1
|
Schmid M, Beyeler R, Caldelari R, Rehmann R, Heussler V, Roques M. Generation of a genetically double-attenuated Plasmodium berghei parasite that fully arrests growth during late liver stage development. PLoS One 2024; 19:e0316164. [PMID: 39739824 DOI: 10.1371/journal.pone.0316164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/06/2024] [Indexed: 01/02/2025] Open
Abstract
Malaria caused by Plasmodium parasites remains a large health burden. One approach to combat this disease involves vaccinating individuals with whole sporozoites that have been genetically modified to arrest their development at a specific stage in the liver by targeted gene deletion, resulting in a genetically attenuated parasite (GAP). Through a comprehensive phenotyping screen, we identified the hscb gene, encoding a putative iron-sulfur protein assembly chaperone, as crucial for liver stage development, making it a suitable candidate gene for GAP generation. Parasites lacking Plasmodium berghei HscB (PbHscB) exhibited normal sporozoite production in mosquitoes, but their liver stage development was severely impaired, characterized by slow growth and delayed expression of merozoite surface protein 1 (MSP1). In vivo experiments demonstrated that PbHscB-deficient parasites exhibited a delay in prepatency of 2-4 days, emphasizing the significance of PbHscB for exo-erythrocytic development. Although knockout of PbHscB alone allowed breakthrough infections, it is a potent candidate for a dual gene deletion strategy. PlasMei2, an RNA-binding protein, was previously found to be crucial for the completion of liver stage development. We generated a PbHscB-PbMei2-double attenuated parasite line, serving as a late liver stage-arresting replication-competent (LARC) GAP, providing a solid block of liver-to-blood stage transition.
Collapse
Affiliation(s)
- Melanie Schmid
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Raphael Beyeler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Reto Caldelari
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Ruth Rehmann
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Magali Roques
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| |
Collapse
|
2
|
Singer M, Kanatani S, Castillo SG, Frischknecht F, Sinnis P. The Plasmodium circumsporozoite protein. Trends Parasitol 2024; 40:1124-1134. [PMID: 39572325 DOI: 10.1016/j.pt.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 12/07/2024]
Abstract
The circumsporozoite protein (CSP) is one of the most studied proteins of the malaria parasite. It is the target of the only licensed malaria vaccines and is essential for sporozoite formation and infectivity. Yet, the mechanisms by which CSP functions and its interactions with other proteins are only beginning to be understood. Here we review the current state of knowledge of CSP structure and function, as sporozoites develop in the mosquito and establish infection in the mammalian host, and outline outstanding questions that need to be addressed.
Collapse
Affiliation(s)
- Mirko Singer
- Parasitology, Center for Infectious Diseases, Heidelberg University Medical Faculty, Heidelberg, Germany
| | - Sachie Kanatani
- Johns Hopkins School of Public Health and Johns Hopkins Malaria Research Institute, 615 North Wolfe Street, Baltimore, MD, USA
| | - Stefano Garcia Castillo
- Johns Hopkins School of Public Health and Johns Hopkins Malaria Research Institute, 615 North Wolfe Street, Baltimore, MD, USA
| | - Friedrich Frischknecht
- Parasitology, Center for Infectious Diseases, Heidelberg University Medical Faculty, Heidelberg, Germany; German Center for Infection Research, partner site Heidelberg, Heidelberg, Germany
| | - Photini Sinnis
- Johns Hopkins School of Public Health and Johns Hopkins Malaria Research Institute, 615 North Wolfe Street, Baltimore, MD, USA.
| |
Collapse
|
3
|
Miura K, Flores-Garcia Y, Long CA, Zavala F. Vaccines and monoclonal antibodies: new tools for malaria control. Clin Microbiol Rev 2024; 37:e0007123. [PMID: 38656211 PMCID: PMC11237600 DOI: 10.1128/cmr.00071-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
SUMMARYMalaria remains one of the biggest health problems in the world. While significant reductions in malaria morbidity and mortality had been achieved from 2000 to 2015, the favorable trend has stalled, rather significant increases in malaria cases are seen in multiple areas. In 2022, there were 249 million estimated cases, and 608,000 malaria-related deaths, mostly in infants and children aged under 5 years, globally. Therefore, in addition to the expansion of existing anti-malarial control measures, it is critical to develop new tools, such as vaccines and monoclonal antibodies (mAbs), to fight malaria. In the last 2 years, the first and second malaria vaccines, both targeting Plasmodium falciparum circumsporozoite proteins (PfCSP), have been recommended by the World Health Organization to prevent P. falciparum malaria in children living in moderate to high transmission areas. While the approval of the two malaria vaccines is a considerable milestone in vaccine development, they have much room for improvement in efficacy and durability. In addition to the two approved vaccines, recent clinical trials with mAbs against PfCSP, blood-stage vaccines against P. falciparum or P. vivax, and transmission-blocking vaccine or mAb against P. falciparum have shown promising results. This review summarizes the development of the anti-PfCSP vaccines and mAbs, and recent topics in the blood- and transmission-blocking-stage vaccine candidates and mAbs. We further discuss issues of the current vaccines and the directions for the development of next-generation vaccines.
Collapse
Affiliation(s)
- Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Malaria Research Institute, Baltimore, Maryland, USA
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Malaria Research Institute, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Zhao HD, Qian HB, Wang ZK, Ren RK, Yu TB, Liu HL. Patient with suspected co-infection of hemorrhagic fever with renal syndrome and malaria: a case report. Front Med (Lausanne) 2024; 11:1341015. [PMID: 38751985 PMCID: PMC11094318 DOI: 10.3389/fmed.2024.1341015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/10/2024] [Indexed: 05/18/2024] Open
Abstract
Background Hemorrhagic fever with renal syndrome (HFRS) is a natural epidemic disease that can be caused by the Hantaan virus (HTNV). Malaria is caused by plasmodium and can be transmitted by a mosquito bite. The similar manifestations shared by these disorders pose a challenge for clinicians in differential diagnosis, in particular, coupled with a false-positive serological test. Case presentation A 46-year-old man was admitted for fever and chills for over 10 days and was suspected of being co-infected with HFRS and malaria due to a history of travel to malaria-endemic areas and a positive HTNV-immunoglobulin M (IgM) test. Although leukocytosis, thrombocytopenia, renal injury, lymphocytosis, overexpression of interleukin-6, and procalcitonin were observed during the hospitalization, the hypotensive, oliguria, and polyuria phases of the HFRS course were not observed. Instead, typical symptoms of malaria were found, including a progressive decrease in erythrocytes and hemoglobin levels with signs of anemia. Furthermore, because the patient had no history of exposure to HFRS endemic areas, exposure to an HTNV-infected rodent, or a positive HTNV-IgG test, and false serological tests of IgM can be caused by various factors, the HFRS coinfection with malaria was ruled out. Conclusion Misdiagnosis can be easily induced by a false serological test, in particular the IgM test which can be influenced by various factors. A combination of health history, epidemiology, physical examination, precise application of specific examinations involving tests of conventional laboratory parameters as well as well-accepted methods such as the immunochromatographic (ICG) test, real-time reverse transcription-polymerase chain reaction (PCR), and Western blot (WB), and acquaintance with disorders with similar manifestations will contribute to the precise diagnosis in clinical treatment.
Collapse
Affiliation(s)
- Han-Dong Zhao
- Central Laboratory of Virology, Shaanxi Provincial Hospital of Infectious Diseases, The Eighth Hospital Affiliated to Medical College of Xi’an Jiaotong University, Xi’an, China
- Clinical Laboratory Center, Shaanxi Provincial Hospital of Infectious Diseases, The Eighth Hospital Affiliated to Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Hong-Bo Qian
- Clinical Laboratory Center, Shaanxi Provincial Hospital of Infectious Diseases, The Eighth Hospital Affiliated to Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Ze-Kun Wang
- Department of Radiology, Shaanxi Provincial Hospital of Infectious Diseases, The Eighth Hospital Affiliated to Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Rui-Kang Ren
- Network and Information Center, Shaanxi Provincial Hospital of Infectious Diseases, The Eighth Hospital Affiliated to Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Tong-Bo Yu
- Clinical Laboratory Center, Shaanxi Provincial Hospital of Infectious Diseases, The Eighth Hospital Affiliated to Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Hong-Li Liu
- Clinical Laboratory Center, Xi’an People’s Hospital (Xi’an Fourth Hospital) Guang-Ren Hospital Affiliated to Xi’an Jiaotong University Health Science Center, Xi’an, China
| |
Collapse
|
5
|
Bera S, Amino R, Cockburn IA, Ganusov VV. Heterogeneity in killing efficacy of individual effector CD8 + T cells against Plasmodium liver stages. Proc Biol Sci 2023; 290:20232280. [PMID: 38018100 PMCID: PMC10685130 DOI: 10.1098/rspb.2023.2280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/01/2023] [Indexed: 11/30/2023] Open
Abstract
Vaccination strategies in mice inducing high numbers of memory CD8+ T cells specific to a single epitope are able to provide sterilizing protection against infection with Plasmodium sporozoites. We have recently found that Plasmodium-specific CD8+ T cells cluster around sporozoite-infected hepatocytes but whether such clusters are important in elimination of the parasite remains incompletely understood. Here, we used our previously generated data in which we employed intravital microscopy to longitudinally image 32 green fluorescent protein (GFP)-expressing Plasmodium yoelii parasites in livers of mice that had received activated Plasmodium-specific CD8+ T cells after sporozoite infection. We found significant heterogeneity in the dynamics of the normalized GFP signal from the parasites (termed 'vitality index' or VI) that was weakly correlated with the number of T cells near the parasite. We also found that a simple model assuming mass-action, additive killing by T cells well describes the VI dynamics for most parasites and predicts a highly variable killing efficacy by individual T cells. Given our estimated median per capita kill rate of k = 0.031/h we predict that a single T cell is typically incapable of killing a parasite within the 48 h lifespan of the liver stage in mice. Stochastic simulations of T cell clustering and killing of the liver stage also suggested that: (i) three or more T cells per infected hepatocyte are required to ensure sterilizing protection; (ii) both variability in killing efficacy of individual T cells and resistance to killing by individual parasites may contribute to the observed variability in VI decline, and (iii) the stable VI of some clustered parasites cannot be explained by measurement noise. Taken together, our analysis for the first time provides estimates of efficiency at which individual CD8+ T cells eliminate intracellular parasitic infection in vivo.
Collapse
Affiliation(s)
- Soumen Bera
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | - Rogerio Amino
- Unit of Malaria Infection and Immunity, Institut Pasteur, 75015 Paris, France
| | - Ian A. Cockburn
- Division of Immunology, Inflammation and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra 2600, Australia
| | - Vitaly V. Ganusov
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
- Department of Mathematics, University of Tennessee, Knoxville, TN 37996, USA
- Host-Pathogen Interactions program, Texas Biomedical Research Institute, San Antonio, TX 78258, USA
| |
Collapse
|
6
|
Nakamae S, Miyagawa S, Ogawa K, Kamiya M, Taniguchi M, Ono A, Kawaguchi M, Teklemichael AA, Jian JY, Araki T, Katagami Y, Mukai H, Annoura T, Yui K, Hirayama K, Kawakami S, Mizukami S. Induction of liver-resident memory T cells and protection at liver-stage malaria by mRNA-containing lipid nanoparticles. Front Immunol 2023; 14:1116299. [PMID: 37680630 PMCID: PMC10482405 DOI: 10.3389/fimmu.2023.1116299] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 07/25/2023] [Indexed: 09/09/2023] Open
Abstract
Recent studies have suggested that CD8+ liver-resident memory T (TRM) cells are crucial in the protection against liver-stage malaria. We used liver-directed mRNA-containing lipid nanoparticles (mRNA-LNPs) to induce liver TRM cells in a murine model. Single-dose intravenous injections of ovalbumin mRNA-LNPs effectively induced antigen-specific cytotoxic T lymphocytes in a dose-dependent manner in the liver on day 7. TRM cells (CD8+ CD44hi CD62Llo CD69+ KLRG1-) were induced 5 weeks after immunization. To examine the protective efficacy, mice were intramuscularly immunized with two doses of circumsporozoite protein mRNA-LNPs at 3-week intervals and challenged with sporozoites of Plasmodium berghei ANKA. Sterile immunity was observed in some of the mice, and the other mice showed a delay in blood-stage development when compared with the control mice. mRNA-LNPs therefore induce memory CD8+ T cells that can protect against sporozoites during liver-stage malaria and may provide a basis for vaccines against the disease.
