151
|
Mekonnen ZA, Grubor-Bauk B, English K, Leung P, Masavuli MG, Shrestha AC, Bertolino P, Bowen DG, Lloyd AR, Gowans EJ, Wijesundara DK. Single-Dose Vaccination with a Hepatotropic Adeno-associated Virus Efficiently Localizes T Cell Immunity in the Liver with the Potential To Confer Rapid Protection against Hepatitis C Virus. J Virol 2019; 93:e00202-19. [PMID: 31292249 PMCID: PMC6744243 DOI: 10.1128/jvi.00202-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/03/2019] [Indexed: 12/31/2022] Open
Abstract
Hepatitis C virus (HCV) is a significant contributor to the global disease burden, and development of an effective vaccine is required to eliminate HCV infections worldwide. CD4+ and CD8+ T cell immunity correlates with viral clearance in primary HCV infection, and intrahepatic CD8+ tissue-resident memory T (TRM) cells provide lifelong and rapid protection against hepatotropic pathogens. Consequently, we aimed to develop a vaccine to elicit HCV-specific CD4+ and CD8+ T cells, including CD8+ TRM cells, in the liver, given that HCV primarily infects hepatocytes. To achieve this, we vaccinated wild-type BALB/c mice with a highly immunogenic cytolytic DNA vaccine encoding a model HCV (genotype 3a) nonstructural protein (NS5B) and a mutant perforin (pVAX-NS5B-PRF), as well as a recombinant adeno-associated virus (AAV) encoding NS5B (rAAV-NS5B). A novel fluorescent target array (FTA) was used to map immunodominant CD4+ T helper (TH) cell and cytotoxic CD8+ T cell epitopes of NS5B in vivo, which were subsequently used to design a KdNS5B451-459 tetramer and analyze NS5B-specific T cell responses in vaccinated mice in vivo The data showed that intradermal prime/boost vaccination with pVAX-NS5B-PRF was effective in eliciting TH and cytotoxic CD8+ T cell responses and intrahepatic CD8+ TRM cells, but a single intravenous dose of hepatotropic rAAV-NS5B was significantly more effective. As a T-cell-based vaccine against HCV should ideally result in localized T cell responses in the liver, this study describes primary observations in the context of HCV vaccination that can be used to achieve this goal.IMPORTANCE There are currently at least 71 million individuals with chronic HCV worldwide and almost two million new infections annually. Although the advent of direct-acting antivirals (DAAs) offers highly effective therapy, considerable remaining challenges argue against reliance on DAAs for HCV elimination, including high drug cost, poorly developed health infrastructure, low screening rates, and significant reinfection rates. Accordingly, development of an effective vaccine is crucial to HCV elimination. An HCV vaccine that elicits T cell immunity in the liver will be highly protective for the following reasons: (i) T cell responses against nonstructural proteins of the virus are associated with clearance of primary infection, and (ii) long-lived liver-resident T cells alone can protect against malaria infection of hepatocytes. Thus, in this study we exploit promising vaccination platforms to highlight strategies that can be used to evoke highly functional and long-lived T cell responses in the liver for protection against HCV.
Collapse
Affiliation(s)
- Zelalem A Mekonnen
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| | - Branka Grubor-Bauk
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| | - Kieran English
- Liver Immunology Group and A. W. Morrow Gastroenterology and Liver Centre, Centenary Institute, Royal Prince Alfred Hospital and University of Sydney, Newtown, NSW, Australia
| | - Preston Leung
- Viral Immunology Systems Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Makutiro G Masavuli
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| | - Ashish C Shrestha
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| | - Patrick Bertolino
- Liver Immunology Group and A. W. Morrow Gastroenterology and Liver Centre, Centenary Institute, Royal Prince Alfred Hospital and University of Sydney, Newtown, NSW, Australia
| | - David G Bowen
- Liver Immunology Group and A. W. Morrow Gastroenterology and Liver Centre, Centenary Institute, Royal Prince Alfred Hospital and University of Sydney, Newtown, NSW, Australia
- Collaborative Transplantation Research Group, Bosch Institute, Royal Prince Alfred Hospital and University of Sydney, Newtown, NSW, Australia
| | - Andrew R Lloyd
- Viral Immunology Systems Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Eric J Gowans
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| | - Danushka K Wijesundara
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
152
|
Winkel BMF, de Korne CM, van Oosterom MN, Staphorst D, Meijhuis M, Baalbergen E, Ganesh MS, Dechering KJ, Vos MW, Chevalley-Maurel SC, Franke-Fayard B, van Leeuwen FWB, Roestenberg M. Quantification of wild-type and radiation attenuated Plasmodium falciparum sporozoite motility in human skin. Sci Rep 2019; 9:13436. [PMID: 31530862 PMCID: PMC6748968 DOI: 10.1038/s41598-019-49895-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/31/2019] [Indexed: 11/09/2022] Open
Abstract
Given the number of global malaria cases and deaths, the need for a vaccine against Plasmodium falciparum (Pf) remains pressing. Administration of live, radiation-attenuated Pf sporozoites can fully protect malaria-naïve individuals. Despite the fact that motility of these attenuated parasites is key to their infectivity and ultimately protective efficacy, sporozoite motility in human tissue (e.g. skin) remains wholly uncharacterized to date. We show that the ability to quantitatively address the complexity of sporozoite motility in human tissue provides an additional tool in the development of attenuated sporozoite vaccines. We imaged Pf movement in the skin of its natural host and compared wild-type and radiation-attenuated GFP-expressing Pf sporozoites. Using custom image analysis software and human skin explants we were able to quantitatively study their key motility features. This head-to-head comparison revealed that radiation attenuation impaired the capacity of sporozoites to vary their movement angle, velocity and direction, promoting less refined movement patterns. Understanding and overcoming these changes in motility will contribute to the development of an efficacious attenuated parasite malaria vaccine.
Collapse
Affiliation(s)
- Béatrice M F Winkel
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Clarize M de Korne
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Matthias N van Oosterom
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Diego Staphorst
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Mark Meijhuis
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Els Baalbergen
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Munisha S Ganesh
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Koen J Dechering
- TropIQ Health Sciences, Transistorweg 5, 6534, Nijmegen, The Netherlands
| | - Martijn W Vos
- TropIQ Health Sciences, Transistorweg 5, 6534, Nijmegen, The Netherlands
| | - Séverine C Chevalley-Maurel
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Blandine Franke-Fayard
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
153
|
Zenklusen I, Jongo S, Abdulla S, Ramadhani K, Lee Sim BK, Cardamone H, Flannery EL, Nguyen T, Fishbaugher M, Steel RWJ, Betz W, Carmago N, Mikolajczak S, Kappe SHI, Hoffman SL, Sack BK, Daubenberger C. Immunization of Malaria-Preexposed Volunteers With PfSPZ Vaccine Elicits Long-Lived IgM Invasion-Inhibitory and Complement-Fixing Antibodies. J Infect Dis 2019; 217:1569-1578. [PMID: 29438525 DOI: 10.1093/infdis/jiy080] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/06/2018] [Indexed: 01/17/2023] Open
Abstract
Background The assessment of antibody responses after immunization with radiation-attenuated, aseptic, purified, cryopreserved Plasmodium falciparum sporozoites (Sanaria PfSPZ Vaccine) has focused on IgG isotype antibodies. Here, we aimed to investigate if P. falciparum sporozoite binding and invasion-inhibitory IgM antibodies are induced following immunization of malaria-preexposed volunteers with PfSPZ Vaccine. Methods Using serum from volunteers immunized with PfSPZ, we measured vaccine-induced IgG and IgM antibodies to P. falciparum circumsporozoite protein (PfCSP) via ELISA. Function of this serum as well as IgM antibody fractions was measured via in vitro in an inhibition of sporozoite invasion assay. These IgM antibody fractions were also measured for binding to sporozoites by immunofluorescence assay and complement fixation on whole sporozoites. Results We found that in addition to anti-PfCSP IgG, malaria-preexposed volunteers developed anti-PfCSP IgM antibodies after immunization with PfSPZ Vaccine and that these IgM antibodies inhibited P. falciparum sporozoite invasion of hepatocytes in vitro. These IgM plasma fractions also fixed complement to whole P. falciparum sporozoites. Conclusions This is the first finding that PfCSP and P. falciparum sporozoite-binding IgM antibodies are induced following immunization of PfSPZ Vaccine in malaria-preexposed individuals and that IgM antibodies can inhibit P. falciparum sporozoite invasion into hepatocytes in vitro and fix complement on sporozoites. These findings indicate that the immunological assessment of PfSPZ Vaccine-induced antibody responses could be more sensitive if they include parasite-specific IgM in addition to IgG antibodies. Clinical Trials Registration NCT02132299.
Collapse
Affiliation(s)
- Isabelle Zenklusen
- Clinical Immunology Unit, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | - Said Jongo
- Ifakara Health Institute, Clinical Trial Unit, Bagamoyo, Tanzania
| | - Salim Abdulla
- Ifakara Health Institute, Clinical Trial Unit, Bagamoyo, Tanzania
| | - Kamaka Ramadhani
- Ifakara Health Institute, Clinical Trial Unit, Bagamoyo, Tanzania
| | | | | | | | - Thao Nguyen
- Center for Infectious Disease Research, Seattle, Washington
| | | | - Ryan W J Steel
- Center for Infectious Disease Research, Seattle, Washington
| | - Will Betz
- Center for Infectious Disease Research, Seattle, Washington
| | - Nelly Carmago
- Center for Infectious Disease Research, Seattle, Washington
| | | | - Stefan H I Kappe
- Center for Infectious Disease Research, Seattle, Washington.,Department of Global Health, University of Washington, Seattle
| | | | - Brandon K Sack
- Center for Infectious Disease Research, Seattle, Washington
| | - Claudia Daubenberger
- Clinical Immunology Unit, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| |
Collapse
|
154
|
Julien JP, Wardemann H. Antibodies against Plasmodium falciparum malaria at the molecular level. Nat Rev Immunol 2019; 19:761-775. [DOI: 10.1038/s41577-019-0209-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
|
155
|
Abstract
Malaria vaccine development has rapidly advanced in the past decade. The very first phase 3 clinical trial of the RTS,S vaccine was completed with over 15,000 African infants and children, and pilot implementation studies are underway. Next-generation candidate vaccines using novel antigens, platforms, or approaches targeting different and/or multiple stages of the Plasmodium life cycle are being tested. Many candidates, in various stages of development, promise enhanced efficacy of long duration and broad protection against genetically diverse malaria strains, with a few studies under way in target populations in endemic areas. Malaria vaccines together with other interventions promise interruption and eventual elimination of malaria in endemic areas.
