RNA (mRNA) as a therapeutic. The concept of nucleic acid therapy with mRNA originated in 1990 when Wolff et al. reported successful expression of proteins in target organs by direct injection of either plasmid DNA or mRNA. It took decades to bring the transfection efficiency of mRNA closer to that of DNA. The next few decades were dedicated to turning in vitro-transcribed (IVT) mRNA from a promising delivery tool for gene therapy into a full-blown therapeutic modality, which changed the biotech market rapidly. Hundreds of clinical trials are currently underway using mRNA for prophylaxis and therapy of infectious diseases and cancers, in regenerative medicine, and genome editing. The potential of IVT mRNA to induce an innate immune response favors its use for vaccination and immunotherapy. Nonetheless, in non-immunotherapy applications, the intrinsic immunostimulatory activity of mRNA directly hinders the desired therapeutic effect since it can seriously impair the target protein expression. Targeting the same innate immune factors can increase the effectiveness of mRNA therapeutics for some indications and decrease it for others, and vice versa. The review aims to present the innate immunity-related 'barriers' or 'springboards' that may affect the development of immunotherapies and non-immunotherapy applications of mRNA medicines.
Collapse
Affiliation(s)
- Albert Muslimov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Valeriy Tereshchenko
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Daniil Shevyrev
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Anna Rogova
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- Saint-Petersburg Chemical-Pharmaceutical University, Professora Popova 14, 197376 St. Petersburg, Russia
- School of Physics and Engineering, ITMO University, Lomonosova 9, 191002 St. Petersburg, Russia
| | - Kirill Lepik
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Vasiliy Reshetnikov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Roman Ivanov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| |
Collapse
|
6
|
Yanik S, Venkatesh V, Parker ML, Ramaswamy R, Diouf A, Sarkar D, Miura K, Long CA, Boulanger MJ, Srinivasan P. Structure guided mimicry of an essential P. falciparum receptor-ligand complex enhances cross neutralizing antibodies. Nat Commun 2023; 14:5879. [PMID: 37735574 PMCID: PMC10514071 DOI: 10.1038/s41467-023-41636-5] [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: 04/04/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
Invasion of human erythrocytes by Plasmodium falciparum (Pf) merozoites relies on the interaction between two parasite proteins: apical membrane antigen 1 (AMA1) and rhoptry neck protein 2 (RON2). While antibodies to AMA1 provide limited protection against Pf in non-human primate malaria models, clinical trials using recombinant AMA1 alone (apoAMA1) yielded no protection due to insufficient functional antibodies. Immunization with AMA1 bound to RON2L, a 49-amino acid peptide from its ligand RON2, has shown superior protection by increasing the proportion of neutralizing antibodies. However, this approach relies on the formation of a complex in solution between the two vaccine components. To advance vaccine development, here we engineered chimeric antigens by replacing the AMA1 DII loop, displaced upon ligand binding, with RON2L. Structural analysis confirmed that the fusion chimera (Fusion-FD12) closely mimics the binary AMA1-RON2L complex. Immunization studies in female rats demonstrated that Fusion-FD12 immune sera, but not purified IgG, neutralized vaccine-type parasites more efficiently compared to apoAMA1, despite lower overall anti-AMA1 titers. Interestingly, Fusion-FD12 immunization enhanced antibodies targeting conserved epitopes on AMA1, leading to increased neutralization of non-vaccine type parasites. Identifying these cross-neutralizing antibody epitopes holds promise for developing an effective, strain-transcending malaria vaccine.
Collapse
Affiliation(s)
- Sean Yanik
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Varsha Venkatesh
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Michelle L Parker
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Raghavendran Ramaswamy
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Deepti Sarkar
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Martin J Boulanger
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 3P6, Canada
| | - Prakash Srinivasan
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA.
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA.
| |
Collapse
|
7
|
González-Sanz M, Berzosa P, Norman FF. Updates on Malaria Epidemiology and Prevention Strategies. Curr Infect Dis Rep 2023; 25:1-9. [PMID: 37361492 PMCID: PMC10248987 DOI: 10.1007/s11908-023-00805-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2023] [Indexed: 06/28/2023]
Abstract
Purpose of Review The objective of this review was to provide an update on recent malaria epidemiology, both globally and in non-endemic areas, to identify the current distribution and repercussions of genetically diverse Plasmodium species and summarize recently implemented intervention and prevention tools. Recent Findings Notable changes in malaria epidemiology have occurred in recent years, with an increase in the number of total cases and deaths globally during 2020-2021, in part attributed to the COVID-19 pandemic. The emergence of artemisinin-resistant species in new areas and the expanding distribution of parasites harbouring deletions of the pfhrp2/3 genes have been concerning. New strategies to curb the burden of this infection, such as vaccination, have been implemented in certain endemic areas and their performance is currently being evaluated. Summary Inadequate control of malaria in endemic regions may have an effect on imported malaria and measures to prevent re-establishment of transmission in malaria-free areas are essential. Enhanced surveillance and investigation of Plasmodium spp. genetic variations will contribute to the successful diagnosis and treatment of malaria in future. Novel strategies for an integrated One Health approach to malaria control should also be strengthened.
Collapse
Affiliation(s)
- Marta González-Sanz
- Infectious Diseases Department, National Referral Unit for Tropical Diseases, Ramón y Cajal University Hospital, IRYCIS, Universidad de Alcalá, CIBER de Enfermedades Infecciosas, Madrid, Spain
| | - Pedro Berzosa
- Malaria and Neglected Tropical Diseases Laboratory, National Centre for Tropical Medicine, Carlos III Health Institute, CIBER de Enfermedades Infecciosas, Madrid, Spain
| | - Francesca F. Norman
- Infectious Diseases Department, National Referral Unit for Tropical Diseases, Ramón y Cajal University Hospital, IRYCIS, Universidad de Alcalá, CIBER de Enfermedades Infecciosas, Madrid, Spain
| |
Collapse
|
8
|
Srinivasan P, Yanik S, Venkatesh V, Parker M, Diouf A, Sarkar D, Miura K, Long C, Boulanger M. Structure guided mimicry of an essential P. falciparum receptor-ligand complex enhances cross neutralizing antibodies. RESEARCH SQUARE 2023:rs.3.rs-2733434. [PMID: 37131813 PMCID: PMC10153359 DOI: 10.21203/rs.3.rs-2733434/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Invasion of human red blood cells (RBCs) by Plasmodium falciparum (Pf) merozoites relies on the interaction between two parasite proteins, apical membrane antigen 1 (AMA1) and rhoptry neck protein 2 (RON2) 1,2 . Antibodies to AMA1 confer limited protection against P. falciparum in non-human primate malaria models 3,4 . However, clinical trials with recombinant AMA1 alone (apoAMA1) saw no protection, likely due to inadequate levels of functional antibodies 5-8 . Notably, immunization with AMA1 in its ligand bound conformation using RON2L, a 49 amino acid peptide from RON2, confers superior protection against P. falciparum malaria by enhancing the proportion of neutralizing antibodies 9,10 . A limitation of this approach, however, is that it requires the two vaccine components to form a complex in solution. To facilitate vaccine development, we engineered chimeric antigens by strategically replacing the AMA1 DII loop that is displaced upon ligand binding with RON2L. Structural characterization of the fusion chimera, Fusion-F D12 to 1.55 Å resolution showed that it closely mimics the binary receptor-ligand complex. Immunization studies showed that Fusion-F D12 immune sera neutralized parasites more efficiently than apoAMA1 immune sera despite having an overall lower anti-AMA1 titer, suggesting improvement in antibody quality. Furthermore, immunization with Fusion-F D12 enhanced antibodies targeting conserved epitopes on AMA1 resulting in greater neutralization of non-vaccine type parasites. Identifying epitopes of such cross-neutralizing antibodies will help in the development of an effective, strain-transcending malaria vaccine. Our fusion protein design is a robust vaccine platform that can be enhanced by incorporating polymorphisms in AMA1 to effectively neutralize all P. falciparum parasites.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Carole Long
- Laboratory of Malaria and Vector Resarch, NIAID/NIH
| | | |
Collapse
|
9
|
Odera DO, Tuju J, Mwai K, Nkumama IN, Fürle K, Chege T, Kimathi R, Diehl S, Musasia FK, Rosenkranz M, Njuguna P, Hamaluba M, Kapulu MC, Frank R, Osier FHA. Anti-merozoite antibodies induce natural killer cell effector function and are associated with immunity against malaria. Sci Transl Med 2023; 15:eabn5993. [PMID: 36753561 PMCID: PMC7616656 DOI: 10.1126/scitranslmed.abn5993] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/19/2023] [Indexed: 02/10/2023]
Abstract
Natural killer (NK) cells are potent immune effectors that can be activated via antibody-mediated Fc receptor engagement. Using multiparameter flow cytometry, we found that NK cells degranulate and release IFN-γ upon stimulation with antibody-opsonized Plasmodium falciparum merozoites. Antibody-dependent NK (Ab-NK) activity was largely strain transcending and enhanced invasion inhibition into erythrocytes. Ab-NK was associated with the successful control of parasitemia after experimental malaria challenge in African adults. In an independent cohort study in children, Ab-NK increased with age, was boosted by concurrent P. falciparum infections, and was associated with a lower risk of clinical episodes of malaria. Nine of the 14 vaccine candidates tested induced Ab-NK, including some less well-characterized antigens: P41, P113, MSP11, RHOPH3, and Pf_11363200. These data highlight an important role of Ab-NK activity in immunity against malaria and provide a potential mechanism for evaluating vaccine candidates.
Collapse
Affiliation(s)
- Dennis O. Odera
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
| | - James Tuju
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kennedy Mwai
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
- Epidemiology and Biostatistics Division, School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Irene N. Nkumama
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Kristin Fürle
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Timothy Chege
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
| | - Rinter Kimathi
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
| | - Stefan Diehl
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Fauzia K. Musasia
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Micha Rosenkranz
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Patricia Njuguna
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
| | - Mainga Hamaluba
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
| | - Melissa C. Kapulu
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
| | - Roland Frank
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Faith H. A. Osier
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Life Sciences, Imperial College London, UK
| |
Collapse
|
10
|
Takashima E, Nagaoka H, Correia R, Alves PM, Roldão A, Christensen D, Guderian JA, Fukushima A, Viebig NK, Depraetere H, Tsuboi T. A novel asexual blood-stage malaria vaccine candidate: PfRipr5 formulated with human-use adjuvants induces potent growth inhibitory antibodies. Front Immunol 2022; 13:1002430. [PMID: 36389677 PMCID: PMC9647036 DOI: 10.3389/fimmu.2022.1002430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2023] Open
Abstract
PfRipr is a highly conserved asexual-blood stage malaria vaccine candidate against Plasmodium falciparum. PfRipr5, a protein fragment of PfRipr inducing the most potent inhibitory antibodies, is a promising candidate for the development of next-generation malaria vaccines, requiring validation of its potential when formulated with adjuvants already approved for human use. In this study, PfRipr5 antigen was efficiently produced in a tank bioreactor using insect High Five cells and the baculovirus expression vector system; purified PfRipr5 was thermally stable in its monomeric form, had high purity and binding capacity to functional monoclonal anti-PfRipr antibody. The formulation of purified PfRipr5 with Alhydrogel®, GLA-SE or CAF®01 adjuvants accepted for human use showed acceptable compatibility. Rabbits immunized with these formulations induced comparable levels of anti-PfRipr5 antibodies, and significantly higher than the control group immunized with PfRipr5 alone. To investigate the efficacy of the antibodies, we used an in vitro parasite growth inhibition assay (GIA). The highest average GIA activity amongst all groups was attained with antibodies induced by immunization with PfRipr5 formulated with CAF®01. Overall, this study validates the potential of adjuvanted PfRipr5 as an asexual blood-stage malaria vaccine candidate, with PfRipr5/CAF®01 being a promising formulation for subsequent pre-clinical and clinical development.
Collapse
Affiliation(s)
- Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Ricardo Correia
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - António Roldão
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut (SSI), Copenhagen, Denmark
| | | | | | - Nicola K. Viebig
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - Hilde Depraetere
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
11
|
Somanathan A, Mian SY, Chaddha K, Uchoi S, Bharti PK, Tandon R, Gaur D, Chauhan VS. Process development and preclinical evaluation of a major Plasmodium falciparum blood stage vaccine candidate, Cysteine-Rich Protective Antigen (CyRPA). Front Immunol 2022; 13:1005332. [PMID: 36211427 PMCID: PMC9535676 DOI: 10.3389/fimmu.2022.1005332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium falciparum Cysteine-Rich Protective Antigen (CyRPA) is an essential, highly conserved merozoite antigen that forms an important multi-protein complex (RH5/Ripr/CyRPA) necessary for erythrocyte invasion. CyRPA is a promising blood-stage vaccine target that has been shown to elicit potent strain-transcending parasite neutralizing antibodies. Recently, we demonstrated that naturally acquired immune anti-CyRPA antibodies are invasion-inhibitory and therefore a correlate of protection against malaria. Here, we describe a process for the large-scale production of tag-free CyRPA vaccine in E. coli and demonstrate its parasite neutralizing efficacy with commonly used adjuvants. CyRPA was purified from inclusion bodies using a one-step purification method with high purity (>90%). Biochemical and biophysical characterization showed that the purified tag-free CyRPA interacted with RH5, readily detected by a conformation-specific CyRPA monoclonal antibody and recognized by sera from malaria infected individuals thus indicating that the recombinant antigen was correctly folded and retained its native conformation. Tag-free CyRPA formulated with Freund’s adjuvant elicited highly potent parasite neutralizing antibodies achieving inhibition of >90% across diverse parasite strains. Importantly, we identified tag-free CyRPA/Alhydrogel formulation as most effective in inducing a highly immunogenic antibody response that exhibited efficacious, cross-strain in vitro parasite neutralization achieving ~80% at 10 mg/ml. Further, CyRPA/Alhydrogel vaccine induced anti-parasite cytokine response in mice. In summary, our study provides a simple, scalable, cost-effective process for the production of tag-free CyRPA that in combination with human-compatible adjuvant induces efficacious humoral and cell-mediated immune response.