Collapse
Affiliation(s)
- Sayuri Nakamae
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Satoshi Miyagawa
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
- Laboratory for Drug Discovery and Disease Research, SHIONOGI & CO., LTD., Osaka, Japan
| | - Koki Ogawa
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Mariko Kamiya
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Mayumi Taniguchi
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Akari Ono
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Maho Kawaguchi
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Awet Alem Teklemichael
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Jiun-Yu Jian
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Tamasa Araki
- Department of Parasitology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Yukimi Katagami
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Hidefumi Mukai
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Takeshi Annoura
- Department of Parasitology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Katsuyuki Yui
- Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Kenji Hirayama
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Shusaku Mizukami
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Nagasaki, Japan
| |
Collapse
|
7
|
Hassert M, Arumugam S, Harty JT. Memory CD8+ T cell-mediated protection against liver-stage malaria. Immunol Rev 2023; 316:84-103. [PMID: 37014087 PMCID: PMC10524177 DOI: 10.1111/imr.13202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/09/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023]
Abstract
Nearly half of the world's population is at risk of malaria, a disease caused by the protozoan parasite Plasmodium, which is estimated to cause more than 240,000,000 infections and kill more than 600,000 people annually. The emergence of Plasmodia resistant to chemoprophylactic treatment highlights the urgency to develop more effective vaccines. In this regard, whole sporozoite vaccination approaches in murine models and human challenge studies have provided substantial insight into the immune correlates of protection from malaria. From these studies, CD8+ T cells have come to the forefront, being identified as critical for vaccine-mediated liver-stage immunity that can prevent the establishment of the symptomatic blood stages and subsequent transmission of infection. However, the unique biological characteristics required for CD8+ T cell protection from liver-stage malaria dictate that more work must be done to design effective vaccines. In this review, we will highlight a subset of studies that reveal basic aspects of memory CD8+ T cell-mediated protection from liver-stage malaria infection.
Collapse
Affiliation(s)
- Mariah Hassert
- Department of Pathology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
| | - Sahaana Arumugam
- Department of Pathology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
- Medical Scientist Training Program, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
| | - John T. Harty
- Department of Pathology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
8
|
Xia M, Vago F, Han L, Huang P, Nguyen L, Boons GJ, Klassen JS, Jiang W, Tan M. The αTSR Domain of Plasmodium Circumsporozoite Protein Bound Heparan Sulfates and Elicited High Titers of Sporozoite Binding Antibody After Displayed by Nanoparticles. Int J Nanomedicine 2023; 18:3087-3107. [PMID: 37312932 PMCID: PMC10259582 DOI: 10.2147/ijn.s406314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
Introduction Malaria is a devastating infectious illness caused by protozoan Plasmodium parasites. The circumsporozoite protein (CSP) on Plasmodium sporozoites binds heparan sulfate proteoglycan (HSPG) receptors for liver invasion, a critical step for prophylactic and therapeutic interventions. Methods In this study, we characterized the αTSR domain that covers region III and the thrombospondin type-I repeat (TSR) of the CSP using various biochemical, glycobiological, bioengineering, and immunological approaches. Results We found for the first time that the αTSR bound heparan sulfate (HS) glycans through support by a fused protein, indicating that the αTSR is a key functional domain and thus a vaccine target. When the αTSR was fused to the S domain of norovirus VP1, the fusion protein self-assembled into uniform S60-αTSR nanoparticles. Three-dimensional structure reconstruction revealed that each nanoparticle consists of an S60 nanoparticle core and 60 surface displayed αTSR antigens. The nanoparticle displayed αTSRs retained the binding function to HS glycans, indicating that they maintained authentic conformations. Both tagged and tag-free S60-αTSR nanoparticles were produced via the Escherichia coli system at high yield by scalable approaches. They are highly immunogenic in mice, eliciting high titers of αTSR-specific antibody that bound specifically to the CSPs of Plasmodium falciparum sporozoites at high titer. Discussion and Conclusion Our data demonstrated that the αTSR is an important functional domain of the CSP. The S60-αTSR nanoparticle displaying multiple αTSR antigens is a promising vaccine candidate potentially against attachment and infection of Plasmodium parasites.
Collapse
Affiliation(s)
- Ming Xia
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Frank Vago
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Ling Han
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Pengwei Huang
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Linh Nguyen
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Geert-Jan Boons
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Department of Chemistry, University of Georgia, Athens, GA, USA
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - John S Klassen
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Wen Jiang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
9
|
Markwalter CF, Petersen JEV, Zeno EE, Sumner KM, Freedman E, Mangeni JN, Abel L, Obala AA, Prudhomme-O’Meara W, Taylor SM. Symptomatic malaria enhances protection from reinfection with homologous Plasmodium falciparum parasites. PLoS Pathog 2023; 19:e1011442. [PMID: 37307293 PMCID: PMC10289385 DOI: 10.1371/journal.ppat.1011442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/23/2023] [Accepted: 05/24/2023] [Indexed: 06/14/2023] Open
Abstract
A signature remains elusive of naturally-acquired immunity against Plasmodium falciparum. We identified P. falciparum in a 14-month cohort of 239 people in Kenya, genotyped at immunogenic parasite targets expressed in the pre-erythrocytic (circumsporozoite protein, CSP) and blood (apical membrane antigen 1, AMA-1) stages, and classified into epitope type based on variants in the DV10, Th2R, and Th3R epitopes in CSP and the c1L region of AMA-1. Compared to asymptomatic index infections, symptomatic malaria was associated with reduced reinfection by parasites bearing homologous CSP-Th2R (adjusted hazard ratio [aHR]:0.63; 95% CI:0.45-0.89; p = 0.008) CSP-Th3R (aHR:0.71; 95% CI:0.52-0.97; p = 0.033), and AMA-1 c1L (aHR:0.63; 95% CI:0.43-0.94; p = 0.022) epitope types. The association of symptomatic malaria with reduced hazard of homologous reinfection was strongest for rare epitope types. Symptomatic malaria provides more durable protection against reinfection with parasites bearing homologous epitope types. The phenotype represents a legible molecular epidemiologic signature of naturally-acquired immunity by which to identify new antigen targets.
Collapse
Affiliation(s)
- Christine F. Markwalter
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
| | - Jens E. V. Petersen
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Erica E. Zeno
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Kelsey M. Sumner
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Elizabeth Freedman
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Judith N. Mangeni
- School of Public Health, College of Health Sciences, Moi University, Eldoret, Kenya
| | - Lucy Abel
- Academic Model Providing Access to Healthcare, Moi Teaching and Referral Hospital, Eldoret, Kenya
| | - Andrew A. Obala
- School of Medicine, College of Health Sciences, Moi University, Eldoret, Kenya
| | - Wendy Prudhomme-O’Meara
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
- School of Public Health, College of Health Sciences, Moi University, Eldoret, Kenya
| | - Steve M. Taylor
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
10
|
Markwalter CF, Petersen JEV, Zeno EE, Sumner KM, Freedman E, Mangeni JN, Abel L, Obala AA, Prudhomme-O’Meara W, Taylor SM. Symptomatic malaria enhances protection from reinfection with homologous Plasmodium falciparum parasites. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.04.23284198. [PMID: 36711685 PMCID: PMC9882554 DOI: 10.1101/2023.01.04.23284198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A signature remains elusive of naturally-acquired immunity against Plasmodium falciparum . We identified P. falciparum in a 14-month cohort of 239 people in Kenya, genotyped at immunogenic parasite targets expressed in the pre-erythrocytic (circumsporozoite protein, CSP) and blood (apical membrane antigen 1, AMA-1) stages, and classified into epitope type based on variants in the DV10, Th2R, and Th3R epitopes in CSP and the c1L region of AMA-1. Compared to asymptomatic index infections, symptomatic malaria was associated with a reduced reinfection by parasites bearing homologous CSP-Th2R (adjusted hazard ratio [aHR]:0.63; 95% CI:0.45-0.89; p=0.008) CSP-Th3R (aHR:0.71; 95% CI:0.52-0.97; p=0.033), and AMA-1 c1L (aHR:0.63; 95% CI:0.43-0.94; p=0.022) epitope types. The association of symptomatic malaria with reduced risk of homologous reinfection was strongest for rare epitope types. Symptomatic malaria more effectively promotes functional immune responses. The phenotype represents a legible molecular epidemiologic signature of naturally-acquired immunity by which to identify new antigen targets.
Collapse
Affiliation(s)
| | - Jens E. V. Petersen
- Division of Infectious Diseases, School of Medicine, Duke University, Durham NC USA
| | - Erica E. Zeno
- Division of Infectious Diseases, School of Medicine, Duke University, Durham NC USA,Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill NC USA
| | - Kelsey M. Sumner
- Division of Infectious Diseases, School of Medicine, Duke University, Durham NC USA,Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill NC USA
| | - Elizabeth Freedman
- Division of Infectious Diseases, School of Medicine, Duke University, Durham NC USA
| | - Judith N. Mangeni
- School of Public Health, College of Health Sciences, Moi University, Eldoret, Kenya
| | - Lucy Abel
- Academic Model Providing Access to Healthcare, Moi Teaching and Referral Hospital, Eldoret Kenya
| | - Andrew A. Obala
- School of Medicine, College of Health Sciences, Moi University, Eldoret Kenya
| | - Wendy Prudhomme-O’Meara
- Duke Global Health Institute, Duke University, Durham NC USA,Division of Infectious Diseases, School of Medicine, Duke University, Durham NC USA,School of Public Health, College of Health Sciences, Moi University, Eldoret, Kenya
| | - Steve M. Taylor
- Duke Global Health Institute, Duke University, Durham NC USA,Division of Infectious Diseases, School of Medicine, Duke University, Durham NC USA,Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill NC USA,Corresponding author: Steve M Taylor ,
| |
Collapse
|
11
|
Infection and Immunity. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
12
|
Richie TL, Church LWP, Murshedkar T, Billingsley PF, James ER, Chen MC, Abebe Y, KC N, Chakravarty S, Dolberg D, Healy SA, Diawara H, Sissoko MS, Sagara I, Cook DM, Epstein JE, Mordmüller B, Kapulu M, Kreidenweiss A, Franke-Fayard B, Agnandji ST, López Mikue MSA, McCall MBB, Steinhardt L, Oneko M, Olotu A, Vaughan AM, Kublin JG, Murphy SC, Jongo S, Tanner M, Sirima SB, Laurens MB, Daubenberger C, Silva JC, Lyke KE, Janse CJ, Roestenberg M, Sauerwein RW, Abdulla S, Dicko A, Kappe SHI, Lee Sim BK, Duffy PE, Kremsner PG, Hoffman SL. Sporozoite immunization: innovative translational science to support the fight against malaria. Expert Rev Vaccines 2023; 22:964-1007. [PMID: 37571809 PMCID: PMC10949369 DOI: 10.1080/14760584.2023.2245890] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
INTRODUCTION Malaria, a devastating febrile illness caused by protozoan parasites, sickened 247,000,000 people in 2021 and killed 619,000, mostly children and pregnant women in sub-Saharan Africa. A highly effective vaccine is urgently needed, especially for Plasmodium falciparum (Pf), the deadliest human malaria parasite. AREAS COVERED Sporozoites (SPZ), the parasite stage transmitted by Anopheles mosquitoes to humans, are the only vaccine immunogen achieving >90% efficacy against Pf infection. This review describes >30 clinical trials of PfSPZ vaccines in the U.S.A., Europe, Africa, and Asia, based on first-hand knowledge of the trials and PubMed searches of 'sporozoites,' 'malaria,' and 'vaccines.' EXPERT OPINION First generation (radiation-attenuated) PfSPZ vaccines are safe, well tolerated, 80-100% efficacious against homologous controlled human malaria infection (CHMI) and provide 18-19 months protection without boosting in Africa. Second generation chemo-attenuated PfSPZ are more potent, 100% efficacious against stringent heterologous (variant strain) CHMI, but require a co-administered drug, raising safety concerns. Third generation, late liver stage-arresting, replication competent (LARC), genetically-attenuated PfSPZ are expected to be both safe and highly efficacious. Overall, PfSPZ vaccines meet safety, tolerability, and efficacy requirements for protecting pregnant women and travelers exposed to Pf in Africa, with licensure for these populations possible within 5 years. Protecting children and mass vaccination programs to block transmission and eliminate malaria are long-term objectives.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sara A. Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Halimatou Diawara
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Mahamadou S. Sissoko
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Issaka Sagara
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - David M. Cook
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Judith E. Epstein
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Mordmüller
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Melissa Kapulu
- Biosciences Department, Kenya Medical Research Institute KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrea Kreidenweiss
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | | | - Selidji T. Agnandji
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | | - Matthew B. B. McCall
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Laura Steinhardt
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Martina Oneko
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Ally Olotu
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Ashley M. Vaughan
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - James G. Kublin
- Department of Global Health, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases and Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Said Jongo
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Marcel Tanner
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Matthew B. Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Claudia Daubenberger
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Joana C. Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kirsten E. Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chris J. Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert W. Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Salim Abdulla
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Alassane Dicko
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Stefan H. I. Kappe
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter G. Kremsner
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | |
Collapse
|
13
|
Padron-Regalado E, Ulaszewska M, Douglas AD, Hill AVS, Spencer AJ. STING-pathway modulation to enhance the immunogenicity of adenoviral-vectored vaccines. Sci Rep 2022; 12:14464. [PMID: 36002507 PMCID: PMC9401198 DOI: 10.1038/s41598-022-18750-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/18/2022] [Indexed: 11/09/2022] Open
Abstract
Traditional chemical adjuvants remain a practical means of enhancing the immunogenicity of vaccines. Nevertheless, it is recognized that increasing the immunogenicity of viral vectors is challenging. Recently, STING ligands have been shown to enhance the efficacy of different vaccine platforms, but their affectivity on viral-vectored vaccination has not been fully assessed. In this study we used a multi-pronged approach to shed light on the immunological properties and potential mechanisms of action of this type of adjuvant and focused our study on replication-deficient human adenovirus serotype 5 (AdHu5). When the STING ligand 2'3'-cGAMP was mixed with AdHu5, the adjuvant enhanced anti-vector immune responses while decreasing the transgene-specific CD8+ T cell response. Studies employing STING-knockout mice and a 2'3'-cGAMP inactive analogue confirmed the aforementioned effects were STING dependent. In vitro assays demonstrated 2'3'-cGAMP induced the production of IFN-β which in turn negatively affected AdHu5 transgene expression and CD8+ T cell immunogenicity. In an effort to overcome the negative impact of early 2'3'-cGAMP signaling on AdHu5 transgene immunogenicity, we generated a bicistronic vector encoding the 2'3'-cGAMP together with a model antigen. Intracellular production of 2'3'-cGAMP after AdHu5 infection was able to enhance transgene-specific CD8+ T cell immunogenicity, although not to a level that would warrant progression of this adjuvant to clinical assessment. This work highlights the importance of timing of 2'3'-cGAMP administration when assessing its adjuvant capacity with different vaccine modalities.
