1
|
Goswami D, Arredondo SA, Betz W, Armstrong J, Kumar S, Zanghi G, Patel H, Camargo N, Oualim KMZ, Seilie AM, Schneider S, Murphy SC, Kappe SHI, Vaughan AM. A conserved Plasmodium nuclear protein is critical for late liver stage development. Commun Biol 2024; 7:1387. [PMID: 39455824 PMCID: PMC11511937 DOI: 10.1038/s42003-024-07063-y] [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: 12/01/2023] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Malaria, caused by Plasmodium parasites, imposes a significant health burden and live-attenuated parasites are being pursued as vaccines. Here, we report on the creation of a genetically attenuated parasite by the deletion of Plasmodium LINUP, encoding a liver stage nuclear protein. In the rodent parasite Plasmodium yoelii, LINUP expression was restricted to liver stage nuclei after the onset of liver stage schizogony. Compared to wildtype P. yoelii, P. yoelii LINUP gene deletion parasites (linup-) exhibited no phenotype in blood stages and mosquito stages but suffered developmental arrest late in liver stage schizogony with a pronounced defect in exo-erythrocytic merozoite formation. This defect caused severe attenuation of the liver stage-to-blood stage transition and immunization of mice with linup - parasites conferred robust protection against infectious sporozoite challenge. LINUP gene deletion in the human parasite Plasmodium falciparum also caused a severe defect in late liver stage differentiation. Importantly, P. falciparum linup - liver stages completely failed to transition from the liver stage to a viable blood stage infection in a humanized mouse model. These results suggest that P. falciparum LINUP is an ideal target for late liver stage attenuation that can be incorporated into a late liver stage-arresting replication competent whole parasite vaccine.
Collapse
Affiliation(s)
- Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
| | - Silvia A Arredondo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - William Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Janna Armstrong
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Gigliola Zanghi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Hardik Patel
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Kenza M Z Oualim
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Annette M Seilie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Sophia Schneider
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - 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.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - 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.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
2
|
Kinyua AW, Turner L, Kimingi HW, Mwai K, Mwikali K, Andisi C, Sim BKL, Bejon P, Kapulu MC, Kinyanjui SM, Lavstsen T, Abdi AI. Antibodies to PfEMP1 and variant surface antigens: Protection after controlled human malaria infection in semi-immune Kenyan adults. J Infect 2024; 89:106252. [PMID: 39182654 PMCID: PMC11409615 DOI: 10.1016/j.jinf.2024.106252] [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: 06/12/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
OBJECTIVES Acquisition of antibodies to Plasmodium falciparum variant surface antigens (VSA) expressed on infected red blood cells (iRBCs) is associated with naturally acquired immunity to malaria. We have previously shown that antibodies to VSA on iRBCs are associated with protection against parasite growth in the context of controlled human malaria infection (CHMI). This study explored whether antibodies to recombinant antigens derived from PfEMP1 domains were independently associated with protection during CHMI in semi-immune Kenyan adults. METHODS We used a multiplex bead assay to measure levels of IgG antibody against a panel of 27 recombinant PfEMP1 antigens derived from the PfEMP1 repertoire of the 3D7 parasite clone. We measured IgG levels in plasma samples collected from the CHMI participants before inoculation with Sanaria® PfSPZ Challenge, on the day of diagnosis, and 35 days post-inoculation. Univariable and multivariable Cox regression analysis was used to evaluate the relationship between the levels of antibodies to the antigens and CHMI outcome. We also adjusted for previous data including antibodies to VSA on iRBCs, and we assessed the kinetics of antibody acquisition to the different PfEMP1 recombinant antigens over time. RESULTS All study participants had detectable antibodies to multiple PfEMP1 proteins before inoculation. All PfEMP1 antigens were associated with protection against parasite growth to the threshold criteria for treatment in CHMI, albeit with substantial collinearity. However, individual PfEMP1 antigens were not independently associated with protection following adjustment for breadth of reactivity to VSA on iRBCs and schizont extract. In addition, antibodies to PfEMP1 antigens derived from group B PfEMP1 were induced and sustained in the participants who could not control parasite growth. CONCLUSION This study shows that the breadth of antibody response to VSA on iRBCs, and not to specific PfEMP1 antigens, is predictive of protection against malaria in CHMI.
Collapse
Affiliation(s)
- Ann W Kinyua
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Louise Turner
- Centre for translational Medicine & Parasitology, Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Righospitalet, Copenhagen, Denmark
| | - Hannah W Kimingi
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kennedy Mwai
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kioko Mwikali
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Cheryl Andisi
- Pwani University Bioscience Research Centre, Pwani University, Kilifi, Kenya
| | | | - Philip Bejon
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, United Kingdom
| | - Melissa C Kapulu
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, United Kingdom
| | - Samson M Kinyanjui
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya; Pwani University Bioscience Research Centre, Pwani University, Kilifi, Kenya; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, United Kingdom; School of Business Studies, Strathmore University, Nairobi, Kenya
| | - Thomas Lavstsen
- Centre for translational Medicine & Parasitology, Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Righospitalet, Copenhagen, Denmark
| | - Abdirahman I Abdi
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya; Pwani University Bioscience Research Centre, Pwani University, Kilifi, Kenya; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, United Kingdom.
| |
Collapse
|
3
|
Moita D, Prudêncio M. Whole-sporozoite malaria vaccines: where we are, where we are going. EMBO Mol Med 2024; 16:2279-2289. [PMID: 39284948 PMCID: PMC11473726 DOI: 10.1038/s44321-024-00131-0] [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: 05/12/2024] [Revised: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 10/16/2024] Open
Abstract
The malaria vaccination landscape has seen significant advancements with the recent endorsement of RTS,S/AS01 and R21/Matrix-M vaccines, which target the pre-erythrocytic stages of Plasmodium falciparum (Pf) infection. However, several challenges remain to be addressed, including the incomplete protection afforded by these vaccines, their dependence on a single Pf antigen, and the fact that they were not designed to protect against P. vivax (Pv) malaria. Injectable formulations of whole-sporozoite (WSpz) malaria vaccines offer a promising alternative to existing subunit vaccines, with recent developments including genetically engineered parasites and optimized administration regimens. Clinical evaluations demonstrate varying efficacy, influenced by factors, such as immune status, prior exposure to malaria, and age. Despite significant progress, a few hurdles persist in vaccine production, deployment, and efficacy in malaria-endemic regions, particularly in children. Concurrently, transgenic parasites expressing Pv antigens emerge as potential solutions for PvWSpz vaccine development. Ongoing clinical studies and advancements in vaccine technology, including the recently described PfSPZ-LARC2 candidate, signify a hopeful future for WSpz malaria vaccines, which hold great promise in the global fight against malaria.
Collapse
Affiliation(s)
- Diana Moita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal.
| |
Collapse
|
4
|
Tian X, Janes HE, Kublin JG. Statistical design and analysis of controlled human malaria infection trials. Malar J 2024; 23:133. [PMID: 38702775 PMCID: PMC11068571 DOI: 10.1186/s12936-024-04959-2] [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: 05/01/2023] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Malaria is a potentially life-threatening disease caused by Plasmodium protozoa transmitted by infected Anopheles mosquitoes. Controlled human malaria infection (CHMI) trials are used to assess the efficacy of interventions for malaria elimination. The operating characteristics of statistical methods for assessing the ability of interventions to protect individuals from malaria is uncertain in small CHMI studies. This paper presents simulation studies comparing the performance of a variety of statistical methods for assessing efficacy of intervention in CHMI trials. METHODS Two types of CHMI designs were investigated: the commonly used single high-dose design (SHD) and the repeated low-dose design (RLD), motivated by simian immunodeficiency virus (SIV) challenge studies. In the context of SHD, the primary efficacy endpoint is typically time to infection. Using a continuous time survival model, five statistical tests for assessing the extent to which an intervention confers partial or full protection under single dose CHMI designs were evaluated. For RLD, the primary efficacy endpoint is typically the binary infection status after a specific number of challenges. A discrete time survival model was used to study the characteristics of RLD versus SHD challenge studies. RESULTS In a SHD study with the continuous time survival model, log-rank test and t-test are the most powerful and provide more interpretable results than Wilcoxon rank-sum tests and Lachenbruch tests, while the likelihood ratio test is uniformly most powerful but requires knowledge of the underlying probability model. In the discrete time survival model setting, SHDs are more powerful for assessing the efficacy of an intervention to prevent infection than RLDs. However, additional information can be inferred from RLD challenge designs, particularly using a likelihood ratio test. CONCLUSIONS Different statistical methods can be used to analyze controlled human malaria infection (CHMI) experiments, and the choice of method depends on the specific characteristics of the experiment, such as the sample size allocation between the control and intervention groups, and the nature of the intervention. The simulation results provide guidance for the trade off in statistical power when choosing between different statistical methods and study designs.
Collapse
Affiliation(s)
- Xiaowen Tian
- Department of Biostatistics, University of Washington, 3980 15th Ave NE, Seattle, WA, 98195, USA.
| | - Holly E Janes
- Department of Biostatistics, University of Washington, 3980 15th Ave NE, Seattle, WA, 98195, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
- Department of Global Health, University of Washington, 3980 15th Ave NE, Seattle, WA, 98195, USA
| |
Collapse
|
5
|
Goswami D, Patel H, Betz W, Armstrong J, Camargo N, Patil A, Chakravarty S, Murphy SC, Sim BKL, Vaughan AM, Hoffman SL, Kappe SH. A replication competent Plasmodium falciparum parasite completely attenuated by dual gene deletion. EMBO Mol Med 2024; 16:723-754. [PMID: 38514791 PMCID: PMC11018819 DOI: 10.1038/s44321-024-00057-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Vaccination with infectious Plasmodium falciparum (Pf) sporozoites (SPZ) administered with antimalarial drugs (PfSPZ-CVac), confers superior sterilizing protection against infection when compared to vaccination with replication-deficient, radiation-attenuated PfSPZ. However, the requirement for drug administration constitutes a major limitation for PfSPZ-CVac. To obviate this limitation, we generated late liver stage-arresting replication competent (LARC) parasites by deletion of the Mei2 and LINUP genes (mei2-/linup- or LARC2). We show that Plasmodium yoelii (Py) LARC2 sporozoites did not cause breakthrough blood stage infections and engendered durable sterilizing immunity against various infectious sporozoite challenges in diverse strains of mice. We next genetically engineered a PfLARC2 parasite strain that was devoid of extraneous DNA and produced cryopreserved PfSPZ-LARC2. PfSPZ-LARC2 liver stages replicated robustly in liver-humanized mice but displayed severe defects in late liver stage differentiation and did not form liver stage merozoites. This resulted in complete abrogation of parasite transition to viable blood stage infection. Therefore, PfSPZ-LARC2 is the next-generation vaccine strain expected to unite the safety profile of radiation-attenuated PfSPZ with the superior protective efficacy of PfSPZ-CVac.