Collapse
Affiliation(s)
- Anjali Somanathan
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Syed Yusuf Mian
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Kritika Chaddha
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Seemalata Uchoi
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Praveen K. Bharti
- ICMR-National Institute of Research in Tribal Health (NIRTH), Jabalpur, India
| | - Ravi Tandon
- Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Deepak Gaur
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Virander Singh Chauhan
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
- *Correspondence: Virander Singh Chauhan,
| |
Collapse
|
12
|
Zolfaghari Emameh R, Barker HR, Turpeinen H, Parkkila S, Hytönen VP. A reverse vaccinology approach on transmembrane carbonic anhydrases from Plasmodium species as vaccine candidates for malaria prevention. Malar J 2022; 21:189. [PMID: 35706028 PMCID: PMC9199335 DOI: 10.1186/s12936-022-04186-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 05/19/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Malaria is a significant parasitic infection, and human infection is mediated by mosquito (Anopheles) biting and subsequent transmission of protozoa (Plasmodium) to the blood. Carbonic anhydrases (CAs) are known to be highly expressed in the midgut and ectoperitrophic space of Anopheles gambiae. Transmembrane CAs (tmCAs) in Plasmodium may be potential vaccine candidates for the control and prevention of malaria. METHODS In this study, two groups of transmembrane CAs, including α-CAs and one group of η-CAs were analysed by immunoinformatics and computational biology methods, such as predictions on transmembrane localization of CAs from Plasmodium spp., affinity and stability of different HLA classes, antigenicity of tmCA peptides, epitope and proteasomal cleavage of Plasmodium tmCAs, accessibility of Plasmodium tmCAs MHC-ligands, allergenicity of Plasmodium tmCAs, disulfide-bond of Plasmodium tmCAs, B cell epitopes of Plasmodium tmCAs, and Cell type-specific expression of Plasmodium CAs. RESULTS Two groups of α-CAs and one group of η-CAs in Plasmodium spp. were identified to contain tmCA sequences, having high affinity towards MHCs, high stability, and strong antigenicity. All putative tmCAs were predicted to contain sequences for proteasomal cleavage in antigen presenting cells (APCs). CONCLUSIONS The predicted results revealed that tmCAs from Plasmodium spp. can be potential targets for vaccination against malaria.
Collapse
Affiliation(s)
- Reza Zolfaghari Emameh
- Department of Energy and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), 14965/161, Tehran, Iran.
| | - Harlan R Barker
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab Laboratories Ltd and Tampere University Hospital, Tampere, Finland
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab Laboratories Ltd and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
13
|
Kumar S, Li X, McDew-White M, Reyes A, Delgado E, Sayeed A, Haile MT, Abatiyow BA, Kennedy SY, Camargo N, Checkley LA, Brenneman KV, Button-Simons KA, Duraisingh MT, Cheeseman IH, Kappe SHI, Nosten F, Ferdig MT, Vaughan AM, Anderson TJC. A Malaria Parasite Cross Reveals Genetic Determinants of Plasmodium falciparum Growth in Different Culture Media. Front Cell Infect Microbiol 2022; 12:878496. [PMID: 35711667 PMCID: PMC9197316 DOI: 10.3389/fcimb.2022.878496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/28/2022] [Indexed: 12/21/2022] Open
Abstract
What genes determine in vitro growth and nutrient utilization in asexual blood-stage malaria parasites? Competition experiments between NF54, clone 3D7, a lab-adapted African parasite, and a recently isolated Asian parasite (NHP4026) reveal contrasting outcomes in different media: 3D7 outcompetes NHP4026 in media containing human serum, while NHP4026 outcompetes 3D7 in media containing AlbuMAX, a commercial lipid-rich bovine serum formulation. To determine the basis for this polymorphism, we conducted parasite genetic crosses using humanized mice and compared genome-wide allele frequency changes in three independent progeny populations cultured in media containing human serum or AlbuMAX. This bulk segregant analysis detected three quantitative trait loci (QTL) regions [on chromosome (chr) 2 containing aspartate transaminase AST; chr 13 containing EBA-140; and chr 14 containing cysteine protease ATG4] linked with differential growth in serum or AlbuMAX in each of the three independent progeny pools. Selection driving differential growth was strong (s = 0.10 – 0.23 per 48-hour lifecycle). We conducted validation experiments for the strongest QTL on chr 13: competition experiments between ΔEBA-140 and 3D7 wildtype parasites showed fitness reversals in the two medium types as seen in the parental parasites, validating this locus as the causative gene. These results (i) demonstrate the effectiveness of bulk segregant analysis for dissecting fitness traits in P. falciparum genetic crosses, and (ii) reveal intimate links between red blood cell invasion and nutrient composition of growth media. Use of parasite crosses combined with bulk segregant analysis will allow systematic dissection of key nutrient acquisition/metabolism and red blood cell invasion pathways in P. falciparum.
Collapse
Affiliation(s)
- Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Xue Li
- Program in Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Marina McDew-White
- Program in Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Ann Reyes
- Program in Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Elizabeth Delgado
- Program in Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Abeer Sayeed
- Program in Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Meseret T. Haile
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Biley A. Abatiyow
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Spencer Y. Kennedy
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Lisa A. Checkley
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, United States
| | - Katelyn V. Brenneman
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, United States
| | - Katrina A. Button-Simons
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, United States
| | - Manoj T. Duraisingh
- Immunology and Infectious Diseases Department, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Ian H. Cheeseman
- Program in Host Pathogen Interactions, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Stefan H. I. Kappe
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine Research building, University of Oxford, Oxford, United Kingdom
| | - Michael T. Ferdig
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, United States
| | - Ashley M. Vaughan
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- *Correspondence: Ashley M. Vaughan, ; Tim J. C. Anderson,
| | - Tim J. C. Anderson
- Program in Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX, United States
- *Correspondence: Ashley M. Vaughan, ; Tim J. C. Anderson,
| |
Collapse
|
14
|
Partey FD, Frimpong A, Ofori MF. Collection and Cryopreservation of Plasmodium falciparum Clinical Isolates in the Field. Methods Mol Biol 2022; 2470:11-17. [PMID: 35881334 DOI: 10.1007/978-1-0716-2189-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
P. falciparum causes the most severe form of malaria in younger children and pregnant women. In vitro culture systems allow researchers to understand parasite biology, elucidate mechanism of host immunity and test efficacy of antimalarial agents or vaccines in preclinical studies. Most laboratory-adapted parasite strains predate the emergence of artemisinin-based drug combinations and mainly originate from Asia or Europe. To fully understand the biochemical and phenotypic characteristics of parasites, it is imperative that researchers are able to culture parasites circulating in an area to unravel any geographical differences at the population level. Ex vivo culturing of clinical isolates can be challenging when collecting samples in the field and requires technical expertise and equipment. To overcome this challenge, clinical isolates are cryopreserved in the field and transported to a laboratory for in vitro studies. In this protocol, we describe different methods of cryopreserving P. falciparum isolates in the field and thawing them for subsequent in vitro culture.
Collapse
Affiliation(s)
| | - Augustina Frimpong
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Michael Fokuo Ofori
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana.
| |
Collapse
|
15
|
Quintana MDP. Expression of Single-Domain Soluble and Disulfide-Folded PfEMP1 Antigens in the Escherichia coli SHuffle Expression System. Methods Mol Biol 2022; 2470:273-282. [PMID: 35881352 DOI: 10.1007/978-1-0716-2189-9_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The genome of Plasmodium falciparum has an A/T content of around 81%. This, together with a high cysteine content and the high molecular weight of several proteins, make the expression of recombinant parasite proteins in heterologous systems challenging. P. falciparum erythrocyte membrane protein 1 (PfEMP1) is a family of proteins composed of several Duffy-binding like (DBL) and cysteine-rich inter-domain region (CIDR) domains involved in cytoadhesion to human host receptors and development of severe malaria. Expression of correctly folded single- and multiple-domain PfEMP1 fragment regions containing cysteines forming disulfide bonds, remains particularly difficult. Nevertheless, expression of single DBL and CIDR domains has been successful and this protocol describes the expression and purification of single-domain soluble PfEMP1 fragments using the Escherichia coli SHuffle expression system.
Collapse
Affiliation(s)
- Maria Del Pilar Quintana
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
16
|
Alum Pickering Emulsion as Effective Adjuvant to Improve Malaria Vaccine Efficacy. Vaccines (Basel) 2021; 9:vaccines9111244. [PMID: 34835175 PMCID: PMC8624716 DOI: 10.3390/vaccines9111244] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/27/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
Malaria is a life-threatening global epidemic disease and has caused more than 400,000 deaths in 2019. To control and prevent malaria, the development of a vaccine is a potential method. An effective malaria vaccine should either combine antigens from all stages of the malaria parasite’s life cycle, or epitopes of multiple key antigens due to the complexity of the Plasmodium parasite. Malaria’s random constructed antigen-1 (M.RCAg-1) is one of the recombinant vaccines, which was selected from a DNA library containing thousands of diverse multi-epitope chimeric antigen genes. Moreover, besides selecting an antigen, using an adjuvant is another important procedure for most vaccine development procedures. Freund’s adjuvant is considered an effective vaccine adjuvant for malaria vaccine, but it cannot be used in clinical settings because of its serious side effects. Traditional adjuvants, such as alum adjuvant, are limited by their unsatisfactory immune effects in malaria vaccines, hence there is an urgent need to develop a novel, safe and efficient adjuvant. In recent years, Pickering emulsions have attracted increasing attention as novel adjuvant. In contrast to classical emulsions, Pickering emulsions are stabilized by solid particles instead of surfactant, having pliability and lateral mobility. In this study, we selected aluminum hydroxide gel (termed as “alum”) as a stabilizer to prepare alum-stabilized Pickering emulsions (ALPE) as a malaria vaccine adjuvant. In addition, monophosphoryl lipid A (MPLA) as an immunostimulant was incorporated into the Pickering emulsion (ALMPE) to further enhance the immune response. In vitro tests showed that, compared with alum, ALPE and ALMPE showed higher antigen load rates and could be effectively endocytosed by J774a.1 cells. In vivo studies indicated that ALMPE could induce as high antibody titers as Freund’s adjuvant. The biocompatibility study also proved ALMPE with excellent biocompatibility. These results suggest that ALMPE is a potential adjuvant for a malaria vaccine.
Collapse
|
17
|
Cui X, Snapper CM. Epstein Barr Virus: Development of Vaccines and Immune Cell Therapy for EBV-Associated Diseases. Front Immunol 2021; 12:734471. [PMID: 34691042 PMCID: PMC8532523 DOI: 10.3389/fimmu.2021.734471] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/15/2021] [Indexed: 11/13/2022] Open
Abstract
Epstein-Barr virus (EBV) is the first human tumor virus discovered and is strongly implicated in the etiology of multiple lymphoid and epithelial cancers. Each year EBV associated cancers account for over 200,000 new cases of cancer and cause 150,000 deaths world-wide. EBV is also the primary cause of infectious mononucleosis, and up to 70% of adolescents and young adults in developed countries suffer from infectious mononucleosis. In addition, EBV has been shown to play a critical role in the pathogenesis of multiple sclerosis. An EBV prophylactic vaccine that induces neutralizing antibodies holds great promise for prevention of EBV associated diseases. EBV envelope proteins including gH/gL, gB and gp350 play key roles in EBV entry and infection of target cells, and neutralizing antibodies elicited by each of these proteins have shown to prevent EBV infection of target cells and markedly decrease EBV titers in the peripheral blood of humanized mice challenged with lethal dose EBV. Recent studies demonstrated that immunization with the combination of gH/gL, gB and/or gp350 induced markedly increased synergistic EBV neutralizing activity compared to immunization with individual proteins. As previous clinical trials focused on gp350 alone were partially successful, the inclusion of gH/gL and gB in a vaccine formulation with gp350 represents a promising approach of EBV prophylactic vaccine development. Therapeutic EBV vaccines have also been tested clinically with encouraging results. Immunization with various vaccine platforms expressing the EBV latent proteins EBNA1, LMP1, and/or LMP2 promoted specific CD4+ and CD8+ cytotoxic responses with anti-tumor activity. The addition of EBV envelope proteins gH/gL, gB and gp350 has the potential to increase the efficacy of a therapeutic EBV vaccine. The immune system plays a critical role in the control of tumors, and immune cell therapy has emerged as a promising treatment of cancers. Adoptive T-cell therapy has been successfully used in the prevention and treatment of post-transplant lymphoproliferative disorder. Chimeric antigen receptor T cell therapy and T cell receptor engineered T cell therapy targeting EBV latent proteins LMP1, LMP2 and/or EBNA1 have been in development, with the goal to increase the specificity and efficacy of treatment of EBV associated cancers.