Collapse
Affiliation(s)
- Eriko Padron-Regalado
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Marta Ulaszewska
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexander D Douglas
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Adrian V S Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexandra J Spencer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
14
|
Schroeder EA, Chirgwin ME, Derbyshire ER. Plasmodium's fight for survival: escaping elimination while acquiring nutrients. Trends Parasitol 2022; 38:544-557. [PMID: 35534377 PMCID: PMC9187605 DOI: 10.1016/j.pt.2022.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/10/2022] [Accepted: 04/10/2022] [Indexed: 01/08/2023]
Abstract
Plasmodium parasites extensively alter their host hepatocyte to evade host detection and support an unprecedented replication rate. Host cell manipulation includes association with the host early and late endomembrane systems, where Plasmodium accesses nutrients while suppressing cellular immune processes. Early endomembrane organelles provide an opportunity to sequester an abundance of lipids and proteins, but the association with late endomembrane organelles also risks autophagy-mediated elimination. While not all parasites survive, those that do benefit from a plethora of nutrients provided through this pathway. In this review, we discuss recent advances in our understanding of how Plasmodium parasites balance the need for host nutrients while avoiding elimination during the liver stage.
Collapse
Affiliation(s)
- Erin A Schroeder
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | | | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA; Department of Chemistry, Duke University, Durham, NC, USA.
| |
Collapse
|
15
|
Noé A, Datoo MS, Flaxman A, Husainy MA, Jenkin D, Bellamy D, Makinson RA, Morter R, Ramos Lopez F, Sheridan J, Voukantsis D, Prasad N, Hill AVS, Ewer KJ, Spencer AJ. Deep Immune Phenotyping and Single-Cell Transcriptomics Allow Identification of Circulating TRM-Like Cells Which Correlate With Liver-Stage Immunity and Vaccine-Induced Protection From Malaria. Front Immunol 2022; 13:795463. [PMID: 35197971 PMCID: PMC8859435 DOI: 10.3389/fimmu.2022.795463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
Protection from liver-stage malaria requires high numbers of CD8+ T cells to find and kill Plasmodium-infected cells. A new malaria vaccine strategy, prime-target vaccination, involves sequential viral-vectored vaccination by intramuscular and intravenous routes to target cellular immunity to the liver. Liver tissue-resident memory (TRM) CD8+ T cells have been shown to be necessary and sufficient for protection against rodent malaria by this vaccine regimen. Ultimately, to most faithfully assess immunotherapeutic responses by these local, specialised, hepatic T cells, periodic liver sampling is necessary, however this is not feasible at large scales in human trials. Here, as part of a phase I/II P. falciparum challenge study of prime-target vaccination, we performed deep immune phenotyping, single-cell RNA-sequencing and kinetics of hepatic fine needle aspirates and peripheral blood samples to study liver CD8+ TRM cells and circulating counterparts. We found that while these peripheral ‘TRM-like’ cells differed to TRM cells in terms of previously described characteristics, they are similar phenotypically and indistinguishable in terms of key T cell residency transcriptional signatures. By exploring the heterogeneity among liver CD8+ TRM cells at single cell resolution we found two main subpopulations that each share expression profiles with blood T cells. Lastly, our work points towards the potential for using TRM−like cells as a correlate of protection by liver-stage malaria vaccines and, in particular, those adopting a prime-target approach. A simple and reproducible correlate of protection would be particularly valuable in trials of liver-stage malaria vaccines as they progress to phase III, large-scale testing in African infants. We provide a blueprint for understanding and monitoring liver TRM cells induced by a prime-target malaria vaccine approach.
Collapse
Affiliation(s)
- Andrés Noé
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- *Correspondence: Andrés Noé, ; ; Alexandra J. Spencer,
| | - Mehreen S. Datoo
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Amy Flaxman
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Daniel Jenkin
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Duncan Bellamy
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Richard Morter
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Dimitrios Voukantsis
- Bioinformatics Hub, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Naveen Prasad
- Bioinformatics Hub, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Katie J. Ewer
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Alexandra J. Spencer
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- *Correspondence: Andrés Noé, ; ; Alexandra J. Spencer,
| |
Collapse
|
16
|
Immunoprofiling Identifies Functional B and T Cell Subsets Induced by an Attenuated Whole Parasite Malaria Vaccine as Correlates of Sterile Immunity. Vaccines (Basel) 2022; 10:vaccines10010124. [PMID: 35062785 PMCID: PMC8780163 DOI: 10.3390/vaccines10010124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 02/05/2023] Open
Abstract
Immune correlates of protection remain elusive for most vaccines. An identified immune correlate would accelerate the down-selection of vaccine formulations by reducing the need for human pathogen challenge studies that are currently required to determine vaccine efficacy. Immunization via mosquito-delivered, radiation-attenuated P. falciparum sporozoites (IMRAS) is a well-established model for efficacious malaria vaccines, inducing greater than 90% sterile immunity. The current immunoprofiling study utilized samples from a clinical trial in which vaccine dosing was adjusted to achieve only 50% protection, thus enabling a comparison between protective and non-protective immune signatures. In-depth immunoprofiling was conducted by assessing a wide range of antigen-specific serological and cellular parameters and applying our newly developed computational tools, including machine learning. The computational component of the study pinpointed previously un-identified cellular T cell subsets (namely, TNFα-secreting CD8+CXCR3−CCR6− T cells, IFNγ-secreting CD8+CCR6+ T cells and TNFα/FNγ-secreting CD4+CXCR3−CCR6− T cells) and B cell subsets (i.e., CD19+CD24hiCD38hiCD69+ transitional B cells) as important factors predictive of protection (92% accuracy). Our study emphasizes the need for in-depth immunoprofiling and subsequent data integration with computational tools to identify immune correlates of protection. The described process of computational data analysis is applicable to other disease and vaccine models.
Collapse
|
17
|
Lefebvre MN, Surette FA, Anthony SM, Vijay R, Jensen IJ, Pewe LL, Hancox LS, Van Braeckel-Budimir N, van de Wall S, Urban SL, Mix MR, Kurup SP, Badovinac VP, Butler NS, Harty JT. Expeditious recruitment of circulating memory CD8 T cells to the liver facilitates control of malaria. Cell Rep 2021; 37:109956. [PMID: 34731605 PMCID: PMC8628427 DOI: 10.1016/j.celrep.2021.109956] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/08/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Circulating memory CD8 T cell trafficking and protective capacity during liver-stage malaria infection remains undefined. We find that effector memory CD8 T cells (Tem) infiltrate the liver within 6 hours after malarial or bacterial infections and mediate pathogen clearance. Tem recruitment coincides with rapid transcriptional upregulation of inflammatory genes in Plasmodium-infected livers. Recruitment requires CD8 T cell-intrinsic LFA-1 expression and the presence of liver phagocytes. Rapid Tem liver infiltration is distinct from recruitment to other non-lymphoid tissues in that it occurs both in the absence of liver tissue resident memory "sensing-and-alarm" function and ∼42 hours earlier than in lung infection by influenza virus. These data demonstrate relevance for Tem in protection against malaria and provide generalizable mechanistic insights germane to control of liver infections.
Collapse
Affiliation(s)
- Mitchell N Lefebvre
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Medical Scientist Training Program, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA
| | - Fionna A Surette
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA; Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Scott M Anthony
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Rahul Vijay
- Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Isaac J Jensen
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA
| | - Lecia L Pewe
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Lisa S Hancox
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | | | - Stephanie van de Wall
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Stina L Urban
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Madison R Mix
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Medical Scientist Training Program, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA
| | - Samarchith P Kurup
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA; Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Noah S Butler
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA; Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - John T Harty
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA.
| |
Collapse
|
18
|
Gibbins MP, Müller K, Matuschewski K, Silvie O, Hafalla JCR. Inferior T cell immunogenicity of a Plasmodium berghei model liver stage antigen expressed throughout pre-erythrocytic maturation. Parasite Immunol 2021; 43:e12877. [PMID: 34515999 DOI: 10.1111/pim.12877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/01/2022]
Abstract
Sporozoite antigens are the basis of a number of malaria vaccines being tested, but the contribution of antigens expressed during subsequent liver stage development to pre-erythrocytic stage immunity is poorly understood. We previously showed that, following immunisation with radiation attenuated sporozoites (RAS), a model epitope embedded in a sporozoite surface protein elicited robust CD8+ T cell responses, whilst the same epitope in a liver stage antigen induced inferior responses. Since RAS arrest early in their development in host hepatocytes, we hypothesised that extending parasite maturation in the liver could considerably improve the epitope-specific CD8+ T cell response. Here, we employed a late liver stage arrested parasite model, azithromycin prophylaxis alongside live sporozoites, to increase expression of the model epitope until full liver stage maturation. Strikingly, this alternative immunisation strategy, which has been shown to elicit superior protection, failed to improve the resulting epitope-specific CD8+ T cell responses. Our findings support the notion that liver stage antigens are poorly immunogenic and provide additional caution about prioritising antigens for vaccine development based solely on immunogenicity.