Collapse
Affiliation(s)
- Matthew B Laurens
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore 21201, Maryland, USA; .,Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.,Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore 21201, Maryland, USA
| |
Collapse
|
156
|
Muhindo MK, Jagannathan P, Kakuru A, Opira B, Olwoch P, Okiring J, Nalugo N, Clark TD, Ruel T, Charlebois E, Feeney ME, Havlir DV, Dorsey G, Kamya MR. Intermittent preventive treatment with dihydroartemisinin-piperaquine and risk of malaria following cessation in young Ugandan children: a double-blind, randomised, controlled trial. THE LANCET. INFECTIOUS DISEASES 2019; 19:962-972. [PMID: 31307883 DOI: 10.1016/s1473-3099(19)30299-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/21/2019] [Accepted: 04/24/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Intermittent preventive treatment (IPT) of malaria with dihydroartemisinin-piperaquine is a promising strategy for malaria prevention in young African children. However, the optimal dosing strategy is unclear and conflicting evidence exists regarding the risk of malaria after cessation of chemoprevention. We aimed to compare two dosing strategies of IPT with dihydroartemisinin-piperaquine in young Ugandan children, and to evaluate the risk of malaria after cessation of IPT. METHODS In this double-blind, randomised controlled phase 2 trial, women and their unborn children were recruited at Tororo District Hospital (Tororo, Uganda). Eligible participants were HIV-negative women aged 16 years or older with a viable pregnancy (gestational age 12-20 weeks). Women and their unborn children were randomly assigned (1:1:1:1) to one of four treatment groups, all receiving dihydroartemisinin-piperaquine, on the basis of the IPT intervention received by the woman during pregnancy: women every 8 weeks, children every 4 weeks; women every 4 weeks, children every 4 weeks; women every 8 weeks, children every 12 weeks; and women every 4 weeks, children every 12 weeks. Block randomisation was done by an independent investigator using a computer-generated randomisation list (permuted block sizes of six and 12). We analysed children on the basis of their random assignment to receive dihydroartemisinin-piperaquine (20 mg/160 mg tablets) once daily for 3 consecutive days every 4 weeks or 12 weeks. Children received study drugs from age 8 weeks to 24 months and were followed-up to age 36 months. Participants and investigators were masked to treatment allocation. The primary outcome was the incidence of symptomatic malaria during the intervention and following cessation of the intervention, adjusted for potential confounders. The primary outcome and safety were assessed in the modified intention-to-treat population, which included all children who reached 8 weeks of age and received at least one dose of study drug. This trial is registered with ClinicalTrials.gov, number NCT02163447. FINDINGS Between Oct 21, 2014, and May 18, 2015, 191 children were born, of whom 183 reached 8 weeks of age and received at least one dose of study drug and thus were included in the primary analysis (96 children in the 4-week group and 87 in the 12-week group). During the intervention, the incidence of symptomatic malaria was significantly lower among children treated every 4 weeks than children treated every 12 weeks; three episodes occurred among children treated every 4 weeks (incidence 0·018 episodes per person-year) compared with 61 episodes among children treated every 12 weeks (incidence 0·39 episodes per person-year; adjusted incidence rate ratio [aIRR] 0·041, 95% CI 0·012-0·150, p<0·0001). After cessation of IPT, children who had previously received dihydroartemisinin-piperaquine every 4 weeks had a lower incidence of symptomatic malaria than children who were treated every 12 weeks; 62 episodes occurred among children previously treated every 4 weeks (incidence 0·73 episodes per person-year) compared with 83 episodes among children treated every 12 weeks (incidence 1·1 episodes per person-year; aIRR 0·62, 0·40-0·95, p=0·028). In the 4-week group, 94 (98%) of 96 children had adverse events versus 87 (100%) of 87 children in the 12-week group. The most commonly reported adverse event was cough in both treatment groups (94 [98%] in the 4-week group vs 87 [100%] in the 12-week group). 16 children had severe adverse events (seven [7%] children in the 4-week group vs nine [10%] children in the 12-week group). No severe adverse events were thought to be related to study drug administration. One death occurred during the intervention (age 8 weeks to 24 months), which was due to respiratory failure unrelated to malaria. INTERPRETATION IPT with dihydroartemisinin-piperaquine given every 4 weeks was superior to treatment every 12 weeks for the prevention of malaria during childhood, and this protection was extended for up to 1 year after cessation of IPT. FUNDING Eunice Kennedy Shriver National Institute of Child Health and Human Development.
Collapse
Affiliation(s)
- Mary K Muhindo
- Infectious Diseases Research Collaboration, Kampala, Uganda.
| | | | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Bishop Opira
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Peter Olwoch
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Jaffer Okiring
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Noeline Nalugo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Tamara D Clark
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Theodore Ruel
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Edwin Charlebois
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Margaret E Feeney
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Diane V Havlir
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Moses R Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda; School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| |
Collapse
|
157
|
Yu KKQ, Wilburn DB, Hackney JA, Darrah PA, Foulds KE, James CA, Smith MT, Jing L, Seder RA, Roederer M, Koelle DM, Swanson WJ, Seshadri C. Conservation of molecular and cellular phenotypes of invariant NKT cells between humans and non-human primates. Immunogenetics 2019; 71:465-478. [PMID: 31123763 PMCID: PMC6647187 DOI: 10.1007/s00251-019-01118-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/26/2019] [Accepted: 04/30/2019] [Indexed: 10/27/2022]
Abstract
Invariant NKT (iNKT) cells in both humans and non-human primates are activated by the glycolipid antigen, α-galactosylceramide (α-GalCer). However, the extent to which the molecular mechanisms of antigen recognition and in vivo phenotypes of iNKT cells are conserved among primate species has not been determined. Using an evolutionary genetic approach, we found a lack of diversifying selection in CD1 genes over 45 million years of evolution, which stands in stark contrast to the history of the MHC system for presenting peptide antigens to T cells. The invariant T cell receptor (TCR)-α chain was strictly conserved across all seven primate clades. Invariant NKT cells from rhesus macaques (Macaca mulatta) bind human CD1D-α-GalCer tetramer and are activated by α-GalCer-loaded human CD1D transfectants. The dominant TCR-β chain cloned from a rhesus-derived iNKT cell line is nearly identical to that found in the human iNKT TCR, and transduction of the rhesus iNKT TCR into human Jurkat cells show that it is sufficient for binding human CD1D-α-GalCer tetramer. Finally, we used a 20-color flow cytometry panel to probe tissue phenotypes of iNKT cells in a cohort of rhesus macaques. We discovered several tissue-resident iNKT populations that have not been previously described in non-human primates but are known in humans, such as TCR-γδ iNKTs. These data reveal a diversity of iNKT cell phenotypes despite convergent evolution of the genes required for lipid antigen presentation and recognition in humans and non-human primates.
Collapse
Affiliation(s)
- Krystle K Q Yu
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Damien B Wilburn
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Joshua A Hackney
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patricia A Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Charlotte A James
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Pathology, Molecular Medicine and Mechanisms of Disease Program, University of Washington, Seattle, WA, USA
| | - Malisa T Smith
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Lichen Jing
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David M Koelle
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Benaroya Research Institute, Seattle, WA, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Willie J Swanson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Chetan Seshadri
- Department of Medicine, University of Washington, Seattle, WA, USA.
- Tuberculosis Research & Training Center, University of Washington, Seattle, WA, USA.
| |
Collapse
|
158
|
Patel H, Althubaiti N, Parmar R, Yadav N, Joshi U, Tyagi RK, Krzych U, Dalai SK. Parasite load stemming from immunization route determines the duration of liver-stage immunity. Parasite Immunol 2019; 41:e12622. [PMID: 30854655 PMCID: PMC6584043 DOI: 10.1111/pim.12622] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 12/28/2018] [Accepted: 02/22/2019] [Indexed: 02/05/2023]
Abstract
Immunization with radiation-attenuated Plasmodium sporozoites (RAS) induces sterile and long-lasting protective immunity. Although intravenous (IV) route of RAS immunization is reported to induce superior immunity compared to intradermal (ID) injection, its role in the maintenance of sterile immunity is yet to be understood. We investigated whether the route of homologous sporozoite challenge of Plasmodium berghei (Pb) RAS-immunized mice would influence the longevity of protection. C57BL/6 mice immunized with Pb-RAS by IV were 100% protected upon primary IV/ID sporozoite challenge. In contrast, ID immunization resulted in 80% protection, regardless of primary challenge route. Interestingly, the route of primary challenge was found to bring difference in the maintenance of sterile protection. While IV Pb RAS-immunized mice remained protected at all challenges regardless of the route of primary challenge, ID Pb-RAS-immunized mice receiving ID primary challenge became parasitaemic upon secondary IV challenge. Significantly, primary IV challenge of Pb RAS ID-immunized mice resulted in 80% and 50% survival at secondary and tertiary challenges, respectively. According to phenotypically diverse liver CD8+ T cells, the percentages and the numbers of both CD8+ T effector memory and resident memory cells were significantly higher in IV than in ID Pb RAS-immunized mice. IFN-γ-producing CD8+ T cells specific to Pb TRAP130 and MIP-4-Kb-17 were also found significantly higher in IV mice than in ID mice. The enhanced T-cell generation and the longevity of protection appear to be dependent on the parasite load during challenge when infection is tolerated under suboptimal CD8+ T-cell response.
Collapse
Affiliation(s)
- Hardik Patel
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Nouf Althubaiti
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Rajesh Parmar
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Naveen Yadav
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Urja Joshi
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Rajeev K Tyagi
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Urszula Krzych
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sarat K Dalai
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
159
|
Martin MD, Jensen IJ, Ishizuka AS, Lefebvre M, Shan Q, Xue HH, Harty JT, Seder RA, Badovinac VP. Bystander responses impact accurate detection of murine and human antigen-specific CD8 T cells. J Clin Invest 2019; 129:3894-3908. [PMID: 31219804 DOI: 10.1172/jci124443] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Induction of memory CD8 T cells is important for controlling infections such as malaria HIV/AIDS, and for cancer immunotherapy. Accurate assessment of antigen (Ag)-specific CD8 T-cells is critical for vaccine optimization and defining correlates of protection. However, conditions for determining Ag-specific CD8 T-cell responses ex-vivo using ICS may be variable, especially in humans with complex antigens. Here, we used an attenuated whole parasite malaria vaccine model in humans and various experimental infections in mice to show that the duration of antigenic stimulation and timing of brefeldin A (BFA) addition influences the magnitude of Ag-specific and bystander T cell responses. Indeed, following immunization with an attenuated whole sporozoite malaria vaccine in humans, significantly higher numbers of IFN-γ producing memory CD8 T-cells comprised of antigen specific and bystander responses were detected by increasing the duration of Ag-stimulation prior to addition of BFA. Mechanistic analyses of virus-specific CD8 T-cells in mice revealed that the increase in IFNg producing CD8 T-cells was due to bystander activation of Ag-experienced memory CD8 T-cells, and correlated with the proportion of Ag-experienced CD8 T-cells in the stimulated populations. Incubation with anti-cytokine antibodies (ex. IL-12) improved accuracy in detecting bona-fide memory CD8 T-cell responses suggesting this as the mechanism for the bystander activation. These data have important implications for accurate assessment of immune responses generated by vaccines intended to elicit protective memory CD8 T-cells.
Collapse
Affiliation(s)
| | - Isaac J Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Andrew S Ishizuka
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Mitchell Lefebvre
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Qiang Shan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Hai-Hui Xue
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA.,Iowa City Veterans Affairs Health Care System, Iowa City, Iowa, USA
| | - John T Harty
- Department of Pathology and.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Vladimir P Badovinac
- Department of Pathology and.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
160
|
Goh YS, McGuire D, Rénia L. Vaccination With Sporozoites: Models and Correlates of Protection. Front Immunol 2019; 10:1227. [PMID: 31231377 PMCID: PMC6560154 DOI: 10.3389/fimmu.2019.01227] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Despite continuous efforts, the century-old goal of eradicating malaria still remains. Multiple control interventions need to be in place simultaneously to achieve this goal. In addition to effective control measures, drug therapies and insecticides, vaccines are critical to reduce mortality and morbidity. Hence, there are numerous studies investigating various malaria vaccine candidates. Most of the malaria vaccine candidates are subunit vaccines. However, they have shown limited efficacy in Phase II and III studies. To date, only whole parasite formulations have been shown to induce sterile immunity in human. In this article, we review and discuss the recent developments in vaccination with sporozoites and the mechanisms of protection involved.
Collapse
Affiliation(s)
- Yun Shan Goh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore
| | - Daniel McGuire
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Laurent Rénia
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
161
|
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
|
162
|
Zhou D, Zheng H, Liu Q, Lu X, Deng X, Jiang L, Hou B, Fu Y, Zhu F, Ding Y, Xu W, Dai J. Attenuated plasmodium sporozoite expressing MAGE-A3 induces antigen-specific CD8+ T cell response against lung cancer in mice. Cancer Biol Med 2019; 16:288-298. [PMID: 31516749 PMCID: PMC6713645 DOI: 10.20892/j.issn.2095-3941.2018.0309] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/12/2018] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Cancer vaccines that rely on tumor antigen-specific CD8+ T cell responses, are promising anti-cancer adjuvant immunotherapies. This study investigated whether genetically attenuated Plasmodium sporozoite (GAS) could be used as a novel vector to induce antigen-specific CD8+ T cell responses against lung cancer. METHODS We constructed GAS/MAGE-A3, a recombinant GAS engineered to express the lung cancer-specific antigen, melanoma-associated antigen 3 (MAGE-A3), and assessed its therapeutic effects against lung cancer. RESULTS Robust parasite-specific CD8αlowCD11ahigh and CD49dhighCD11ahigh CD4+ T cell responses as well as a MAGE-A3-specific CD8+ T cell response were induced in GAS/MAGE-A3-immunized mice. Adoptive transfer of GAS/MAGE-A3-induced CD8+ T cells from HLA-A2 transgenic mice into lung cancer-bearing nude mice inhibited tumor growth and prolonged survival. CONCLUSIONS These findings demonstrate that GAS/MAGE-A3 induces a strong MAGE-A3-specific CD8+ T cell response against lung cancer in vivo, and indicate that GAS is a novel and efficacious antigen delivery vector for antitumor immunotherapy.