Collapse
Affiliation(s)
- Xinle Cui
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.,The Institute for Vaccine Research and Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Clifford M Snapper
- The Institute for Vaccine Research and Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Citranvi Biosciences LLC, Chapel Hill, NC, United States
| |
Collapse
|
18
|
Bonam SR, Rénia L, Tadepalli G, Bayry J, Kumar HMS. Plasmodium falciparum Malaria Vaccines and Vaccine Adjuvants. Vaccines (Basel) 2021; 9:1072. [PMID: 34696180 PMCID: PMC8541031 DOI: 10.3390/vaccines9101072] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 12/02/2022] Open
Abstract
Malaria-a parasite vector-borne disease-is a global health problem, and Plasmodium falciparum has proven to be the deadliest among Plasmodium spp., which causes malaria in humans. Symptoms of the disease range from mild fever and shivering to hemolytic anemia and neurological dysfunctions. The spread of drug resistance and the absence of effective vaccines has made malaria disease an ever-emerging problem. Although progress has been made in understanding the host response to the parasite, various aspects of its biology in its mammalian host are still unclear. In this context, there is a pressing demand for the development of effective preventive and therapeutic strategies, including new drugs and novel adjuvanted vaccines that elicit protective immunity. The present article provides an overview of the current knowledge of anti-malarial immunity against P. falciparum and different options of vaccine candidates in development. A special emphasis has been made on the mechanism of action of clinically used vaccine adjuvants.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, F-75006 Paris, France;
| | - Laurent Rénia
- A*STAR Infectious Diseases Labs, 8A Biomedical Grove, Singapore 138648, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
| | - Ganesh Tadepalli
- Vaccine Immunology Laboratory, Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India;
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, F-75006 Paris, France;
- Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678623, India
| | - Halmuthur Mahabalarao Sampath Kumar
- Vaccine Immunology Laboratory, Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India;
| |
Collapse
|
19
|
Michelow IC, Park S, Tsai SW, Rayta B, Pasaje CFA, Nelson S, Early AM, Frosch AP, Ayodo G, Raj DK, Nixon CE, Nixon CP, Pond-Tor S, Friedman JF, Fried M, Duffy PE, Le Roch KG, Niles JC, Kurtis JD. A newly characterized malaria antigen on erythrocyte and merozoite surfaces induces parasite inhibitory antibodies. J Exp Med 2021; 218:e20200170. [PMID: 34342640 PMCID: PMC8340565 DOI: 10.1084/jem.20200170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/11/2021] [Accepted: 06/24/2021] [Indexed: 11/13/2022] Open
Abstract
We previously identified a Plasmodium falciparum (Pf) protein of unknown function encoded by a single-copy gene, PF3D7_1134300, as a target of antibodies in plasma of Tanzanian children in a whole-proteome differential screen. Here we characterize this protein as a blood-stage antigen that localizes to the surface membranes of both parasitized erythrocytes and merozoites, hence its designation as Pf erythrocyte membrane and merozoite antigen 1 (PfEMMA1). Mouse anti-PfEMMA1 antisera and affinity-purified human anti-PfEMMA1 antibodies inhibited growth of P. falciparum strains by up to 68% in growth inhibition assays. Following challenge with uniformly fatal Plasmodium berghei (Pb) ANKA, up to 40% of mice immunized with recombinant PbEMMA1 self-cured, and median survival of lethally infected mice was up to 2.6-fold longer than controls (21 vs. 8 d, P = 0.005). Furthermore, high levels of naturally acquired human anti-PfEMMA1 antibodies were associated with a 46% decrease in parasitemia over 2.5 yr of follow-up of Tanzanian children. Together, these findings suggest that antibodies to PfEMMA1 mediate protection against malaria.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan/immunology
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Antigens, Protozoan/metabolism
- Child, Preschool
- Erythrocyte Membrane/parasitology
- Female
- Host-Parasite Interactions/physiology
- Humans
- Infant
- Malaria Vaccines/genetics
- Malaria Vaccines/immunology
- Malaria, Falciparum/immunology
- Malaria, Falciparum/mortality
- Malaria, Falciparum/parasitology
- Merozoites/immunology
- Merozoites/metabolism
- Mice, Inbred BALB C
- Plasmodium falciparum/immunology
- Plasmodium falciparum/pathogenicity
- Plasmodium falciparum/physiology
- Polymorphism, Single Nucleotide
- Protozoan Proteins/chemistry
- Protozoan Proteins/genetics
- Protozoan Proteins/immunology
- Protozoan Proteins/metabolism
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Recombinant Proteins/metabolism
- Tanzania
- Mice
Collapse
Affiliation(s)
- Ian C. Michelow
- Department of Pediatrics, Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI
- Center for International Health Research, Rhode Island Hospital, Providence, RI
| | - Sangshin Park
- Center for International Health Research, Rhode Island Hospital, Providence, RI
- Graduate School of Urban Public Health & Department of Urban Big Data Convergence, University of Seoul, Seoul, Republic of Korea
| | - Shu-Whei Tsai
- Department of Pediatrics, Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI
- Center for International Health Research, Rhode Island Hospital, Providence, RI
| | - Bonnie Rayta
- Department of Pediatrics, Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI
- Center for International Health Research, Rhode Island Hospital, Providence, RI
| | | | - Sara Nelson
- Department of Pediatrics, Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI
- Center for International Health Research, Rhode Island Hospital, Providence, RI
| | - Angela M. Early
- Infectious Disease and Microbiome Program, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Anne P. Frosch
- Department of Medicine, Hennepin Healthcare Research Institute, University of Minnesota, Minneapolis, MN
| | - George Ayodo
- Kenya Medical Research Institute, Centre of Global Health Research, Kisumu, Kenya
- Jaramogi Oginga Odinga University of Science and Technology, Bondo, Kenya
| | - Dipak K. Raj
- Center for International Health Research, Rhode Island Hospital, Providence, RI
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Christina E. Nixon
- Center for International Health Research, Rhode Island Hospital, Providence, RI
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Christian P. Nixon
- Center for International Health Research, Rhode Island Hospital, Providence, RI
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Sunthorn Pond-Tor
- Center for International Health Research, Rhode Island Hospital, Providence, RI
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Jennifer F. Friedman
- Center for International Health Research, Rhode Island Hospital, Providence, RI
- Department of Pediatrics, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Karine G. Le Roch
- Department of Molecular, Cell and Systems Biology, Center for Infectious Disease and Vector Research, University of California, Riverside, Riverside, CA
| | - Jacquin C. Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Jonathan D. Kurtis
- Center for International Health Research, Rhode Island Hospital, Providence, RI
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
20
|
Aguttu C, Okech BA, Mukisa A, Lubega GW. Screening and characterization of hypothetical proteins of Plasmodium falciparum as novel vaccine candidates in the fight against malaria using reverse vaccinology. J Genet Eng Biotechnol 2021; 19:103. [PMID: 34269931 PMCID: PMC8283385 DOI: 10.1186/s43141-021-00199-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/16/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Plasmodium falciparum is the most deadly and leading cause of morbidity and mortality in Africa. About 90% of all malaria deaths in the world today occur in Sub-Saharan Africa especially in children aged < 5 years. In 2018, it was reported that there were 228 million malaria cases that resulted in 405,000 deaths from 91 countries. Currently, a fully effective and long-lasting preventive malaria vaccine is still elusive therefore more effort is needed to identify better effective vaccine candidates. The aim of this study was to identify and characterize hypothetical proteins as vaccine candidates derived from Plasmodium falciparum 3D7 genome by reverse vaccinology. RESULTS Of the 23 selected hypothetical proteins, 5 were predicted on the extracellular localization by WoLFPSORTv.2.0 program and all the 5 had less than 2 transmembrane regions that were predicted by TMHMMv2.0 and HMMTOP programs at default settings. Two out of the five proteins lacked secretory signal peptides as predicted by SignalP program. Among the 5 extracellular proteins, 3 were predicted to be antigenic by VaxiJen (score ≥ 0.5) and had negative GRAVY values ranging from - 1.156 to - 0.440. B cell epitope prediction by ABCpred and BCpred programs revealed a total of 15 antigenic epitopes. A total of 13 cytotoxic T cells were predicted from the 3 proteins using CTLPred online server. Only 2 out of the 13 CTL were antigenic, immunogenic, non-allergenic, and non-toxic using VaxiJen, IEDB, AllergenFp, and Toxinpred servers respectively in that order. Five HTL peptides from XP_001351030.1 protein are predicted inducers of all the three cytokines. STRING protein-protein network analysis of HPs revealed XP_001350955.1 closely interacts with nucleoside diphosphate kinase (PF13-0349) at 0.704, XP_001351030.1 interacts with male development protein1 (Mdv-1) at 0.645, and XP_001351047.1 with an uncharacterized protein (MAL8P1.53) at 0.400. CONCLUSION Reverse vaccinology is a promising strategy for the screening and identification of antigenic antigens with potential capacity to elicit cellular and humoral immune responses against P. falciparum infection. In this study, potential vaccine candidates of Plasmodium falciparum were identified and screened using standard bioinformatics tools. The vaccine candidates contained antigenic and immunogenic epitopes which could be considered for novel and effective vaccine targets. However, we strongly recommend in vivo and in vitro experiments to validate their immunogenicity and protective efficacy to completely decipher the vaccine targets against malaria.
Collapse
Affiliation(s)
- Claire Aguttu
- Department of Biochemistry and Sports Science, College of Natural Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda
| | | | - Ambrose Mukisa
- Department of Biochemistry and Sports Science, College of Natural Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda
| | - George William Lubega
- Department of Bio-molecular Resources and Bio-lab Sciences, School of Biosecurity, Biotechnology and Laboratory Sciences (SBLS), College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, P.O Box 7062, Kampala, Uganda
| |
Collapse
|
21
|
Production and Immunogenicity of a Tag-Free Recombinant Chimera Based on PfMSP-1 and PfMSP-3 Using Alhydrogel and Dipeptide-Based Hydrogels. Vaccines (Basel) 2021; 9:vaccines9070782. [PMID: 34358198 PMCID: PMC8310097 DOI: 10.3390/vaccines9070782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
A fusion chimeric vaccine comprising multiple protective domains of different blood-stage Plasmodium falciparum antigens is perhaps necessary for widening the protective immune responses and reducing the morbidity caused by the disease. Here we continue to build upon the prior work of developing a recombinant fusion chimera protein, His-tagged PfMSP-Fu24, by producing it as a tag-free recombinant protein. In this study, tag-free recombinant PfMSPFu24 (rFu24) was expressed in Escherichia coli, and the soluble protein was purified using a three-step purification involving ammonium sulphate precipitation followed by 2-step ion exchange chromatography procedures and shown that it was highly immunogenic with the human-compatible adjuvant Alhydrogel. We further investigated two dipeptides, phenylalanine-α, β-dehydrophenylalanine (FΔF) and Leucine-α, β-dehydrophenylalanine (LΔF) based hydrogels as effective delivery platforms for rFu24. These dipeptides self-assembled spontaneously to form a highly stable hydrogel under physiological conditions. rFu24 was efficiently entrapped in both the F∆F and L∆F hydrogels, and the three-dimensional (3D) mesh-like structures of the hydrogels remained intact after the entrapment of the antigen. The two hydrogels significantly stimulated rFu24-specific antibody titers, and the sera from the immunized mice showed an invasion inhibitory activity comparable to that of Alhydrogel. Easily synthesized dipeptide hydrogels can be used as an effective antigen delivery platform to induce immune responses.
Collapse
|
22
|
Sadri Najafabadi Z, Nazarian S, Kargar M, Kafilzadeh F. Designing of a chimeric protein contains StxB, intimin and EscC against toxicity and adherence of enterohemorrhagic Escherichia coli O157:H7 and evaluation of serum antibody titers against it. Mol Immunol 2021; 134:218-227. [PMID: 33823320 DOI: 10.1016/j.molimm.2021.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/08/2021] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 strain is known as one of the major human foodborne pathogens. Lack of effective clinical treatment for human diarrheal diseases confirms the need for vaccine production against enteric bacteria such as E.coli O157:H7. Shiga-like toxin (Stx), EscC, and Intimin are the main important virulent factors of this enteric pathogen. In the present study, a comparative Omics analysis was conducted to identify most invasion EHEC antigenic factors as a potential immunogen. SEI (Stx-EscC-Intimin) trivalent chimeric protein was designed from the exposed and epitope rich part of these virulence factors. Sequence optimization, physicochemical properties, mRNA folding, three-dimensional structure and immunoinformatics data were investigated. The chimeric gene was synthesized with codon bias of E. coli. Recombinant protein was expressed and confirmed by western blot analysis. To evaluate the immunogenicity of the designed protein, the protein was administered to BALB/c mice and the serum IgG was determined by ELISA. Based on the Ramachandran plot, the validation data showed that 90.1 % of residues lie in the favored region. The high antigenicity of the multimeric protein was predicted by the immunoinformatic analysis. Epitope prediction had shown the proper distribution of linear and conformational B-cell epitopes and the competition of T-cell epitopes to bind MHC molecules too. Recombinant ESI Protein with 74.5 kDa was expressed in E. coli. Western blot analysis by anti-Stx antibody, confirmed a single band of chimeric protein. Consequently, the chimeric gene was designed and constructed after assessments. From in silico approach, the protein deduced from this cassette can be an immunogen candidate, and act against toxicity and adherence of EHEC.