Collapse
Affiliation(s)
- Matthew P Gibbins
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Katja Müller
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Institute of Biology, Humboldt University, Berlin, Germany
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Institute of Biology, Humboldt University, Berlin, Germany
| | - Olivier Silvie
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Julius Clemence R Hafalla
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
19
|
Hon C, Friesen J, Ingmundson A, Scheppan D, Hafalla JCR, Müller K, Matuschewski K. Conservation of S20 as an Ineffective and Disposable IFNγ-Inducing Determinant of Plasmodium Sporozoites Indicates Diversion of Cellular Immunity. Front Microbiol 2021; 12:703804. [PMID: 34421862 PMCID: PMC8377727 DOI: 10.3389/fmicb.2021.703804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/07/2021] [Indexed: 11/19/2022] Open
Abstract
Despite many decades of research to develop a malaria vaccine, only one vaccine candidate has been explored in pivotal phase III clinical trials. This candidate subunit vaccine consists of a portion of a single Plasmodium antigen, circumsporozoite protein (CSP). This antigen was initially identified in the murine malaria model and shown to contain an immunodominant and protective CD8+ T cell epitope specific to the H-2Kd (BALB/c)-restricted genetic background. A high-content screen for CD8+ epitopes in the H2Kb/Db (C57BL/6)-restricted genetic background, identified two distinct dominant epitopes. In this study, we present a characterization of one corresponding antigen, the Plasmodium sporozoite-specific protein S20. Plasmodium berghei S20 knockout sporozoites and liver stages developed normally in vitro and in vivo. This potent infectivity of s20(-) sporozoites permitted comparative analysis of knockout and wild-type parasites in cell-based vaccination. Protective immunity of irradiation-arrested s20(-) sporozoites in single, double and triple immunizations was similar to irradiated unaltered sporozoites in homologous challenge experiments. These findings demonstrate the presence of an immunogenic Plasmodium pre-erythrocytic determinant, which is not essential for eliciting protection. Although S20 is not needed for colonization of the mammalian host and for initiation of a blood infection, it is conserved amongst Plasmodium species. Malarial parasites express conserved, immunogenic proteins that are not required to establish infection but might play potential roles in diverting cellular immune responses.
Collapse
Affiliation(s)
- Calvin Hon
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - Johannes Friesen
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Medical Care Unit Labor 28 GmbH, Berlin, Germany
| | - Alyssa Ingmundson
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Diana Scheppan
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Julius C R Hafalla
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Katja Müller
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
20
|
Collins KA, Brod F, Snaith R, Ulaszewska M, Longley RJ, Salman AM, Gilbert SC, Spencer AJ, Franco D, Ballou WR, Hill AVS. Ultra-low dose immunization and multi-component vaccination strategies enhance protection against malaria in mice. Sci Rep 2021; 11:10792. [PMID: 34031479 PMCID: PMC8144388 DOI: 10.1038/s41598-021-90290-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/10/2021] [Indexed: 11/10/2022] Open
Abstract
An effective vaccine would be a valuable tool for malaria control and elimination; however, the leading malaria vaccine in development, RTS,S/AS01, provided only partial protection in a Phase 3 trial. R21 is a next-generation RTS,S-like vaccine. We have previously shown in mice that R21 administered in Matrix-M is highly immunogenic, able to elicit complete protection against sporozoite challenge, and can be successfully administered with TRAP based viral-vectors resulting in enhanced protection. In this study, we developed a novel, GMP-compatible purification process for R21, and evaluated the immunogenicity and protective efficacy of ultra-low doses of both R21 and RTS,S when formulated in AS01. We demonstrated that both vaccines are highly immunogenic and also elicit comparable high levels of protection against transgenic parasites in BALB/c mice. By lowering the vaccine dose there was a trend for increased immunogenicity and sterile protection, with the highest dose vaccine groups achieving the lowest efficacy (50% sterile protection). We also evaluated the ability to combine RTS,S/AS01 with TRAP based viral-vectors and observed concurrent induction of immune responses to both antigens with minimal interference when mixing the vaccines prior to administration. These studies suggest that R21 or RTS,S could be combined with viral-vectors for a multi-component vaccination approach and indicate that low dose vaccination should be fully explored in humans to maximize potential efficacy.
Collapse
Affiliation(s)
- Katharine A Collins
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK. .,Radboud Institute for Health Science, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Florian Brod
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rebecca Snaith
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Marta Ulaszewska
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rhea J Longley
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ahmed M Salman
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah C Gilbert
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexandra J Spencer
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - Adrian V S Hill
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
21
|
Müller K, Gibbins MP, Roberts M, Reyes‐Sandoval A, Hill AVS, Draper SJ, Matuschewski K, Silvie O, Hafalla JCR. Low immunogenicity of malaria pre-erythrocytic stages can be overcome by vaccination. EMBO Mol Med 2021; 13:e13390. [PMID: 33709544 PMCID: PMC8033512 DOI: 10.15252/emmm.202013390] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 11/09/2022] Open
Abstract
Immunogenicity is considered one important criterion for progression of candidate vaccines to further clinical evaluation. We tested this assumption in an infection and vaccination model for malaria pre-erythrocytic stages. We engineered Plasmodium berghei parasites that harbour a well-characterised epitope for stimulation of CD8+ T cells, either as an antigen in the sporozoite surface-expressed circumsporozoite protein or the parasitophorous vacuole membrane associated protein upregulated in sporozoites 4 (UIS4) expressed in exo-erythrocytic forms (EEFs). We show that the antigen origin results in profound differences in immunogenicity with a sporozoite antigen eliciting robust, superior antigen-specific CD8+ T-cell responses, whilst an EEF antigen evokes poor responses. Despite their contrasting immunogenic properties, both sporozoite and EEF antigens gain access to antigen presentation pathways in hepatocytes, as recognition and targeting by vaccine-induced effector CD8+ T cells results in high levels of protection when targeting either antigen. Our study is the first demonstration that poorly immunogenic EEF antigens do not preclude their susceptibility to antigen-specific CD8+ T-cell killing, which has wide-ranging implications on antigen prioritisation for next-generation pre-erythrocytic malaria vaccines.
Collapse
Affiliation(s)
- Katja Müller
- Parasitology UnitMax Planck Institute for Infection BiologyBerlinGermany
- Department of Molecular ParasitologyInstitute of BiologyHumboldt UniversityBerlinGermany
| | - Matthew P Gibbins
- Department of Infection BiologyFaculty of Infectious and Tropical DiseasesLondon School of Hygiene and Tropical MedicineLondonUK
- Present address:
Wellcome Centre for Integrative ParasitologyInstitute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
| | - Mark Roberts
- Department of Infection BiologyFaculty of Infectious and Tropical DiseasesLondon School of Hygiene and Tropical MedicineLondonUK
| | - Arturo Reyes‐Sandoval
- Jenner InstituteUniversity of OxfordOxfordUK
- Present address:
Instituto Politécnico NacionalIPN. Av. Luis Enrique Erro s/n, Unidad Adolfo López MateosMexico CityMexico
| | | | | | - Kai Matuschewski
- Parasitology UnitMax Planck Institute for Infection BiologyBerlinGermany
- Department of Molecular ParasitologyInstitute of BiologyHumboldt UniversityBerlinGermany
| | - Olivier Silvie
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies InfectieusesCIMI‐ParisParisFrance
| | - Julius Clemence R Hafalla
- Department of Infection BiologyFaculty of Infectious and Tropical DiseasesLondon School of Hygiene and Tropical MedicineLondonUK
| |
Collapse
|
22
|
Chatterjee D, Cockburn IA. The challenges of a circumsporozoite protein-based malaria vaccine. Expert Rev Vaccines 2021; 20:113-125. [PMID: 33554669 DOI: 10.1080/14760584.2021.1874924] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION A safe and effective vaccine will likely be necessary for the control or eradication of malaria which kills 400,000 annually. Our most advanced vaccine candidate to date is RTS,S which is based on the Plasmodium falciparum circumsporozoite protein (PfCSP) of the malaria parasite. However, protection by RTS,S is incomplete and short-lived. AREAS COVERED Here we summarize results from recent clinical trials of RTS,S and critically evaluate recent studies that aim to understand the correlates of protective immunity and why vaccine-induced protection is short-lived. In particular, recent systems serology studies have highlighted a key role for the necessity of inducing functional antibodies. In-depth analyses of immune responses to CSP in both mouse models and vaccinated humans have also highlighted difficulties in generating the maintaining high-quality antibody responses. Finally, in recent years biophysical and structural studies of antibody binding to PfCSP have led to a better understanding of how highly potent antibodies can block infection, which can inform vaccine design. EXPERT OPINION We highlight how both structure-guided vaccine design and a better understanding of the immune response to PfCSP can inform a second generation of PfCSP-based vaccines stimulating a broader range of protective targets within PfCSP.
Collapse
Affiliation(s)
- Deepyan Chatterjee
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, the Australian National University, Canberra, Australia
| | - Ian Andrew Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, the Australian National University, Canberra, Australia
| |
Collapse
|
23
|
Umthong S, Dunn JR, Cheng HH. Depletion of CD8αβ + T Cells in Chickens Demonstrates Their Involvement in Protective Immunity towards Marek's Disease with Respect to Tumor Incidence and Vaccinal Protection. Vaccines (Basel) 2020; 8:E557. [PMID: 32987648 PMCID: PMC7712963 DOI: 10.3390/vaccines8040557] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/17/2020] [Accepted: 09/20/2020] [Indexed: 01/12/2023] Open
Abstract
Marek's disease (MD) is a lymphoproliferative disease in chickens caused by Marek's disease virus (MDV), a highly oncogenic alphaherpesvirus. Since 1970, MD has been controlled through widespread vaccination of commercial flocks. However, repeated and unpredictable MD outbreaks continue to occur in vaccinated flocks, indicating the need for a better understanding of MDV pathogenesis to guide improved or alternative control measures. As MDV is an intracellular pathogen that infects and transforms CD4+ T cells, the host cell-mediated immune response is considered to be vital for controlling MDV replication and tumor formation. In this study, we addressed the role of CD8+ T cells in vaccinal protection by widely-used monovalent (SB-1 and HVT) and bivalent (SB-1+HVT) MD vaccines. We established a method to deplete CD8+ T cells in chickens and found that their depletion through injection of anti-CD8 monoclonal antibodies (mAb) increased tumor induction and MD pathology, and reduced vaccinal protection to MD, which supports the important role of CD8+ T cells for both MD and vaccinal protection.
Collapse
Affiliation(s)
- Supawadee Umthong
- Microbiology and Molecular Genetics Program, Michigan State University, East Lansing, MI 48823, USA;
- USDA, ARS, US National Poultry Research Center, Avian Disease and Oncology Laboratory, East Lansing, MI 48823, USA;
| | - John R. Dunn
- USDA, ARS, US National Poultry Research Center, Avian Disease and Oncology Laboratory, East Lansing, MI 48823, USA;
| | - Hans H. Cheng
- USDA, ARS, US National Poultry Research Center, Avian Disease and Oncology Laboratory, East Lansing, MI 48823, USA;
| |
Collapse
|
24
|
Importance of the Immunodominant CD8 + T Cell Epitope of Plasmodium berghei Circumsporozoite Protein in Parasite- and Vaccine-Induced Protection. Infect Immun 2020; 88:IAI.00383-20. [PMID: 32719159 DOI: 10.1128/iai.00383-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022] Open
Abstract
The circumsporozoite protein (CSP) builds up the surface coat of sporozoites and is the leading malaria pre-erythrocytic-stage vaccine candidate. CSP has been shown to induce robust CD8+ T cell responses that are capable of eliminating developing parasites in hepatocytes, resulting in protective immunity. In this study, we characterized the importance of the immunodominant CSP-derived epitope SYIPSAEKI of Plasmodium berghei in both sporozoite- and vaccine-induced protection in murine infection models. In BALB/c mice, where SYIPSAEKI is efficiently presented in the context of the major histocompatibility complex class I (MHC-I) molecule H-2-Kd, we established that epitope-specific CD8+ T cell responses contribute to parasite killing following sporozoite immunization. Yet, sterile protection was achieved in the absence of this epitope, substantiating the concept that other antigens can be sufficient for parasite-induced protective immunity. Furthermore, we demonstrated that SYIPSAEKI-specific CD8+ T cell responses elicited by viral-vectored CSP-expressing vaccines effectively targeted parasites in hepatocytes. The resulting sterile protection strictly relied on the expression of SYIPSAEKI. In C57BL/6 mice, which are unable to present the immunodominant epitope, CSP-based vaccines did not confer complete protection, despite the induction of high levels of CSP-specific antibodies. These findings underscore the significance of CSP in protection against malaria pre-erythrocytic stages and demonstrate that a significant proportion of the protection against the parasite is mediated by CD8+ T cells specific for the immunodominant CSP-derived epitope.