Collapse
Affiliation(s)
- Dong Zhou
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Quanxing Liu
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xiao Lu
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xufeng Deng
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Department of Cardiothoracic Surgery, The First People's Hospital of Zunyi, Zunyi 563000, China
| | - Li Jiang
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Bing Hou
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yong Fu
- Department of Pathogenic Biology, Third Military Medical University, Chongqing 400037, China
| | - Feng Zhu
- Department of Pathogenic Biology, Third Military Medical University, Chongqing 400037, China
| | - Yan Ding
- Department of Pathogenic Biology, Third Military Medical University, Chongqing 400037, China
| | - Wenyue Xu
- Department of Pathogenic Biology, Third Military Medical University, Chongqing 400037, China
| | - Jigang Dai
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| |
Collapse
|
163
|
Winkel BM, de Korne CM, van Oosterom MN, Staphorst D, Bunschoten A, Langenberg MC, Chevalley-Maurel SC, Janse CJ, Franke-Fayard B, van Leeuwen FW, Roestenberg M. A tracer-based method enables tracking of Plasmodium falciparum malaria parasites during human skin infection. Theranostics 2019; 9:2768-2778. [PMID: 31244921 PMCID: PMC6568182 DOI: 10.7150/thno.33467] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 02/22/2019] [Indexed: 11/10/2022] Open
Abstract
Introduction: The skin stage of malaria is a vital and vulnerable migratory life stage of the parasite. It has been characterised in rodent models, but remains wholly uninvestigated for human malaria parasites. To enable in depth analysis of not genetically modified (non-GMO) Plasmodium falciparum (Pf) sporozoite behaviour in human skin, we devised a labelling technology (Cy5M2, targeting the sporozoite mitochondrion) that supports tracking of individual non-GMO sporozoites in human skin. Methods: Sporozoite labelling with Cy5M2 was performed in vitro as well as via the feed of infected Anopheles mosquitos. Labelling was validated using confocal microscopy and flow cytometry and the fitness of labelled sporozoites was determined by analysis of infectivity to human hepatocytes in vitro, and in vivo in a rodent infection model. Using confocal video microscopy and custom software, single-sporozoite tracking studies in human skin-explants were performed. Results: Both in vitro and in mosquito labelling strategies yielded brightly fluorescent sporozoites of three different Plasmodium species. Cy5M2 uptake colocalized with MitoTracker® green and could be blocked using the known Translocator protein (TSPO)-inhibitor PK11195. This method supported the visualization and subsequent quantitative analysis of the migration patterns of individual non-GMO Pf sporozoites in human skin and did not affect the fitness of sporozoites. Conclusions: The ability to label and image non-GMO Plasmodium sporozoites provides the basis for detailed studies on the human skin stage of malaria with potential for in vivo translation. As such, it is an important tool for development of vaccines based on attenuated sporozoites and their route of administration.
Collapse
Affiliation(s)
- Béatrice M.F. Winkel
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Clarize M. de Korne
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Matthias N. van Oosterom
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Diego Staphorst
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Anton Bunschoten
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Laboratory of BioNanoTechnology Wageningen University and Research, Droevendaalsesteeg 4, 6708 PB Wageningen, The Netherlands
| | - Marijke C.C. Langenberg
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | | | - Chris J. Janse
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Blandine Franke-Fayard
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Fijs W.B. van Leeuwen
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
164
|
Mekonnen ZA, Grubor-Bauk B, Masavuli MG, Shrestha AC, Ranasinghe C, Bull RA, Lloyd AR, Gowans EJ, Wijesundara DK. Toward DNA-Based T-Cell Mediated Vaccines to Target HIV-1 and Hepatitis C Virus: Approaches to Elicit Localized Immunity for Protection. Front Cell Infect Microbiol 2019; 9:91. [PMID: 31001491 PMCID: PMC6456646 DOI: 10.3389/fcimb.2019.00091] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 03/14/2019] [Indexed: 01/07/2023] Open
Abstract
Human immunodeficiency virus (HIV)-1 and hepatitis C virus (HCV) are major contributors to the global disease burden with many experts recognizing the requirement of an effective vaccine to bring a durable end to these viral epidemics. The most promising vaccine candidates that have advanced into pre-clinical models and the clinic to eliminate or provide protection against these chronic viruses are viral vectors [e.g., recombinant cytomegalovirus, Adenovirus, and modified vaccinia Ankara (MVA)]. This raises the question, is there a need to develop DNA vaccines against HIV-1 and HCV? Since the initial study from Wolff and colleagues which showed that DNA represents a vector that can be used to express transgenes durably in vivo, DNA has been regularly evaluated as a vaccine vector albeit with limited success in large animal models and humans. However, several recent studies in Phase I-IIb trials showed that vaccination of patients with recombinant DNA represents a feasible therapeutic intervention to even cure cervical cancer, highlighting the potential of using DNA for human vaccinations. In this review, we will discuss the limitations and the strategies of using DNA as a vector to develop prophylactic T cell-mediated vaccines against HIV-1 and HCV. In particular, we focus on potential strategies exploiting DNA vectors to elicit protective localized CD8+ T cell immunity in the liver for HCV and in the cervicovaginal mucosa for HIV-1 as localized immunity will be an important, if not critical component, of an efficacious vaccine against these viral infections.
Collapse
Affiliation(s)
- Zelalem A. Mekonnen
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Branka Grubor-Bauk
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Makutiro G. Masavuli
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Ashish C. Shrestha
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Charani Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Rowena A. Bull
- Viral Immunology Systems Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Andrew R. Lloyd
- Viral Immunology Systems Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Eric J. Gowans
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Danushka K. Wijesundara
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia,*Correspondence: Danushka K. Wijesundara
| |
Collapse
|
165
|
Heide J, Vaughan KC, Sette A, Jacobs T, Schulze Zur Wiesch J. Comprehensive Review of Human Plasmodium falciparum-Specific CD8+ T Cell Epitopes. Front Immunol 2019; 10:397. [PMID: 30949162 PMCID: PMC6438266 DOI: 10.3389/fimmu.2019.00397] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/15/2019] [Indexed: 12/16/2022] Open
Abstract
Control of malaria is an important global health issue and there is still an urgent need for the development of an effective prophylactic vaccine. Multiple studies have provided strong evidence that Plasmodium falciparum-specific MHC class I-restricted CD8+ T cells are important for sterile protection against Plasmodium falciparum infection. Here, we present an interactive epitope map of all P. falciparum-specific CD8+ T cell epitopes published to date, based on a comprehensive data base (IEDB), and literature search. The majority of the described P. falciparum-specific CD8+ T cells were directed against the antigens CSP, TRAP, AMA1, and LSA1. Notably, most of the epitopes were discovered in vaccine trials conducted with malaria-naïve volunteers. Only few immunological studies of P. falciparum-specific CD8+ T cell epitopes detected in patients suffering from acute malaria or in people living in malaria endemic areas have been published. Further detailed immunological mappings of P. falciparum-specific epitopes of a broader range of P. falciparum proteins in different settings and with different disease status are needed to gain a more comprehensive understanding of the role of CD8+ T cell responses for protection, and to better guide vaccine design and to study their efficacy.
Collapse
Affiliation(s)
- Janna Heide
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Kerrie C Vaughan
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States.,Department of Medicine, Division of Infectious Diseases, University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Thomas Jacobs
- Protozoa Immunology, Bernhard-Nocht-Institute of Tropical Medicine, Hamburg, Germany
| | - Julian Schulze Zur Wiesch
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
166
|
Friedman-Klabanoff DJ, Laurens MB, Berry AA, Travassos MA, Adams M, Strauss KA, Shrestha B, Levine MM, Edelman R, Lyke KE. The Controlled Human Malaria Infection Experience at the University of Maryland. Am J Trop Med Hyg 2019; 100:556-565. [PMID: 30675854 PMCID: PMC6402913 DOI: 10.4269/ajtmh.18-0476] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 12/03/2018] [Indexed: 11/07/2022] Open
Abstract
Controlled human malaria infection (CHMI) is a powerful tool to evaluate the efficacy of malaria vaccines and pharmacologics. Investigators at the University of Maryland, Baltimore, Center for Vaccine Development (UMB-CVD) pioneered the technique in the 1970s and continue to advance the frontiers of CHMI research. We reviewed the records of 338 malaria-naive volunteers who underwent CHMI at UMB-CVD with Plasmodium falciparum from 1971 until 2017. These 338 volunteers underwent 387 CHMI events, including 60 via intradermal injection or direct venous inoculation (DVI) of purified, cryopreserved sporozoites. No volunteer suffered an unplanned hospitalization or required intravenous therapy related to CHMI. Median prepatency period was longer in challenges using NF54 (9 days) than in those using 7G8 (8 days), P = 0.0006 by the log-rank test. With dose optimization of DVI, the prepatent period did not differ between DVI and mosquito bite challenge (log-rank test, P = 0.66). Polymerase chain reaction (PCR) detected P. falciparum infection 3 days earlier than thick smears (P < 0.001), and diagnosis by ultrasensitive PCR was associated with less severe symptoms than smear-based diagnosis (39% versus 0%, P = 0.0003). Historical studies with NF54 showed a shorter median prepatency period of 10.3 days than more recent studies (median 11.0 days, P = 0.02) despite significantly lower salivary gland scores in earlier studies, P = 0.0001. The 47-year experience of CHMI at UMB-CVD has led to advancements in sporozoite delivery, diagnostics, and use of heterologous challenge. Additional studies on new challenge strains and genomic data to reflect regional heterogeneity will help advance the use of CHMI as supporting data for vaccine licensure.
Collapse
Affiliation(s)
- DeAnna J. Friedman-Klabanoff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Matthew B. Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrea A. Berry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mark A. Travassos
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Matthew Adams
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kathy A. Strauss
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Biraj Shrestha
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Myron M. Levine
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Robert Edelman
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kirsten E. Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
167
|
Phenotypic Screening of Small Molecules with Antimalarial Activity for Three Different Parasitic Life Stages. Methods Mol Biol 2019. [PMID: 29736708 DOI: 10.1007/978-1-4939-7847-2_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Malaria remains one of the deadliest infectious diseases globally. Available therapeutic agents are already limited in their efficacy, and drug resistance threatens to diminish further our ability to prevent and treat the disease. Despite a renewed effort to identify compounds with antimalarial activity, the drug discovery and development pipeline lacks target diversity and availability of compounds that target liver- and gametocyte-stage parasites. Phenotypic screens are a powerful and valuable tool for identifying new chemical compounds with antimalarial activity. This chapter highlights recent phenotypic screening methodologies for all three parasitic life stages.
Collapse
|
168
|
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
|
169
|
Walk J, Stok JE, Sauerwein RW. Can Patrolling Liver-Resident T Cells Control Human Malaria Parasite Development? Trends Immunol 2019; 40:186-196. [PMID: 30713008 DOI: 10.1016/j.it.2019.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/08/2019] [Accepted: 01/13/2019] [Indexed: 12/23/2022]
Abstract
Recently, a population of non-recirculating, tissue-resident memory CD8+ T cells has been identified; cells that seems to act as key sentinels for invading microorganisms with enhanced effector functions. In malaria, the liver represents the first site for parasite development before a definite infection is established in circulating red blood cells. Here, we discuss the evidence obtained from animal models on several diseases and hypothesize that liver-resident memory CD8+ T cells (hepatic TRM) play a critical role in providing protective liver-stage immunity against Plasmodium malaria parasites. Although observations in human malaria trials are limited to peripheral blood, we propose recommendations for the translation of some of these findings to human malaria research.
Collapse
Affiliation(s)
- Jona Walk
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Jorn E Stok
- University Medical Center Utrecht, PO Box 85500, Utrecht, The Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
170
|
Wilson KL, Flanagan KL, Prakash MD, Plebanski M. Malaria vaccines in the eradication era: current status and future perspectives. Expert Rev Vaccines 2019; 18:133-151. [PMID: 30601095 DOI: 10.1080/14760584.2019.1561289] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The challenge to eradicate malaria is an enormous task that will not be achieved by current control measures, thus an efficacious and long-lasting malaria vaccine is required. The licensing of RTS, S/AS01 is a step forward in providing some protection, but a malaria vaccine that protects across multiple transmission seasons is still needed. To achieve this, inducing beneficial immune responses while minimising deleterious non-targeted effects will be essential. AREAS COVERED This article discusses the current challenges and advances in malaria vaccine development and reviews recent human clinical trials for each stage of infection. Pubmed and ScienceDirect were searched, focusing on cell mediated immunity and how T cell subsets might be targeted in future vaccines using novel adjuvants and emerging vaccine technologies. EXPERT COMMENTARY Despite decades of research there is no highly effective licensed malaria vaccine. However, there is cause for optimism as new adjuvants and vaccine systems emerge, and our understanding of correlates of protection increases, especially regarding cellular immunity. The new field of heterologous (non-specific) effects of vaccines also highlights the broader consequences of immunization. Importantly, the WHO led Malaria Vaccine Technology Roadmap illustrates that there is a political will among the global health community to make it happen.