Collapse
Affiliation(s)
| | - Shahram Nazarian
- Department of Biological Sciences, Faculty of Science, Imam Hossein University, Tehran, Iran.
| | - Mohammad Kargar
- Department of Microbiology, Jahrom Branch, Islamic Azad University, Jahrom, Iran
| | - Farshid Kafilzadeh
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom, Iran
| |
Collapse
|
23
|
Immunization with Epstein-Barr Virus Core Fusion Machinery Envelope Proteins Elicit High Titers of Neutralizing Activities and Protect Humanized Mice from Lethal Dose EBV Challenge. Vaccines (Basel) 2021; 9:vaccines9030285. [PMID: 33808755 PMCID: PMC8003492 DOI: 10.3390/vaccines9030285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/26/2021] [Accepted: 03/11/2021] [Indexed: 12/18/2022] Open
Abstract
Epstein–Barr virus (EBV) is the primary cause of infectious mononucleosis and is strongly implicated in the etiology of multiple lymphoid and epithelial cancers. EBV core fusion machinery envelope proteins gH/gL and gB coordinately mediate EBV fusion and entry into its target cells, B lymphocytes and epithelial cells, suggesting these proteins could induce antibodies that prevent EBV infection. We previously reported that the immunization of rabbits with recombinant EBV gH/gL or trimeric gB each induced markedly higher serum EBV-neutralizing titers for B lymphocytes than that of the leading EBV vaccine candidate gp350. In this study, we demonstrated that immunization of rabbits with EBV core fusion machinery proteins induced high titer EBV neutralizing antibodies for both B lymphocytes and epithelial cells, and EBV gH/gL in combination with EBV trimeric gB elicited strong synergistic EBV neutralizing activities. Furthermore, the immune sera from rabbits immunized with EBV gH/gL or trimeric gB demonstrated strong passive immune protection of humanized mice from lethal dose EBV challenge, partially or completely prevented death respectively, and markedly decreased the EBV load in peripheral blood of humanized mice. These data strongly suggest the combination of EBV core fusion machinery envelope proteins gH/gL and trimeric gB is a promising EBV prophylactic vaccine.
Collapse
|
24
|
Moon JE, Ockenhouse C, Regules JA, Vekemans J, Lee C, Chuang I, Traskine M, Jongert E, Ivinson K, Morelle D, Komisar JL, Lievens M, Sedegah M, Garver LS, Sikaffy AK, Waters NC, Ballou WR, Ofori-Anyinam O. A Phase IIa Controlled Human Malaria Infection and Immunogenicity Study of RTS,S/AS01E and RTS,S/AS01B Delayed Fractional Dose Regimens in Malaria-Naive Adults. J Infect Dis 2021; 222:1681-1691. [PMID: 32687161 PMCID: PMC7552430 DOI: 10.1093/infdis/jiaa421] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/17/2020] [Indexed: 01/04/2023] Open
Abstract
Background A previous RTS,S/AS01B vaccine challenge trial demonstrated that a 3-dose (0-1-7–month) regimen with a fractional third dose can produce high vaccine efficacy (VE) in adults challenged 3 weeks after vaccination. This study explored the VE of different delayed fractional dose regimens of adult and pediatric RTS,S/AS01 formulations. Methods A total of 130 participants were randomized into 5 groups. Four groups received 3 doses of RTS,S/AS01B or RTS,S/AS01E on a 0-1-7–month schedule, with the final 1 or 2 doses being fractional (one-fifth dose volume). One group received 1 full (month 0) and 1 fractional (month 7) dose of RTS,S/AS01E. Immunized and unvaccinated control participants underwent Plasmodium falciparum–infected mosquito challenge (controlled human malaria infection) 3 months after immunization, a timing chosen to potentially discriminate VEs between groups. Results The VE of 3-dose formulations ranged from 55% (95% confidence interval, 27%–72%) to 76% (48%–89%). Groups administered equivalent formulations of RTS,S/AS01E and RTS,S/AS01B demonstrated comparable VE. The 2-dose group demonstrated lower VE (29% [95% confidence interval, 6%–46%]). All regimens were well tolerated and immunogenic, with trends toward higher anti-circumsporozoite antibody titers in participants protected against infection. Conclusions RTS,S/AS01E can provide VE comparable to an equivalent RTS,S/AS01B regimen in adults, suggesting a universal formulation may be considered. Results also suggest that the 2-dose regimen is inferior to the 3-dose regimens evaluated. Clinical Trial Registration NCT03162614
Collapse
Affiliation(s)
- James E Moon
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | - Jason A Regules
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | - Cynthia Lee
- PATH-Malaria Vaccine Initiative, Washington, DC, USA
| | - Ilin Chuang
- Naval Medical Research Center, Silver Spring, Maryland, USA
| | | | | | - Karen Ivinson
- PATH-Malaria Vaccine Initiative, Washington, DC, USA
| | | | - Jack L Komisar
- Naval Medical Research Center, Silver Spring, Maryland, USA
| | | | - Martha Sedegah
- Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Lindsey S Garver
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - April K Sikaffy
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Norman C Waters
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | | | | |
Collapse
|
25
|
Doritchamou JYA, Suurbaar J, Tuikue Ndam N. Progress and new horizons toward a VAR2CSA-based placental malaria vaccine. Expert Rev Vaccines 2021; 20:215-226. [PMID: 33472449 DOI: 10.1080/14760584.2021.1878029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Several malaria vaccines are under various phases of development with some promising results. In placental malaria (PM) a deliberately anti-disease approach is considered as many studies have underlined the key role of VAR2CSA protein, which therefore represents the leading vaccine candidate. However, evidence indicates that VAR2CSA antigenic polymorphism remains an obstacle to overcome.Areas covered: This review analyzes the progress made thus far in developing a VAR2CSA-based vaccine, and addresses the current issues and challenges that must be overcome to develop an effective PM vaccine.Expert opinion: Phase I trials of PAMVAC and PRIMVAC VAR2CSA vaccines have shown more or less satisfactory results with regards to safety and immunogenicity. The second generation of VAR2CSA-based vaccines could benefit from optimization approaches to broaden the activity spectrum against various placenta-binding isolates through continued advances in the structural understanding of the interaction with CSA.
Collapse
Affiliation(s)
- Justin Yai Alamou Doritchamou
- Laboratory of Malaria Immunology & Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Suurbaar
- Université de Paris, MERIT, IRD, F-75006 Paris, France.,Noguchi Memorial Institute for Medical Research, Department of Immunology, University of Ghana, Accra, Ghana
| | - Nicaise Tuikue Ndam
- Université de Paris, MERIT, IRD, F-75006 Paris, France.,Noguchi Memorial Institute for Medical Research, Department of Immunology, University of Ghana, Accra, Ghana
| |
Collapse
|
26
|
Lozano JM, Rodríguez Parra Z, Hernández-Martínez S, Yasnot-Acosta MF, Rojas AP, Marín-Waldo LS, Rincón JE. The Search of a Malaria Vaccine: The Time for Modified Immuno-Potentiating Probes. Vaccines (Basel) 2021; 9:vaccines9020115. [PMID: 33540947 PMCID: PMC7913233 DOI: 10.3390/vaccines9020115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/25/2022] Open
Abstract
Malaria is a deadly disease that takes the lives of more than 420,000 people a year and is responsible for more than 229 million clinical cases globally. In 2019, 95% of malaria morbidity occurred in African countries. The development of a highly protective vaccine is an urgent task that remains to be solved. Many vaccine candidates have been developed, from the use of the entire attenuated and irradiated pre-erythrocytic parasite forms (or recombinantly expressed antigens thereof) to synthetic candidates formulated in a variety of adjuvants and delivery systems, however these have unfortunately proven a limited efficacy. At present, some vaccine candidates are finishing safety and protective efficacy trials, such as the PfSPZ and the RTS,S/AS01 which are being introduced in Africa. We propose a strategy for introducing non-natural elements into target antigens representing key epitopes of Plasmodium spp. Accordingly, chemical strategies and knowledge of host immunity to Plasmodium spp. have served as the basis. Evidence is obtained after being tested in experimental rodent models for malaria infection and recognized for human sera from malaria-endemic regions. This encourages us to propose such an immune-potentiating strategy to be further considered in the search for new vaccine candidates.
Collapse
Affiliation(s)
- José Manuel Lozano
- Grupo de Investigación Mimetismo Molecular de los Agentes Infecciosos, Departamento de Farmacia, Universidad Nacional de Colombia—Sede Bogotá, 111321 Bogota, Colombia;
- Correspondence: ; Tel.: +57-3102-504-657
| | - Zully Rodríguez Parra
- Grupo de Investigación Mimetismo Molecular de los Agentes Infecciosos, Departamento de Farmacia, Universidad Nacional de Colombia—Sede Bogotá, 111321 Bogota, Colombia;
| | - Salvador Hernández-Martínez
- Dirección de Infección e Inmunidad, Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, 62508 Cuernavaca, Morelos, Mexico;
| | - Maria Fernanda Yasnot-Acosta
- Grupo de Investigaciones Microbiológicas y Biomédicas de Córdoba, Universidad de Córdoba, 230002 Monteria, Colombia;
| | - Angela Patricia Rojas
- Grupo de Investigación Biología Celular y Autoinmuniad, Departamento de Farmacia, Universidad Nacional de Colombia-Sede Bogotá, 111321 Bogota, Colombia;
| | | | - Juan Edilberto Rincón
- Departamento de Ingeniería y Mecatrónica, Universidad Nacional de Colombia-Sede Bogotá, 111321 Bogota, Colombia;
| |
Collapse
|
27
|
Federizon J, Feugmo CGT, Huang WC, He X, Miura K, Razi A, Ortega J, Karttunen M, Lovell JF. Experimental and Computational Observations of Immunogenic Cobalt Porphyrin Lipid Bilayers: Nanodomain-Enhanced Antigen Association. Pharmaceutics 2021; 13:pharmaceutics13010098. [PMID: 33466686 PMCID: PMC7828809 DOI: 10.3390/pharmaceutics13010098] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/18/2022] Open
Abstract
Cobalt porphyrin phospholipid (CoPoP) can incorporate within bilayers to enable non-covalent surface-display of antigens on liposomes by mixing with proteins bearing a polyhistidine tag (his-tag); however, the mechanisms for how this occurs are poorly understood. These were investigated using the his-tagged model antigen Pfs25, a protein antigen candidate for malaria transmission-blocking vaccines. Pfs25 was found to associate with the small molecule aquocobalamin, a form of vitamin B12 and a cobalt-containing corrin macrocycle, but without particle formation, enabling comparative assessment. Relative to CoPoP liposomes, binding and serum stability studies indicated a weaker association of Pfs25 to aquocobalamin or cobalt nitrilotriacetic acid (Co-NTA) liposomes, which have cobalt displayed in the aqueous phase on lipid headgroups. Antigen internalization by macrophages was enhanced with Pfs25 bound to CoPoP liposomes. Immunization in mice with Pfs25 bound to CoPoP liposomes elicited antibodies that recognized ookinetes and showed transmission-reducing activity. To explore the physical mechanisms involved, we employed molecular dynamics (MD) simulations of bilayers containing phospholipid, cholesterol, as well as either CoPoP or NTA-functionalized lipids. The results show that the CoPoP-containing bilayer creates nanodomains that allow access for a limited but sufficient amount of water molecules that could be replaced by his-tags due to their favorable free energy properties allowing for stabilization. The position of the metal center within the NTA liposomes was much more exposed to the aqueous environment, which could explain its limited capacity for stabilizing Pfs25. This study illustrates the impact of CoPoP-induced antigen particleization in enhancing vaccine efficacy, and provides molecular insights into the CoPoP bilayer properties that enable this.
Collapse
Affiliation(s)
- Jasmin Federizon
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA; (J.F.); (W.-C.H.); (X.H.)
| | | | - Wei-Chiao Huang
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA; (J.F.); (W.-C.H.); (X.H.)
| | - Xuedan He
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA; (J.F.); (W.-C.H.); (X.H.)
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA;
| | - Aida Razi
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada; (A.R.); (J.O.)
| | - Joaquin Ortega
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada; (A.R.); (J.O.)
| | - Mikko Karttunen
- Department of Chemistry, the University of Western Ontario, London, ON N6A 3K7, Canada;
- Centre for Advanced Materials and Biomaterials Research, the University of Western Ontario, London, ON N6A 3K7, Canada
- Department of Applied Mathematics, the University of Western Ontario, London, ON N6A 5B7, Canada
- Correspondence: (M.K.); (J.F.L.)
| | - Jonathan F. Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA; (J.F.); (W.-C.H.); (X.H.)