Collapse
|
25
|
Phares TW, Kotraiah V, Karunarathne DS, Huang J, Browne CD, Buontempo P, Mansour M, Noe AR, Wykes MN, Pannucci J, Tsuji M, Gutierrez GM. A Peptide-Based PD1 Antagonist Enhances T-Cell Priming and Efficacy of a Prophylactic Malaria Vaccine and Promotes Survival in a Lethal Malaria Model. Front Immunol 2020; 11:1377. [PMID: 32733457 PMCID: PMC7363839 DOI: 10.3389/fimmu.2020.01377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/29/2020] [Indexed: 12/14/2022] Open
Abstract
The blockade of programmed cell death-1 (PD1) and its ligand PDL1 has been proven to be a successful immunotherapy against several cancers. Similar to cancer, PD1 contributes to the establishment of several chronic infectious diseases, including malaria. While monoclonal antibodies (mAbs) targeting checkpoint receptors are revolutionary in cancer treatment, the immune-related adverse events (irAEs) may prevent their utilization in prophylactic and therapeutic treatments of infectious diseases. The irAEs are, in part, due to the prolonged half-life of mAbs resulting in prolonged activation of the immune system. As an alternative modality to mAbs, peptides represent a viable option because they possess a shorter pharmacokinetic half-life and offer more formulation and delivery options. Here, we report on a 22-amino acid immunomodulatory peptide, LD01, derived from a Bacillus bacteria. When combined prophylactically with an adenovirus-based or irradiated sporozoite-based malaria vaccine, LD01 significantly enhanced antigen-specific CD8+ T cell expansion. Therapeutically, LD01 treatment of mice infected with a lethal malaria strain resulted in survival that was associated with lower numbers of FOXP3+Tbet+CD4+ regulatory T cells. Taken together, our results demonstrate that LD01 is a potent immunomodulator that acts upon the adaptive immune system to stimulate T cell responses both prophylactically and therapeutically.
Collapse
Affiliation(s)
- Timothy W Phares
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | - Vinayaka Kotraiah
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | | | - Jing Huang
- The Aaron Diamond AIDS Research Center, New York, NY, United States
| | | | - Peter Buontempo
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | - Marc Mansour
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | - Amy R Noe
- Leidos Life Sciences, Leidos Inc., Frederick, MD, United States
| | | | - James Pannucci
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | - Moriya Tsuji
- The Aaron Diamond AIDS Research Center, New York, NY, United States
| | - Gabriel M Gutierrez
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| |
Collapse
|
26
|
Abstract
Immunity to malaria has been linked to the availability and function of helper CD4+ T cells, cytotoxic CD8+ T cells and γδ T cells that can respond to both the asymptomatic liver stage and the symptomatic blood stage of Plasmodium sp. infection. These T cell responses are also thought to be modulated by regulatory T cells. However, the precise mechanisms governing the development and function of Plasmodium-specific T cells and their capacity to form tissue-resident and long-lived memory populations are less well understood. The field has arrived at a point where the push for vaccines that exploit T cell-mediated immunity to malaria has made it imperative to define and reconcile the mechanisms that regulate the development and functions of Plasmodium-specific T cells. Here, we review our current understanding of the mechanisms by which T cell subsets orchestrate host resistance to Plasmodium infection on the basis of observational and mechanistic studies in humans, non-human primates and rodent models. We also examine the potential of new experimental strategies and human infection systems to inform a new generation of approaches to harness T cell responses against malaria.
Collapse
|
27
|
Mitran CJ, Yanow SK. The Case for Exploiting Cross-Species Epitopes in Malaria Vaccine Design. Front Immunol 2020; 11:335. [PMID: 32174924 PMCID: PMC7056716 DOI: 10.3389/fimmu.2020.00335] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/10/2020] [Indexed: 12/21/2022] Open
Abstract
The infection dynamics between different species of Plasmodium that infect the same human host can both suppress and exacerbate disease. This could arise from inter-parasite interactions, such as competition, from immune regulation, or both. The occurrence of protective, cross-species (heterologous) immunity is an unlikely event, especially considering that strain-transcending immunity within a species is only partial despite lifelong exposure to that species. Here we review the literature in humans and animal models to identify the contexts where heterologous immunity can arise, and which antigens may be involved. From the perspective of vaccine design, understanding the mechanisms by which exposure to an antigen from one species can elicit a protective response to another species offers an alternative strategy to conventional approaches that focus on immunodominant antigens within a single species. The underlying hypothesis is that certain epitopes are conserved across evolution, in sequence or in structure, and shared in antigens from different species. Vaccines that focus on conserved epitopes may overcome the challenges posed by polymorphic immunodominant antigens; but to uncover these epitopes requires approaches that consider the evolutionary history of protein families across species. The key question for vaccinologists will be whether vaccines that express these epitopes can elicit immune responses that are functional and contribute to protection against Plasmodium parasites.
Collapse
Affiliation(s)
| | - Stephanie K. Yanow
- School of Public Health, University of Alberta, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
28
|
Bettencourt P. Current Challenges in the Identification of Pre-Erythrocytic Malaria Vaccine Candidate Antigens. Front Immunol 2020; 11:190. [PMID: 32153565 PMCID: PMC7046804 DOI: 10.3389/fimmu.2020.00190] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/24/2020] [Indexed: 12/27/2022] Open
Abstract
Plasmodium spp.-infected mosquitos inject sporozoites into the skin of a mammalian host during a blood meal. These enter the host's circulatory system and establish an infection in the liver. After a silent metamorphosis, merozoites invade the blood leading to the symptomatic and transmissible stages of malaria. The silent pre-erythrocytic malaria stage represents a bottleneck in the disease which is ideal to block progression to clinical malaria, through chemotherapeutic and immunoprophylactic interventions. RTS,S/AS01, the only malaria vaccine close to licensure, although with poor efficacy, blocks the sporozoite invasion mainly through the action of antibodies against the CSP protein, a major component of the pellicle of the sporozoite. Strikingly, sterile protection against malaria can be obtained through immunization with radiation-attenuated sporozoites, genetically attenuated sporozoites or through chemoprophylaxis with infectious sporozoites in animals and humans, but the deployability of sporozoite-based live vaccines pose tremendous challenges. The protection induced by sporozoites occurs in the pre-erythrocytic stages and is mediated mainly by antibodies against the sporozoite and CD8+ T cells against peptides presented by MHC class I molecules in infected hepatocytes. Thus, the identification of malaria antigens expressed in the sporozoite and liver-stage may provide new vaccine candidates to be included, alone or in combination, as recombinant protein-based, virus-like particles or sub-unit virally-vectored vaccines. Here I review the efforts being made to identify Plasmodium falciparum antigens expressed during liver-stage with focus on the development of parasite, hepatocyte, mouse models, and resulting rate of infection in order to identify new vaccine candidates and to improve the efficacy of the current vaccines. Finally, I propose new approaches for the identification of liver-stage antigens based on immunopeptidomics.
Collapse
|
29
|
Preclinical Development and Assessment of Viral Vectors Expressing a Fusion Antigen of Plasmodium falciparum LSA1 and LSAP2 for Efficacy against Liver-Stage Malaria. Infect Immun 2020; 88:IAI.00573-19. [PMID: 31740525 PMCID: PMC6977128 DOI: 10.1128/iai.00573-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
Despite promising progress in malaria vaccine development in recent years, an efficacious subunit vaccine against Plasmodium falciparum remains to be licensed and deployed. Cell-mediated protection from liver-stage malaria relies on a sufficient number of antigen-specific T cells reaching the liver during the time that parasites are present. A single vaccine expressing two antigens could potentially increase both the size and breadth of the antigen-specific response while halving vaccine production costs. Despite promising progress in malaria vaccine development in recent years, an efficacious subunit vaccine against Plasmodium falciparum remains to be licensed and deployed. Cell-mediated protection from liver-stage malaria relies on a sufficient number of antigen-specific T cells reaching the liver during the time that parasites are present. A single vaccine expressing two antigens could potentially increase both the size and breadth of the antigen-specific response while halving vaccine production costs. In this study, we investigated combining two liver-stage antigens, P. falciparum LSA1 (PfLSA1) and PfLSAP2, and investigated the induction of protective efficacy by coadministration of single-antigen vectors or vaccination with dual-antigen vectors, using simian adenovirus and modified vaccinia virus Ankara vectors. The efficacy of these vaccines was assessed in mouse malaria challenge models using chimeric P. berghei parasites expressing the relevant P. falciparum antigens and challenging mice at the peak of the T cell response. Vaccination with a combination of the single-antigen vectors expressing PfLSA1 or PfLSAP2 was shown to improve protective efficacy compared to vaccination with each single-antigen vector alone. Vaccination with dual-antigen vectors expressing both PfLSA1 and PfLSAP2 resulted in responses to both antigens, particularly in outbred mice, and most importantly, the efficacy was equivalent to that of vaccination with a mixture of single-antigen vectors. Based on these promising data, dual-antigen vectors expressing PfLSA1 and PfLSAP2 will now proceed to manufacturing and clinical assessment under good manufacturing practice (GMP) guidelines.
Collapse
|
30
|
Lefebvre MN, Harty JT. You Shall Not Pass: Memory CD8 T Cells in Liver-Stage Malaria. Trends Parasitol 2019; 36:147-157. [PMID: 31843536 DOI: 10.1016/j.pt.2019.11.004] [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] [Received: 10/02/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022]
Abstract
Each year over 200 million malaria infections occur, with over 400 000 associated deaths. Vaccines formed with attenuated whole parasites can induce protective memory CD8 T cell responses against liver-stage malaria; however, widespread administration of such vaccines is logistically challenging. Recent scientific findings are delineating how protective memory CD8 T cell populations are primed and maintained and how such cells mediate immunity to liver-stage malaria. Memory CD8 T cell anatomic localization and expression of transcription factors, homing receptors, and signaling molecules appear to play integral roles in protective immunity to liver-stage malaria. Further investigation of how such factors contribute to optimal protective memory CD8 T cell generation and maintenance in humans will inform efforts for improved vaccines.
Collapse
Affiliation(s)
- Mitchell N Lefebvre
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - John T Harty
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA; Department of Pathology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
31
|
Singh SK, Plieskatt J, Chourasia BK, Singh V, Bolscher JM, Dechering KJ, Adu B, López-Méndez B, Kaviraj S, Locke E, King CR, Theisen M. The Plasmodium falciparum circumsporozoite protein produced in Lactococcus lactis is pure and stable. J Biol Chem 2019; 295:403-414. [PMID: 31792057 DOI: 10.1074/jbc.ra119.011268] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/03/2019] [Indexed: 12/20/2022] Open
Abstract
The Plasmodium falciparum circumsporozoite protein (PfCSP) is a sporozoite surface protein whose role in sporozoite motility and cell invasion has made it the leading candidate for a pre-erythrocytic malaria vaccine. However, production of high yields of soluble recombinant PfCSP, including its extensive NANP and NVDP repeats, has proven problematic. Here, we report on the development and characterization of a secreted, soluble, and stable full-length PfCSP (containing 4 NVDP and 38 NANP repeats) produced in the Lactococcus lactis expression system. The recombinant full-length PfCSP, denoted PfCSP4/38, was produced initially with a histidine tag and purified by a simple two-step procedure. Importantly, the recombinant PfCSP4/38 retained a conformational epitope for antibodies as confirmed by both in vivo and in vitro characterizations. We characterized this complex protein by HPLC, light scattering, MS analysis, differential scanning fluorimetry, CD, SDS-PAGE, and immunoblotting with conformation-dependent and -independent mAbs, which confirmed it to be both pure and soluble. Moreover, we found that the recombinant protein is stable at both frozen and elevated-temperature storage conditions. When we used L. lactis-derived PfCSP4/38 to immunize mice, it elicited high levels of functional antibodies that had the capacity to modify sporozoite motility in vitro We concluded that the reported yield, purity, results of biophysical analyses, and stability of PfCSP4/38 warrant further consideration of using the L. lactis system for the production of circumsporozoite proteins for preclinical and clinical applications in malaria vaccine development.