Collapse
Affiliation(s)
- K L Wilson
- a Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences , Monash University , Melbourne , Australia.,b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| | - K L Flanagan
- a Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences , Monash University , Melbourne , Australia.,b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia.,c School of Medicine, Faculty of Health Sciences , University of Tasmania , Launceston , Australia
| | - M D Prakash
- b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| | - M Plebanski
- b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| |
Collapse
|
171
|
Hansen SG, Womack J, Scholz I, Renner A, Edgel KA, Xu G, Ford JC, Grey M, St Laurent B, Turner JM, Planer S, Legasse AW, Richie TL, Aguiar JC, Axthelm MK, Villasante ED, Weiss W, Edlefsen PT, Picker LJ, Früh K. Cytomegalovirus vectors expressing Plasmodium knowlesi antigens induce immune responses that delay parasitemia upon sporozoite challenge. PLoS One 2019; 14:e0210252. [PMID: 30673723 PMCID: PMC6343944 DOI: 10.1371/journal.pone.0210252] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 12/19/2018] [Indexed: 12/12/2022] Open
Abstract
The development of a sterilizing vaccine against malaria remains one of the highest priorities for global health research. While sporozoite vaccines targeting the pre-erythrocytic stage show great promise, it has not been possible to maintain efficacy long-term, likely due to an inability of these vaccines to maintain effector memory T cell responses in the liver. Vaccines based on human cytomegalovirus (HCMV) might overcome this limitation since vectors based on rhesus CMV (RhCMV), the homologous virus in rhesus macaques (RM), elicit and indefinitely maintain high frequency, non-exhausted effector memory T cells in extralymphoid tissues, including the liver. Moreover, RhCMV strain 68-1 elicits CD8+ T cells broadly recognizing unconventional epitopes exclusively restricted by MHC-II and MHC-E. To evaluate the potential of these unique immune responses to protect against malaria, we expressed four Plasmodium knowlesi (Pk) antigens (CSP, AMA1, SSP2/TRAP, MSP1c) in RhCMV 68-1 or in Rh189-deleted 68-1, which additionally elicits canonical MHC-Ia-restricted CD8+ T cells. Upon inoculation of RM with either of these Pk Ag expressing RhCMV vaccines, we obtained T cell responses to each of the four Pk antigens. Upon challenge with Pk sporozoites we observed a delayed appearance of blood stage parasites in vaccinated RM consistent with a 75-80% reduction of parasite release from the liver. Moreover, the Rh189-deleted RhCMV/Pk vectors elicited sterile protection in one RM. Once in the blood, parasite growth was not affected. In contrast to T cell responses induced by Pk infection, RhCMV vectors maintained sustained T cell responses to all four malaria antigens in the liver post-challenge. The delayed appearance of blood stage parasites is thus likely due to a T cell-mediated inhibition of liver stage parasite development. As such, this vaccine approach can be used to efficiently test new T cell antigens, improve current vaccines targeting the liver stage and complement vaccines targeting erythrocytic antigens.
Collapse
Affiliation(s)
- Scott G Hansen
- Oregon Health & Science University, Vaccine & Gene Therapy Institute, Beaverton, OR, United States of America
- Oregon Health & Science University, Oregon National Primate Research Center, Beaverton, OR, United States of America
| | - Jennie Womack
- Oregon Health & Science University, Vaccine & Gene Therapy Institute, Beaverton, OR, United States of America
| | - Isabel Scholz
- Oregon Health & Science University, Vaccine & Gene Therapy Institute, Beaverton, OR, United States of America
| | - Andrea Renner
- US Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Kimberly A Edgel
- US Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Guangwu Xu
- Oregon Health & Science University, Vaccine & Gene Therapy Institute, Beaverton, OR, United States of America
| | - Julia C Ford
- Oregon Health & Science University, Vaccine & Gene Therapy Institute, Beaverton, OR, United States of America
| | - Mikayla Grey
- Oregon Health & Science University, Vaccine & Gene Therapy Institute, Beaverton, OR, United States of America
| | - Brandyce St Laurent
- National Institutes of Health, Laboratory of Malaria and Vector Research, Malaria Pathogenesis and Human Immunity Unit, Rockville, MD, United States of America
| | - John M Turner
- Oregon Health & Science University, Oregon National Primate Research Center, Beaverton, OR, United States of America
| | - Shannon Planer
- Oregon Health & Science University, Oregon National Primate Research Center, Beaverton, OR, United States of America
| | - Al W Legasse
- Oregon Health & Science University, Oregon National Primate Research Center, Beaverton, OR, United States of America
| | - Thomas L Richie
- US Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Joao C Aguiar
- US Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Michael K Axthelm
- Oregon Health & Science University, Vaccine & Gene Therapy Institute, Beaverton, OR, United States of America
- Oregon Health & Science University, Oregon National Primate Research Center, Beaverton, OR, United States of America
| | - Eileen D Villasante
- US Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Walter Weiss
- US Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Paul T Edlefsen
- Statistical Center for HIV/AIDS Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Louis J Picker
- Oregon Health & Science University, Vaccine & Gene Therapy Institute, Beaverton, OR, United States of America
- Oregon Health & Science University, Oregon National Primate Research Center, Beaverton, OR, United States of America
| | - Klaus Früh
- Oregon Health & Science University, Vaccine & Gene Therapy Institute, Beaverton, OR, United States of America
- Oregon Health & Science University, Oregon National Primate Research Center, Beaverton, OR, United States of America
| |
Collapse
|
172
|
O'Brochta DA, Alford R, Harrell R, Aluvihare C, Eappen AG, Li T, Chakravarty S, Sim BKL, Hoffman SL, Billingsley PF. Is Saglin a mosquito salivary gland receptor for Plasmodium falciparum? Malar J 2019; 18:2. [PMID: 30602380 PMCID: PMC6317240 DOI: 10.1186/s12936-018-2634-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/20/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Saglin, a 100 kDa protein composed of two 50 kDa homodimers, is present in the salivary glands of Anopheles gambiae and has been considered an essential receptor for sporozoites (SPZ) of Plasmodium berghei and Plasmodium falciparum (Pf), allowing SPZ to recognize, bind to, and infect mosquito salivary glands. Spatial and temporal patterns of Saglin expression reported here, however, suggest that this model does not fully describe the Saglin-SPZ interaction. RESULTS Saglin protein was detected by indirect immunofluorescence microscopy only in the medial and proximal-lateral lobes, but not in the distal-lateral lobes, of the salivary glands of An. gambiae; the pattern of expression was independent of mosquito age or physiological state. These results were confirmed by steady-state Saglin transcript and protein expression using qRT-PCR and Western-blot analysis, respectively. Saglin was localized to the basal surface of the cells of the medial lobes and was undetectable elsewhere (intracellularly, on the lateral or apical membranes, the cells' secretory vacuoles, or in the salivary duct). In the cells of the proximal lateral lobes of the salivary glands, Saglin was distinctly intracellular and was not localized to any of the cell surfaces. Transgenic Anopheles stephensi were produced that expressed An. gambiae Saglin in the distal lateral lobes of the salivary gland. Additional Saglin expression did not enhance infection by PfSPZ compared to non-transgenic siblings fed on the same gametocyte-containing blood meal. CONCLUSIONS The absence of Saglin in the distal lateral lobes of the salivary glands, a primary destination for SPZ, suggests Saglin is not an essential receptor for Plasmodium SPZ. The lack of any correlation between increased Saglin expression in the distal lateral lobes of the salivary glands of transgenic An. stephensi and PfSPZ infection is also consistent with Saglin not being an essential salivary gland receptor for Plasmodium SPZ.
Collapse
Affiliation(s)
- David A O'Brochta
- Department of Entomology and The Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, 9600 Gudelsky Drive, Rockville, MD, 20850, USA.,Foundation for the National Institutes of Health, 11400 Rockville Pike, Suite 600, North Bethesda, MD, 20852, USA
| | - Robert Alford
- Department of Entomology and The Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Robert Harrell
- University of Maryland Insect Transformation Facility, The Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Channa Aluvihare
- University of Maryland Insect Transformation Facility, The Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Abraham G Eappen
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Tao Li
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Sumana Chakravarty
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - B Kim Lee Sim
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Stephen L Hoffman
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Peter F Billingsley
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA.
| |
Collapse
|
173
|
Ricotta E, Kwan J. Artemisinin-Resistant Malaria as a Global Catastrophic Biological Threat. Curr Top Microbiol Immunol 2019; 424:33-57. [PMID: 31218504 DOI: 10.1007/82_2019_163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The global spread of artemisinin resistance brings with it the threat of incurable malaria. Already, this disease threatens over 219 million lives per year and causes 5-6% losses in GDP in endemic areas, even with current advances in prevention and treatment. This chapter discusses the currently tenuous position we are in globally, and the impact that could be seen if artemisinin treatment is lost, whether due to the unchecked spread of K13 mutations or poor global investment in treatment and prevention advances. Artemisinin is the backbone of current ACT treatment programs and severe malarial treatment; without it, the success of future malaria eradication programs will be in jeopardy.
Collapse
Affiliation(s)
- Emily Ricotta
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Quarters 15B-1, 8 West Dr, Bethesda, MD, 20892, USA.
- Kelly Government Solutions, Bethesda, USA.
| | - Jennifer Kwan
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Quarters 15B-1, 8 West Dr, Bethesda, MD, 20892, USA
| |
Collapse
|
174
|
Abstract
Vaccines that target the preerythrocytic phase of malaria hold great promise as elimination tools since they are the sole vaccines that can achieve sterile protection against a challenge. This chapter focuses on preerythrocytic stage vaccines based on live attenuated parasites. It first summarizes the main conclusions that have emerged from studies in rodents, which compared various parasite attenuation methods, and then presents the vaccination regimens that are currently being tested in humans.
Collapse
|
175
|
Itsara LS, Zhou Y, Do J, Grieser AM, Vaughan AM, Ghosh AK. The Development of Whole Sporozoite Vaccines for Plasmodium falciparum Malaria. Front Immunol 2018; 9:2748. [PMID: 30619241 PMCID: PMC6297750 DOI: 10.3389/fimmu.2018.02748] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/08/2018] [Indexed: 12/20/2022] Open
Abstract
Each year malaria kills hundreds of thousands of people and infects hundreds of millions of people despite current control measures. An effective malaria vaccine will likely be necessary to aid in malaria eradication. Vaccination using whole sporozoites provides an increased repertoire of immunogens compared to subunit vaccines across at least two life cycle stages of the parasite, the extracellular sporozoite, and intracellular liver stage. Three potential whole sporozoite vaccine approaches are under development and include genetically attenuated parasites, radiation attenuated sporozoites, and wild-type sporozoites administered in combination with chemoprophylaxis. Pre-clinical and clinical studies have demonstrated whole sporozoite vaccine immunogenicity, including humoral and cellular immunity and a range of vaccine efficacy that depends on the pre-exposure of vaccinated individuals. While whole sporozoite vaccines can provide protection against malaria in some cases, more recent studies in malaria-endemic regions demonstrate the need for improvements. Moreover, challenges remain in manufacturing large quantities of sporozoites for vaccine commercialization. A promising solution to the whole sporozoite manufacturing challenge is in vitro culturing methodology, which has been described for several Plasmodium species, including the major disease-causing human malaria parasite, Plasmodium falciparum. Here, we review whole sporozoite vaccine immunogenicity and in vitro culturing platforms for sporozoite production.
Collapse
Affiliation(s)
| | | | - Julie Do
- MalarVx, Inc., Seattle, WA, United States
| | | | - Ashley M Vaughan
- Seattle Children's Research Institute, Seattle, WA, United States
| | | |
Collapse
|
176
|
Frimpong A, Kusi KA, Ofori MF, Ndifon W. Novel Strategies for Malaria Vaccine Design. Front Immunol 2018; 9:2769. [PMID: 30555463 PMCID: PMC6281765 DOI: 10.3389/fimmu.2018.02769] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022] Open
Abstract
The quest for a licensed effective vaccine against malaria remains a global priority. Even though classical vaccine design strategies have been successful for some viral and bacterial pathogens, little success has been achieved for Plasmodium falciparum, which causes the deadliest form of malaria due to its diversity and ability to evade host immune responses. Nevertheless, recent advances in vaccinology through high throughput discovery of immune correlates of protection, lymphocyte repertoire sequencing and structural design of immunogens, provide a comprehensive approach to identifying and designing a highly efficacious vaccine for malaria. In this review, we discuss novel vaccine approaches that can be employed in malaria vaccine design.