- Correspondence: (M.K.); (J.F.L.)
| |
Collapse
|
28
|
Davoodi S, Bolhassani A, Namazi F. In vivo delivery of a multiepitope peptide and Nef protein using novel cell-penetrating peptides for development of HIV-1 vaccine candidate. Biotechnol Lett 2021; 43:547-559. [PMID: 33386500 PMCID: PMC7775797 DOI: 10.1007/s10529-020-03060-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES A potent HIV vaccine should overcome some limitations such as polymorphism of human HLA, the diversity of HIV-1 virus, and the lack of an effective delivery system. In this study, a DNA construct encoding Nef60-84, Nef126-144, Vpr34-47, Vpr60-75, Gp16030-53, Gp160308-323, and P248-151 epitopes was designed using bioinformatics tools. The pcDNA3.1-nef-vpr-gp160-p24 and pcDNA3.1-nef constructs were prepared in large scale as endotoxin-free form. Moreover, the recombinant Nef-Vpr-Gp160-p24 polypeptide and Nef protein were generated inE. coli. These constructs were delivered using cell penetrating peptides (CPPs) in vivo, and immune responses were assessed for different modalities in BALB/c mice. RESULTS The recombinant DNA constructs were confirmed as the ~ 867 bp and ~ 648 bp bands related tonef-vpr-gp160-p24 andnef genes on agarose gel. Moreover, the purified Nef-Vpr-Gp160-p24 polypeptide and Nef protein showed the ~ 32 kDa and ~ 30 kDa bands on SDS-PAGE, respectively. The results of immune responses indicated that the heterologous prime/boost regimens using both Nef-Vpr-Gp160-P24 and Nef antigens induced significantly the secretion of IgG2a, IgG2b, IFN-γ and Granzyme B compared to other groups. The levels of Granzyme B in mice immunized with Nef antigen were higher than those immunized with Nef-Vpr-Gp160-P24 antigen. The CPPs showed the same potency with Montanide adjuvant for eliciting immune responses. CONCLUSIONS The heterologous prime/boost regimens for both antigens could significantly direct immune responses toward Th1 and CTL activity compared to other regimens. Comparing the efficiency of Nef-Vpr-Gp160-P24 and Nef constructs, the Nef-Vpr-Gp160-P24 constructs delivered by CPPs showed promising results as an HIV vaccine candidate.
Collapse
Affiliation(s)
- Saba Davoodi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| | - Fatemeh Namazi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
29
|
Breda LCD, Menezes IG, Paulo LNM, de Almeida SR. Immune Sensing and Potential Immunotherapeutic Approaches to Control Chromoblastomycosis. J Fungi (Basel) 2020; 7:jof7010003. [PMID: 33375204 PMCID: PMC7822212 DOI: 10.3390/jof7010003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/30/2022] Open
Abstract
Chromoblastomycosis (CBM) is a neglected, chronic, and progressive subcutaneous mycosis caused by different species of fungi from the Herpotrichiellaceae family. CBM disease is usually associated with agricultural activities, and its infection is characterized by verrucous, erythematous papules, and atrophic lesions on the upper and lower limbs, leading to social stigma and impacts on patients' welfare. The economic aspect of disease treatment is another relevant issue. There is no specific treatment for CBM, and different anti-fungal drug associations are used to treat the patients. However, the long period of the disease and the high cost of the treatment lead to treatment interruption and, consequently, relapse of the disease. In previous years, great progress had been made in the comprehension of the CBM pathophysiology. In this review, we discuss the differences in the cell wall composition of conidia, hyphae, and muriform cells, with a particular focus on the activation of the host immune response. We also highlight the importance of studies about the host skin immunology in CBM. Finally, we explore different immunotherapeutic studies, highlighting the importance of these approaches for future treatment strategies for CBM.
Collapse
|
30
|
Gogoi P, Shakya A, Ghosh SK, Gogoi N, Gahtori P, Singh N, Bhattacharyya DR, Singh UP, Bhat HR. In silico study, synthesis, and evaluation of the antimalarial activity of hybrid dimethoxy pyrazole 1,3,5-triazine derivatives. J Biochem Mol Toxicol 2020; 35:e22682. [PMID: 33332673 DOI: 10.1002/jbt.22682] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/13/2020] [Accepted: 11/26/2020] [Indexed: 01/06/2023]
Abstract
Malaria continues to become a major global health problem, particularly in Sub-Saharan Africa, Asia, and Latin America. The widespread emergence of resistance to first-line drugs has further bolstered an urgent need for a new and cost-effective antimalarial(s). Thus, the present study enumerates the synthesis of novel hybrid dimethoxy pyrazole 1,3,5-triazine derivatives 7(a-j) and their in silico results short-listed three compounds with good binding energies and dock scores. Docking analysis shows that hydrogen-bonding predominates and typically involves key residues, such as Asp54, Tyr170, Ile164, and Arg122. The in vitro antimalarial evaluation of three top-ranked compounds (7e, 7g, and 7h) showed half-maximal inhibitory concentration values range from 53.85 to 100 μg/ml against chloroquine-sensitive strain 3D7 of Plasmodium falciparum. Compound 7e may be utilized as a lead for further optimization work in drug discovery due to good antimalarial activity.
Collapse
Affiliation(s)
- Pinku Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, India
| | - Anshul Shakya
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, India
| | - Surajit K Ghosh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, India
| | - Neelutpal Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, India
| | - Prashant Gahtori
- School of Pharmacy, Graphic Era Hill University, Dehradun, Uttarakhand, India
| | - Nardev Singh
- School of Pharmacy, Graphic Era Hill University, Dehradun, Uttarakhand, India
| | - Dibya R Bhattacharyya
- Regional Medical Research Centre, Indian Council of Medical Research (ICMR), Dibrugarh, Assam, India
| | - Udaya P Singh
- Department of Pharmaceutical Sciences, Drug Design and Discovery Laboratory, Sam Higginbottom University of Agriculture Technology and Sciences, Allahabad, India
| | - Hans R Bhat
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, India
| |
Collapse
|
31
|
Mordmüller B, Sulyok M, Egger-Adam D, Resende M, de Jongh WA, Jensen MH, Smedegaard HH, Ditlev SB, Soegaard M, Poulsen L, Dyring C, Calle CL, Knoblich A, Ibáñez J, Esen M, Deloron P, Ndam N, Issifou S, Houard S, Howard RF, Reed SG, Leroy O, Luty AJF, Theander TG, Kremsner PG, Salanti A, Nielsen MA. First-in-human, Randomized, Double-blind Clinical Trial of Differentially Adjuvanted PAMVAC, A Vaccine Candidate to Prevent Pregnancy-associated Malaria. Clin Infect Dis 2020; 69:1509-1516. [PMID: 30629148 PMCID: PMC6792113 DOI: 10.1093/cid/ciy1140] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 01/03/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Malaria in pregnancy has major impacts on mother and child health. To complement existing interventions, such as intermittent preventive treatment and use of impregnated bed nets, we developed a malaria vaccine candidate with the aim of reducing sequestration of asexual "blood-stage" parasites in the placenta, the major virulence mechanism. METHODS The vaccine candidate PAMVAC is based on a recombinant fragment of VAR2CSA, the Plasmodium falciparum protein responsible for binding to the placenta via chondroitin sulfate A (CSA). Healthy, adult malaria-naive volunteers were immunized with 3 intramuscular injections of 20 μg (n = 9) or 50 μg (n = 27) PAMVAC, adjuvanted with Alhydrogel or glucopyranosyl lipid adjuvant in stable emulsion (GLA-SE) or in a liposomal formulation with QS21 (GLA-LSQ). Allocation was random and double blind. The vaccine was given every 4 weeks. Volunteers were observed for 6 months following last immunization. RESULTS All PAMVAC formulations were safe and well tolerated. A total of 262 adverse events (AEs) occurred, 94 (10 grade 2 and 2 grade 3) at least possibly related to the vaccine. No serious AEs occurred. Distribution and severity of AEs were similar in all arms. PAMVAC was immunogenic in all participants. PAMVAC-specific antibody levels were highest with PAMVAC-GLA-SE. The antibodies inhibited binding of VAR2CSA expressing P. falciparum-infected erythrocytes to CSA in a standardized functional assay. CONCLUSIONS PAMVAC formulated with Alhydrogel or GLA-based adjuvants was safe, well tolerated, and induced functionally active antibodies. Next, PAMVAC will be assessed in women before first pregnancies in an endemic area. CLINICAL TRIALS REGISTRATION EudraCT 2015-001827-21; ClinicalTrials.gov NCT02647489.
Collapse
Affiliation(s)
- Benjamin Mordmüller
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany.,Centre de Recherches Médicales de Lambaréné, Gabon
| | - Mihály Sulyok
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany
| | - Diane Egger-Adam
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany
| | - Mafalda Resende
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | | | - Mette H Jensen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | - Helle Holm Smedegaard
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | - Sisse B Ditlev
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | | | | | | | - Carlos Lamsfus Calle
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany
| | - Annette Knoblich
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany
| | - Javier Ibáñez
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany
| | - Meral Esen
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany.,Centre de Recherches Médicales de Lambaréné, Gabon
| | - Philippe Deloron
- Mère et Enfant face aux Infections Tropicales, Institut de Recherche pour le Développement, Université Paris 5, Sorbonne Paris Cité, France
| | - Nicaise Ndam
- Mère et Enfant face aux Infections Tropicales, Institut de Recherche pour le Développement, Université Paris 5, Sorbonne Paris Cité, France
| | - Saadou Issifou
- Fondation pour la Recherche Scientifique and Institut de Recherche Clinique du Bénin, Cotonou
| | | | | | - Steven G Reed
- Infectious Disease Research Institute, Seattle, Washington
| | - Odile Leroy
- European Vaccine Initiative, Heidelberg, Germany
| | - Adrian J F Luty
- Mère et Enfant face aux Infections Tropicales, Institut de Recherche pour le Développement, Université Paris 5, Sorbonne Paris Cité, France
| | - Thor G Theander
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | - Peter G Kremsner
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany.,Centre de Recherches Médicales de Lambaréné, Gabon
| | - Ali Salanti
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | - Morten A Nielsen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| |
Collapse
|
32
|
Skwarczynski M, Chandrudu S, Rigau-Planella B, Islam MT, Cheong YS, Liu G, Wang X, Toth I, Hussein WM. Progress in the Development of Subunit Vaccines against Malaria. Vaccines (Basel) 2020; 8:vaccines8030373. [PMID: 32664421 PMCID: PMC7563759 DOI: 10.3390/vaccines8030373] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/02/2022] Open
Abstract
Malaria is a life-threatening disease and one of the main causes of morbidity and mortality in the human population. The disease also results in a major socio-economic burden. The rapid spread of malaria epidemics in developing countries is exacerbated by the rise in drug-resistant parasites and insecticide-resistant mosquitoes. At present, malaria research is focused mainly on the development of drugs with increased therapeutic effects against Plasmodium parasites. However, a vaccine against the disease is preferable over treatment to achieve long-term control. Trials to develop a safe and effective immunization protocol for the control of malaria have been occurring for decades, and continue on today; still, no effective vaccines are available on the market. Recently, peptide-based vaccines have become an attractive alternative approach. These vaccines utilize short protein fragments to induce immune responses against malaria parasites. Peptide-based vaccines are safer than traditional vaccines, relatively inexpensive to produce, and can be composed of multiple T- and B-cell epitopes integrated into one antigenic formulation. Various combinations, based on antigen choice, peptide epitope modification and delivery mechanism, have resulted in numerous potential malaria vaccines candidates; these are presently being studied in both preclinical and clinical trials. This review describes the current landscape of peptide-based vaccines, and addresses obstacles and opportunities in the production of malaria vaccines.
Collapse
Affiliation(s)
- Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Saranya Chandrudu
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Berta Rigau-Planella
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Md. Tanjir Islam
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Yee S. Cheong
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Genan Liu
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Xiumin Wang
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- Correspondence: (I.T.); (W.M.H.)
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
- Correspondence: (I.T.); (W.M.H.)
| |
Collapse
|
33
|
Ragotte RJ, Higgins MK, Draper SJ. The RH5-CyRPA-Ripr Complex as a Malaria Vaccine Target. Trends Parasitol 2020; 36:545-559. [PMID: 32359873 PMCID: PMC7246332 DOI: 10.1016/j.pt.2020.04.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 11/04/2022]
Abstract
Despite ongoing efforts, a highly effective vaccine against Plasmodium falciparum remains elusive. Vaccines targeting the pre-erythrocytic stages of the P. falciparum life cycle are the most advanced to date, affording moderate levels of efficacy in field trials. However, the discovery that the members of the merozoite PfRH5-PfCyRPA-PfRipr (RCR) complex are capable of inducing strain-transcendent neutralizing antibodies has renewed enthusiasm for the possibility of preventing disease by targeting the parasite during the blood stage of infection. With Phase I/II clinical trials now underway using first-generation vaccines against PfRH5, and more on the horizon for PfCyRPA and PfRipr, this review explores the rationale and future potential of the RCR complex as a P. falciparum vaccine target.