Collapse
Affiliation(s)
- Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Hardik Patel
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - William Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Janna Armstrong
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Asha Patil
- Sanaria Inc., 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | | | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - B Kim Lee Sim
- Sanaria Inc., 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Stefan Hi Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
6
|
Kibwana E, Kapulu M. Controlled Human Malaria Infection Studies in Africa-Past, Present, and Future. Curr Top Microbiol Immunol 2024; 445:337-365. [PMID: 35704094 PMCID: PMC7616462 DOI: 10.1007/82_2022_256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Controlled human infection studies have contributed significantly to the understanding of pathogeneses and treatment of infectious diseases. In malaria, deliberately infecting humans with malaria parasites was used as a treatment for neurosyphilis in the early 1920s. More recently, controlled human malaria infection (CHMI) has become a valuable, cost-effective tool to fast-track the development and evaluation of new anti-malarial drugs and/or vaccines. CHMI studies have also been used to define host/parasite interactions and immunological correlates of protection. CHMI involves infecting a small number of healthy volunteers with malaria parasites, monitoring their parasitemia and providing anti-malarial treatment when a set threshold is reached. In this review we discuss the introduction, development, and challenges of modern-day Plasmodium falciparum CHMI studies conducted in Africa, and the impact of naturally acquired immunity on infectivity and vaccine efficacy. CHMIs have shown to be an invaluable tool particularly in accelerating malaria vaccine research. Although there are limitations of CHMI studies for estimating public health impacts and for regulatory purposes, their strength lies in proof-of-concept efficacy data at an early stage of development, providing a faster way to select vaccines for further development and providing valuable insights in understanding the mechanisms of immunity to malarial infection.
Collapse
Affiliation(s)
- Elizabeth Kibwana
- Bioscience Department, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | - Melissa Kapulu
- Bioscience Department, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| |
Collapse
|
7
|
van der Plas JL, Kuiper VP, Bagchus WM, Bödding M, Yalkinoglu Ö, Tappert A, Seitzinger A, Spangenberg T, Bezuidenhout D, Wilkins J, Oeuvray C, Dhingra SK, Thathy V, Fidock DA, Smidt LCA, Roozen GVT, Koopman JPR, Lamers OAC, Sijtsma J, van Schuijlenburg R, Wessels E, Meij P, Kamerling IMC, Roestenberg M, Khandelwal A. Causal chemoprophylactic activity of cabamiquine against Plasmodium falciparum in a controlled human malaria infection: a randomised, double-blind, placebo-controlled study in the Netherlands. THE LANCET. INFECTIOUS DISEASES 2023; 23:1164-1174. [PMID: 37414066 DOI: 10.1016/s1473-3099(23)00212-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/05/2023] [Accepted: 03/24/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND Cabamiquine is a novel antimalarial that inhibits Plasmodium falciparum translation elongation factor 2. We investigated the causal chemoprophylactic activity and dose-exposure-response relationship of single oral doses of cabamiquine following the direct venous inoculation (DVI) of P falciparum sporozoites in malaria-naive, healthy volunteers. METHODS This was a phase 1b, randomised, double-blind, placebo-controlled, adaptive, dose-finding, single-centre study performed in Leiden, Netherlands. Malaria-naive, healthy adults aged 18-45 years were divided into five cohorts and randomly assigned (3:1) to receive cabamiquine or placebo. Randomisation was done by an independent statistician using codes in a permuted block schedule with a block size of four. Participants, investigators, and study personnel were masked to treatment allocation. A single, oral dose regimen of cabamiquine (200, 100, 80, 60, or 30 mg) or matching placebo was administered either at 2 h (early liver-stage) or 96 h (late liver-stage) after DVI. The primary endpoints based on a per-protocol analysis set were the number of participants who developed parasitaemia within 28 days of DVI, time to parasitaemia, number of participants with documented parasite blood-stage growth, clinical symptoms of malaria, and exposure-efficacy modelling. The impact of cabamiquine on liver stages was evaluated indirectly by the appearance of parasitaemia in the blood. The Clopper-Pearson CI (nominal 95%) was used to express the protection rate. The secondary outcomes were safety and tolerability, assessed in those who had received DVI and were administered one dose of the study intervention. The trial was prospectively registered on ClinicalTrials.gov (NCT04250363). FINDINGS Between Feb 17, 2020 and April 29, 2021, 39 healthy participants were enrolled (early liver-stage: 30 mg [n=3], 60 mg [n=6], 80 mg [n=6], 100 mg [n=3], 200 mg [n=3], pooled placebo [n=6]; late liver-stage: 60 mg [n=3], 100 mg [n=3], 200 mg [n=3], pooled placebo [n=3]). A dose-dependent causal chemoprophylactic effect was observed, with four (67%) of six participants in the 60 mg, five (83%) of six participants in the 80 mg, and all three participants in the 100 and 200 mg cabamiquine dose groups protected from parasitaemia up to study day 28, whereas all participants in the pooled placebo and 30 mg cabamiquine dose group developed parasitaemia. A single, oral dose of 100 mg cabamiquine or higher provided 100% protection against parasitaemia when administered during early or late liver-stage malaria. The median time to parasitaemia in those with early liver-stage malaria was prolonged to 15, 22, and 24 days for the 30, 60, and 80 mg dose of cabamiquine, respectively, compared with 10 days for the pooled placebo. All participants with positive parasitaemia showed documented blood-stage parasite growth, apart from one participant in the pooled placebo group and one participant in the 30 mg cabamiquine group. Most participants did not exhibit any malaria symptoms in both the early and late liver-stage groups, and those reported were mild in severity. A positive dose-exposure-efficacy relationship was established across exposure metrics. The median maximum concentration time was 1-6 h, with a secondary peak observed between 6 h and 12 h in all cabamiquine dose groups (early liver-stage). All cabamiquine doses were safe and well tolerated. Overall, 26 (96%) of 27 participants in the early liver-stage group and ten (83·3%) of 12 participants in the late liver-stage group reported at least one treatment-emergent adverse event (TEAE) with cabamiquine or placebo. Most TEAEs were of mild severity, transient, and resolved without sequelae. The most frequently reported cabamiquine-related TEAE was headache. No dose-related trends were observed in the incidence, severity, or causality of TEAEs. INTERPRETATION The results from this study show that cabamiquine has a dose-dependent causal chemoprophylactic activity. Together with previously demonstrated activity against the blood stages combined with a half-life of more than 150 h, these results indicate that cabamiquine could be developed as a single-dose monthly regimen for malaria prevention. FUNDING The healthcare business of Merck KGaA, Darmstadt, Germany.
Collapse
Affiliation(s)
- Johan L van der Plas
- Centre for Human Drug Research, Leiden, Netherlands; Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Vincent P Kuiper
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Wilhelmina M Bagchus
- Merck Institute for Pharmacometrics, Merck Serono (an affiliate of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| | | | | | - Aliona Tappert
- The healthcare business of Merck KGaA, Darmstadt, Germany
| | | | - Thomas Spangenberg
- Global Health Institute of Merck, Ares Trading (a subsidiary of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Deon Bezuidenhout
- Merck (Pty) (an affiliate of Merck KGaA, Darmstadt, Germany), Modderfontein, South Africa
| | | | - Claude Oeuvray
- Global Health Institute of Merck, Ares Trading (a subsidiary of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | | | - Vandana Thathy
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Fidock
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Geert V T Roozen
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Jan Pieter R Koopman
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Olivia A C Lamers
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Jeroen Sijtsma
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Roos van Schuijlenburg
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Els Wessels
- Department of Medical Microbiology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Pauline Meij
- Center for Cell and Gene Therapy, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Ingrid M C Kamerling
- Centre for Human Drug Research, Leiden, Netherlands; Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Meta Roestenberg
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.
| | | |
Collapse
|
8
|
Jiménez-Díaz MB, Möhrle JJ, Angulo-Barturen I, Demarta-Gatsi C. Using Cryopreserved Plasmodium falciparum Sporozoites in a Humanized Mouse Model to Study Early Malaria Infection Processes and Test Prophylactic Treatments. Microorganisms 2023; 11:2209. [PMID: 37764054 PMCID: PMC10536749 DOI: 10.3390/microorganisms11092209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 09/29/2023] Open
Abstract
In addition to vector control, long-lasting insecticidal nets and case management, the prevention of infection through vaccination and/or chemoprevention are playing an increasing role in the drive to eradicate malaria. These preventative approaches represent opportunities for improvement: new drugs may be discovered that target the early infectious stages of the Plasmodium parasite in the liver (rather than the symptomatic, abundant blood stage), and new, exciting vaccination technologies have recently been validated (using mRNA or novel adjuvants). Exploiting these possibilities requires the availability of humanized mouse models that support P. falciparum infection yet avoid the hazardous use of infectious mosquitoes. Here, we show that commercially available P. falciparum sporozoites and FRG mice carrying human hepatocytes and red blood cells faithfully recapitulate the early human malaria disease process, presenting an opportunity to use this model for the evaluation of prophylactic treatments with a novel mode of action.
Collapse
|
9
|
You H, Jones MK, Gordon CA, Arganda AE, Cai P, Al-Wassiti H, Pouton CW, McManus DP. The mRNA Vaccine Technology Era and the Future Control of Parasitic Infections. Clin Microbiol Rev 2023; 36:e0024121. [PMID: 36625671 PMCID: PMC10035331 DOI: 10.1128/cmr.00241-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Despite intensive long-term efforts, with very few exceptions, the development of effective vaccines against parasitic infections has presented considerable challenges, given the complexity of parasite life cycles, the interplay between parasites and their hosts, and their capacity to escape the host immune system and to regulate host immune responses. For many parasitic diseases, conventional vaccine platforms have generally proven ill suited, considering the complex manufacturing processes involved and the costs they incur, the inability to posttranslationally modify cloned target antigens, and the absence of long-lasting protective immunity induced by these antigens. An effective antiparasite vaccine platform is required to assess the effectiveness of novel vaccine candidates at high throughput. By exploiting the approach that has recently been used successfully to produce highly protective COVID mRNA vaccines, we anticipate a new wave of research to advance the use of mRNA vaccines to prevent parasitic infections in the near future. This article considers the characteristics that are required to develop a potent antiparasite vaccine and provides a conceptual foundation to promote the development of parasite mRNA-based vaccines. We review the recent advances and challenges encountered in developing antiparasite vaccines and evaluate the potential of developing mRNA vaccines against parasites, including those causing diseases such as malaria and schistosomiasis, against which vaccines are currently suboptimal or not yet available.