Collapse
Affiliation(s)
- Augustina Frimpong
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.,African Institute for Mathematical Sciences, Cape Coast, Ghana
| | - Kwadwo Asamoah Kusi
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Michael Fokuo Ofori
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Wilfred Ndifon
- African Institute for Mathematical Sciences, Cape Coast, Ghana.,African Institute for Mathematical Sciences, University of Stellenbosch, Cape Town, South Africa
| |
Collapse
|
177
|
Pre-clinical evaluation of a P. berghei-based whole-sporozoite malaria vaccine candidate. NPJ Vaccines 2018; 3:54. [PMID: 30510775 PMCID: PMC6258718 DOI: 10.1038/s41541-018-0091-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/17/2018] [Indexed: 11/11/2022] Open
Abstract
Whole-sporozoite vaccination/immunization induces high levels of protective immunity in both rodent models of malaria and in humans. Recently, we generated a transgenic line of the rodent malaria parasite P. berghei (Pb) that expresses the P. falciparum (Pf) circumsporozoite protein (PfCS), and showed that this parasite line (PbVac) was capable of (1) infecting and developing in human hepatocytes but not in human erythrocytes, and (2) inducing neutralizing antibodies against the human Pf parasite. Here, we analyzed PbVac in detail and developed tools necessary for its use in clinical studies. A microbiological contaminant-free Master Cell Bank of PbVac parasites was generated through a process of cyclic propagation and clonal expansion in mice and mosquitoes and was genetically characterized. A highly sensitive qRT-PCR-based method was established that enables PbVac parasite detection and quantification at low parasite densities in vivo. This method was employed in a biodistribution study in a rabbit model, revealing that the parasite is only present at the site of administration and in the liver up to 48 h post infection and is no longer detectable at any site 10 days after administration. An extensive toxicology investigation carried out in rabbits further showed the absence of PbVac-related toxicity. In vivo drug sensitivity assays employing rodent models of infection showed that both the liver and the blood stage forms of PbVac were completely eliminated by Malarone® treatment. Collectively, our pre-clinical safety assessment demonstrates that PbVac possesses all characteristics necessary to advance into clinical evaluation. PbVac is a transgenic malaria parasite expressing circumsporozoite antigen from the human parasite Plasmodium falciparum. PbVac elicits neutralizing P. falciparum antibodies and can infect human hepatocytes but not erythrocytes, suggesting that humans would be non-permissive. Miguel Prudêncio and colleagues at the Institute of Molecular Medicine in Lisbon perform a detailed in vivo analysis and toxicology of PbVac. Extensive biodistribution analysis using a highly sensitive qPCR in non-permissive rabbit hosts shows PbVac are present at the initial bite site early on with later appearance in the liver, but by day 10 is undetectable. Importantly no PbVac could be detected in the blood at any time-point. PbVac was well tolerated with no apparent pathological signatures. In permissive mouse hosts PbVac could be effectively eliminated from both the blood and liver and could thereby act as a potential clinical ‘safety net’ in the event of an erythrocytic stage or persistence in the liver.
Collapse
|
178
|
Abstract
CD8 T cells comprising the memory pool display considerable heterogeneity, with individual cells differing in phenotype and function. This review will focus on our current understanding of heterogeneity within the antigen-specific memory CD8 T cell compartment and classifications of memory CD8 T cell subsets with defined and discrete functionalities. Recent data suggest that phenotype and/or function of numerically stable circulatory memory CD8 T cells are defined by the age of memory CD8 T cell (or time after initial antigen-encounter). In addition, history of antigen stimulations has a profound effect on memory CD8 T cell populations, suggesting that repeated infections (or vaccination) have the capacity to further shape the memory CD8 T cell pool. Finally, genetic background of hosts and history of exposure to diverse microorganisms likely contribute to the observed heterogeneity in the memory CD8 T cell compartment. Extending our tool box and exploring alternative mouse models (i.e., "dirty" and/or outbred mice) to encompass and better model diversity observed in humans will remain an important goal for the near future that will likely shed new light into the mechanisms that govern biology of memory CD8 T cells.
Collapse
Affiliation(s)
- Matthew D Martin
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA, United States.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
179
|
Islam MA, Firdous J, Badruddoza AZM, Reesor E, Azad M, Hasan A, Lim M, Cao W, Guillemette S, Cho CS. M cell targeting engineered biomaterials for effective vaccination. Biomaterials 2018; 192:75-94. [PMID: 30439573 DOI: 10.1016/j.biomaterials.2018.10.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/09/2018] [Accepted: 10/28/2018] [Indexed: 02/08/2023]
Abstract
Vaccines are one of the greatest medical interventions of all time and have been successful in controlling and eliminating a myriad of diseases over the past two centuries. Among several vaccination strategies, mucosal vaccines have wide clinical applications and attract considerable interest in research, showing potential as innovative and novel therapeutics. In mucosal vaccination, targeting (microfold) M cells is a frontline prerequisite for inducing effective antigen-specific immunostimulatory effects. In this review, we primarily focus on materials engineered for use as vaccine delivery platforms to target M cells. We also describe potential M cell targeting areas, methods to overcome current challenges and limitations of the field. Furthermore, we present the potential of biomaterials engineering as well as various natural and synthetic delivery technologies to overcome the challenges of M cell targeting, all of which are absent in current literature. Finally, we briefly discuss manufacturing and regulatory processes to bring a robust perspective on the feasibility and potential of this next-generation vaccine technology.
Collapse
Affiliation(s)
- Mohammad Ariful Islam
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Jannatul Firdous
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Abu Zayed Md Badruddoza
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Emma Reesor
- Department of Nanotechnology Engineering, University of Waterloo, Waterloo, Canada
| | - Mohammad Azad
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Michael Lim
- Department of Nanotechnology Engineering, University of Waterloo, Waterloo, Canada
| | - Wuji Cao
- Department of Nanotechnology Engineering, University of Waterloo, Waterloo, Canada
| | - Simon Guillemette
- Department of Nanotechnology Engineering, University of Waterloo, Waterloo, Canada
| | - Chong Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
180
|
Cockburn IA, Seder RA. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat Immunol 2018; 19:1199-1211. [PMID: 30333613 DOI: 10.1038/s41590-018-0228-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/31/2018] [Indexed: 02/08/2023]
Abstract
Development of a malaria vaccine remains a critical priority to decrease clinical disease and mortality and facilitate eradication. Accordingly, RTS,S, a protein-subunit vaccine, has completed phase III clinical trials and confers ~30% protection against clinical infection over 4 years. Whole-attenuated-sporozoite and viral-subunit vaccines induce between 20% and 100% protection against controlled human malaria infection, but there is limited published evidence to date for durable, high-level efficacy (>50%) against natural exposure. Importantly, fundamental scientific advances related to the potency, durability, breadth and location of immune responses will be required for improving vaccine efficacy with these and other vaccine approaches. In this Review, we focus on the current understanding of immunological mechanisms of protection from animal models and human vaccine studies, and on how these data should inform the development of next-generation vaccines. Furthermore, we introduce the concept of using passive immunization with monoclonal antibodies as a new approach to prevent and eliminate malaria.
Collapse
Affiliation(s)
- Ian A Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
181
|
Emran TB, Iyori M, Ono Y, Amelia F, Yusuf Y, Islam A, Alam A, Tamura M, Ogawa R, Matsuoka H, Yamamoto DS, Yoshida S. Baculovirus-Induced Fast-Acting Innate Immunity Kills Liver-Stage Plasmodium. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2441-2451. [PMID: 30209187 DOI: 10.4049/jimmunol.1800908] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/15/2018] [Indexed: 12/14/2022]
Abstract
Baculovirus (BV), an enveloped insect virus with a circular dsDNA genome, possesses unique characteristics that induce strong innate immune responses in mammalian cells. In this study, we show that BV administration in BALB/c mice not only provides complete protection against a subsequent Plasmodium berghei sporozoite infection for up to 7 d after the injection but also eliminates existing liver-stage parasites completely. The elimination of sporozoites by BV was superior to that by primaquine, and this effect occurred in a TLR9-independent manner. At 6 h after BV administration, IFN-α and IFN-γ were robustly produced in the serum, and RNA transcripts of IFN-stimulated genes were markedly upregulated in the liver compared with control mice. The in vivo passive transfer of serum after BV administration effectively eliminated liver-stage parasites, and IFN-α neutralization abolished this effect, indicating that the BV liver-stage parasite-killing mechanism is downstream of the type I IFN signaling pathway. These findings provide evidence that BV-induced, fast-acting innate immunity completely kills liver-stage parasites and, thus, may lead to new malaria drug and vaccine strategies.
Collapse
Affiliation(s)
- Talha Bin Emran
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yuki Ono
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Fitri Amelia
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Ashekul Islam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Asrar Alam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Megumi Tamura
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Ryohei Ogawa
- Department of Radiological Sciences, University of Toyama, Toyama 930-0194, Japan; and
| | - Hiroyuki Matsuoka
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0431, Japan
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0431, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan;
| |
Collapse
|
182
|
Gola A, Silman D, Walters AA, Sridhar S, Uderhardt S, Salman AM, Halbroth BR, Bellamy D, Bowyer G, Powlson J, Baker M, Venkatraman N, Poulton I, Berrie E, Roberts R, Lawrie AM, Angus B, Khan SM, Janse CJ, Ewer KJ, Germain RN, Spencer AJ, Hill AVS. Prime and target immunization protects against liver-stage malaria in mice. Sci Transl Med 2018; 10:10/460/eaap9128. [DOI: 10.1126/scitranslmed.aap9128] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 03/08/2018] [Accepted: 08/20/2018] [Indexed: 12/24/2022]
Abstract
Despite recent advances in treatment and vector control, malaria is still a leading cause of death, emphasizing the need for an effective vaccine. The malaria life cycle can be subdivided into three stages: the invasion and growth within liver hepatocytes (pre-erythrocytic stage), the blood stage (erythrocytic stage), and, finally, the sexual stage (occurring within the mosquito vector). Antigen (Ag)-specific CD8+ T cells are effectively induced by heterologous prime-boost viral vector immunization and known to correlate with liver-stage protection. However, liver-stage malaria vaccines have struggled to generate and maintain the high numbers of Plasmodium-specific circulating T cells necessary to confer sterile protection. We describe an alternative “prime and target” vaccination strategy aimed specifically at inducing high numbers of tissue-resident memory T cells present in the liver at the time of hepatic infection. This approach bypasses the need for very high numbers of circulating T cells and markedly increases the efficacy of subunit immunization against liver-stage malaria with clinically relevant Ags and clinically tested viral vectors in murine challenge models. Translation to clinical use has begun, with encouraging results from a pilot safety and feasibility trial of intravenous chimpanzee adenovirus vaccination in humans. This work highlights the value of a prime-target approach for immunization against malaria and suggests that this strategy may represent a more general approach for prophylaxis or immunotherapy of other liver infections and diseases.
Collapse
|
183
|
McCall MBB, Kremsner PG, Mordmüller B. Correlating efficacy and immunogenicity in malaria vaccine trials. Semin Immunol 2018; 39:52-64. [PMID: 30219621 DOI: 10.1016/j.smim.2018.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 08/06/2018] [Indexed: 12/19/2022]
Abstract
The availability of an effective and appropriately implemented malaria vaccine would form a crucial cornerstone of public health efforts to fight this disease. Despite many decades of research, however, no malaria vaccine has yet shown satisfactory protective efficacy or been rolled-out. Validated immunological substitute endpoints have the potential to accelerate clinical vaccine development by reducing the required complexity, size, duration and cost of clinical trials. Besides facilitating clinical development of existing vaccine candidates, understanding immunological mechanisms of protection may drive the development of fundamentally new vaccination approaches. In this review we focus on correlates of protection in malaria vaccine development: Does immunogenicity predict malaria vaccine efficacy and why is this question particularly difficult? Have immunological correlates accelerated malaria vaccine development in the past and will they facilitate it in the future? Does Controlled Human Malaria Infection represent a valid model for identifying such immunological correlates, or a correlate of protection against naturally-acquired malaria in itself?