Collapse
Affiliation(s)
- Robert J Ragotte
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK.
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
34
|
Parzych EM, Miura K, Long CA, Burns JM. Maintaining immunogenicity of blood stage and sexual stage subunit malaria vaccines when formulated in combination. PLoS One 2020; 15:e0232355. [PMID: 32348377 PMCID: PMC7190115 DOI: 10.1371/journal.pone.0232355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/13/2020] [Indexed: 11/18/2022] Open
Abstract
Background Eradication of Plasmodium falciparum malaria will likely require a multivalent vaccine, but the development of a highly efficacious subunit-based formulation has been challenging. We previously showed that production and immunogenicity of two leading vaccine targets, PfMSP119 (blood-stage) and Pfs25 (sexual stage), could be enhanced upon genetic fusion to merozoite surface protein 8 (PfMSP8). Here, we sought to optimize a Pfs25-based formulation for use in combination with rPfMSP1/8 with the goal of maintaining the immunogenicity of each subunit. Methods Comparative mouse studies were conducted to assess the effects of adjuvant selection (Alhydrogel vs. glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE)) and antigen dose (2.5 vs. 0.5 μg) on the induction of anti-Pfs25 immune responses. The antibody response (magnitude, IgG subclass profile, and transmission-reducing activity (TRA)) and cellular responses (proliferation, cytokine production) generated in response to each formulation were assessed. Similarly, immunogenicity of a bivalent vaccine containing rPfMSP1/8 and rPfs25/8 was evaluated. Results Alum-based formulations elicited strong and comparable humoral and cellular responses regardless of antigen form (unfused rPfs25 or chimeric rPfs25/8) or dose. In contrast, GLA-SE based formulations elicited differential responses as a function of both parameters, with 2.5 μg of rPfs25/8 inducing the highest titers of functional anti-Pfs25 antibodies. Based on these data, chimeric rPfs25/8 was selected and tested in a bivalent formulation with rPfMSP1/8. Strong antibody titers against Pfs25 and PfMSP119 domains were induced with GLA-SE based formulations, with no indication of antigenic competition. Conclusions We were able to generate an immunogenic bivalent vaccine designed to target multiple parasite stages that could reduce both clinical disease and parasite transmission. The use of the same PfMSP8 carrier for two different vaccine components was effective in this bivalent formulation. As such, the incorporation of additional protective targets fused to the PfMSP8 carrier into the formulation should be feasible, further broadening the protective response.
Collapse
Affiliation(s)
- Elizabeth M. Parzych
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Kazutoyo Miura
- Malaria Immunology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Carole A. Long
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- Malaria Immunology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - James M. Burns
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
35
|
Brown DD, Solomon S, Lerner D, Del Rio M. Malaria and acute kidney injury. Pediatr Nephrol 2020; 35:603-608. [PMID: 30706124 DOI: 10.1007/s00467-018-4191-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/19/2018] [Accepted: 12/28/2018] [Indexed: 10/27/2022]
Abstract
Malaria is a parasitic infection transmitted by mosquitos, resulting in significant morbidity and mortality. It affects 212 million worldwide, causing death in up to 303,000 children annually. In the USA, up to 1700 people are affected yearly. Although the prevalence in developed countries is less than in developing countries, travelers from low transmission areas, and those from endemic areas who later return, are very susceptible to malaria and its complications. Severe malaria can cause significant multiorgan dysfunction including acute kidney injury (AKI). The pathogenesis is not clearly understood but proposed mechanisms include acute tubular necrosis (ATN) due to impediments in renal microcirculation, infection-triggered proinflammatory reactions within the kidney, and metabolic disturbances. Providers must consider malarial infection in cases of AKI in someone with a travel history, as early recognition and treatment are crucial to improving outcomes. This article will review malaria-induced AKI in order to provide a better understanding of this infection's effect on the kidneys.
Collapse
Affiliation(s)
- Denver D Brown
- Department of Pediatric Nephrology at The Children's Hospital at Montefiore, 3326 Bainbridge Ave, Bronx, NY, 10029, USA.
| | - Sonia Solomon
- Department of Pediatric Nephrology at The Children's Hospital at Montefiore, 3326 Bainbridge Ave, Bronx, NY, 10029, USA
| | - Daniele Lerner
- Department of Pediatric Nephrology at The Children's Hospital at Montefiore, 3326 Bainbridge Ave, Bronx, NY, 10029, USA
| | - Marcela Del Rio
- Department of Pediatric Nephrology at The Children's Hospital at Montefiore, 3326 Bainbridge Ave, Bronx, NY, 10029, USA
| |
Collapse
|
36
|
Warszawski S, Dekel E, Campeotto I, Marshall JM, Wright KE, Lyth O, Knop O, Regev-Rudzki N, Higgins MK, Draper SJ, Baum J, Fleishman SJ. Design of a basigin-mimicking inhibitor targeting the malaria invasion protein RH5. Proteins 2020; 88:187-195. [PMID: 31325330 PMCID: PMC6904230 DOI: 10.1002/prot.25786] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/02/2019] [Accepted: 07/12/2019] [Indexed: 11/07/2022]
Abstract
Many human pathogens use host cell-surface receptors to attach and invade cells. Often, the host-pathogen interaction affinity is low, presenting opportunities to block invasion using a soluble, high-affinity mimic of the host protein. The Plasmodium falciparum reticulocyte-binding protein homolog 5 (RH5) provides an exciting candidate for mimicry: it is highly conserved and its moderate affinity binding to the human receptor basigin (KD ≥1 μM) is an essential step in erythrocyte invasion by this malaria parasite. We used deep mutational scanning of a soluble fragment of human basigin to systematically characterize point mutations that enhance basigin affinity for RH5 and then used Rosetta to design a variant within the sequence space of affinity-enhancing mutations. The resulting seven-mutation design exhibited 1900-fold higher affinity (KD approximately 1 nM) for RH5 with a very slow binding off rate (0.23 h-1 ) and reduced the effective Plasmodium growth-inhibitory concentration by at least 10-fold compared to human basigin. The design provides a favorable starting point for engineering on-rate improvements that are likely to be essential to reach therapeutically effective growth inhibition.
Collapse
Affiliation(s)
- Shira Warszawski
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elya Dekel
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ivan Campeotto
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Jennifer M. Marshall
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Katherine E. Wright
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK
| | - Oliver Lyth
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK
| | - Orli Knop
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Neta Regev-Rudzki
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Simon J Draper
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK
| | - Sarel J Fleishman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
37
|
Characterization of drug resistance and genetic diversity of Plasmodium falciparum parasites from Tripura, Northeast India. Sci Rep 2019; 9:13704. [PMID: 31548652 PMCID: PMC6757058 DOI: 10.1038/s41598-019-50152-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 09/06/2019] [Indexed: 01/23/2023] Open
Abstract
Monitoring of anti-malarial drug resistance is vital in Northeast India as this region shares its international border with Southeast Asia. Genetic diversity of Plasmodium parasites regulates transmission dynamics, disease severity and vaccine efficacy. P. falciparum chloroquine resistance transporter (Pfcrt), multidrug resistance-1 (Pfmdr-1) and kelch 13 propeller (PfK-13) genes which govern antimalarial drug resistance and three genetic diversity markers, merozoite surface protein 1 and 2 (Pfmsp-1, Pfmsp-2) and glutamate rich protein (Pfglurp) were evaluated from Tripura, Northeast India using molecular tools. In the Pfcrt gene, 87% isolates showed triple mutations at codons M74I, N75E and K76T. 12.5% isolates in Pfmdr-1 gene showed mutation at N86Y. No polymorphism in PfK-13 propeller was found. Polyclonal infections were observed in 53.85% isolates and more commonly in adults (p = 0.0494). In the Pfmsp-1 locus, the K1 allelic family was predominant (71.2%) followed by the 3D7/IC family (69.2%) in the Pfmsp-2 locus. RII region of Pfglurp exhibited nine alleles with expected heterozygosity of 0.85. The multiplicity of infection for Pfmsp-1, Pfmsp-2 and Pfglurp were 1.56, 1.31 and 1.06 respectively. Overall, the study demonstrated a high level of chloroquine resistance and extensive parasite diversity in the region, necessitating regular surveillance in this population group.
Collapse
|
38
|
Chan JA, Wetzel D, Reiling L, Miura K, Drew DR, Gilson PR, Anderson DA, Richards JS, Long CA, Suckow M, Jenzelewski V, Tsuboi T, Boyle MJ, Piontek M, Beeson JG. Malaria vaccine candidates displayed on novel virus-like particles are immunogenic and induce transmission-blocking activity. PLoS One 2019; 14:e0221733. [PMID: 31504038 PMCID: PMC6736250 DOI: 10.1371/journal.pone.0221733] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/13/2019] [Indexed: 01/23/2023] Open
Abstract
The development of effective malaria vaccines remains a global health priority. Currently, the most advanced vaccine, known as RTS,S, has only shown modest efficacy in clinical trials. Thus, the development of more efficacious vaccines by improving the formulation of RTS,S for increased efficacy or to interrupt malaria transmission are urgently needed. The RTS,S vaccine is based on the presentation of a fragment of the sporozoite antigen on the surface of virus-like particles (VLPs) based on human hepatitis B virus (HBV). In this study, we have developed and evaluated a novel VLP platform based on duck HBV (known as Metavax) for malaria vaccine development. This platform can incorporate large and complex proteins into VLPs and is produced in a Hansenula cell line compatible with cGMP vaccine production. Here, we have established the expression of leading P. falciparum malaria vaccine candidates as VLPs. This includes Pfs230 and Pfs25, which are candidate transmission-blocking vaccine antigens. We demonstrated that the VLPs effectively induce antibodies to malaria vaccine candidates with minimal induction of antibodies to the duck-HBV scaffold antigen. Antibodies to Pfs230 also recognised native protein on the surface of gametocytes, and antibodies to both Pfs230 and Pfs25 demonstrated transmission-reducing activity in standard membrane feeding assays. These results establish the potential utility of this VLP platform for malaria vaccines, which may be suitable for the development of multi-component vaccines that achieve high vaccine efficacy and transmission-blocking immunity.
Collapse
Affiliation(s)
- Jo-Anne Chan
- Burnet Institute, Life Sciences, Melbourne, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, VIC, Australia
| | - David Wetzel
- ARTES Biotechnology GmbH, Langenfeld, Germany
- Technical University of Dortmund, Laboratory of Plant and Process Design, Dortmund, Germany
| | - Linda Reiling
- Burnet Institute, Life Sciences, Melbourne, VIC, Australia
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, United States of America
| | - Damien R. Drew
- Burnet Institute, Life Sciences, Melbourne, VIC, Australia
| | - Paul R. Gilson
- Burnet Institute, Life Sciences, Melbourne, VIC, Australia
| | | | - Jack S. Richards
- Burnet Institute, Life Sciences, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, VIC, Australia
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, United States of America
| | | | | | - Takafumi Tsuboi
- Proteo-Science Centre, Ehime University, Matsuyama, Ehime, Japan
| | - Michelle J. Boyle
- Immunology Department, QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | | | - James G. Beeson
- Burnet Institute, Life Sciences, Melbourne, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, VIC, Australia
- Department of Medicine, University of Melbourne, VIC, Australia
- * E-mail:
| |
Collapse
|
39
|
Labbé GM, Miura K, Silk SE, Gu W, Moon JE, Jin J, Payne RO, Fay MP, Dutta S, Long CA, Draper SJ. Harmonization study between three laboratories for expression of malaria vaccine clinical trial IgG antibody ELISA data in µg/mL. Malar J 2019; 18:300. [PMID: 31477111 PMCID: PMC6721210 DOI: 10.1186/s12936-019-2935-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 08/24/2019] [Indexed: 11/11/2022] Open
Abstract
Background The ability to report vaccine-induced IgG responses in terms of µg/mL, as opposed arbitrary units (AU), enables a more informed interpretation of the magnitude of the immune response, and better comparison between vaccines targeting different antigens. However, these interpretations rely on the accuracy of the methodology, which is used to generate ELISA data in µg/mL. In a previous clinical trial of a vaccine targeting the apical membrane antigen 1 (AMA1) from Plasmodium falciparum, three laboratories (Oxford, NIH and WRAIR) reported ELISA data in µg/mL that were correlated but not concordant. This current study sought to harmonize the methodology used to generate a conversion factor (CF) for ELISA analysis of human anti-AMA1 IgG responses across the three laboratories. Methods Purified IgG was distributed to the three laboratories and, following a set protocol provided by NIH, AMA1-specific human IgG was affinity purified. A new “harmonized CF” was generated by each laboratory using their in-house ELISA, and the original clinical trial ELISA data were re-analysed accordingly. Results Statistical analysis showed that the data remained highly correlated across all three laboratories, although only Oxford and NIH were able to harmonize their CF for ELISA and generate concordant data. Conclusions This study enabled two out of the three laboratories to harmonize their µg/mL readouts for the human anti-AMA1 IgG ELISA, but results reported from WRAIR are ~ twofold higher. Given the need to validate such information for each species and antigen of interest, it is important to bear in mind these likely differences when interpreting µg/mL ELISA data in the future.