Collapse
Affiliation(s)
- Susheel K Singh
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | | | - Bishwanath Kumar Chourasia
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Vandana Singh
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | | | | | - Bright Adu
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Blanca López-Méndez
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | | | - Emily Locke
- PATH's Malaria Vaccine Initiative, Washington, D. C. 20001
| | - C Richter King
- PATH's Malaria Vaccine Initiative, Washington, D. C. 20001
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark.
| |
Collapse
|
32
|
Kelemen RK, Rajakaruna H, Cockburn IA, Ganusov VV. Clustering of Activated CD8 T Cells Around Malaria-Infected Hepatocytes Is Rapid and Is Driven by Antigen-Specific Cells. Front Immunol 2019; 10:2153. [PMID: 31616407 PMCID: PMC6764016 DOI: 10.3389/fimmu.2019.02153] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/28/2019] [Indexed: 01/23/2023] Open
Abstract
Malaria, a disease caused by parasites of the Plasmodium genus, begins when Plasmodium-infected mosquitoes inject malaria sporozoites while searching for blood. Sporozoites migrate from the skin via blood to the liver, infect hepatocytes, and form liver stages which in mice 48 h later escape into blood and cause clinical malaria. Vaccine-induced activated or memory CD8 T cells are capable of locating and eliminating all liver stages in 48 h, thus preventing the blood-stage disease. However, the rules of how CD8 T cells are able to locate all liver stages within a relatively short time period remains poorly understood. We recently reported formation of clusters consisting of variable numbers of activated CD8 T cells around Plasmodium yoelii (Py)-infected hepatocytes. Using a combination of experimental data and mathematical models we now provide additional insights into mechanisms of formation of these clusters. First, we show that a model in which cluster formation is driven exclusively by T-cell-extrinsic factors, such as variability in "attractiveness" of different liver stages, cannot explain distribution of cluster sizes in different experimental conditions. In contrast, the model in which cluster formation is driven by the positive feedback loop (i.e., larger clusters attract more CD8 T cells) can accurately explain the available data. Second, while both Py-specific CD8 T cells and T cells of irrelevant specificity (non-specific CD8 T cells) are attracted to the clusters, we found no evidence that non-specific CD8 T cells play a role in cluster formation. Third and finally, mathematical modeling suggested that formation of clusters occurs rapidly, within few hours after adoptive transfer of CD8 T cells, thus illustrating high efficiency of CD8 T cells in locating their targets in complex peripheral organs, such as the liver. Taken together, our analysis provides novel insights into and attempts to discriminate between alternative mechanisms driving the formation of clusters of antigen-specific CD8 T cells in the liver.
Collapse
Affiliation(s)
- Reka K. Kelemen
- Institute of Science and Technology, Vienna, Austria
- Genome Science and Technology Program, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Harshana Rajakaruna
- Department of Microbiology, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Ian A. Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Vitaly V. Ganusov
- Genome Science and Technology Program, University of Tennessee, Knoxville, Knoxville, TN, United States
- Department of Microbiology, University of Tennessee, Knoxville, Knoxville, TN, United States
| |
Collapse
|
33
|
Vom Steeg LG, Flores-Garcia Y, Zavala F, Klein SL. Irradiated sporozoite vaccination induces sex-specific immune responses and protection against malaria in mice. Vaccine 2019; 37:4468-4476. [PMID: 31262583 PMCID: PMC7862922 DOI: 10.1016/j.vaccine.2019.06.075] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 10/26/2022]
Abstract
In both preclinical animal studies and human clinical trials, adult females tend to develop greater adaptive immune responses than males following receipt of either viral or bacterial vaccines. While there is currently no approved malaria vaccine, several anti-sporozoite vaccines, including RTS,S/AS01 and attenuated sporozoite vaccines, are in development, but the impact of sex and age on their efficacy remains undefined. To examine sex differences in the efficacy of anti-sporozoite stage malaria vaccination, adult (10 weeks of age) or juvenile (11 days of age) male and female C3H mice were twice vaccinated with irradiated transgenic Plasmodium berghei sporozoites expressing the P. falciparum circumsporozoite (CSP) protein and 45 days post boost vaccination, mice were challenged with transgenic P. berghei via mosquito bite or intradermal challenge. Immunization with irradiated sporozoites resulted in greater protection against challenge in adult females, which was associated with greater anti-CSP antibody production and avidity, as well as greater hepatic, but not splenic, CD8+ T cell IFNƴ production in adult females than adult males. No sex differences in adaptive immune responses or protection were observed in mice vaccinated prior to puberty, suggesting a role for sex steroid hormones. Depletion of testosterone in males increased, whereas rescue of testosterone decreased, anti-CSP antibody production, the number of antigen-specific CD8+ T cells isolated from the liver, and protection following parasite challenge. Conversely, depletion of sex steroids in female mice did not alter vaccine-induced responses or protection following challenge. These data suggest that elevated testosterone concentrations in males reduce adaptive immunity and contribute to sex differences in malaria vaccine efficacy.
Collapse
Affiliation(s)
- Landon G Vom Steeg
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, MD, USA
| | - Yevel Flores-Garcia
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, MD, USA
| | - Fidel Zavala
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, MD, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, MD, USA; Department of Biochemistry and Molecular Biology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
34
|
Shears MJ, Sekhar Nirujogi R, Swearingen KE, Renuse S, Mishra S, Jaipal Reddy P, Moritz RL, Pandey A, Sinnis P. Proteomic Analysis of Plasmodium Merosomes: The Link between Liver and Blood Stages in Malaria. J Proteome Res 2019; 18:3404-3418. [PMID: 31335145 DOI: 10.1021/acs.jproteome.9b00324] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The pre-erythrocytic liver stage of the malaria parasite, comprising sporozoites and the liver stages into which they develop, remains one of the least understood parts of the lifecycle, in part owing to the low numbers of parasites. Nonetheless, it is recognized as an important target for antimalarial drugs and vaccines. Here we provide the first proteomic analysis of merosomes, which define the final phase of the liver stage and are responsible for initiating the blood stage of infection. We identify a total of 1879 parasite proteins, and a core set of 1188 proteins quantitatively detected in every biological replicate, providing an extensive picture of the protein repertoire of this stage. This unique data set will allow us to explore key questions about the biology of merosomes and hepatic merozoites.
Collapse
Affiliation(s)
- Melanie J Shears
- Department of Molecular Microbiology & Immunology , Johns Hopkins Bloomberg School of Public Health , 615 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Raja Sekhar Nirujogi
- Department of Biological Chemistry , Johns Hopkins School of Medicine , 733 N. Broadway , Baltimore , Maryland 21205 , United States.,Institute of Bioinformatics , International Tech Park , Bangalore 560 066 , India
| | - Kristian E Swearingen
- Institute for Systems Biology , 401 Terry Avenue , North Seattle , Washington 98109 , United States
| | - Santosh Renuse
- Department of Biological Chemistry , Johns Hopkins School of Medicine , 733 N. Broadway , Baltimore , Maryland 21205 , United States
| | - Satish Mishra
- Department of Molecular Microbiology & Immunology , Johns Hopkins Bloomberg School of Public Health , 615 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Panga Jaipal Reddy
- Institute for Systems Biology , 401 Terry Avenue , North Seattle , Washington 98109 , United States
| | - Robert L Moritz
- Institute for Systems Biology , 401 Terry Avenue , North Seattle , Washington 98109 , United States
| | - Akhilesh Pandey
- Department of Biological Chemistry , Johns Hopkins School of Medicine , 733 N. Broadway , Baltimore , Maryland 21205 , United States
| | - Photini Sinnis
- Department of Molecular Microbiology & Immunology , Johns Hopkins Bloomberg School of Public Health , 615 North Wolfe Street , Baltimore , Maryland 21205 , United States
| |
Collapse
|
35
|
Yusuf Y, Yoshii T, Iyori M, Yoshida K, Mizukami H, Fukumoto S, Yamamoto DS, Alam A, Emran TB, Amelia F, Islam A, Otsuka H, Takashima E, Tsuboi T, Yoshida S. Adeno-Associated Virus as an Effective Malaria Booster Vaccine Following Adenovirus Priming. Front Immunol 2019; 10:730. [PMID: 31024558 PMCID: PMC6460511 DOI: 10.3389/fimmu.2019.00730] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
An ideal malaria vaccine platform should potently induce protective immune responses and block parasite transmission from mosquito to human, and it should maintain these effects for an extended period. Here, we have focused on vaccine development based on adeno-associated virus serotype 1 (AAV1), a viral vector widely studied in the field of clinical gene therapy that is able to induce long-term transgene expression without causing toxicity in vivo. Our results show the potential utility of AAV1 vectors as an extremely potent booster vaccine to induce durable immunity when combined with an adenovirus-priming vaccine in a rodent malaria model. We generated a series of recombinant AAV1s and human adenovirus type 5 (AdHu5) expressing either Plasmodium falciparum circumsporozoite protein (PfCSP) or P25 (Pfs25) protein. Heterologous two-dose immunization with an AdHu5-prime and AAV1-boost (AdHu5-AAV1) elicited robust and durable PfCSP- or Pfs25-specific functional antibodies over 280 days. Regarding protective efficacy, AdHu5-AAV1 PfCSP achieved high sterile protection (up to 80% protection rate) against challenge with transgenic Plasmodium berghei sporozoites expressing PfCSP. When examining transmission-blocking (TB) efficacy, we found that immunization with AdHu5-AAV1 Pfs25 maintained TB activity in vivo against transgenic P. berghei expressing Pfs25 for 287 days (99% reduction in oocyst intensity, 85% reduction in oocyst prevalence). Our data indicate that AAV1-based malaria vaccines can confer potent and durable protection as well as TB efficacy when administered following an AdHu5 priming vaccine, supporting the further evaluation of this regimen in clinical trials as a next-generation malaria vaccine platform.
Collapse
Affiliation(s)
- Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
- Department of Parasitology, Faculty of Medicine, University of Hasanuddin, Makassar, Indonesia
| | - Tatsuya Yoshii
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Kunitaka Yoshida
- Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroaki Mizukami
- Division of Gene therapy, Jichi Medical University, Shimotsuke, Japan
| | - Shinya Fukumoto
- National Research Centre for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Daisuke S. Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke, Japan
| | - Asrar Alam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Talha Bin Emran
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Fitri Amelia
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Ashekul Islam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Hiromu Otsuka
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
36
|
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
|
37
|
Khan S, Parrillo M, Gutierrez AH, Terry FE, Moise L, Martin WD, De Groot AS. Immune escape and immune camouflage may reduce the efficacy of RTS,S vaccine in Malawi. Hum Vaccin Immunother 2019; 16:214-227. [PMID: 30614773 PMCID: PMC7062414 DOI: 10.1080/21645515.2018.1560772] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The RTS,S/AS01 malaria vaccine will undergo a pilot vaccination study in sub-Saharan Africa beginning in 2019. RTS,S/AS01 Phase III trials reported an efficacy of 28.3% (children 5–17 months) and 18.3% (infants 6–12 weeks), with substantial variability across study sites. We postulated that the relatively low efficacy of the RTS,S vaccine and variability across sites may be due to lack of T-cell epitopes in the vaccine antigen, and due to the HLA distribution of the vaccinated population, and/or due to ‘immune camouflage’, an immune escape mechanism. To examine these hypotheses, we used immunoinformatics tools to compare T helper epitopes contained in RTS,S vaccine antigens with Plasmodium falciparum circumsporozoite protein (CSP) variants isolated from infected individuals in Malawi. The prevalence of epitopes restricted by specific HLA-DRB1 alleles was inversely associated with prevalence of the HLA-DRB1 allele in the Malawi study population, suggesting immune escape. In addition, T-cell epitopes in the CSP of strains circulating in Malawi were more often restricted by low-frequency HLA-DRB1 alleles in the population. Furthermore, T-cell epitopes that were highly conserved across CSP variants in Malawi possessed TCR-facing residues that were highly conserved in the human proteome, potentially reducing T-cell help through tolerance. The CSP component of the RTS,S vaccine also exhibited a low degree of T-cell epitope relatedness to circulating variants. These results suggest that RTS,S vaccine efficacy may be impacted by low T-cell epitope content, reduced presentation of T-cell epitopes by prevalent HLA-DRB1, high potential for human-cross-reactivity, and limited conservation with the CSP of circulating malaria strains.