Collapse
Affiliation(s)
- Matthew B B McCall
- Institut für Tropenmedizin, Universität Tübingen and Deutsches Zentrum für Infektionsforschung, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.
| | - Peter G Kremsner
- Institut für Tropenmedizin, Universität Tübingen and Deutsches Zentrum für Infektionsforschung, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Benjamin Mordmüller
- Institut für Tropenmedizin, Universität Tübingen and Deutsches Zentrum für Infektionsforschung, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|
184
|
Olsen TM, Stone BC, Chuenchob V, Murphy SC. Prime-and-Trap Malaria Vaccination To Generate Protective CD8 + Liver-Resident Memory T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:1984-1993. [PMID: 30127085 DOI: 10.4049/jimmunol.1800740] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/02/2018] [Indexed: 11/19/2022]
Abstract
Tissue-resident memory CD8+ T (Trm) cells in the liver are critical for long-term protection against pre-erythrocytic Plasmodium infection. Such protection can usually be induced with three to five doses of i.v. administered radiation-attenuated sporozoites (RAS). To simplify and accelerate vaccination, we tested a DNA vaccine designed to induce potent T cell responses against the SYVPSAEQI epitope of Plasmodium yoelii circumsporozoite protein. In a heterologous "prime-and-trap" regimen, priming using gene gun-administered DNA and boosting with one dose of RAS attracted expanding Ag-specific CD8+ T cell populations to the liver, where they became Trm cells. Vaccinated in this manner, BALB/c mice were completely protected against challenge, an outcome not reliably achieved following one dose of RAS or following DNA-only vaccination. This study demonstrates that the combination of CD8+ T cell priming by DNA and boosting with liver-homing RAS enhances formation of a completely protective liver Trm cell response and suggests novel approaches for enhancing T cell-based pre-erythrocytic malaria vaccines.
Collapse
Affiliation(s)
- Tayla M Olsen
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98109.,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109
| | - Brad C Stone
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98109.,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109
| | - Vorada Chuenchob
- Center for Infectious Disease Research, University of Washington, Seattle, WA 98109; and
| | - Sean C Murphy
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98109; .,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109.,Department of Microbiology, University of Washington, Seattle, WA 98195
| |
Collapse
|
185
|
Murphy SC, Ishizuka AS, Billman ZP, Olsen TM, Seilie AM, Chang M, Smith N, Chuenchob V, Chakravarty S, Sim BKL, Kappe SHI, Hoffman SL, Seder RA. Plasmodium 18S rRNA of intravenously administered sporozoites does not persist in peripheral blood. Malar J 2018; 17:275. [PMID: 30053881 PMCID: PMC6062992 DOI: 10.1186/s12936-018-2422-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/19/2018] [Indexed: 01/14/2023] Open
Abstract
Background Plasmodium 18S rRNA is a biomarker used to monitor blood-stage infections in malaria clinical trials. Plasmodium sporozoites also express this biomarker, and there is conflicting evidence about how long sporozoite-derived 18S rRNA persists in peripheral blood. If present in blood for an extended timeframe, sporozoite-derived 18S rRNA could complicate use as a blood-stage biomarker. Methods Blood samples from Plasmodium yoelii infected mice were tested for Plasmodium 18S rRNA and their coding genes (rDNA) using sensitive quantitative reverse transcription PCR and quantitative PCR assays, respectively. Blood and tissues from Plasmodium falciparum sporozoite (PfSPZ)-infected rhesus macaques were similarly tested. Results In mice, when P. yoelii sporozoite inoculation and blood collection were performed at the same site (tail vein), low level rDNA positivity persisted for 2 days post-infection. Compared to intact parasites with high rRNA-to-rDNA ratios, this low level positivity was accompanied by no increase in rRNA-to-rDNA, indicating detection of residual, non-viable parasite rDNA. When P. yoelii sporozoites were administered via the retro-orbital vein and blood sampled by cardiac puncture, neither P. yoelii 18S rRNA nor rDNA were detected 24 h post-infection. Similarly, there was no P. falciparum 18S rRNA detected in blood of rhesus macaques 3 days after intravenous injection with extremely high doses of PfSPZ. Plasmodium 18S rRNA in the rhesus livers increased by approximately 101-fold from 3 to 6 days post infection, indicating liver-stage proliferation. Conclusions Beyond the first few hours after injection, sporozoite-derived Plasmodium 18S rRNA was not detected in peripheral blood. Diagnostics based on 18S rRNA are unlikely to be confounded by sporozoite inocula in human clinical trials.
Collapse
Affiliation(s)
- Sean C Murphy
- Departments of Laboratory Medicine and Microbiology, University of Washington, 750 Republican St., E630, Seattle, WA, 98109, USA. .,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, 750 Republican St., Seattle, WA, 98109, USA.
| | - Andrew S Ishizuka
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 40, Room 3512, 40 Convent Drive, Bethesda, MD, 20814, USA
| | - Zachary P Billman
- Departments of Laboratory Medicine and Microbiology, University of Washington, 750 Republican St., E630, Seattle, WA, 98109, USA
| | - Tayla M Olsen
- Departments of Laboratory Medicine and Microbiology, University of Washington, 750 Republican St., E630, Seattle, WA, 98109, USA
| | - Annette M Seilie
- Departments of Laboratory Medicine and Microbiology, University of Washington, 750 Republican St., E630, Seattle, WA, 98109, USA
| | - Ming Chang
- Departments of Laboratory Medicine and Microbiology, University of Washington, 750 Republican St., E630, Seattle, WA, 98109, USA
| | - Nahum Smith
- Departments of Laboratory Medicine and Microbiology, University of Washington, 750 Republican St., E630, Seattle, WA, 98109, USA
| | - Vorada Chuenchob
- Center for Infectious Disease Research, 307 Westlake Ave N #500, Seattle, WA, 98109, USA
| | - Sumana Chakravarty
- Sanaria, Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - B Kim Lee Sim
- Sanaria, Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Stefan H I Kappe
- Center for Infectious Disease Research, 307 Westlake Ave N #500, Seattle, WA, 98109, USA
| | - Stephen L Hoffman
- Sanaria, Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 40, Room 3512, 40 Convent Drive, Bethesda, MD, 20814, USA
| |
Collapse
|
186
|
Flaxman A, Ewer KJ. Methods for Measuring T-Cell Memory to Vaccination: From Mouse to Man. Vaccines (Basel) 2018; 6:E43. [PMID: 30037078 PMCID: PMC6161152 DOI: 10.3390/vaccines6030043] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 12/27/2022] Open
Abstract
The development of effective vaccines continues to be a key goal for public health bodies, governments, funding bodies and pharmaceutical companies. With new vaccines such as Shingrix targeting Shingles and Bexsero for Meningitis B, licensed in recent years, today's population can be protected from more infectious diseases than ever before. Despite this, we are yet to license vaccines for some of the deadliest endemic diseases affecting children, such as malaria. In addition, the threat of epidemics caused by emerging pathogens is very real as exemplified by the 2014⁻2016 Ebola outbreak. Most licensed vaccines provide efficacy through humoral immunity and correlates of protection often quantify neutralising antibody titre. The role of T-cells in vaccine efficacy is less well understood and more complex to quantify. Defining T-cell responses which afford protection also remains a challenge, although more sophisticated assays for assessing cell-mediated immunity with the potential for higher throughput and scalability are now available and warrant review. Here we discuss the benefits of multiparameter cytokine analysis and omics approaches compared with flow cytometric and ELISpot assays. We also review technical challenges unique to clinical trial studies, including assay validation across laboratories and availability of sample type. Measuring T-cell immunogenicity alongside humoral responses provides information on the breadth of immune responses induced by vaccination. Accurately enumerating and phenotyping T-cell immunogenicity to vaccination is key for the determination of immune correlates of protection. However, identifying such T-cell parameters remains challenging without a clear understanding of the immunological mechanisms by which a T-cell-mediated response induces protection.
Collapse
Affiliation(s)
- Amy Flaxman
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| |
Collapse
|
187
|
Jongo SA, Shekalaghe SA, Church LWP, Ruben AJ, Schindler T, Zenklusen I, Rutishauser T, Rothen J, Tumbo A, Mkindi C, Mpina M, Mtoro AT, Ishizuka AS, Kassim KR, Milando FA, Qassim M, Juma OA, Mwakasungula S, Simon B, James ER, Abebe Y, Kc N, Chakravarty S, Saverino E, Bakari BM, Billingsley PF, Seder RA, Daubenberger C, Sim BKL, Richie TL, Tanner M, Abdulla S, Hoffman SL. Safety, Immunogenicity, and Protective Efficacy against Controlled Human Malaria Infection of Plasmodium falciparum Sporozoite Vaccine in Tanzanian Adults. Am J Trop Med Hyg 2018; 99:338-349. [PMID: 29943719 PMCID: PMC6090339 DOI: 10.4269/ajtmh.17-1014] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
We are using controlled human malaria infection (CHMI) by direct venous inoculation (DVI) of cryopreserved, infectious Plasmodium falciparum (Pf) sporozoites (SPZ) (PfSPZ Challenge) to try to reduce time and costs of developing PfSPZ Vaccine to prevent malaria in Africa. Immunization with five doses at 0, 4, 8, 12, and 20 weeks of 2.7 × 105 PfSPZ of PfSPZ Vaccine gave 65% vaccine efficacy (VE) at 24 weeks against mosquito bite CHMI in U.S. adults and 52% (time to event) or 29% (proportional) VE over 24 weeks against naturally transmitted Pf in Malian adults. We assessed the identical regimen in Tanzanians for VE against PfSPZ Challenge. Twenty- to thirty-year-old men were randomized to receive five doses normal saline or PfSPZ Vaccine in a double-blind trial. Vaccine efficacy was assessed 3 and 24 weeks later. Adverse events were similar in vaccinees and controls. Antibody responses to Pf circumsporozoite protein were significantly lower than in malaria-naïve Americans, but significantly higher than in Malians. All 18 controls developed Pf parasitemia after CHMI. Four of 20 (20%) vaccinees remained uninfected after 3 week CHMI (P = 0.015 by time to event, P = 0.543 by proportional analysis) and all four (100%) were uninfected after repeat 24 week CHMI (P = 0.005 by proportional, P = 0.004 by time to event analysis). Plasmodium falciparum SPZ Vaccine was safe, well tolerated, and induced durable VE in four subjects. Controlled human malaria infection by DVI of PfSPZ Challenge appeared more stringent over 24 weeks than mosquito bite CHMI in United States or natural exposure in Malian adults, thereby providing a rigorous test of VE in Africa.
Collapse
Affiliation(s)
- Said A Jongo
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Seif A Shekalaghe
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | | | | | - Tobias Schindler
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - Isabelle Zenklusen
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - Tobias Rutishauser
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - Julian Rothen
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - Anneth Tumbo
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Catherine Mkindi
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Maximillian Mpina
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Ali T Mtoro
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Andrew S Ishizuka
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | | | - Florence A Milando
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Munira Qassim
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Omar A Juma
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Solomon Mwakasungula
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Beatus Simon
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | | | | | | | | | | | - Bakari M Bakari
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | | | - Robert A Seder
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Claudia Daubenberger
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - B Kim Lee Sim
- Protein Potential LLC, Rockville, Maryland.,Sanaria Inc., Rockville, Maryland
| | | | - Marcel Tanner
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - Salim Abdulla
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | | |
Collapse
|
188
|
Frank R, Gabel M, Heiss K, Mueller AK, Graw F. Varying Immunizations With Plasmodium Radiation-Attenuated Sporozoites Alter Tissue-Specific CD8 + T Cell Dynamics. Front Immunol 2018; 9:1137. [PMID: 29892289 PMCID: PMC5985394 DOI: 10.3389/fimmu.2018.01137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/07/2018] [Indexed: 12/12/2022] Open
Abstract
Whole sporozoite vaccines represent one of the most promising strategies to induce protection against malaria. However, the development of efficient vaccination protocols still remains a major challenge. To understand how the generation of immunity is affected by variations in vaccination dosage and frequency, we systematically analyzed intrasplenic and intrahepatic CD8+ T cell responses following varied immunizations of mice with radiation-attenuated sporozoites. By combining experimental data and mathematical modeling, our analysis indicates a reversing role of spleen and liver in the generation of protective liver-resident CD8+ T cells during priming and booster injections: While the spleen acts as a critical source compartment during priming, the increase in vaccine-induced hepatic T cell levels is likely due to local reactivation in the liver in response to subsequent booster injections. Higher dosing accelerates the efficient generation of liver-resident CD8+ T cells by especially affecting their local reactivation. In addition, we determine the differentiation and migration pathway from splenic precursors toward hepatic memory cells thereby presenting a mechanistic framework for the impact of various vaccination protocols on these dynamics. Thus, our work provides important insights into organ-specific CD8+ T cell dynamics and their role and interplay in the formation of protective immunity against malaria.