Collapse
Affiliation(s)
- Geneviève M Labbé
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD, 20852, USA
| | - Sarah E Silk
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Wenjuan Gu
- Biostatistics Research Branch, NIAID/NIH, Rockville, MD, 20852, USA
| | - James E Moon
- Military Malaria Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Jing Jin
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Ruth O Payne
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Michael P Fay
- Biostatistics Research Branch, NIAID/NIH, Rockville, MD, 20852, USA
| | - Sheetij Dutta
- Military Malaria Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD, 20852, USA
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
| |
Collapse
|
40
|
Burns AL, Dans MG, Balbin JM, de Koning-Ward TF, Gilson PR, Beeson JG, Boyle MJ, Wilson DW. Targeting malaria parasite invasion of red blood cells as an antimalarial strategy. FEMS Microbiol Rev 2019; 43:223-238. [PMID: 30753425 PMCID: PMC6524681 DOI: 10.1093/femsre/fuz005] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
Plasmodium spp. parasites that cause malaria disease remain a significant global-health burden. With the spread of parasites resistant to artemisinin combination therapies in Southeast Asia, there is a growing need to develop new antimalarials with novel targets. Invasion of the red blood cell by Plasmodium merozoites is essential for parasite survival and proliferation, thus representing an attractive target for therapeutic development. Red blood cell invasion requires a co-ordinated series of protein/protein interactions, protease cleavage events, intracellular signals, organelle release and engagement of an actin-myosin motor, which provide many potential targets for drug development. As these steps occur in the bloodstream, they are directly susceptible and exposed to drugs. A number of invasion inhibitors against a diverse range of parasite proteins involved in these different processes of invasion have been identified, with several showing potential to be optimised for improved drug-like properties. In this review, we discuss red blood cell invasion as a drug target and highlight a number of approaches for developing antimalarials with invasion inhibitory activity to use in future combination therapies.
Collapse
Affiliation(s)
- Amy L Burns
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia 5005
| | - Madeline G Dans
- Burnet Institute, Melbourne, Victoria, Australia 3004.,Deakin University, School of Medicine, Waurn Ponds, Victoria, Australia 3216
| | - Juan M Balbin
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia 5005
| | | | - Paul R Gilson
- Burnet Institute, Melbourne, Victoria, Australia 3004
| | - James G Beeson
- Burnet Institute, Melbourne, Victoria, Australia 3004.,Central Clinical School and Department of Microbiology, Monash University 3004.,Department of Medicine, University of Melbourne, Australia 3052
| | - Michelle J Boyle
- Burnet Institute, Melbourne, Victoria, Australia 3004.,QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia 4006
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia 5005.,Burnet Institute, Melbourne, Victoria, Australia 3004
| |
Collapse
|
41
|
Outer membrane protein complex as a carrier for malaria transmission blocking antigen Pfs230. NPJ Vaccines 2019; 4:24. [PMID: 31312527 PMCID: PMC6614402 DOI: 10.1038/s41541-019-0121-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/07/2019] [Indexed: 12/12/2022] Open
Abstract
Malaria transmission blocking vaccines (TBV) target the mosquito stage of parasite development by passive immunization of mosquitoes feeding on a vaccinated human. Through uptake of vaccine-induced antibodies in a blood meal, mosquito infection is halted and hence transmission to another human host is blocked. Pfs230 is a gametocyte and gamete surface antigen currently under clinical evaluation as a TBV candidate. We have previously shown that chemical conjugation of poorly immunogenic TBV antigens to Exoprotein A (EPA) can enhance their immunogenicity. Here, we assessed Outer Membrane Protein Complex (OMPC), a membrane vesicle derived from Neisseria meningitidis, as a carrier for Pfs230. We prepared Pfs230-OMPC conjugates with varying levels of antigen load and examined immunogenicity in mice. Chemical conjugation of Pfs230 to OMPC enhanced immunogenicity and functional activity of the Pfs230 antigen, and OMPC conjugates achieved 2-fold to 20-fold higher antibody titers than Pfs230-EPA/AdjuPhos® at different doses. OMPC conjugates were highly immunogenic even at low doses, indicating a dose-sparing effect. EPA conjugates induced an IgG subclass profile biased towards a Th2 response, whereas OMPC conjugates induced a strong Th1-biased immune response with high levels of IgG2, which can benefit Pfs230 antibody functional activity, which depends on complement activation. OMPC is a promising carrier for Pfs230 vaccines. Malaria transmission blocking vaccines (TBV) target Plasmodium stages that transmit between human and mosquitos in order to interrupt the parasite’s life cycle and reduce spread. One TBV antigen currently under clinical development is Pf230, which is expressed on sexual Plasmodium stages. In this study, led by Patrick Duffy from the NIAID, researchers improve immunogenicity of Pf230. They chemically conjugate a part of Pf230 to membrane vesicles derived from bacteria, so-called outer membrane protein complexes (OMPC). Immunization of mice with Pf230-OMPC elicits a higher antibody response and a more balanced IgG subclass profile than control immunizations. Serum from Pf230-OMPC-vaccinated mice efficiently blocks infection of mosquitoes. These results with mice encourage further pre-clinical and clinical characterization of OMPC as a carrier for TBV antigens.
Collapse
|
42
|
Pattinson DJ, Apte SH, Wibowo N, Chuan YP, Rivera-Hernandez T, Groves PL, Lua LH, Middelberg APJ, Doolan DL. Chimeric Murine Polyomavirus Virus-Like Particles Induce Plasmodium Antigen-Specific CD8 + T Cell and Antibody Responses. Front Cell Infect Microbiol 2019; 9:215. [PMID: 31275867 PMCID: PMC6593135 DOI: 10.3389/fcimb.2019.00215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/03/2019] [Indexed: 12/28/2022] Open
Abstract
An effective vaccine against the Plasmodium parasite is likely to require the induction of robust antibody and T cell responses. Chimeric virus-like particles are an effective vaccine platform for induction of antibody responses, but their capacity to induce robust cellular responses and cell-mediated protection against pathogen challenge has not been established. To evaluate this, we produced chimeric constructs using the murine polyomavirus structural protein with surface-exposed CD8+ or CD4+ T cell or B cell repeat epitopes derived from the Plasmodium yoelii circumsporozoite protein, and assessed immunogenicity and protective capacity in a murine model. Robust CD8+ T cell responses were induced by immunization with the chimeric CD8+ T cell epitope virus-like particles, however CD4+ T cell responses were very low. The B cell chimeric construct induced robust antibody responses but there was no apparent synergy when T cell and B cell constructs were administered as a pool. A heterologous prime/boost regimen using plasmid DNA priming followed by a VLP boost was more effective than homologous VLP immunization for cellular immunity and protection. These data show that chimeric murine polyomavirus virus-like particles are a good platform for induction of CD8+ T cell responses as well as antibody responses.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan
- Antibody Formation/immunology
- Antigens, Protozoan/immunology
- B-Lymphocytes
- CD4-Positive T-Lymphocytes
- CD8-Positive T-Lymphocytes/immunology
- Disease Models, Animal
- Epitopes, B-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/immunology
- Immunity, Cellular
- Immunization
- Immunization, Secondary
- Malaria Vaccines
- Mice
- Mice, Inbred BALB C
- Plasmodium yoelii
- Polyomavirus/genetics
- Polyomavirus/immunology
- Protozoan Proteins/immunology
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- David J. Pattinson
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Simon H. Apte
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nani Wibowo
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Yap P. Chuan
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Tania Rivera-Hernandez
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Penny L. Groves
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Linda H. Lua
- Protein Expression Facility, University of Queensland, Brisbane, QLD, Australia
| | - Anton P. J. Middelberg
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Denise L. Doolan
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
43
|
Combining Monophosphoryl Lipid A (MPL), CpG Oligodeoxynucleotide (ODN), and QS-21 Adjuvants Induces Strong and Persistent Functional Antibodies and T Cell Responses against Cell-Traversal Protein for Ookinetes and Sporozoites (CelTOS) of Plasmodium falciparum in BALB/c Mice. Infect Immun 2019; 87:IAI.00911-18. [PMID: 30936155 DOI: 10.1128/iai.00911-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 03/17/2019] [Indexed: 12/22/2022] Open
Abstract
Plasmodium falciparum cell-traversal protein for ookinetes and sporozoites (PfCelTOS) is an advanced vaccine candidate that has a crucial role in the traversal of the malaria parasite in both mosquito and mammalian hosts. As recombinant purified proteins are normally poor immunogens, they require to be admixed with an adjuvant(s); therefore, the objective of the present study was to evaluate the capacity of different vaccine adjuvants, monophosphoryl lipid A (MPL), CpG, and Quillaja saponaria Molina fraction 21 (QS-21), alone or in combination (MCQ [MPL/CpG/QS-21]), to enhance the immunogenicity of Escherichia coli-expressed PfCelTOS in BALB/c mice. This goal was achieved by the assessment of anti-PfCelTOS IgG antibodies (level, titer, IgG isotype profile, avidity, and persistence) and extracellular Th1 cytokines using an enzyme-linked immunosorbent assay (ELISA) on postimmunized BALB/c mouse sera and PfCelTOS-stimulated splenocytes, respectively. Also, an assessment of the transmission-reducing activity (TRA) of anti-PfCelTOS obtained from different vaccine groups was carried out in female Anopheles stephensi mosquitoes by using a standard membrane feeding assay (SMFA). In comparison to PfCelTOS alone, administration of PfCelTOS with three distinct potent Th1 adjuvants in vaccine mouse groups showed enhancement and improvement of PfCelTOS immunogenicity that generated more bias toward a Th1 response with significantly enhanced titers and avidity of the anti-PfCelTOS responses that could impair ookinete development in A. stephensi However, immunization of mice with PfCelTOS with MCQ mixture adjuvants resulted in the highest levels of induction of antibody titers, avidity, and inhibitory antibodies in oocyst development (88%/26.7% reductions in intensity/prevalence) in A. stephensi It could be suggested that adjuvant combinations with different mechanisms stimulate better functional antibody responses than adjuvants individually against challenging diseases such as malaria.
Collapse
|
44
|
Mehrizi AA, Ameri Torzani M, Zakeri S, Jafary Zadeh A, Babaeekhou L. Th1 immune response to Plasmodium falciparum recombinant thrombospondin-related adhesive protein (TRAP) antigen is enhanced by TLR3-specific adjuvant, poly(I:C) in BALB/c mice. Parasite Immunol 2019; 40:e12538. [PMID: 29799636 DOI: 10.1111/pim.12538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 05/21/2018] [Indexed: 12/12/2022]
Abstract
Sporozoite-based malaria vaccines have provided a gold standard for malaria vaccine development, and thrombospondin-related adhesive protein (TRAP) serves as the main vaccine candidate antigen on sporozoites. As recombinant malaria vaccine candidate antigens are poorly immunogenic, additional appropriate immunostimulants, such as an efficient adjuvant, are highly essential to modulate Th1-cell predominance and also to induce a protective and long-lived immune response. In this study, polyinosinic:polycytidylic acid [poly(I:C)], the ligand of TLR3, was considered as the potential adjuvant for vaccines targeting stronger Th1-based immune responses. For this purpose, BALB/c mice were immunized with rPfTRAP delivered in putative poly(I:C) adjuvant, and humoural and cellular immune responses were determined in different immunized mouse groups. Delivery of rPfTRAP with poly(I:C) induced high levels and titres of persisted and also high-avidity anti-rPfTRAP IgG antibodies comparable to complete Freund's adjuvant (CFA)/incomplete Freund's adjuvant (IFA) adjuvant after the second boost. In addition, rPfTRAP formulated with poly(I:C) elicited a higher ratio of IFN-γ/IL-5, IgG2a/IgG1, and IgG2b/IgG1 than with CFA/IFA, indicating that poly(I:C) supports the induction of a stronger Th1-based immune response. This is a first time study which reveals the potential of rPfTRAP delivery in poly(I:C) to increase the level, avidity and durability of both anti-PfTRAP cytophilic antibodies and Th1 cytokines.