Collapse
Affiliation(s)
- Sundos Khan
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA
| | - Matthew Parrillo
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA
| | | | | | - Leonard Moise
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA.,EpiVax, Inc., Providence, RI, USA
| | | | - Anne S De Groot
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA.,EpiVax, Inc., Providence, RI, USA
| |
Collapse
|
38
|
Mura M, Ruffié C, Combredet C, Aliprandini E, Formaglio P, Chitnis CE, Amino R, Tangy F. Recombinant measles vaccine expressing malaria antigens induces long-term memory and protection in mice. NPJ Vaccines 2019; 4:12. [PMID: 30820355 PMCID: PMC6393439 DOI: 10.1038/s41541-019-0106-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 01/22/2019] [Indexed: 11/09/2022] Open
Abstract
Following the RTS,S malaria vaccine, which showed only partial protection with short-term memory, there is strong support to develop second-generation malaria vaccines that yield higher efficacy with longer duration. The use of replicating viral vectors to deliver subunit vaccines is of great interest due to their capacity to induce efficient cellular immune responses and long-term memory. The measles vaccine virus offers an efficient and safe live viral vector that could easily be implemented in the field. Here, we produced recombinant measles viruses (rMV) expressing malaria “gold standard” circumsporozoïte antigen (CS) of Plasmodium berghei (Pb) and Plasmodium falciparum (Pf) to test proof of concept of this delivery strategy. Immunization with rMV expressing PbCS or PfCS induced high antibody responses in mice that did not decrease for at least 22 weeks post-prime, as well as rapid development of cellular immune responses. The observed long-term memory response is key for development of second-generation malaria vaccines. Sterile protection was achieved in 33% of immunized mice, as usually observed with the CS antigen, and all other immunized animals were clinically protected from severe and lethal Pb ANKA-induced cerebral malaria. Further rMV-vectored malaria vaccine candidates expressing additional pre-erythrocytic and blood-stage antigens in combination with rMV expressing PfCS may provide a path to development of next generation malaria vaccines with higher efficacy. Following the limited success of the RTS,S recombinant malaria vaccine there is a pressing need for second generation malaria vaccines. Frédéric Tangy and colleagues at the Pasteur Institute, Paris, generate novel vaccines based on recombinant measles virus (rMV) expressing the major circumsporozoite antigen CS from either Plasmodium berghei (rMV-CSPb) or P. falciparum (rMV-CSPf). rMV is a strong vector candidate because of its widespread use, safety profile and efficacy. Mice permissive to rMV infection show rapid and durable (at least 22 weeks) CS antibody responses as well as activation of cell-mediated immunity and type 1 helper responses following vaccination with rMV-CSPb or rMV-CSPf. rMV-CSPb vaccination protects mice from lethal challenge with Pb sporozoites, and in a subset of mice leads to sterile immunity. The rMV vector offers the potential of incorporating further antigens from other Plasmodium infection stages and thereby enhancement of vaccine efficacy.
Collapse
Affiliation(s)
- Marie Mura
- 1Viral Genomics and Vaccination, Institut Pasteur, CNRS UMR-3569, 28 rue du Dr Roux, 75015 Paris, France.,2Anti-infectious Biotherapies and Immunity, Institut de Recherche Biomédicale des Armées, 1 place du Général Valérie André, BP73 Brétigny-sur-Orge Cedex, France
| | - Claude Ruffié
- 1Viral Genomics and Vaccination, Institut Pasteur, CNRS UMR-3569, 28 rue du Dr Roux, 75015 Paris, France
| | - Chantal Combredet
- 1Viral Genomics and Vaccination, Institut Pasteur, CNRS UMR-3569, 28 rue du Dr Roux, 75015 Paris, France
| | - Eduardo Aliprandini
- 3Malaria Infection and Immunity, Institut Pasteur, CNRS UMR-3569, 28 rue du Dr Roux, 75015 Paris, France
| | - Pauline Formaglio
- 3Malaria Infection and Immunity, Institut Pasteur, CNRS UMR-3569, 28 rue du Dr Roux, 75015 Paris, France
| | - Chetan E Chitnis
- 4Malaria Parasite Biology and Vaccines, Institut Pasteur, CNRS UMR-3569, 28 rue du Dr Roux, 75015 Paris, France
| | - Rogerio Amino
- 3Malaria Infection and Immunity, Institut Pasteur, CNRS UMR-3569, 28 rue du Dr Roux, 75015 Paris, France
| | - Frédéric Tangy
- 1Viral Genomics and Vaccination, Institut Pasteur, CNRS UMR-3569, 28 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
39
|
Abstract
Plasmodium sporozoites are injected into the skin as mosquitoes probe for blood. From here, they migrate through the dermis to find blood vessels which they enter in order to be rapidly carried to the liver, where they invade hepatocytes and develop into the next life cycle stage, the exoerythrocytic stage. Once sporozoites enter the blood circulation, they are found in hepatocytes within minutes. In contrast, sporozoite exit from the inoculation site resembles a slow trickle and occurs over several hours. Thus, sporozoites spend the majority of their extracellular time at the inoculation site, raising the hypothesis that this is when the malarial parasite is most vulnerable to antibody-mediated destruction. Here, we investigate this hypothesis and demonstrate that the neutralizing capacity of circulating antibodies is greater at the inoculation site than in the blood circulation. Furthermore, these antibodies are working, at least in part, by impacting sporozoite motility at the inoculation site. Using actively and passively immunized mice, we found that most parasites are either immobilized at the site of injection or display reduced motility, particularly in their net displacement. We also found that antibodies severely impair the entry of sporozoites into the bloodstream. Overall, our data suggest that antibodies targeting the migratory sporozoite exert a large proportion of their protective effect at the inoculation site.IMPORTANCE Studies in experimental animal models and humans have shown that antibodies against Plasmodium sporozoites abolish parasite infectivity and provide sterile immunity. While it is well documented that these antibodies can be induced after immunization with attenuated parasites or subunit vaccines, the mechanisms by and location in which they neutralize parasites have not been fully elucidated. Here, we report studies indicating that these antibodies display a significant portion of their protective effect in the skin after injection of sporozoites and that one mechanism by which they work is by impairing sporozoite motility, thus diminishing their ability to reach blood vessels. These results suggest that immune protection against malaria begins at the earliest stages of parasite infection and emphasize the need of performing parasite challenge in the skin for the evaluation of protective immunity.
Collapse
|
40
|
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
|
41
|
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
|
42
|
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
|
43
|
Bliss CM, Bowyer G, Anagnostou NA, Havelock T, Snudden CM, Davies H, de Cassan SC, Grobbelaar A, Lawrie AM, Venkatraman N, Poulton ID, Roberts R, Mange PB, Choudhary P, Faust SN, Colloca S, Gilbert SC, Nicosia A, Hill AVS, Ewer KJ. Assessment of novel vaccination regimens using viral vectored liver stage malaria vaccines encoding ME-TRAP. Sci Rep 2018; 8:3390. [PMID: 29467399 PMCID: PMC5821890 DOI: 10.1038/s41598-018-21630-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/07/2018] [Indexed: 11/18/2022] Open
Abstract
Heterologous prime-boost vaccination with viral vectors simian adenovirus 63 (ChAd63) and Modified Vaccinia Ankara (MVA) induces potent T cell and antibody responses in humans. The 8-week regimen demonstrates significant efficacy against malaria when expressing the pre-erythrocytic malaria antigen Thrombospondin-Related Adhesion Protein fused to a multiple epitope string (ME-TRAP). We tested these vaccines in 7 new 4- and 8- week interval schedules to evaluate safety and immunogenicity of multiple ChAd63 ME-TRAP priming vaccinations (denoted A), multiple MVA ME-TRAP boosts (denoted M) and alternating vectors. All regimens exhibited acceptable reactogenicity and CD8+ T cell immunogenicity was enhanced with a 4-week interval (AM) and with incorporation of additional ChAd63 ME-TRAP vaccination at 4- or 8-weeks (AAM or A_A_M). Induction of TRAP antibodies was comparable between schedules. T cell immunity against the ChAd63 hexon did not affect T cell responses to the vaccine insert, however pre-vaccination ChAd63-specific T cells correlated with reduced TRAP antibodies. Vaccine-induced antibodies against MVA did not affect TRAP antibody induction, and correlated positively with ME-TRAP-specific T cells. This study identifies potentially more effective immunisation regimens to assess in Phase IIa trials and demonstrates a degree of flexibility with the timing of vectored vaccine administration, aiding incorporation into existing vaccination programmes.
Collapse
Affiliation(s)
- Carly M Bliss
- The Jenner Institute, University of Oxford, Oxford, UK.
| | | | | | - Tom Havelock
- NIHR Wellcome Trust Clinical Research Facility, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Huw Davies
- Department of Medicine, Division of Infectious Diseases, University of California, Irvine, CA, USA
| | | | | | | | | | - Ian D Poulton
- The Jenner Institute, University of Oxford, Oxford, UK
| | | | - Pooja B Mange
- The Jenner Institute, University of Oxford, Oxford, UK
| | | | - Saul N Faust
- NIHR Wellcome Trust Clinical Research Facility, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | | | - Alfredo Nicosia
- ReiThera (formerly Okairos), 00144, Rome, Italy
- CEINGE, Via Comunale Margherita, 484-538, 80131, Napoli, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | | - Katie J Ewer
- The Jenner Institute, University of Oxford, Oxford, UK
| |
Collapse
|
44
|
Pichugin A, Zarling S, Perazzo L, Duffy PE, Ploegh HL, Krzych U. Identification of a Novel CD8 T Cell Epitope Derived from Plasmodium berghei Protective Liver-Stage Antigen. Front Immunol 2018; 9:91. [PMID: 29434602 PMCID: PMC5796907 DOI: 10.3389/fimmu.2018.00091] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/12/2018] [Indexed: 11/13/2022] Open
Abstract
We recently identified novel Plasmodium berghei (Pb) liver stage (LS) genes that as DNA vaccines significantly reduce Pb LS parasite burden (LPB) in C57Bl/6 (B6) mice through a mechanism mediated, in part, by CD8 T cells. In this study, we sought to determine fine antigen (Ag) specificities of CD8 T cells that target LS malaria parasites. Guided by algorithms for predicting MHC class I-restricted epitopes, we ranked sequences of 32 Pb LS Ags and selected ~400 peptides restricted by mouse H-2Kb and H-2Db alleles for analysis in the high-throughput method of caged MHC class I-tetramer technology. We identified a 9-mer H-2Kb restricted CD8 T cell epitope, Kb-17, which specifically recognized and activated CD8 T cell responses in B6 mice immunized with Pb radiation-attenuated sporozoites (RAS) and challenged with infectious sporozoites (spz). The Kb-17 peptide is derived from the recently described novel protective Pb LS Ag, PBANKA_1031000 (MIF4G-like protein). Notably, immunization with the Kb-17 epitope delivered in the form of a minigene in the adenovirus serotype 5 vector reduced LPB in mice infected with spz. On the basis of our results, Kb-17 peptide was available for CD8 T cell activation and recall following immunization with Pb RAS and challenge with infectious spz. The identification of a novel MHC class I-restricted epitope from the protective Pb LS Ag, MIF4G-like protein, is crucial for advancing our understanding of immune responses to Plasmodium and by extension, toward vaccine development against malaria.