Collapse
Affiliation(s)
- Roland Frank
- Centre for Infectious Diseases, Parasitology Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Gabel
- Centre for Modeling and Simulation in the Biosciences, BioQuant-Center, Heidelberg University, Heidelberg, Germany
| | - Kirsten Heiss
- Centre for Infectious Diseases, Parasitology Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - Ann-Kristin Mueller
- Centre for Infectious Diseases, Parasitology Unit, University Hospital Heidelberg, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Frederik Graw
- Centre for Modeling and Simulation in the Biosciences, BioQuant-Center, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
189
|
Emran TB, Iyori M, Ono Y, Amelia F, Yusuf Y, Islam A, Alam A, Ogawa R, Matsuoka H, Yamamoto D, Yoshida S. Baculovirus-inducing fast-acting innate immunity kills Plasmodium liver stages.. [DOI: 10.1101/320036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
ABSTRACTBaculovirus (BV), an enveloped insect virus with a circular double-stranded DNA genome, possesses unique characteristics that induce strong innate immune responses in mammalian cells. Here, we show that BV administration not only sterilely protects BALB/c mice for at least 7 days from subsequent Plasmodium berghei sporozoite infection but also eliminates existing liver-stage parasites completely, effects superior to those of primaquine, and does so in a TLR9-independent manner. Six hours post-BV administration, IFN-α and IFN-γ were robustly produced in serum, and RNA transcripts of interferon-stimulated genes were drastically upregulated in the liver. The in vivo passive transfer of post-BV administration serum effectively eliminated liver-stage parasites, and IFN-α neutralization abolished this effect, indicating that the BV liver-stage parasite killing mechanism is downstream of the type I IFN signaling pathway. Our results demonstrate that BV is a potent IFN-inducing prophylactic and therapeutic agent with great potential for further development as a new malaria vaccine and/or anti-hypnozoite drug.
Collapse
|
190
|
A human monoclonal antibody prevents malaria infection by targeting a new site of vulnerability on the parasite. Nat Med 2018; 24:408-416. [PMID: 29554083 PMCID: PMC5893371 DOI: 10.1038/nm.4512] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 01/07/2023]
Abstract
Development of a highly effective vaccine or antibodies for the prevention and ultimately elimination of malaria is urgently needed. Here we report the isolation of a number of human monoclonal antibodies directed against the Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP) from several subjects immunized with an attenuated Pf whole-sporozoite (SPZ) vaccine (Sanaria PfSPZ Vaccine). Passive transfer of one of these antibodies, monoclonal antibody CIS43, conferred high-level, sterile protection in two different mouse models of malaria infection. The affinity and stoichiometry of CIS43 binding to PfCSP indicate that there are two sequential multivalent binding events encompassing the repeat domain. The first binding event is to a unique 'junctional' epitope positioned between the N terminus and the central repeat domain of PfCSP. Moreover, CIS43 prevented proteolytic cleavage of PfCSP on PfSPZ. Analysis of crystal structures of the CIS43 antigen-binding fragment in complex with the junctional epitope determined the molecular interactions of binding, revealed the epitope's conformational flexibility and defined Asn-Pro-Asn (NPN) as the structural repeat motif. The demonstration that CIS43 is highly effective for passive prevention of malaria has potential application for use in travelers, military personnel and elimination campaigns and identifies a new and conserved site of vulnerability on PfCSP for next-generation rational vaccine design.
Collapse
|
191
|
Reiman JM, Kumar S, Rodriguez IB, Gnidehou S, Ito K, Stanisic DI, Lee M, McPhun V, Majam V, Willemsen NM, Batzloff MR, Raja AI, Dooley B, Hoffman SL, Yanow SK, Good MF. Induction of immunity following vaccination with a chemically attenuated malaria vaccine correlates with persistent antigenic stimulation. Clin Transl Immunology 2018; 7:e1015. [PMID: 29670745 PMCID: PMC5894007 DOI: 10.1002/cti2.1015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/13/2018] [Accepted: 03/14/2018] [Indexed: 12/12/2022] Open
Abstract
Objectives Blood stage malaria parasites attenuated with seco-cyclopropyl pyrrolo indole (CPI) analogues induce robust immunity in mice to homologous and heterologous malaria parasites and are being considered for the development of a human vaccine. However, it is not understood how attenuated parasites induce immunity. We showed that following vaccination, parasite DNA persisted in blood for several months, raising the possibility that ongoing immune stimulation may be critical. However, parasites were not seen microscopically beyond 24 h postvaccination. We aimed to provide a mechanistic understanding of immune induction. Methods Mice were vaccinated with chemically attenuated Plasmodium chabaudi parasites. PCR and adoptive transfer studies were used to determine the presence of parasites and antigen in vivo. In other experiments, Plasmodium falciparum parasitised red blood cells were attenuated in vitro and RNA and antigen expression studied. Results We show that blood transferred from vaccinated mice into naïve mice activates T cells and induces complete protective immunity in the recipient mice strongly suggesting that there is persistence of parasite antigen postvaccination. This is supported by the presence of parasite RNA in vaccinated mice and both RNA and antigen expression in P. falciparum cultures treated with CPI drugs in vitro. In addition, drugs that block parasite growth also prevent the induction of immunity in vaccinated mice, indicating that some growth of attenuated parasites is required for immune induction. Conclusions Attenuated parasites persist at submicroscopic levels in the blood of mice postvaccination with the ability to activate T cells and induce ongoing protective immune responses.
Collapse
Affiliation(s)
- Jennifer M Reiman
- Institute for Glycomics Griffith University Gold Coast QLD Australia
| | - Sanjai Kumar
- Laboratory of Emerging Pathogens Division of Emerging and Transfusion Transmitted Diseases CBER, FDA Rockville MD USA
| | | | | | - Koichi Ito
- Institute for Glycomics Griffith University Gold Coast QLD Australia
| | | | - Moses Lee
- Georgia State University Atlanta GA USA
| | - Virginia McPhun
- Institute for Glycomics Griffith University Gold Coast QLD Australia
| | - Victoria Majam
- Laboratory of Emerging Pathogens Division of Emerging and Transfusion Transmitted Diseases CBER, FDA Rockville MD USA
| | | | | | - Amber I Raja
- Institute for Glycomics Griffith University Gold Coast QLD Australia
| | - Brad Dooley
- Institute for Glycomics Griffith University Gold Coast QLD Australia
| | | | | | - Michael F Good
- Institute for Glycomics Griffith University Gold Coast QLD Australia
| |
Collapse
|
192
|
Gray JI, Westerhof LM, MacLeod MKL. The roles of resident, central and effector memory CD4 T-cells in protective immunity following infection or vaccination. Immunology 2018; 154:574-581. [PMID: 29570776 PMCID: PMC6050220 DOI: 10.1111/imm.12929] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 03/02/2018] [Accepted: 03/06/2018] [Indexed: 12/25/2022] Open
Abstract
Immunological memory provides rapid protection to pathogens previously encountered through infection or vaccination. CD4 T-cells play a central role in all adaptive immune responses. Vaccines must, therefore, activate CD4 T-cells if they are to generate protective immunity. For many diseases, we do not have effective vaccines. These include human immunodeficiency virus (HIV), tuberculosis and malaria, which are responsible for many millions of deaths each year across the globe. CD4 T-cells play many different roles during the immune response coordinating the actions of many other cells. In order to harness the diverse protective effects of memory CD4 T-cells, we need to understand how memory CD4 T-cells are generated and how they protect the host. Here we review recent findings on the location of different subsets of memory CD4 T-cells that are found in peripheral tissues (tissue resident memory T-cells) and in the circulation (central and effector memory T-cells). We discuss the generation of these cells, and the evidence that demonstrates how they provide immune protection in animal and human challenge models.
Collapse
Affiliation(s)
- Joshua I. Gray
- Centre for ImmunobiologyInstitute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
| | - Lotus M. Westerhof
- Centre for ImmunobiologyInstitute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
- GLAZgo Discovery CentreInstitute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
| | - Megan K. L. MacLeod
- Centre for ImmunobiologyInstitute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
| |
Collapse
|
193
|
Nlinwe ON, Kusi KA, Adu B, Sedegah M. T-cell responses against Malaria: Effect of parasite antigen diversity and relevance for vaccine development. Vaccine 2018; 36:2237-2242. [PMID: 29573877 DOI: 10.1016/j.vaccine.2018.03.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 01/21/2018] [Accepted: 03/07/2018] [Indexed: 11/24/2022]
Abstract
The on-going agenda for global malaria elimination will require the development of additional disease control and prevention measures since currently available tools are showing signs of inadequacy. Malaria vaccines are seen as one such important addition to the control arsenal since vaccines have proven to be highly effective public health tools against important human diseases. Both cell-mediated and antibody responses are generally believed to be important for malaria parasite control, although the exact targets of T and B cell responses against malaria have not been clearly defined. However, our current understanding of the immune response to malaria suggests that T cell responses against multiple antigenic targets may potentially be key for the development of a highly efficacious malaria vaccine. This review takes a comprehensive look at the available literature on T cell-mediated immunity against all human stages of the malaria parasite and the effect of antigen diversity on these responses. The implications of these interrelationships for the development of an effective vaccine for malaria are also highlighted.
Collapse
Affiliation(s)
- Omarine Nfor Nlinwe
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P.O. Box LG 581, Legon, Accra, Ghana.
| | - Kwadwo Asamoah Kusi
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P.O. Box LG 581, Legon, Accra, Ghana.
| | - Bright Adu
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P.O. Box LG 581, Legon, Accra, Ghana.
| | - Martha Sedegah
- Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 209 l0-7500, USA.
| |
Collapse
|
194
|
Zhou J, Kaiser A, Ng C, Karcher R, McConnell T, Paczkowski P, Fernandez C, Zhang M, Mackay S, Tsuji M. CD8+ T-cell mediated anti-malaria protection induced by malaria vaccines; assessment of hepatic CD8+ T cells by SCBC assay. Hum Vaccin Immunother 2018; 13:1625-1629. [PMID: 28362549 DOI: 10.1080/21645515.2017.1304333] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Malaria is a severe infectious disease with relatively high mortality, thus having been a scourge of humanity. There are a few candidate malaria vaccines that have shown a protective efficacy in humans against malaria. One of the candidate human malaria vaccines, which is based on human malaria sporozoites and called PfSPZ Vaccine, has been shown to protect a significant proportion of vaccine recipients from getting malaria. PfSPZ Vaccine elicits a potent response of hepatic CD8+ T cells that are specific for malaria antigens in non-human primates. To further characterize hepatic CD8+ T cells induced by the sporozoite-based malaria vaccine in a mouse model, we have used a cutting-edge Single-cell Barcode (SCBC) assay, a recently emerged approach/method for investigating the nature of T-cells responses during infection or cancer. Using the SCBC technology, we have identified a population of hepatic CD8+ T cells that are polyfunctional at a single cell level only in a group of vaccinated mice upon malaria challenge. The cytokines/chemokines secreted by these polyfunctional CD8+ T-cell subsets include MIP-1α, RANTES, IFN-γ, and/or IL-17A, which have shown to be associated with protective T-cell responses against certain pathogens. Therefore, a successful induction of such polyfunctional hepatic CD8+ T cells may be a key to the development of effective human malaria vaccine. In addition, the SCBC technology could provide a new level of diagnostic that will allow for a more accurate determination of vaccine efficacy.