Collapse
Affiliation(s)
- Hong You
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Malcolm K. Jones
- School of Veterinary Science, The University of Queensland, Brisbane, Australia
| | - Catherine A. Gordon
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Alexa E. Arganda
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Pengfei Cai
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Harry Al-Wassiti
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Colin W. Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Donald P. McManus
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
10
|
Patterns of Heterochromatin Transitions Linked to Changes in the Expression of Plasmodium falciparum Clonally Variant Genes. Microbiol Spectr 2023; 11:e0304922. [PMID: 36515553 PMCID: PMC9927496 DOI: 10.1128/spectrum.03049-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The survival of malaria parasites in the changing human blood environment largely depends on their ability to alter gene expression by epigenetic mechanisms. The active state of Plasmodium falciparum clonally variant genes (CVGs) is associated with euchromatin characterized by the histone mark H3K9ac, whereas the silenced state is characterized by H3K9me3-based heterochromatin. Expression switches are linked to euchromatin-heterochromatin transitions, but these transitions have not been characterized for the majority of CVGs. To define the heterochromatin distribution patterns associated with the alternative transcriptional states of CVGs, we compared H3K9me3 occupancy at a genome-wide level among several parasite subclones of the same genetic background that differed in the transcriptional state of many CVGs. We found that de novo heterochromatin formation or the complete disruption of a heterochromatin domain is a relatively rare event, and for the majority of CVGs, expression switches can be explained by the expansion or retraction of heterochromatin domains. We identified different modalities of heterochromatin changes linked to transcriptional differences, but despite this complexity, heterochromatin distribution patterns generally enable the prediction of the transcriptional state of specific CVGs. We also found that in some subclones, several var genes were simultaneously in an active state. Furthermore, the heterochromatin levels in the putative regulatory region of the gdv1 antisense noncoding RNA, a regulator of sexual commitment, varied between parasite lines with different sexual conversion rates. IMPORTANCE The malaria parasite P. falciparum is responsible for more than half a million deaths every year. P. falciparum clonally variant genes (CVGs) mediate fundamental host-parasite interactions and play a key role in parasite adaptation to fluctuations in the conditions of the human host. The expression of CVGs is regulated at the epigenetic level by changes in the distribution of a type of chromatin called heterochromatin. Here, we describe at a genome-wide level the changes in the heterochromatin distribution associated with the different transcriptional states of CVGs. Our results also reveal a likely role for heterochromatin at a particular locus in determining the parasite investment in transmission to mosquitoes. Additionally, this data set will enable the prediction of the transcriptional state of CVGs from epigenomic data, which is important for the study of parasite adaptation to the conditions of the host in natural malaria infections.
Collapse
|
11
|
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: 4] [Impact Index Per Article: 4.0] [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
|
12
|
Cryopreservation of Plasmodium Sporozoites. Pathogens 2022; 11:pathogens11121487. [PMID: 36558821 PMCID: PMC9784981 DOI: 10.3390/pathogens11121487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Malaria is a deadly disease caused by the parasite, Plasmodium, and impacts the lives of millions of people around the world. Following inoculation into mammalian hosts by infected mosquitoes, the sporozoite stage of Plasmodium undergoes obligate development in the liver before infecting erythrocytes and causing clinical malaria. The most promising vaccine candidates for malaria rely on the use of attenuated live sporozoites to induce protective immune responses. The scope of widespread testing or clinical use of such vaccines is limited by the absence of efficient, reliable, or transparent strategies for the long-term preservation of live sporozoites. Here we outline a method to cryopreserve the sporozoites of various human and murine Plasmodium species. We found that the structural integrity, viability, and in vivo or in vitro infectiousness were conserved in the recovered cryopreserved sporozoites. Cryopreservation using our approach also retained the transgenic properties of sporozoites and immunization with cryopreserved radiation attenuated sporozoites (RAS) elicited strong immune responses. Our work offers a reliable protocol for the long-term storage and recovery of human and murine Plasmodium sporozoites and lays the groundwork for the widespread use of live sporozoites for research and clinical applications.
Collapse
|
13
|
In vitro production of infectious Plasmodium falciparum sporozoites. Nature 2022; 612:534-539. [PMID: 36477528 DOI: 10.1038/s41586-022-05466-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 10/20/2022] [Indexed: 12/12/2022]
Abstract
An effective vaccine is needed for the prevention and elimination of malaria. The only immunogens that have been shown to have a protective efficacy of more than 90% against human malaria are Plasmodium falciparum (Pf) sporozoites (PfSPZ) manufactured in mosquitoes (mPfSPZ)1-7. The ability to produce PfSPZ in vitro (iPfSPZ) without mosquitoes would substantially enhance the production of PfSPZ vaccines and mosquito-stage malaria research, but this ability is lacking. Here we report the production of hundreds of millions of iPfSPZ. iPfSPZ invaded human hepatocytes in culture and developed to mature liver-stage schizonts expressing P. falciparum merozoite surface protein 1 (PfMSP1) in numbers comparable to mPfSPZ. When injected into FRGhuHep mice containing humanized livers, iPfSPZ invaded the human hepatocytes and developed to PfMSP1-expressing late liver stage parasites at 45% the quantity of cryopreserved mPfSPZ. Human blood from FRGhuHep mice infected with iPfSPZ produced asexual and sexual erythrocytic-stage parasites in culture, and gametocytes developed to PfSPZ when fed to mosquitoes, completing the P. falciparum life cycle from infectious gametocyte to infectious gametocyte without mosquitoes or primates.
Collapse
|
14
|
Chakravarty S, Shears MJ, James ER, Rai U, Kc N, Conteh S, Lambert LE, Duffy PE, Murphy SC, Hoffman SL. Efficient infection of non-human primates with purified, cryopreserved Plasmodium knowlesi sporozoites. Malar J 2022; 21:247. [PMID: 36030292 PMCID: PMC9418655 DOI: 10.1186/s12936-022-04261-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/17/2022] [Indexed: 11/29/2022] Open
Abstract
Background Plasmodium falciparum (Pf) sporozoite (SPZ) vaccines are the only candidate malaria vaccines that induce > 90% vaccine efficacy (VE) against controlled human malaria infection and the only malaria vaccines to have achieved reproducible VE against malaria in adults in Africa. The goal is to increase the impact and reduce the cost of PfSPZ vaccines by optimizing vaccine potency and manufacturing, which will benefit from identification of immunological responses contributing to protection in humans. Currently, there is no authentic animal challenge model for assessing P. falciparum malaria VE. Alternatively, Plasmodium knowlesi (Pk), which infects humans and non-human primates (NHPs) in nature, can be used to experimentally infect rhesus macaques (Macaca mulatta) to assess VE. Methods Sanaria has, therefore, produced purified, vialed, cryopreserved PkSPZ and conducted challenge studies in several naïve NHP cohorts. In the first cohort, groups of three rhesus macaques each received doses of 5 × 102, 2.5 × 103, 1.25 × 104 and 2.5 × 104 PkSPZ administered by direct venous inoculation. The infectivity of 1.5 × 103 PkSPZ cryopreserved with an altered method and of 1.5 × 103 PkSPZ cryopreserved for four years was tested in a second and third cohort of rhesus NHPs. The lastly, three pig-tailed macaques (Macaca nemestrina), a natural P. knowlesi host, were challenged with 2.5 × 103 PkSPZ cryopreserved six years earlier. Results In the first cohort, all 12 animals developed P. knowlesi parasitaemia by thick blood smear, and the time to positivity (prepatent period) followed a non-linear 4-parameter logistic sigmoidal model with a median of 11, 10, 8, and 7 days, respectively (r2 = 1). PkSPZ cryopreserved using a modified rapid-scalable method infected rhesus with a pre-patent period of 10 days, as did PkSPZ cryopreserved four years prior to infection, similar to the control group. Cryopreserved PkSPZ infected pig-tailed macaques with median time to positivity by thin smear, of 11 days. Conclusion This study establishes the capacity to consistently infect NHPs with purified, vialed, cryopreserved PkSPZ, providing a foundation for future studies to probe protective immunological mechanisms elicited by PfSPZ vaccines that cannot be established in humans. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04261-z.
Collapse
Affiliation(s)
- Sumana Chakravarty
- Sanaria, Inc, 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Melanie J Shears
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.,Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | - Eric R James
- Sanaria, Inc, 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Urvashi Rai
- Sanaria, Inc, 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Natasha Kc
- Sanaria, Inc, 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Solomon Conteh
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, USA
| | - Lynn E Lambert
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, USA
| | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.,Washington National Primate Research Center, University of Washington, Seattle, WA, USA.,Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Stephen L Hoffman
- Sanaria, Inc, 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA.
| |
Collapse
|
15
|
A PfSPZ vaccine immunization regimen equally protective against homologous and heterologous controlled human malaria infection. NPJ Vaccines 2022; 7:100. [PMID: 35999221 PMCID: PMC9396563 DOI: 10.1038/s41541-022-00510-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 06/24/2022] [Indexed: 11/08/2022] Open
Abstract
Immunization with radiation-attenuated Plasmodium falciparum (Pf) sporozoites (SPZ) in PfSPZ Vaccine, has provided better vaccine efficacy (VE) against controlled human malaria infection (CHMI) with the same parasites as in the vaccine (homologous) than with genetically distant parasites (heterologous). We sought to identify an immunization regimen that provided similar VE against CHMI with homologous and heterologous Pf for at least 9 weeks in malaria-naïve adults. Such a regimen was identified in part 1 (optimization), an open label study, and confirmed in part 2 (verification), a randomized, double-blind, placebo-controlled study in which VE was assessed by cross-over repeat CHMI with homologous (PfNF54) and heterologous (Pf7G8) PfSPZ at 3 and 9–10 weeks. VE was calculated using Bayesian generalized linear regression. In part 1, vaccination with 9 × 105 PfSPZ on days 1, 8, and 29 protected 5/5 (100%) subjects against homologous CHMI at 3 weeks after the last immunization. In part 2, the same 3-dose regimen protected 5/6 subjects (83%) against heterologous CHMI at both 3 and 9–10 weeks after the last immunization. Overall VE was 78% (95% predictive interval: 57–92%), and against heterologous and homologous was 79% (95% PI: 54–95%) and 77% (95% PI: 50–95%) respectively. PfSPZ Vaccine was safe and well tolerated. A 4-week, 3-dose regimen of PfSPZ Vaccine provided similar VE for 9–10 weeks against homologous and heterologous CHMI. The trial is registered with ClinicalTrials.gov, NCT02704533.