Collapse
Affiliation(s)
- Alexander Pichugin
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Stasya Zarling
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Leah Perazzo
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Patrick Emmet Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, (NIH), Rockville, MD, United States
| | - Hidde Lolke Ploegh
- Program in Cellular and Molecular Medicine, Division of Molecular Biology, Department of Medicine, Boston Children's Hospital, Boston, MD, United States
| | - Urszula Krzych
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
45
|
Müller K, Gibbins MP, Matuschewski K, Hafalla JCR. Evidence of cross-stage CD8+ T cell epitopes in malaria pre-erythrocytic and blood stage infections. Parasite Immunol 2017; 39. [PMID: 28380250 DOI: 10.1111/pim.12434] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/28/2017] [Indexed: 12/18/2022]
Abstract
Malaria parasites have a complex, multistage life cycle and there is a widely held view that each stage displays a distinct set of antigens presented to the immune system. Yet, molecular analysis of malaria parasites suggests that many putative antigenic targets are shared amongst the different stages. The specificities of these cross-stage antigens and the functions of the immune responses they elicit are poorly characterized. It is well-known that CD8+ T cells play opposing immune functions following Plasmodium berghei (Pb) infection of C57BL/6 mice. Whilst these cells play a crucial role in protective immunity against pre-erythrocytic stages, they are implicated in the development of severe disease during blood stages. Recently, CD8+ T cell epitopes derived from proteins supposedly specific for either pre-erythrocytic or blood stages have been described. In this brief report, we have compiled and confirmed data that the majority of the mRNAs and/or proteins from which these epitopes are derived display expression across pre-erythrocytic and blood stages. Importantly, we provide evidence of cross-stage immune recognition of the majority of these CD8+ T cell epitopes. Hence, our findings provide a resource to further examine the relevance of antigen-specific cross-stage responses during malaria infections.
Collapse
Affiliation(s)
- K Müller
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - M P Gibbins
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, UK
| | - K Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - J C R Hafalla
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
46
|
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
|
47
|
Scally SW, Murugan R, Bosch A, Triller G, Costa G, Mordmüller B, Kremsner PG, Sim BKL, Hoffman SL, Levashina EA, Wardemann H, Julien JP. Rare PfCSP C-terminal antibodies induced by live sporozoite vaccination are ineffective against malaria infection. J Exp Med 2017; 215:63-75. [PMID: 29167197 PMCID: PMC5748854 DOI: 10.1084/jem.20170869] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/30/2017] [Accepted: 10/11/2017] [Indexed: 11/16/2022] Open
Abstract
Scally et al. show the molecular, structural, and functional characterization of human antibodies against the C-terminal domain of Plasmodium falciparum (Pf) circumsporozoite (CSP [C-PfCSP]) and reveal that its arrangement on the Pf sporozoite surface and epitope polymorphism contribute to poor C-PfCSP immunogenicity and ineffective humoral responses in volunteers protected against Pf malaria. Antibodies against the central repeat of the Plasmodium falciparum (Pf) circumsporozoite protein (CSP) inhibit parasite activity and correlate with protection from malaria. However, the humoral response to the PfCSP C terminus (C-PfCSP) is less well characterized. Here, we describe B cell responses to C-PfCSP from European donors who underwent immunization with live Pf sporozoites (PfSPZ Challenge) under chloroquine prophylaxis (PfSPZ-CVac), and were protected against controlled human malaria infection. Out of 215 PfCSP-reactive monoclonal antibodies, only two unique antibodies were specific for C-PfCSP, highlighting the rare occurrence of C-PfCSP–reactive B cells in PfSPZ-CVac–induced protective immunity. These two antibodies showed poor sporozoite binding and weak inhibition of parasite traversal and development, and did not protect mice from infection with PfCSP transgenic Plasmodium berghei sporozoites. Structural analyses demonstrated that one antibody interacts with a polymorphic region overlapping two T cell epitopes, suggesting that variability in C-PfCSP may benefit parasite escape from humoral and cellular immunity. Our data identify important features underlying C-PfCSP shortcomings as a vaccine target.
Collapse
Affiliation(s)
- Stephen W Scally
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Rajagopal Murugan
- B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Alexandre Bosch
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Gianna Triller
- B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Giulia Costa
- Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Benjamin Mordmüller
- Institute of Tropical Medicine and German Center for Infection Research, University of Tübingen, Tübingen, Germany
| | - Peter G Kremsner
- Institute of Tropical Medicine and German Center for Infection Research, University of Tübingen, Tübingen, Germany
| | | | | | - Elena A Levashina
- Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hedda Wardemann
- B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Jean-Philippe Julien
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada .,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
48
|
Shiratsuchi T, Rai U, Kaneko I, Zhang M, Iwanaga S, Yuda M, Tsuji M. A potent malaria vaccine based on adenovirus with dual modifications at Hexon and pVII. Vaccine 2017; 35:6990-7000. [PMID: 29089194 DOI: 10.1016/j.vaccine.2017.10.066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 09/23/2017] [Accepted: 10/20/2017] [Indexed: 02/08/2023]
Abstract
Adenovirus (Ad) is thought to be one of the most promising platforms for a malaria vaccine targeted against its liver stages, because of its ability to induce a strong T-cell response against a transgene. However, a further improvement of this platform is needed in order to elicit another arm of the immunity, i.e. humoral response, against malaria. In order to augment immunogenicity and protective efficacy of Ad-based malaria vaccine, we inserted B-cell, as well as CD4+ T-cell, epitopes of Plasmodium falciparum circumsporozoite protein (PfCSP) into the capsid protein, Hexon, and the core protein, VII (pVII), of Ad, respectively, in addition to the PfCSP transgene. Insertion of PfCSP-derived B cell epitope to Hexon significantly enhanced the epitope-specific antibody response compared to AdPfCSP, an Ad vaccine expressing only PfCSP transgene. PfCSP-derived CD4+ T-cell epitope insertion into pVII augmented not only PfCSP-specific CD4+ T-cell response but also anti-PfCSP antibody response. Finally, mice immunized with AdPfCSP having both Hexon and pVII modifications were more protected than AdPfCSP or Hexon-modified AdPfCSP against challenge with transgenic rodent malaria parasites expressing the PfCSP. Overall, this study has demonstrated that Hexon and pVII-modified AdPfCSP vaccine is a promising malaria vaccine which induces strong PfCSP-specific humoral, CD4+ T-cell, and CD8+ T-cell responses and protects against infection with transgenic malaria parasites expressing the PfCSP.
Collapse
Affiliation(s)
- Takayuki Shiratsuchi
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, 455 First Avenue, New York, NY 10016, USA; Otsuka Maryland Medicinal Laboratories, Inc., 9900 Medical Center Drive, Rockville, MD 20850, USA
| | - Urvashi Rai
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, 455 First Avenue, New York, NY 10016, USA
| | - Izumi Kaneko
- Department of Medical Zoology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Min Zhang
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, 455 First Avenue, New York, NY 10016, USA; Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Shiroh Iwanaga
- Department of Medical Zoology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan; Department of Environmental Parasitology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Masao Yuda
- Department of Medical Zoology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Moriya Tsuji
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, 455 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
49
|
Venkatraman N, Anagnostou N, Bliss C, Bowyer G, Wright D, Lövgren-Bengtsson K, Roberts R, Poulton I, Lawrie A, Ewer K, V S Hill A. Safety and immunogenicity of heterologous prime-boost immunization with viral-vectored malaria vaccines adjuvanted with Matrix-M™. Vaccine 2017; 35:6208-6217. [PMID: 28941620 DOI: 10.1016/j.vaccine.2017.09.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/09/2017] [Accepted: 09/07/2017] [Indexed: 12/19/2022]
Abstract
The use of viral vectors in heterologous prime-boost regimens to induce potent T cell responses in addition to humoral immunity is a promising vaccination strategy in the fight against malaria. We conducted an open-label, first-in-human, controlled Phase I study evaluating the safety and immunogenicity of Matrix-M adjuvanted vaccination with a chimpanzee adenovirus serotype 63 (ChAd63) prime followed by a modified vaccinia Ankara (MVA) boost eight weeks later, both encoding the malaria ME-TRAP antigenic sequence (a multiple epitope string fused to thrombospondin-related adhesion protein). Twenty-two healthy adults were vaccinated intramuscularly with either ChAd63-MVA ME-TRAP alone (n=6) or adjuvanted with 25μg (n=8) or 50μg (n=8) Matrix-M. Vaccinations appeared to be safe and generally well tolerated, with the majority of local and systemic adverse events being mild in nature. The addition of Matrix-M to the vaccine did not increase local reactogenicity; however, systemic adverse events were reported more frequently by volunteers who received adjuvanted vaccine in comparison to the control group. T cell ELISpot responses peaked at 7-days post boost vaccination with MVA ME-TRAP in all three groups. TRAP-specific IgG responses were highest at 28-days post boost with MVA ME-TRAP in all three groups. There were no differences in cellular and humoral immunogenicity at any of the time points between the control group and the adjuvanted groups. We demonstrate that Matrix-M can be safely used in combination with ChAd63-MVA ME-TRAP heterologous prime-boost immunization without any reduction in cellular or humoral immunogenicity. Clinical Trials Registration NCT01669512.
Collapse
Affiliation(s)
- Navin Venkatraman
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7DQ, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, United Kingdom.
| | - Nicholas Anagnostou
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7DQ, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, United Kingdom
| | - Carly Bliss
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7DQ, UK
| | - Georgina Bowyer
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7DQ, UK
| | - Danny Wright
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7DQ, UK
| | | | - Rachel Roberts
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7DQ, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, United Kingdom
| | - Ian Poulton
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7DQ, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, United Kingdom
| | - Alison Lawrie
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7DQ, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, United Kingdom
| | - Katie Ewer
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7DQ, UK
| | - Adrian V S Hill
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7DQ, UK
| |
Collapse
|
50
|
Bruder JT, Chen P, Ekberg G, Smith EC, Lazarski CA, Myers BA, Bolton J, Sedegah M, Villasante E, Richie TL, King CR, Aguiar JC, Doolan DL, Brough DE. Profiling the Targets of Protective CD8 + T Cell Responses to Infection. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:20-31. [PMID: 28948187 PMCID: PMC5602877 DOI: 10.1016/j.omtm.2017.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 08/14/2017] [Indexed: 11/22/2022]
Abstract
T cells are critical effectors of host immunity that target intracellular pathogens, such as the causative agents of HIV, tuberculosis, and malaria. The development of vaccines that induce effective cell-mediated immunity against such pathogens has proved challenging; for tuberculosis and malaria, many of the antigens targeted by protective T cells are not known. Here, we report a novel approach for screening large numbers of antigens as potential targets of T cells. Malaria provides an excellent model to test this antigen discovery platform because T cells are critical mediators of protection following immunization with live sporozoite vaccines and the specific antigen targets are unknown. We generated an adenovirus array by cloning 312 highly expressed pre-erythrocytic Plasmodium yoelii antigens into adenovirus vectors using high-throughput methodologies. The array was screened to identify antigen-specific CD8+ T cells induced by a live sporozoite vaccine regimen known to provide high levels of sterile protection mediated by CD8+ T cells. We identified 69 antigens that were targeted by CD8+ T cells induced by this vaccine regimen. The antigen that recalled the highest frequency of CD8+ T cells, PY02605, induced protective responses in mice, demonstrating proof of principle for this approach in identifying antigens for vaccine development.
Collapse
Affiliation(s)
- Joseph T. Bruder
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
- Corresponding author: Joseph T. Bruder, Summit Consulting, 567 Chestertown Street, Gaithersburg, MD 20878, USA.
| | - Ping Chen
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
| | - Greg Ekberg
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
| | - Emily C. Smith
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., 6720A Rockledge Drive, Suite 100, Bethesda, MD 20817, USA
| | | | - Bennett A. Myers
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
| | - Jessica Bolton
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., 6720A Rockledge Drive, Suite 100, Bethesda, MD 20817, USA
| | - Martha Sedegah
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Eileen Villasante
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Thomas L. Richie
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - C. Richter King
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
| | - Joao C. Aguiar
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
- Camris International, 3 Bethesda Metro Center, 16th Floor, Bethesda, MD 20814, USA
| | - Denise L. Doolan
- Australian Institute of Tropical Health and Medicine, James Cook University, McGregor Road, Cairns, QLD 4870, Australia
| | - Douglas E. Brough
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
| |
Collapse
|