Collapse
Affiliation(s)
- Jing Zhou
- a IsoPlexis , Branford , CT , USA.,b Aaron Diamond AIDS Research Center , Affiliate of the Rockefeller University , New York , NY , USA
| | - Alaina Kaiser
- a IsoPlexis , Branford , CT , USA.,b Aaron Diamond AIDS Research Center , Affiliate of the Rockefeller University , New York , NY , USA
| | - Colin Ng
- a IsoPlexis , Branford , CT , USA.,b Aaron Diamond AIDS Research Center , Affiliate of the Rockefeller University , New York , NY , USA
| | - Rachel Karcher
- a IsoPlexis , Branford , CT , USA.,b Aaron Diamond AIDS Research Center , Affiliate of the Rockefeller University , New York , NY , USA
| | - Tim McConnell
- a IsoPlexis , Branford , CT , USA.,b Aaron Diamond AIDS Research Center , Affiliate of the Rockefeller University , New York , NY , USA
| | - Patrick Paczkowski
- a IsoPlexis , Branford , CT , USA.,b Aaron Diamond AIDS Research Center , Affiliate of the Rockefeller University , New York , NY , USA
| | - Cristina Fernandez
- a IsoPlexis , Branford , CT , USA.,b Aaron Diamond AIDS Research Center , Affiliate of the Rockefeller University , New York , NY , USA
| | - Min Zhang
- a IsoPlexis , Branford , CT , USA.,b Aaron Diamond AIDS Research Center , Affiliate of the Rockefeller University , New York , NY , USA
| | - Sean Mackay
- a IsoPlexis , Branford , CT , USA.,b Aaron Diamond AIDS Research Center , Affiliate of the Rockefeller University , New York , NY , USA
| | - Moriya Tsuji
- a IsoPlexis , Branford , CT , USA.,b Aaron Diamond AIDS Research Center , Affiliate of the Rockefeller University , New York , NY , USA
| |
Collapse
|
195
|
Yoshida K, Iyori M, Blagborough AM, Salman AM, Dulal P, Sala KA, Yamamoto DS, Khan SM, Janse CJ, Biswas S, Yoshii T, Yusuf Y, Tokoro M, Hill AVS, Yoshida S. Adenovirus-prime and baculovirus-boost heterologous immunization achieves sterile protection against malaria sporozoite challenge in a murine model. Sci Rep 2018; 8:3896. [PMID: 29497047 PMCID: PMC5832798 DOI: 10.1038/s41598-018-21369-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/02/2018] [Indexed: 12/16/2022] Open
Abstract
With the increasing prevalence of artemisinin-resistant malaria parasites, a highly efficacious and durable vaccine for malaria is urgently required. We have developed an experimental virus-vectored vaccine platform based on an envelope-modified baculovirus dual-expression system (emBDES). Here, we show a conceptually new vaccine platform based on an adenovirus-prime/emBDES-boost heterologous immunization regimen expressing the Plasmodium falciparum circumsporozoite protein (PfCSP). A human adenovirus 5-prime/emBDES-boost heterologous immunization regimen consistently achieved higher sterile protection against transgenic P. berghei sporozoites expressing PfCSP after a mosquito-bite challenge than reverse-ordered or homologous immunization. This high protective efficacy was also achieved with a chimpanzee adenovirus 63-prime/emBDES-boost heterologous immunization regimen against an intravenous sporozoite challenge. Thus, we show that the adenovirus-prime/emBDES-boost heterologous immunization regimen confers sterile protection against sporozoite challenge by two individual routes, providing a promising new malaria vaccine platform for future clinical use.
Collapse
Affiliation(s)
- Kunitaka Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan.,Kanazawa University Graduate School of Medical Sciences, 13 Takara-machi, Kanazawa, 920-0934, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Andrew M Blagborough
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, Imperial College Road, South Kensington, London, SW7 2AZ, UK
| | - Ahmed M Salman
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK.,Leiden Malaria Research Group, Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, (LUMC, L4-Q), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Pawan Dulal
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Katarzyna A Sala
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, Imperial College Road, South Kensington, London, SW7 2AZ, UK
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, 329-0431, Tochigi, Japan
| | - Shahid M Khan
- Leiden Malaria Research Group, Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, (LUMC, L4-Q), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Chris J Janse
- Leiden Malaria Research Group, Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, (LUMC, L4-Q), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Sumi Biswas
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Tatsuya Yoshii
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Masaharu Tokoro
- Kanazawa University Graduate School of Medical Sciences, 13 Takara-machi, Kanazawa, 920-0934, Japan
| | - Adrian V S Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan.
| |
Collapse
|
196
|
Protection from experimental cerebral malaria with a single intravenous or subcutaneous whole-parasite immunization. Sci Rep 2018; 8:3085. [PMID: 29449638 PMCID: PMC5814423 DOI: 10.1038/s41598-018-21551-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 02/05/2018] [Indexed: 12/20/2022] Open
Abstract
Cerebral malaria is a life-threatening complication of Plasmodia infection and a major cause of child mortality in Sub-Saharan Africa. We report that protection from experimental cerebral malaria in the rodent model is obtained by a single intravenous or subcutaneous whole-parasite immunization. Whole-parasite immunization with radiation-attenuated sporozoites was equally protective as immunization with non-attenuated sporozoites under chemoprophylaxis. Both immunization regimens delayed the development of blood-stage parasites, but differences in cellular and humoral immune mechanisms were observed. Single-dose whole-parasite vaccination might serve as a relatively simple and feasible immunization approach to prevent life-threatening cerebral malaria.
Collapse
|
197
|
Abstract
PURPOSE OF REVIEW Great progress has been made in reducing malaria morbidity and mortality, yet the parasite continues to cause a startling 200 million infections and 500 000 deaths annually. Malaria vaccine development is pushing new boundaries by steady advancement toward a licensed product. RECENT FINDINGS Despite 50 years of research, the complexity of Plasmoidum falciparum confounds all attempts to eradicate the organism. This very complexity has pushed the boundaries of vaccine development to new heights, yet it remains to be seen if an affordable vaccine can provide durable and high-level protection. Novel vaccines such as RTS,S/AS01E are on the edge of licensure, but old techniques have resurged with the ability to deliver vialed, whole organism vaccines. Novel adjuvants, multistage/multiantigen approaches and transmission blocking vaccines all contribute to a multipronged battle plan to conquer malaria. SUMMARY Vaccines are the most cost-effective tools to control infectious diseases, yet the complexity of malaria has frustrated all attempts to develop an effective product. This review concentrates on recent advances in malaria vaccine development that lend hope that a vaccine can be produced and malaria eradicated.
Collapse
|
198
|
Olotu A, Urbano V, Hamad A, Eka M, Chemba M, Nyakarungu E, Raso J, Eburi E, Mandumbi DO, Hergott D, Maas CD, Ayekaba MO, Milang DN, Rivas MR, Schindler T, Embon OM, Ruben AJ, Saverino E, Abebe Y, Kc N, James ER, Murshedkar T, Manoj A, Chakravarty S, Li M, Adams M, Schwabe C, Segura JL, Daubenberger C, Tanner M, Richie TL, Billingsley PF, Lee Sim BK, Abdulla S, Hoffman SL. Advancing Global Health through Development and Clinical Trials Partnerships: A Randomized, Placebo-Controlled, Double-Blind Assessment of Safety, Tolerability, and Immunogenicity of PfSPZ Vaccine for Malaria in Healthy Equatoguinean Men. Am J Trop Med Hyg 2018; 98:308-318. [PMID: 29141739 PMCID: PMC5928718 DOI: 10.4269/ajtmh.17-0449] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Equatorial Guinea (EG) has implemented a successful malaria control program on Bioko Island. A highly effective vaccine would be an ideal complement to this effort and could lead to halting transmission and eliminating malaria. Sanaria® PfSPZ Vaccine (Plasmodium falciparum sporozoite Vaccine) is being developed for this purpose. To begin the process of establishing the efficacy of and implementing a PfSPZ Vaccine mass vaccination program in EG, we decided to conduct a series of clinical trials of PfSPZ Vaccine on Bioko Island. Because no clinical trial had ever been conducted in EG, we first successfully established the ethical, regulatory, quality, and clinical foundation for conducting trials. We now report the safety, tolerability, and immunogenicity results of the first clinical trial in the history of the country. Thirty adult males were randomized in the ratio 2:1 to receive three doses of 2.7 × 105 PfSPZ of PfSPZ Vaccine (N = 20) or normal saline placebo (N = 10) by direct venous inoculation at 8-week intervals. The vaccine was safe and well tolerated. Seventy percent, 65%, and 45% of vaccinees developed antibodies to Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP) by enzyme-linked immunosorbent assay, PfSPZ by automated immunofluorescence assay, and PfSPZ by inhibition of sporozoite invasion assay, respectively. Antibody responses were significantly lower than responses in U.S. adults who received the same dosage regimen, but not significantly different than responses in young adult Malians. Based on these results, a clinical trial enrolling 135 subjects aged 6 months to 65 years has been initiated in EG; it includes PfSPZ Vaccine and first assessment in Africa of PfSPZ-CVac. ClinicalTrials.gov identifier: NCT02418962.
Collapse
Affiliation(s)
- Ally Olotu
- Equatorial Guinea Malaria Vaccine Initiative, Malabo, Equatorial Guinea
- Ifakara Health Institute, Dar es Salaam, Tanzania
| | - Vicente Urbano
- Ministry of Health and Social Welfare, Malabo, Bioko Norte, Equatorial Guinea
- Equatorial Guinea Malaria Vaccine Initiative, Malabo, Equatorial Guinea
| | - Ali Hamad
- Equatorial Guinea Malaria Vaccine Initiative, Malabo, Equatorial Guinea
- Ifakara Health Institute, Dar es Salaam, Tanzania
| | - Martin Eka
- Ministry of Health and Social Welfare, Malabo, Bioko Norte, Equatorial Guinea
- Equatorial Guinea Malaria Vaccine Initiative, Malabo, Equatorial Guinea
| | - Mwajuma Chemba
- Equatorial Guinea Malaria Vaccine Initiative, Malabo, Equatorial Guinea
- Ifakara Health Institute, Dar es Salaam, Tanzania
| | - Elizabeth Nyakarungu
- Equatorial Guinea Malaria Vaccine Initiative, Malabo, Equatorial Guinea
- Ifakara Health Institute, Dar es Salaam, Tanzania
| | - Jose Raso
- Ministry of Health and Social Welfare, Malabo, Bioko Norte, Equatorial Guinea
- Equatorial Guinea Malaria Vaccine Initiative, Malabo, Equatorial Guinea
| | - Esther Eburi
- Medical Care Development International, Silver Spring, Maryland
- Equatorial Guinea Malaria Vaccine Initiative, Malabo, Equatorial Guinea
| | - Dolores O Mandumbi
- Ministry of Health and Social Welfare, Malabo, Bioko Norte, Equatorial Guinea
- Equatorial Guinea Malaria Vaccine Initiative, Malabo, Equatorial Guinea
| | - Dianna Hergott
- Medical Care Development International, Silver Spring, Maryland
| | - Carl D Maas
- Marathon EG Production Ltd, Punta Europa, Bioko Norte, Malabo, Equatorial Guinea
| | - Mitoha O Ayekaba
- Marathon EG Production Ltd, Punta Europa, Bioko Norte, Malabo, Equatorial Guinea
| | - Diosdado N Milang
- Ministry of Health and Social Welfare, Malabo, Bioko Norte, Equatorial Guinea
- Equatorial Guinea Malaria Vaccine Initiative, Malabo, Equatorial Guinea
| | - Matilde R Rivas
- Ministry of Health and Social Welfare, Malabo, Bioko Norte, Equatorial Guinea
- Equatorial Guinea Malaria Vaccine Initiative, Malabo, Equatorial Guinea
| | | | - Oscar M Embon
- La Paz Medical Center, Sipopo, Bioko Island, Equatorial Guinea
| | | | | | | | | | | | | | | | | | | | - Matthew Adams
- Division of Malaria Research, Institute for Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | | | - J Luis Segura
- Medical Care Development International, Silver Spring, Maryland
| | | | - Marcel Tanner
- University of Basel, Basel, Switzerland
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
199
|
|
200
|
Silvie O, Amino R, Hafalla JC. Tissue-specific cellular immune responses to malaria pre-erythrocytic stages. Curr Opin Microbiol 2017; 40:160-167. [PMID: 29217460 DOI: 10.1016/j.mib.2017.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 11/30/2022]
Abstract
Complete and long-lasting protective immunity against malaria can be achieved through vaccination with invasive live attenuated Plasmodium sporozoites, the motile stage inoculated in the host skin during a mosquito bite. Protective immunity relies primarily on effector CD8+ T cells targeting the parasite in the liver. Understanding the tissue-specific features of the immune response is emerging as a vital requirement for understanding protective immunity. The small parasite inoculum, the scarcity of infected cells and the tolerogenic properties of the liver represent hurdles for the establishment of protective immunity in endemic areas. In this review, we discuss recent advances on liver-specific features of immunity including innate recognition of malaria pre-erythrocytic stages, CD8+ T cell interactions with infected hepatocytes, antigen presentation for effective CD8+ T cell responses and generation of liver-resident memory CD8+ T cells. A better understanding of the factors involved in the induction and maintenance of effector CD8+ T cell immunity against malaria pre-erythrocytic stages is crucial for the development of an effective vaccine targeting the initial phase of malaria infection.
Collapse
Affiliation(s)
- Olivier Silvie
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, U1135, ERL8255, Paris, France.
| | - Rogerio Amino
- Unit of Malaria Infection and Immunity, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.
| | - Julius Clemence Hafalla
- Immunology and Infection Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom.
| |
Collapse
|