Collapse
|
16
|
Mpina M, Stabler TC, Schindler T, Raso J, Deal A, Acuche Pupu L, Nyakarungu E, Del Carmen Ovono Davis M, Urbano V, Mtoro A, Hamad A, Lopez MSA, Pasialo B, Eyang MAO, Rivas MR, Falla CC, García GA, Momo JC, Chuquiyauri R, Saverino E, Preston Church LW, Kim Lee Sim B, Manguire B, Tanner M, Maas C, Abdulla S, Billingsley PF, Hoffman SL, Jongo S, Richie TL, Daubenberger CA. Diagnostic performance and comparison of ultrasensitive and conventional rapid diagnostic test, thick blood smear and quantitative PCR for detection of low-density Plasmodium falciparum infections during a controlled human malaria infection study in Equatorial Guinea. Malar J 2022; 21:99. [PMID: 35331251 PMCID: PMC8943516 DOI: 10.1186/s12936-022-04103-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/23/2022] [Indexed: 12/02/2022] Open
Abstract
Background Progress towards malaria elimination has stagnated, partly because infections persisting at low parasite densities comprise a large reservoir contributing to ongoing malaria transmission and are difficult to detect. This study compared the performance of an ultrasensitive rapid diagnostic test (uRDT) designed to detect low density infections to a conventional RDT (cRDT), expert microscopy using Giemsa-stained thick blood smears (TBS), and quantitative polymerase chain reaction (qPCR) during a controlled human malaria infection (CHMI) study conducted in malaria exposed adults (NCT03590340). Methods Blood samples were collected from healthy Equatoguineans aged 18–35 years beginning on day 8 after CHMI with 3.2 × 103 cryopreserved, infectious Plasmodium falciparum sporozoites (PfSPZ Challenge, strain NF54) administered by direct venous inoculation. qPCR (18s ribosomal DNA), uRDT (Alere™ Malaria Ag P.f.), cRDT [Carestart Malaria Pf/PAN (PfHRP2/pLDH)], and TBS were performed daily until the volunteer became TBS positive and treatment was administered. qPCR was the reference for the presence of Plasmodium falciparum parasites. Results 279 samples were collected from 24 participants; 123 were positive by qPCR. TBS detected 24/123 (19.5% sensitivity [95% CI 13.1–27.8%]), uRDT 21/123 (17.1% sensitivity [95% CI 11.1–25.1%]), cRDT 10/123 (8.1% sensitivity [95% CI 4.2–14.8%]); all were 100% specific and did not detect any positive samples not detected by qPCR. TBS and uRDT were more sensitive than cRDT (TBS vs. cRDT p = 0.015; uRDT vs. cRDT p = 0.053), detecting parasitaemias as low as 3.7 parasites/µL (p/µL) (TBS and uRDT) compared to 5.6 p/µL (cRDT) based on TBS density measurements. TBS, uRDT and cRDT did not detect any of the 70/123 samples positive by qPCR below 5.86 p/µL, the qPCR density corresponding to 3.7 p/µL by TBS. The median prepatent periods in days (ranges) were 14.5 (10–20), 18.0 (15–28), 18.0 (15–20) and 18.0 (16–24) for qPCR, TBS, uRDT and cRDT, respectively; qPCR detected parasitaemia significantly earlier (3.5 days) than the other tests. Conclusions TBS and uRDT had similar sensitivities, both were more sensitive than cRDT, and neither matched qPCR for detecting low density parasitaemia. uRDT could be considered an alternative to TBS in selected applications, such as CHMI or field diagnosis, where qualitative, dichotomous results for malaria infection might be sufficient. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04103-y.
Collapse
Affiliation(s)
- Maxmillian Mpina
- Swiss Tropical and Public Health Institute, Basel, Switzerland. .,University of Basel, Basel, Switzerland. .,Ifakara Health Institute, Ifakara, Tanzania.
| | - Thomas C Stabler
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Tobias Schindler
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Jose Raso
- Medical Care Development International, Malabo, Equatorial Guinea.,Equatorial Guinea Ministry of Health and Social Welfare, Malabo, Equatorial Guinea
| | - Anna Deal
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | | | - Elizabeth Nyakarungu
- Ifakara Health Institute, Ifakara, Tanzania.,Medical Care Development International, Malabo, Equatorial Guinea
| | | | - Vicente Urbano
- Medical Care Development International, Malabo, Equatorial Guinea.,Equatorial Guinea Ministry of Health and Social Welfare, Malabo, Equatorial Guinea
| | - Ali Mtoro
- Ifakara Health Institute, Ifakara, Tanzania.,Medical Care Development International, Malabo, Equatorial Guinea
| | - Ali Hamad
- Ifakara Health Institute, Ifakara, Tanzania.,Medical Care Development International, Malabo, Equatorial Guinea
| | - Maria Silvia A Lopez
- Medical Care Development International, Malabo, Equatorial Guinea.,Equatorial Guinea Ministry of Health and Social Welfare, Malabo, Equatorial Guinea
| | - Beltran Pasialo
- Medical Care Development International, Malabo, Equatorial Guinea.,Equatorial Guinea Ministry of Health and Social Welfare, Malabo, Equatorial Guinea
| | - Marta Alene Owono Eyang
- Medical Care Development International, Malabo, Equatorial Guinea.,Equatorial Guinea Ministry of Health and Social Welfare, Malabo, Equatorial Guinea
| | - Matilde Riloha Rivas
- Equatorial Guinea Ministry of Health and Social Welfare, Malabo, Equatorial Guinea
| | | | | | - Juan Carlos Momo
- Medical Care Development International, Malabo, Equatorial Guinea.,Equatorial Guinea Ministry of Health and Social Welfare, Malabo, Equatorial Guinea
| | - Raul Chuquiyauri
- Medical Care Development International, Malabo, Equatorial Guinea.,Sanaria Inc., 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | | | | | - B Kim Lee Sim
- Sanaria Inc., 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | | | - Marcel Tanner
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Carl Maas
- Marathon EG production Ltd., Houston, USA
| | | | | | | | - Said Jongo
- Ifakara Health Institute, Ifakara, Tanzania.,Medical Care Development International, Malabo, Equatorial Guinea
| | - Thomas L Richie
- Sanaria Inc., 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Claudia A Daubenberger
- Swiss Tropical and Public Health Institute, Basel, Switzerland. .,University of Basel, Basel, Switzerland.
| |
Collapse
|
17
|
Sissoko MS, Healy SA, Katile A, Zaidi I, Hu Z, Kamate B, Samake Y, Sissoko K, Mwakingwe-Omari A, Lane J, Imeru A, Mohan R, Thera I, Guindo CO, Dolo A, Niare K, Koïta F, Niangaly A, Rausch KM, Zeguime A, Guindo MA, Bah A, Abebe Y, James ER, Manoj A, Murshedkar T, Kc N, Sim BKL, Billingsley PF, Richie TL, Hoffman SL, Doumbo O, Duffy PE. Safety and efficacy of a three-dose regimen of Plasmodium falciparum sporozoite vaccine in adults during an intense malaria transmission season in Mali: a randomised, controlled phase 1 trial. THE LANCET. INFECTIOUS DISEASES 2022; 22:377-389. [PMID: 34801112 PMCID: PMC8981424 DOI: 10.1016/s1473-3099(21)00332-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/28/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND WHO recently approved a partially effective vaccine that reduces clinical malaria in children, but increased vaccine activity is required to pursue malaria elimination. A phase 1 clinical trial was done in Mali, west Africa, to assess the safety, immunogenicity, and protective efficacy of a three-dose regimen of Plasmodium falciparum sporozoite (PfSPZ) Vaccine (a metabolically active, non-replicating, whole malaria sporozoite vaccine) against homologous controlled human malaria infection (CHMI) and natural P falciparum infection. METHODS We recruited healthy non-pregnant adults aged 18-50 years in Donéguébougou, Mali, and surrounding villages (Banambani, Toubana, Torodo, Sirababougou, Zorokoro) for an open-label, dose-escalation pilot study and, thereafter, a randomised, double-blind, placebo-controlled main trial. Pilot study participants were enrolled on an as-available basis to one group of CHMI infectivity controls and three staggered vaccine groups receiving: one dose of 4·5 × 105, one dose of 9 × 105, or three doses of 1·8 × 106 PfSPZ via direct venous inoculation at approximately 8 week intervals, followed by homologous CHMI 5 weeks later with infectious PfSPZ by direct venous inoculation (PfSPZ Challenge). Main cohort participants were stratified by village and randomly assigned (1:1) to receive three doses of 1·8 × 106 PfSPZ or normal saline at 1, 13, and 19 week intervals using permuted block design by the study statistician. The primary outcome was safety and tolerability of at least one vaccine dose; the secondary outcome was vaccine efficacy against homologous PfSPZ CHMI (pilot study) or against naturally transmitted P falciparum infection (main study) measured by thick blood smear. Combined artesunate and amodiaquine was administered to eliminate pre-existing parasitaemia. Outcomes were analysed by modified intention to treat (mITT; including all participants who received at least one dose of investigational product; safety and vaccine efficacy) and per protocol (vaccine efficacy). This trial is registered with ClinicalTrials.gov, number NCT02627456. FINDINGS Between Dec 20, 2015, and April 30, 2016, we enrolled 56 participants into the pilot study (five received the 4·5 × 105 dose, five received 9 × 105, 30 received 1·8 × 106, 15 were CHMI controls, and one withdrew before vaccination) and 120 participants into the main study cohort with 60 participants assigned PfSPZ Vaccine and 60 placebo in the main study. Adverse events and laboratory abnormalities post-vaccination in all dosing groups were few, mainly mild, and did not differ significantly between vaccine groups (all p>0·05). Unexpected severe transaminitis occured in four participants: one participant in pilot phase that received 1·8 × 106 PfSPZ Vaccine, one participant in main phase that received 1·8 × 106 PfSPZ Vaccine, and two participants in the main phase placebo group. During PfSPZ CHMI, approximately 5 weeks after the third dose of 1·8 × 106 PfSPZ, none of 29 vaccinees and one of 15 controls became positive on thick blood smear; subsequent post-hoc PCR analysis for submicroscopic blood stage infections detected P falciparum parasites in none of the 29 vaccine recipients and eight of 15 controls during CHMI. In the main trial, 32 (58%) of 55 vaccine recipients and 42 (78%) of 54 controls became positive on thick blood smear during 24-week surveillance after vaccination. Vaccine efficacy (1-hazard ratio) was 0·51 per protocol (95% CI 0·20-0·70; log-rank p=0·0042) and 0·39 by mITT (0·04-0·62; p=0·033); vaccine efficacy (1-risk ratio) was 0·24 per-protocol (0·02-0·41; p=0·031) and 0·22 mITT (0·01-0·39; p=0·041). INTERPRETATION A three-dose regimen of PfSPZ Vaccine was safe, well tolerated, and conferred 51% vaccine efficacy against intense natural P falciparum transmission, similar to 52% vaccine efficacy reported for a five-dose regimen in a previous trial. FUNDING US National Institute of Allergy and Infectious Diseases, National Institutes of Health, Sanaria. TRANSLATION For the French translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Mahamadou S Sissoko