Collapse
Affiliation(s)
- A A Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - M Ameri Torzani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.,Department of Biology, Islamshahr Branch, Islamic Azad University, Islamshahr, Iran
| | - S Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - A Jafary Zadeh
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - L Babaeekhou
- Department of Biology, Islamshahr Branch, Islamic Azad University, Islamshahr, Iran
| |
Collapse
|
45
|
Pirahmadi S, Zakeri S, A Mehrizi A, D Djadid N, Raz AA, J Sani J, Abbasi R, Ghorbanzadeh Z. Cell-traversal protein for ookinetes and sporozoites (CelTOS) formulated with potent TLR adjuvants induces high-affinity antibodies that inhibit Plasmodium falciparum infection in Anopheles stephensi. Malar J 2019; 18:146. [PMID: 31014347 PMCID: PMC6480871 DOI: 10.1186/s12936-019-2773-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/11/2019] [Indexed: 02/02/2023] Open
Abstract
Background Plasmodium falciparum parasite is the most deadly species of human malaria, and the development of an effective vaccine that prevents P. falciparum infection and transmission is a key target for malarial elimination and eradication programmes. P. falciparum cell-traversal protein for ookinetes and sporozoites (PfCelTOS) is an advanced vaccine candidate. A comparative study was performed to characterize the immune responses in BALB/c mouse immunized with Escherichia coli-expressed recombinant PfCelTOS (rPfCelTOS) in toll-like receptor (TLR)-based adjuvants, CpG and Poly I:C alone or in combination (CpG + Poly I:C), followed by the assessment of transmission-reducing activity (TRA) of anti-rPfCelTOS antibodies obtained from different vaccine groups in Anopheles stephensi. Methods The aim of the current work was achieved by head-to-head comparison of the vaccine groups using conventional and avidity enzyme-linked immunosorbent assay (ELISA), immunofluorescence test (IFAT), and standard membrane feeding assay (SMFA). Results Comparing to rPfCelTOS alone, administration of rPfCelTOS with two distinct TLR-based adjuvants in vaccine mouse groups showed a significant increase in responses (antibody level, IgG subclass analysis, avidity, and Th1 cytokines) and was able to induce reasonable transmission-reducing activity. Also, comparable functional activity of anti-rPfCelTOS antibodies was found in group that received antigen in either CpG or Poly I:C (69.9%/20% and 73.5%/24.4%, respectively, reductions in intensity/prevalence). However, the vaccine group receiving rPfCelTOS in combination with CpG + Poly I:C showed a significant induction in antibody titers and inhibitory antibodies in oocysts development (78.3%/19.6% reductions in intensity/prevalence) in An. stephensi. Conclusions A key finding in this investigation is that rPfCelTOS administered alone in BALB/c mouse is poorly immunogenic, with relatively low IgG level, avidity, inhibitory antibodies, and mixed Th1/Th2 responses. However, immunological characteristic (IgG level, cytophilic IgG2a and IgG2b, avidity, and Th1 cytokines) and TRA of anti-rPfCelTOS significantly enhanced in the presence of co-administration of TLR-based adjuvants, confirming that targeting TLRs would be an effective means for the enhancement of inducing TRA against rPfCelTOS. Electronic supplementary material The online version of this article (10.1186/s12936-019-2773-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran.
| | - Akram A Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran
| | - Navid D Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran
| | - Abbas-Ali Raz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran
| | - Jafar J Sani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran
| | - Ronak Abbasi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran
| | - Zahra Ghorbanzadeh
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran
| |
Collapse
|
46
|
Yusuf Y, Yoshii T, Iyori M, Yoshida K, Mizukami H, Fukumoto S, Yamamoto DS, Alam A, Emran TB, Amelia F, Islam A, Otsuka H, Takashima E, Tsuboi T, Yoshida S. Adeno-Associated Virus as an Effective Malaria Booster Vaccine Following Adenovirus Priming. Front Immunol 2019; 10:730. [PMID: 31024558 PMCID: PMC6460511 DOI: 10.3389/fimmu.2019.00730] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
An ideal malaria vaccine platform should potently induce protective immune responses and block parasite transmission from mosquito to human, and it should maintain these effects for an extended period. Here, we have focused on vaccine development based on adeno-associated virus serotype 1 (AAV1), a viral vector widely studied in the field of clinical gene therapy that is able to induce long-term transgene expression without causing toxicity in vivo. Our results show the potential utility of AAV1 vectors as an extremely potent booster vaccine to induce durable immunity when combined with an adenovirus-priming vaccine in a rodent malaria model. We generated a series of recombinant AAV1s and human adenovirus type 5 (AdHu5) expressing either Plasmodium falciparum circumsporozoite protein (PfCSP) or P25 (Pfs25) protein. Heterologous two-dose immunization with an AdHu5-prime and AAV1-boost (AdHu5-AAV1) elicited robust and durable PfCSP- or Pfs25-specific functional antibodies over 280 days. Regarding protective efficacy, AdHu5-AAV1 PfCSP achieved high sterile protection (up to 80% protection rate) against challenge with transgenic Plasmodium berghei sporozoites expressing PfCSP. When examining transmission-blocking (TB) efficacy, we found that immunization with AdHu5-AAV1 Pfs25 maintained TB activity in vivo against transgenic P. berghei expressing Pfs25 for 287 days (99% reduction in oocyst intensity, 85% reduction in oocyst prevalence). Our data indicate that AAV1-based malaria vaccines can confer potent and durable protection as well as TB efficacy when administered following an AdHu5 priming vaccine, supporting the further evaluation of this regimen in clinical trials as a next-generation malaria vaccine platform.
Collapse
Affiliation(s)
- Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
- Department of Parasitology, Faculty of Medicine, University of Hasanuddin, Makassar, Indonesia
| | - Tatsuya Yoshii
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Kunitaka Yoshida
- Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroaki Mizukami
- Division of Gene therapy, Jichi Medical University, Shimotsuke, Japan
| | - Shinya Fukumoto
- National Research Centre for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Daisuke S. Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke, Japan
| | - Asrar Alam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Talha Bin Emran
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Fitri Amelia
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Ashekul Islam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Hiromu Otsuka
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
47
|
Charlton Hume HK, Vidigal J, Carrondo MJT, Middelberg APJ, Roldão A, Lua LHL. Synthetic biology for bioengineering virus-like particle vaccines. Biotechnol Bioeng 2019; 116:919-935. [PMID: 30597533 PMCID: PMC7161758 DOI: 10.1002/bit.26890] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/08/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Vaccination is the most effective method of disease prevention and control. Many viruses and bacteria that once caused catastrophic pandemics (e.g., smallpox, poliomyelitis, measles, and diphtheria) are either eradicated or effectively controlled through routine vaccination programs. Nonetheless, vaccine manufacturing remains incredibly challenging. Viruses exhibiting high antigenic diversity and high mutation rates cannot be fairly contested using traditional vaccine production methods and complexities surrounding the manufacturing processes, which impose significant limitations. Virus-like particles (VLPs) are recombinantly produced viral structures that exhibit immunoprotective traits of native viruses but are noninfectious. Several VLPs that compositionally match a given natural virus have been developed and licensed as vaccines. Expansively, a plethora of studies now confirms that VLPs can be designed to safely present heterologous antigens from a variety of pathogens unrelated to the chosen carrier VLPs. Owing to this design versatility, VLPs offer technological opportunities to modernize vaccine supply and disease response through rational bioengineering. These opportunities are greatly enhanced with the application of synthetic biology, the redesign and construction of novel biological entities. This review outlines how synthetic biology is currently applied to engineer VLP functions and manufacturing process. Current and developing technologies for the identification of novel target-specific antigens and their usefulness for rational engineering of VLP functions (e.g., presentation of structurally diverse antigens, enhanced antigen immunogenicity, and improved vaccine stability) are described. When applied to manufacturing processes, synthetic biology approaches can also overcome specific challenges in VLP vaccine production. Finally, we address several challenges and benefits associated with the translation of VLP vaccine development into the industry.
Collapse
Affiliation(s)
- Hayley K. Charlton Hume
- The University of Queensland, Australian Institute of Bioengineering and NanotechnologySt LuciaQueenslandAustralia
| | - João Vidigal
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da RepúblicaOeirasPortugal
| | - Manuel J. T. Carrondo
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
| | - Anton P. J. Middelberg
- Faculty of Engineering, Computer and Mathematical Sciences, The University of AdelaideAdelaideSouth AustraliaAustralia
| | - António Roldão
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da RepúblicaOeirasPortugal
| | | |
Collapse
|
48
|
Abstract
The development of highly effective and durable vaccines against the human malaria parasites Plasmodium falciparum and P. vivax remains a key priority. Decades of endeavor have taught that achieving this goal will be challenging; however, recent innovation in malaria vaccine research and a diverse pipeline of novel vaccine candidates for clinical assessment provides optimism. With first-generation pre-erythrocytic vaccines aiming for licensure in the coming years, it is important to reflect on how next-generation approaches can improve on their success. Here we review the latest vaccine approaches that seek to prevent malaria infection, disease, and transmission and highlight some of the major underlying immunological and molecular mechanisms of protection. The synthesis of rational antigen selection, immunogen design, and immunization strategies to induce quantitatively and qualitatively improved immune effector mechanisms offers promise for achieving sustained high-level protection.
Collapse
|
49
|
Yman V, White MT, Asghar M, Sundling C, Sondén K, Draper SJ, Osier FHA, Färnert A. Antibody responses to merozoite antigens after natural Plasmodium falciparum infection: kinetics and longevity in absence of re-exposure. BMC Med 2019; 17:22. [PMID: 30696449 PMCID: PMC6352425 DOI: 10.1186/s12916-019-1255-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 01/08/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Antibodies against merozoite antigens are key components of malaria immunity. The naturally acquired antibody response to these antigens is generally considered short-lived; however, the underlying mechanisms remain unclear. Prospective studies of travellers with different levels of prior exposure, returning to malaria-free countries with Plasmodium infection, offer a unique opportunity to investigate the kinetics and composition of the antibody response after natural infection. METHODS Adults diagnosed with P. falciparum malaria in Stockholm, Sweden (20 likely malaria naïve and 41 with repeated previous exposure during residency in sub-Saharan Africa) were sampled at diagnosis and 10 days and 1, 3, 6, and 12 months after treatment. Total and subclass-specific IgG responses to P. falciparum merozoite antigens (AMA-1, MSP-119, MSP-2, MSP-3, and RH5) and tetanus toxoid were measured by multiplex bead-based immunoassays and ELISA. Mathematical modelling was used to estimate the exposure-dependent longevity of antibodies and antibody-secreting cells (ASCs). RESULTS A majority of individuals mounted detectable antibody responses towards P. falciparum merozoite antigens at diagnosis; however, the magnitude and breadth were greater in individuals with prior exposure. In both exposure groups, antibody levels increased rapidly for 2 weeks and decayed thereafter. Previously exposed individuals maintained two- to ninefold greater antibody levels throughout the 1-year follow-up. The half-lives of malaria-specific long-lived ASCs, responsible for maintaining circulating antibodies, ranged from 1.8 to 3.7 years for merozoite antigens and were considerably short compared to tetanus-specific ASCs. Primary infected individuals did acquire a long-lived component of the antibody response; however, the total proportion of long-lived ASCs generated in response to infection was estimated not to exceed 10%. In contrast, previously exposed individuals maintained substantially larger numbers of long-lived ASCs (10-56% of total ASCs). CONCLUSION The short-lived nature of the naturally acquired antibody response, to all tested merozoite antigens, following primary malaria infection can be attributed to a combination of a poor acquisition and short half-life of long-lived ASCs. Greater longevity is acquired with repeated infections and can be explained by the maintenance of larger numbers of long-lived ASCs. These insights advance our understanding of naturally acquired malaria immunity and will guide strategies for further development of both vaccines and serological tools to monitor exposure.
Collapse
Affiliation(s)
- Victor Yman
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 76, Stockholm, Sweden.
| | - Michael T White
- Department of Parasites and Insect Vectors, Institut Pasteur, 25-28 Rue du Dr Roux, 75015, Paris, France
| | - Muhammad Asghar
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Klara Sondén
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Simon J Draper
- Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Faith H A Osier
- Kenya Medical Research Institute - Wellcome Trust Research Program, Centre for Geographic Medicine Research-Coast, PO Box 230-80108, Kilifi, Kenya
- Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Anna Färnert
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, 171 76, Stockholm, Sweden
| |
Collapse
|
50
|
Garrido-Cardenas JA, Manzano-Agugliaro F, González-Cerón L, Gil-Montoya F, Alcayde-Garcia A, Novas N, Mesa-Valle C. The Identification of Scientific Communities and Their Approach to Worldwide Malaria Research. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:ijerph15122703. [PMID: 30513616 PMCID: PMC6313382 DOI: 10.3390/ijerph15122703] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022]
Abstract
It is essential to establish a pattern to detect the strengths and weaknesses of working groups publishing on malaria, to promote coordination to facilitate the eradication of the disease. Given the complexity of the scientific network of groups and institutions studying malaria, it is necessary to use a mathematical algorithm that allows us to know the real structure of research on the disease in the world. In this work, articles with the word “malaria” in the title or author keywords gathered from Elsevier Scopus database were analyzed. By means of specific software, graphs were created. The analysis of the data allowed established different scientific communities, among which two were very diverse: one formed by those groups concerned about the vector transmission and control, and another one focused on the drug resistance of the parasite. Basic, applied, and operational research to eradicate malaria is an ambitious goal of the international institutions and the scientific community. The combination of effort and the establishment of a worldwide-scientific network that allows an effective interconnection (exchange) of knowledge, infrastructure technology, collaborators, financial resources, and datasets will contribute more effectively to end the disease.
Collapse
Affiliation(s)
| | | | - Lilia González-Cerón
- Regional Center for Public Health Research, National Institute of Public Health, Tapachula 30700, Chiapas, Mexico.
| | | | | | - Nuria Novas
- Department of Engineering, University of Almeria, CeiA3. 04120 Almeria, Spain.
| | | |
Collapse
|