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Abdoulaye Katile
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Irfan Zaidi
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zonghui Hu
- Biostatistical Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bourama Kamate
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Yacouba Samake
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Kourane Sissoko
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Agnes Mwakingwe-Omari
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Center for Vaccine Research, GlaxoSmithKline, Rockville, MD, USA
| | - Jacquelyn Lane
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alemush Imeru
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rathy Mohan
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ismaila Thera
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Cheick Oumar Guindo
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Amagana Dolo
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Karamoko Niare
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Fanta Koïta
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Amadou Niangaly
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Kelly M Rausch
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Amatigue Zeguime
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Merepen A Guindo
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Aissatou Bah
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | - Natasha Kc
- Sanaria, Rockville, MD, USA; Protein Potential, Rockville, MD, USA
| | - B Kim Lee Sim
- Sanaria, Rockville, MD, USA; Protein Potential, Rockville, MD, USA
| | | | | | | | - Ogobara Doumbo
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Mali
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
18
|
Shibeshi W, Bagchus W, Yalkinoglu Ö, Tappert A, Engidawork E, Oeuvray C. Reproducibility of malaria sporozoite challenge model in humans for evaluating efficacy of vaccines and drugs: a systematic review. BMC Infect Dis 2021; 21:1274. [PMID: 34930178 PMCID: PMC8686662 DOI: 10.1186/s12879-021-06953-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The development of novel malaria vaccines and antimalarial drugs is limited partly by emerging challenges to conduct field trials in malaria endemic areas, including unknown effects of existing immunity and a reported fall in malaria incidence. As a result, Controlled Human Malaria Infection (CHMI) has become an important approach for accelerated development of malarial vaccines and drugs. We conducted a systematic review of the literature to establish aggregate evidence on the reproducibility of a malaria sporozoite challenge model. METHODS A systematic review of research articles published between 1990 and 2018 on efficacy testing of malaria vaccines and drugs using sporozoite challenge and sporozoite infectivity studies was conducted using Pubmed, Scopus, Embase and Cochrane Library, ClinicalTrials.gov and Trialtrove. The inclusion criteria were randomized and non-randomized, controlled or open-label trials using P. falciparum or P. vivax sporozoite challenges. The data were extracted from articles using standardized data extraction forms and descriptive analysis was performed for evidence synthesis. The endpoints considered were infectivity, prepatent period, parasitemia and safety of sporozoite challenge. RESULTS Seventy CHMI trials conducted with a total of 2329 adult healthy volunteers were used for analysis. CHMI was induced by bites of mosquitoes infected with P. falciparum or P. vivax in 52 trials and by direct venous inoculation of P. falciparum sporozoites (PfSPZ challenge) in 18 trials. Inoculation with P. falciparum-infected mosquitoes produced 100% infectivity in 40 studies and the mean/median prepatent period assessed by thick blood smear (TBS) microscopy was ≤ 12 days in 24 studies. On the other hand, out of 12 infectivity studies conducted using PfSPZ challenge, 100% infection rate was reproduced in 9 studies with a mean or median prepatent period of 11 to 15.3 days as assessed by TBS and 6.8 to 12.6 days by PCR. The safety profile of P. falciparum and P.vivax CHMI was characterized by consistent features of malaria infection. CONCLUSION There is ample evidence on consistency of P. falciparum CHMI models in terms of infectivity and safety endpoints, which supports applicability of CHMI in vaccine and drug development. PfSPZ challenge appears more feasible for African trials based on current evidence of safety and efficacy.
Collapse
Affiliation(s)
- Workineh Shibeshi
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia.
- Global Health Institute of Merck, Ares Trading S.A., A subsidiary of Merck KGaA, Darmstadt, Germany.
| | - Wilhelmina Bagchus
- Translational Medicine, Merck Serono S.A., An Affiliate of Merck KGaA, Darmstadt, Germany
| | - Özkan Yalkinoglu
- Translational Medicine, Merck Healthcare KGaA, Darmstadt, Germany
| | - Aliona Tappert
- Global Patient Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Ephrem Engidawork
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Claude Oeuvray
- Global Health Institute of Merck, Ares Trading S.A., A subsidiary of Merck KGaA, Darmstadt, Germany
| |
Collapse
|
19
|
Kapulu MC, Njuguna P, Hamaluba M, Kimani D, Ngoi JM, Musembi J, Ngoto O, Otieno E, Billingsley PF. Safety and PCR monitoring in 161 semi-immune Kenyan adults following controlled human malaria infection. JCI Insight 2021; 6:e146443. [PMID: 34264864 PMCID: PMC8492329 DOI: 10.1172/jci.insight.146443] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 07/14/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUNDNaturally acquired immunity to malaria is incompletely understood. We used controlled human malaria infection (CHMI) to study the impact of past exposure on malaria in Kenyan adults in relation to infection with a non-Kenyan parasite strain.METHODSWe administered 3.2 × 103 aseptic, purified, cryopreserved Plasmodium falciparum sporozoites (Sanaria PfSPZ Challenge, NF54 West African strain) by direct venous inoculation and undertook clinical monitoring and serial quantitative PCR (qPCR) of the 18S ribosomal RNA gene. The study endpoint was met when parasitemia reached 500 or more parasites per μL blood, clinically important symptoms were seen, or at 21 days after inoculation. All volunteers received antimalarial drug treatment upon meeting the endpoint.RESULTSOne hundred and sixty-one volunteers underwent CHMI between August 4, 2016, and February 14, 2018. CHMI was well tolerated, with no severe or serious adverse events. Nineteen volunteers (11.8%) were excluded from the analysis based on detection of antimalarial drugs above the minimal inhibitory concentration or parasites genotyped as non-NF54. Of the 142 volunteers who were eligible for analysis, 26 (18.3%) had febrile symptoms and were treated; 30 (21.1%) reached 500 or more parasites per μL and were treated; 53 (37.3%) had parasitemia without meeting thresholds for treatment; and 33 (23.2%) remained qPCR negative.CONCLUSIONWe found that past exposure to malaria, as evidenced by location of residence, in some Kenyan adults can completely suppress in vivo growth of a parasite strain originating from outside Kenya.TRIAL REGISTRATIONClinicalTrials.gov NCT02739763.FUNDINGWellcome Trust.
Collapse
Affiliation(s)
- Melissa C. Kapulu
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Patricia Njuguna
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Mainga Hamaluba
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Domtila Kimani
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Joyce M. Ngoi
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Janet Musembi
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Omar Ngoto
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Edward Otieno
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | | |
Collapse
|
20
|
Mwakingwe-Omari A, Healy SA, Lane J, Cook DM, Kalhori S, Wyatt C, Kolluri A, Marte-Salcedo O, Imeru A, Nason M, Ding LK, Decederfelt H, Duan J, Neal J, Raiten J, Lee G, Hume JCC, Jeon JE, Ikpeama I, Kc N, Chakravarty S, Murshedkar T, Church LWP, Manoj A, Gunasekera A, Anderson C, Murphy SC, March S, Bhatia SN, James ER, Billingsley PF, Sim BKL, Richie TL, Zaidi I, Hoffman SL, Duffy PE. Two chemoattenuated PfSPZ malaria vaccines induce sterile hepatic immunity. Nature 2021; 595:289-294. [PMID: 34194041 PMCID: PMC11127244 DOI: 10.1038/s41586-021-03684-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 06/01/2021] [Indexed: 02/06/2023]
Abstract
The global decline in malaria has stalled1, emphasizing the need for vaccines that induce durable sterilizing immunity. Here we optimized regimens for chemoprophylaxis vaccination (CVac), for which aseptic, purified, cryopreserved, infectious Plasmodium falciparum sporozoites (PfSPZ) were inoculated under prophylactic cover with pyrimethamine (PYR) (Sanaria PfSPZ-CVac(PYR)) or chloroquine (CQ) (PfSPZ-CVac(CQ))-which kill liver-stage and blood-stage parasites, respectively-and we assessed vaccine efficacy against homologous (that is, the same strain as the vaccine) and heterologous (a different strain) controlled human malaria infection (CHMI) three months after immunization ( https://clinicaltrials.gov/ , NCT02511054 and NCT03083847). We report that a fourfold increase in the dose of PfSPZ-CVac(PYR) from 5.12 × 104 to 2 × 105 PfSPZs transformed a minimal vaccine efficacy (low dose, two out of nine (22.2%) participants protected against homologous CHMI), to a high-level vaccine efficacy with seven out of eight (87.5%) individuals protected against homologous and seven out of nine (77.8%) protected against heterologous CHMI. Increased protection was associated with Vδ2 γδ T cell and antibody responses. At the higher dose, PfSPZ-CVac(CQ) protected six out of six (100%) participants against heterologous CHMI three months after immunization. All homologous (four out of four) and heterologous (eight out of eight) infectivity control participants showed parasitaemia. PfSPZ-CVac(CQ) and PfSPZ-CVac(PYR) induced a durable, sterile vaccine efficacy against a heterologous South American strain of P. falciparum, which has a genome and predicted CD8 T cell immunome that differs more strongly from the African vaccine strain than other analysed African P. falciparum strains.
Collapse
Affiliation(s)
- Agnes Mwakingwe-Omari
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Center for Vaccine Research, GlaxoSmithKline, Rockville, MD, USA
| | - Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jacquelyn Lane
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David M Cook
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sahand Kalhori
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Charles Wyatt
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aarti Kolluri
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Omely Marte-Salcedo
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alemush Imeru
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Martha Nason
- Biostatistical Research Branch, National Institute of Allergy and Infectious Diseases, Rockville, MD, USA
| | - Lei K Ding
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hope Decederfelt
- Clinical Center Pharmacy Department, National Institutes of Health, Bethesda, MD, USA
| | - Junhui Duan
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jillian Neal
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jacob Raiten
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Grace Lee
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jen C C Hume
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jihyun E Jeon
- Clinical Center Pharmacy Department, National Institutes of Health, Bethesda, MD, USA
| | - Ijeoma Ikpeama
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Natasha Kc
- Sanaria, Rockville, MD, USA
- Protein Potential, Rockville, MD, USA
| | | | | | | | | | | | - Charles Anderson
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Seattle Malaria Clinical Trials Center, Fred Hutch Cancer Research Center, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Broad Institute, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | - B Kim Lee Sim
- Sanaria, Rockville, MD, USA
- Protein Potential, Rockville, MD, USA
| | | | - Irfan Zaidi
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
21
|
Billingsley PF, George KI, Eappen AG, Harrell RA, Alford R, Li T, Chakravarty S, Sim BKL, Hoffman SL, O'Brochta DA. Transient knockdown of Anopheles stephensi LRIM1 using RNAi increases Plasmodium falciparum sporozoite salivary gland infections. Malar J 2021; 20:284. [PMID: 34174879 PMCID: PMC8235909 DOI: 10.1186/s12936-021-03818-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Plasmodium falciparum (Pf) sporozoites (PfSPZ) can be administered as a highly protective vaccine conferring the highest protection seen to date. Sanaria® PfSPZ vaccines are produced using aseptically reared Anopheles stephensi mosquitoes. The bionomics of sporogonic development of P. falciparum in A. stephensi to fully mature salivary gland PfSPZ is thought to be modulated by several components of the mosquito innate immune system. In order to increase salivary gland PfSPZ infections in A. stephensi and thereby increase vaccine production efficiency, a gene knock down approach was used to investigate the activity of the immune deficiency (IMD) signaling pathway downstream effector leucine-rich repeat immune molecule 1 (LRIM1), an antagonist to Plasmodium development. METHODS Expression of LRIM1 in A. stephensi was reduced following injection of double stranded (ds) RNA into mosquitoes. By combining the Gal4/UAS bipartite system with in vivo expression of short hairpin (sh) RNA coding for LRIM1 reduced expression of LRIM1 was targeted in the midgut, fat body, and salivary glands. RT-qPCR was used to demonstrate fold-changes in gene expression in three transgenic crosses and the effects on P. falciparum infections determined in mosquitoes showing the greatest reduction in LRIM1 expression. RESULTS LRIM1 expression could be reduced, but not completely silenced, by expression of LRIM1 dsRNA. Infections of P. falciparum oocysts and PfSPZ were consistently and significantly higher in transgenic mosquitoes than wild type controls, with increases in PfSPZ ranging from 2.5- to tenfold. CONCLUSIONS Plasmodium falciparum infections in A. stephensi can be increased following reduced expression of LRIM1. These data provide the springboard for more precise knockout of LRIM1 for the eventual incorporation of immune-compromised A. stephensi into manufacturing of Sanaria's PfSPZ products.
Collapse
Affiliation(s)
- Peter F Billingsley
- Sanaria Inc, Suite A209, 9800 Medical Center Drive, Rockville, MD, 20850, USA.
| | - Kasim I George
- Institute for Bioscience and Biotechnology Research and Department of Entomology, University of Maryland, Gudelsky Drive, Rockville, MD, 20850, USA
- Qiagen Inc, 19300 Germantown Road, Germantown, MD, 20874, USA
| | - Abraham G Eappen
- Sanaria Inc, Suite A209, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Robert A Harrell
- Institute for Bioscience and Biotechnology Research and Department of Entomology, University of Maryland, Gudelsky Drive, Rockville, MD, 20850, USA
- Insect Transformation Facility, Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Robert Alford
- Institute for Bioscience and Biotechnology Research and Department of Entomology, University of Maryland, Gudelsky Drive, Rockville, MD, 20850, USA
- Insect Transformation Facility, Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Tao Li
- Sanaria Inc, Suite A209, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Sumana Chakravarty
- Sanaria Inc, Suite A209, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - B Kim Lee Sim
- Sanaria Inc, Suite A209, 9800 Medical Center Drive, Rockville, MD, 20850, USA
- Protein Potential, Suite A209, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Stephen L Hoffman
- Sanaria Inc, Suite A209, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - David A O'Brochta
- Institute for Bioscience and Biotechnology Research and Department of Entomology, University of Maryland, Gudelsky Drive, Rockville, MD, 20850, USA
- Foundation for the National Institutes of Health, 11400 Rockville Pike, Suite 600, North Bethesda, MD, 20852, USA
| |
Collapse
|
22
|
Murphy SC, Deye GA, Sim BKL, Galbiati S, Kennedy JK, Cohen KW, Chakravarty S, KC N, Abebe Y, James ER, Kublin JG, Hoffman SL, Richie TL, Jackson LA. PfSPZ-CVac efficacy against malaria increases from 0% to 75% when administered in the absence of erythrocyte stage parasitemia: A randomized, placebo-controlled trial with controlled human malaria infection. PLoS Pathog 2021; 17:e1009594. [PMID: 34048504 PMCID: PMC8191919 DOI: 10.1371/journal.ppat.1009594] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/10/2021] [Accepted: 04/29/2021] [Indexed: 11/18/2022] Open
Abstract
PfSPZ-CVac combines 'PfSPZ Challenge', which consists of infectious Plasmodium falciparum sporozoites (PfSPZ), with concurrent antimalarial chemoprophylaxis. In a previously-published PfSPZ-CVac study, three doses of 5.12x104 PfSPZ-CVac given 28 days apart had 100% vaccine efficacy (VE) against controlled human malaria infection (CHMI) 10 weeks after the last immunization, while the same dose given as three injections five days apart had 63% VE. Here, we conducted a dose escalation trial of similarly condensed schedules. Of the groups proceeding to CHMI, the first study group received three direct venous inoculations (DVIs) of a dose of 5.12x104 PfSPZ-CVac seven days apart and the next full dose group received three DVIs of a higher dose of 1.024x105 PfSPZ-CVac five days apart. CHMI (3.2x103 PfSPZ Challenge) was performed by DVI 10 weeks after the last vaccination. In both CHMI groups, transient parasitemia occurred starting seven days after each vaccination. For the seven-day interval group, the second and third vaccinations were therefore administered coincident with parasitemia from the prior vaccination. Parasitemia was associated with systemic symptoms which were severe in 25% of subjects. VE in the seven-day group was 0% (7/7 infected) and in the higher-dose, five-day group was 75% (2/8 infected). Thus, the same dose of PfSPZ-CVac previously associated with 63% VE when given on a five-day schedule in the prior study had zero VE here when given on a seven-day schedule, while a double dose given on a five-day schedule here achieved 75% VE. The relative contributions of the five-day schedule and/or the higher dose to improved VE warrant further investigation. It is notable that administration of PfSPZ-CVac on a schedule where vaccine administration coincided with blood-stage parasitemia was associated with an absence of sterile protective immunity. Clinical trials registration: NCT02773979.
Collapse
Affiliation(s)
- Sean C. Murphy
- Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, Seattle, Washington, United States of America
| | - Gregory A. Deye
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - B. Kim Lee Sim
- Sanaria Inc., Rockville, Maryland, United States of America
| | - Shirley Galbiati
- The Emmes Company, Rockville, Maryland, United States of America
| | | | - Kristen W. Cohen
- Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | | | - Natasha KC
- Sanaria Inc., Rockville, Maryland, United States of America
| | - Yonas Abebe
- Sanaria Inc., Rockville, Maryland, United States of America
| | - Eric R. James
- Sanaria Inc., Rockville, Maryland, United States of America
| | - James G. Kublin
- Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | | | | | - Lisa A. Jackson
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
23
|
Systems analysis and controlled malaria infection in Europeans and Africans elucidate naturally acquired immunity. Nat Immunol 2021; 22:654-665. [PMID: 33888898 DOI: 10.1038/s41590-021-00911-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/02/2021] [Indexed: 01/31/2023]
Abstract
Controlled human infections provide opportunities to study the interaction between the immune system and malaria parasites, which is essential for vaccine development. Here, we compared immune signatures of malaria-naive Europeans and of Africans with lifelong malaria exposure using mass cytometry, RNA sequencing and data integration, before and 5 and 11 days after venous inoculation with Plasmodium falciparum sporozoites. We observed differences in immune cell populations, antigen-specific responses and gene expression profiles between Europeans and Africans and among Africans with differing degrees of immunity. Before inoculation, an activated/differentiated state of both innate and adaptive cells, including elevated CD161+CD4+ T cells and interferon-γ production, predicted Africans capable of controlling parasitemia. After inoculation, the rapidity of the transcriptional response and clusters of CD4+ T cells, plasmacytoid dendritic cells and innate T cells were among the features distinguishing Africans capable of controlling parasitemia from susceptible individuals. These findings can guide the development of a vaccine effective in malaria-endemic regions.
Collapse
|
24
|
Jongo SA, Church LWP, Mtoro AT, Schindler T, Chakravarty S, Ruben AJ, Swanson PA, Kassim KR, Mpina M, Tumbo AM, Milando FA, Qassim M, Juma OA, Bakari BM, Simon B, James ER, Abebe Y, Kc N, Saverino E, Fink M, Cosi G, Gondwe L, Studer F, Styers D, Seder RA, Schindler T, Billingsley PF, Daubenberger C, Sim BKL, Tanner M, Richie TL, Abdulla S, Hoffman SL. Increase of Dose Associated With Decrease in Protection Against Controlled Human Malaria Infection by PfSPZ Vaccine in Tanzanian Adults. Clin Infect Dis 2021; 71:2849-2857. [PMID: 31782768 DOI: 10.1093/cid/ciz1152] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND A vaccine would be an ideal tool for reducing malaria's impact. PfSPZ Vaccine (radiation attenuated, aseptic, purified, cryopreserved Plasmodium falciparum [Pf] sporozoites [SPZ]) has been well tolerated and safe in >1526 malaria-naive and experienced 6-month to 65-year-olds in the United States, Europe, and Africa. When vaccine efficacy (VE) of 5 doses of 2.7 × 105 PfSPZ of PfSPZ Vaccine was assessed in adults against controlled human malaria infection (CHMI) in the United States and Tanzania and intense field transmission of heterogeneous Pf in Mali, Tanzanians had the lowest VE (20%). METHODS To increase VE in Tanzania, we increased PfSPZ/dose (9 × 105 or 1.8 × 106) and decreased numbers of doses to 3 at 8-week intervals in a double blind, placebo-controlled trial. RESULTS All 22 CHMIs in controls resulted in parasitemia by quantitative polymerase chain reaction. For the 9 × 105 PfSPZ group, VE was 100% (5/5) at 3 or 11 weeks (P < .000l, Barnard test, 2-tailed). For 1.8 × 106 PfSPZ, VE was 33% (2/6) at 7.5 weeks (P = .028). VE of dosage groups (100% vs 33%) was significantly different (P = .022). Volunteers underwent repeat CHMI at 37-40 weeks after last dose. 6/6 and 5/6 volunteers developed parasitemia, but time to first parasitemia was significantly longer than controls in the 9 × 105 PfSPZ group (10.89 vs 7.80 days) (P = .039), indicating a significant reduction in parasites in the liver. Antibody and T-cell responses were higher in the 1.8 × 106 PfSPZ group. CONCLUSIONS In Tanzania, increasing the dose from 2.7 × 105 to 9 × 105 PfSPZ increased VE from 20% to 100%, but increasing to 1.8 × 106 PfSPZ significantly reduced VE. CLINICAL TRIALS REGISTRATION NCT02613520.
Collapse
Affiliation(s)
- Said A Jongo
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | - Ali T Mtoro
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Tobias Schindler
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | | | | | - Phillip A Swanson
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kamaka R Kassim
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Maximillian Mpina
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | - Anneth-Mwasi Tumbo
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | - Florence A Milando
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Munira Qassim
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Omar A Juma
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Bakari M Bakari
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Beatus Simon
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | | | | | | | - Martina Fink
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | - Glenda Cosi
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | - Linda Gondwe
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | - Fabian Studer
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | | | - Robert A Seder
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Tobias Schindler
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | | | - Claudia Daubenberger
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | - B Kim Lee Sim
- Sanaria Inc., Rockville, Maryland, USA.,Protein Potential LLC, Rockville, Maryland, USA
| | - Marcel Tanner
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | | | - Salim Abdulla
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | |
Collapse
|
25
|
Vekemans J, Schellenberg D, Benns S, O'Brien K, Alonso P. Meeting report: WHO consultation on malaria vaccine development, Geneva, 15-16 July 2019. Vaccine 2021; 39:2907-2916. [PMID: 33931251 DOI: 10.1016/j.vaccine.2021.03.093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 01/25/2023]
Abstract
Considerable progress has been made in malaria control in the last two decades, but progress has stalled in the last few years. New tools are needed to achieve public health goals in malaria control and elimination. A first generation vaccine, RTS,S/AS01, is currently being evaluated as it undergoes pilot implementation through routine health systems in parts of three African countries. The development of this vaccine took over 30 years and has been full of uncertainties. Even now, important unknowns remain as to its future role in public health. Lessons need to be learnt for second generation and future vaccines, including how to facilitate early planning of investments, streamlining of development, regulatory and policy pathways. A number of candidate vaccines populate the current development pipeline, some of which have the potential to contribute to burden reduction if efficacy is confirmed in conditions of natural exposure, and if they are amenable to affordable supply and programmatic implementation. New, innovative technologies will be needed if future malaria vaccines are to overcome important scientific hurdles and induce durable, high level protection. WHO convened a stakeholder consultation on the status of malaria vaccine research and development to inform the recently reconstituted Malaria Vaccine Advisory Committee (MALVAC) which will assist WHO in updating its current guidance and recommendations about priorities and product preferences for malaria vaccines.
Collapse
Affiliation(s)
- Johan Vekemans
- World Health Organization, 20 Av Appia, 1211 Geneva 27, Switzerland
| | | | | | - Kate O'Brien
- World Health Organization, 20 Av Appia, 1211 Geneva 27, Switzerland
| | - Pedro Alonso
- World Health Organization, 20 Av Appia, 1211 Geneva 27, Switzerland
| |
Collapse
|
26
|
Nevagi RJ, Good MF, Stanisic DI. Plasmodium infection and drug cure for malaria vaccine development. Expert Rev Vaccines 2021; 20:163-183. [PMID: 33428505 DOI: 10.1080/14760584.2021.1874923] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Despite decades of research into the development of a vaccine to combat the malaria parasite, a highly efficacious malaria vaccine is not yet available. Different whole parasite-based vaccine approaches, including deliberate Plasmodium infection and drug cure (IDC), have been evaluated in pre-clinical and early phase clinical trials. The advantage of whole parasite vaccines is that they induce immune responses against multiple parasite antigens, thus lowering the impact of antigenic diversity. Deliberate Plasmodium IDC, as a vaccine approach, involves administering infectious, live parasites in combination with an anti-malarial drug, which controls the infection and enables induction of protective immune responses.
Collapse
Affiliation(s)
- Reshma J Nevagi
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| | - Michael F Good
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| | - Danielle I Stanisic
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| |
Collapse
|
27
|
Jongo SA, Urbano V, Church LWP, Olotu A, Manock SR, Schindler T, Mtoro A, Kc N, Hamad A, Nyakarungu E, Mpina M, Deal A, Bijeri JR, Ondo Mangue ME, Ntutumu Pasialo BE, Nguema GN, Owono SN, Rivas MR, Chemba M, Kassim KR, James ER, Stabler TC, Abebe Y, Saverino E, Sax J, Hosch S, Tumbo AM, Gondwe L, Segura JL, Falla CC, Phiri WP, Hergott DEB, García GA, Schwabe C, Maas CD, Murshedkar T, Billingsley PF, Tanner M, Ayekaba MO, Sim BKL, Daubenberger C, Richie TL, Abdulla S, Hoffman SL. Immunogenicity and Protective Efficacy of Radiation-Attenuated and Chemo-Attenuated PfSPZ Vaccines in Equatoguinean Adults. Am J Trop Med Hyg 2021; 104:283-293. [PMID: 33205741 PMCID: PMC7790068 DOI: 10.4269/ajtmh.20-0435] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Plasmodium falciparum sporozoite (PfSPZ) Vaccine (radiation-attenuated, aseptic, purified, cryopreserved PfSPZ) and PfSPZ-CVac (infectious, aseptic, purified, cryopreserved PfSPZ administered to subjects taking weekly chloroquine chemoprophylaxis) have shown vaccine efficacies (VEs) of 100% against homologous controlled human malaria infection (CHMI) in nonimmune adults. Plasmodium falciparum sporozoite-CVac has never been assessed against CHMI in African vaccinees. We assessed the safety, immunogenicity, and VE against homologous CHMI of three doses of 2.7 × 106 PfSPZ of PfSPZ Vaccine at 8-week intervals and three doses of 1.0 × 105 PfSPZ of PfSPZ-CVac at 4-week intervals with each arm randomized, double-blind, placebo-controlled, and conducted in parallel. There were no differences in solicited adverse events between vaccinees and normal saline controls, or between PfSPZ Vaccine and PfSPZ-CVac recipients during the 6 days after administration of investigational product. However, from days 7–13, PfSPZ-CVac recipients had significantly more AEs, probably because of Pf parasitemia. Antibody responses were 2.9 times higher in PfSPZ Vaccine recipients than PfSPZ-CVac recipients at time of CHMI. Vaccine efficacy at a median of 14 weeks after last PfSPZ-CVac dose was 55% (8 of 13, P = 0.051) and at a median of 15 weeks after last PfSPZ Vaccine dose was 27% (5 of 15, P = 0.32). The higher VE in PfSPZ-CVac recipients of 55% with a 27-fold lower dose was likely a result of later stage parasite maturation in the liver, leading to induction of cellular immunity against a greater quantity and broader array of antigens.
Collapse
Affiliation(s)
- Said A Jongo
- 1Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Vicente Urbano
- 2Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | | | - Ally Olotu
- 1Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | | | - Ali Mtoro
- 1Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Natasha Kc
- 3Sanaria Inc., Rockville, Maryland.,5Protein Potential LLC, Rockville, Maryland
| | - Ali Hamad
- 1Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Elizabeth Nyakarungu
- 1Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | - Anna Deal
- 4Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - José Raso Bijeri
- 2Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | - Martin Eka Ondo Mangue
- 2Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | | | - Genaro Nsue Nguema
- 2Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | - Salomon Nguema Owono
- 2Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | - Matilde Riloha Rivas
- 2Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | - Mwajuma Chemba
- 1Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Kamaka R Kassim
- 1Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | | | | | | | - Julian Sax
- 4Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Salome Hosch
- 4Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | - Linda Gondwe
- 4Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - J Luis Segura
- 6Medical Care Development International, Silver Spring, Maryland
| | | | | | | | | | | | - Carl D Maas
- 7Marathon EG Production, Ltd., Bioko Norte, Equatorial Guinea
| | | | | | - Marcel Tanner
- 4Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Mitoha Ondo'o Ayekaba
- 2Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | - B Kim Lee Sim
- 3Sanaria Inc., Rockville, Maryland.,5Protein Potential LLC, Rockville, Maryland
| | | | | | - Salim Abdulla
- 1Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | |
Collapse
|
28
|
Wockner LF, Hoffmann I, Webb L, Mordmüller B, Murphy SC, Kublin JG, O'Rourke P, McCarthy JS, Marquart L. Growth Rate of Plasmodium falciparum: Analysis of Parasite Growth Data From Malaria Volunteer Infection Studies. J Infect Dis 2020; 221:963-972. [PMID: 31679015 PMCID: PMC7198127 DOI: 10.1093/infdis/jiz557] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/29/2019] [Indexed: 12/16/2022] Open
Abstract
Background Growth rate of malaria parasites in the blood of infected subjects is an important
measure of efficacy of drugs and vaccines. Methods We used log-linear and sine-wave models to estimate the parasite growth rate of the 3D7
strain of Plasmodium falciparum using data from 177 subjects from 14
induced blood stage malaria (IBSM) studies conducted at QIMR Berghofer. We estimated
parasite multiplication rate per 48 hour (PMR48), PMR per life-cycle
(PMRLC), and parasite life-cycle duration. We compared these parameters to
those from studies conducted elsewhere with infections induced by IBSM (n=66),
sporozoites via mosquito bite (n=336) or injection (n=51). Results The parasite growth rate of 3D7 in QIMR Berghofer studies was 0.75/day (95% CI:
0.73–0.77/day), PMR48 was 31.9 (95% CI: 28.7–35.4),
PMRLC was 16.4 (95% CI: 15.1–17.8) and parasite life-cycle was 38.8
hour (95% CI: 38.3–39.2 hour). These parameters were similar to estimates from
IBSM studies elsewhere (0.71/day, 95% CI: 0.67–0.75/day; PMR48 26.6,
95% CI: 22.2–31.8), but significantly higher (P < 0.001)
than in sporozoite studies (0.47/day, 95% CI: 0.43–0.50/day; PMR48
8.6, 95% CI: 7.3–10.1). Conclusions Parasite growth rates were similar across different IBSM studies and higher than
infections induced by sporozoite.
Collapse
Affiliation(s)
- Leesa F Wockner
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Isabell Hoffmann
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Lachlan Webb
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Sean C Murphy
- Departments of Laboratory Medicine and Microbiology, University of Washington, Seattle, Washington, USA
| | - James G Kublin
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Peter O'Rourke
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
29
|
Billingsley PF, Maas CD, Olotu A, Schwabe C, García GA, Rivas MR, Hergott DEB, Daubenberger C, Saverino E, Chaouch A, Embon O, Chemba M, Nyakarungu E, Hamad A, Cortes C, Schindler T, Mpina M, Mtoro A, Sim BKL, Richie TL, McGhee K, Tanner M, Obiang Lima GM, Abdulla S, Hoffman SL, Ayekaba MO. The Equatoguinean Malaria Vaccine Initiative: From the Launching of a Clinical Research Platform to Malaria Elimination Planning in Central West Africa. Am J Trop Med Hyg 2020; 103:947-954. [PMID: 32458790 PMCID: PMC7470544 DOI: 10.4269/ajtmh.19-0966] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Fifteen years of investment in malaria control on Bioko Island, Equatorial Guinea (EG), dramatically reduced malaria-associated morbidity and mortality, but the impact has plateaued. To progress toward elimination, EG is investing in the development of a malaria vaccine. We assessed the unique public–private partnership that has had such a significant impact on malaria on Bioko Island and now added a major effort on malaria vaccine development. As part of a $79M commitment, the EG government (75%) and three American energy companies (25%) have invested since 2012 greater than $55M in the Equatoguinean Malaria Vaccine Initiative (EGMVI) to support clinical development of Sanaria® PfSPZ vaccines (Sanaria Inc., Rockville, MD). In turn, the vaccine development program is building human capital and physical capacity. The EGMVI established regulatory and ethical oversight to ensure compliance with the International Conference on Harmonization and Good Clinical Practices for the first importation of investigational product, ethical approval, and conduct of a clinical trial in Equatoguinean history. The EGMVI has completed three vaccine trials in EG, two vaccine trials in Tanzania, and a malaria incidence study, and initiated preparations for a 2,100-volunteer clinical trial. Personnel are training for advanced degrees abroad and have been trained in Good Clinical Practices and protocol-specific methods. A new facility has established the foundation for a national research institute. Biomedical research and development within this visionary, ambitious public–private partnership is fostering major improvements in EG. The EGMVI plans to use a PfSPZ Vaccine alongside standard malaria control interventions to eliminate Pf malaria from Bioko, becoming a potential model for elimination campaigns elsewhere.
Collapse
Affiliation(s)
| | - Carl D Maas
- Marathon Oil, Malabo Dos, Bioko Norte, Equatorial Guinea
| | - Ally Olotu
- KEMRI Wellcome Trust Research Programme, Kilifi, Kenya.,Ifakara Health Institute, Bagamoyo, Tanzania
| | | | | | - Matilde Riloha Rivas
- Ministry of Health and Social Welfare, Government of Equatorial Guinea, Malabo, Equatorial Guinea
| | | | - Claudia Daubenberger
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | - Adel Chaouch
- Marathon Oil, Malabo Dos, Bioko Norte, Equatorial Guinea
| | - Oscar Embon
- La Paz Hospital Medical Center, Sipopo, Equatorial Guinea
| | | | | | - Ali Hamad
- Ifakara Health Institute, Bagamoyo, Tanzania
| | - Carlos Cortes
- Medical Care Development International, Silver Spring, Maryland
| | - Tobias Schindler
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Maximillian Mpina
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,Ifakara Health Institute, Bagamoyo, Tanzania
| | - Ali Mtoro
- Ifakara Health Institute, Bagamoyo, Tanzania
| | | | | | - Ken McGhee
- Noble Energy, Malabo Dos, Equatorial Guinea
| | - Marcel Tanner
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | | | | | - Mitoha Ondo'o Ayekaba
- Ministry of Health and Social Welfare, Government of Equatorial Guinea, Malabo, Equatorial Guinea.,Marathon Oil, Malabo Dos, Bioko Norte, Equatorial Guinea
| |
Collapse
|
30
|
Shears MJ, Seilie AM, Kim Lee Sim B, Hoffman SL, Murphy SC. Quantification of Plasmodium knowlesi versus Plasmodium falciparum in the rhesus liver: implications for malaria vaccine studies in rhesus models. Malar J 2020; 19:313. [PMID: 32867784 PMCID: PMC7457220 DOI: 10.1186/s12936-020-03385-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Background Rhesus macaques are valuable pre-clinical models for malaria vaccine development. The Plasmodium knowlesi/rhesus and Plasmodium falciparum/rhesus models are two established platforms for malaria vaccine testing, and both have previously been used to assess live-attenuated sporozoite vaccines. However, there is evidence that the susceptibility of the rhesus liver to P. knowlesi versus P. falciparum sporozoites likely differs, potentially complicating comparisons between these two platforms. Methods To quantify the differing susceptibility of rhesus to P. knowlesi and P. falciparum sporozoites, animals were infected by direct venous inoculation of purified, cryopreserved wild-type P. knowlesi sporozoites (PkSPZ) or P. falciparum sporozoites (PfSPZ). The entire liver was collected 5 days post-infection, and parasite burden in each liver lobe was quantified using an ultrasensitive Plasmodium 18S rRNA RT-PCR biomarker assay. The potential of using 18S rRNA copy number in the rhesus liver to directly measure the efficacy of vaccines targeting P. falciparum sporozoites and liver stages was also theoretically evaluated. Results Infection of rhesus with a high dose of PkSPZ led to consistently high burden liver stage infections (range 9.5–10.1 log10 copies 18S rRNA/g of liver), with similar amounts of parasite 18S rRNA detected in every liver lobe. Inoculation of rhesus with high doses of PfSPZ led to more variable, lower liver burdens (range 4.9–6.6 log10 copies 18S rRNA/g of liver in infected lobes), with parasite 18S rRNA below the limit of detection in some liver lobes. The low signal and heterogeneity of liver burden in the PfSPZ-infected animals indicates that even this extremely sensitive molecular assay cannot be used to assess reliably vaccine efficacy in the P. falciparum/rhesus platform. Conclusions Detection of 18S rRNA in the liver following high dose intravenous PfSPZ confirmed that rhesus are modestly susceptible to wild-type P. falciparum sporozoites. However, comparison of 18S rRNA RT-PCR biomarker signal indicates that the P. falciparum liver burden was 3–5 logs lower than in PkSPZ-infected animals. Quantification of this difference in liver stage burden will help guide and interpret data from pre-clinical studies of live-attenuated sporozoite vaccines in rhesus models.
Collapse
Affiliation(s)
- Melanie J Shears
- Department of Laboratory Medicine and Pathology, University of Washington, 750 Republican St., F870, Seattle, WA, 98109, USA.,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, 750 Republican St., Seattle, WA, 98109, USA
| | - Annette M Seilie
- Department of Laboratory Medicine and Pathology, University of Washington, 750 Republican St., F870, Seattle, WA, 98109, USA.,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, 750 Republican St., Seattle, WA, 98109, USA
| | - B Kim Lee Sim
- Sanaria, Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Stephen L Hoffman
- Sanaria, Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, 750 Republican St., F870, Seattle, WA, 98109, USA. .,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, 750 Republican St., Seattle, WA, 98109, USA. .,Washington National Primate Research Center, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA.
| |
Collapse
|
31
|
Abstract
The increasing recent interest in human challenge studies or controlled human infection model studies for accelerating vaccine development has been driven by the recognition of the unique ability of these studies to contribute to the understanding of response to infection and the performance of vaccines. With streamlining of ethical processes, conduct and supervision and the availability of new investigative tools from immunophenotyping to glycobiology, the potential to derive valuable data to inform vaccine testing and development has never been greater. However, issues of availability and standardization of challenge strains, conduct of studies in disease endemic locations and the iteration between clinical and laboratory studies still need to be addressed to gain maximal value for vaccine development.
Collapse
Affiliation(s)
- Amrita Sekhar
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Gagandeep Kang
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India.
| |
Collapse
|
32
|
Schmaler M, Orlova-Fink N, Rutishauser T, Abdulla S, Daubenberger C. Human unconventional T cells in Plasmodium falciparum infection. Semin Immunopathol 2020; 42:265-277. [PMID: 32076813 PMCID: PMC7223888 DOI: 10.1007/s00281-020-00791-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 02/07/2020] [Indexed: 12/22/2022]
Abstract
Malaria is an old scourge of humankind and has a large negative impact on the economic development of affected communities. Recent success in malaria control and reduction of mortality seems to have stalled emphasizing that our current intervention tools need to be complemented by malaria vaccines. Different populations of unconventional T cells such as mucosal-associated invariant T (MAIT) cells, invariant natural killer T (iNKT) cells and γδ T cells are gaining attention in the field of malaria immunology. Significant advances in our basic understanding of unconventional T cell biology in rodent malaria models have been made, however, their roles in humans during malaria are less clear. Unconventional T cells are abundant in skin, gut and liver tissues, and long-lasting expansions and functional alterations were observed upon malaria infection in malaria naïve and malaria pre-exposed volunteers. Here, we review the current understanding of involvement of unconventional T cells in anti-Plasmodium falciparum immunity and highlight potential future research avenues.
Collapse
Affiliation(s)
- Mathias Schmaler
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Nina Orlova-Fink
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Tobias Rutishauser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Salim Abdulla
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Claudia Daubenberger
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland.
| |
Collapse
|
33
|
Chughlay MF, Akakpo S, Odedra A, Csermak-Renner K, Djeriou E, Winnips C, Leboulleux D, Gaur AH, Shanks GD, McCarthy J, Chalon S. Liver Enzyme Elevations in Plasmodium falciparum Volunteer Infection Studies: Findings and Recommendations. Am J Trop Med Hyg 2020; 103:378-393. [PMID: 32314694 PMCID: PMC7356411 DOI: 10.4269/ajtmh.19-0846] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Malaria volunteer infection studies (VISs) accelerate new drug and vaccine development. In the induced blood-stage malaria (IBSM) model, volunteers are inoculated with erythrocytes infected with Plasmodium falciparum. Observations of elevated liver enzymes in the IBSM model with new chemical entities (NCEs) promoted an analysis of available data. Data were reviewed from eight IBSM studies of seven different NCEs, plus two studies with the registered antimalarial piperaquine conducted between June 2013 and January 2017 at QIMR Berghofer, Brisbane, Australia. Alanine aminotransferase (ALT) was elevated (> 2.5 times the upper limit of normal [×ULN]) in 20/114 (17.5%) participants. Of these, 8.9% (10/114) had moderate increases (> 2.5–5 × ULN), noted in seven studies of six different NCEs ± piperaquine or piperaquine alone, and 8.9% (10/114) had severe elevations (> 5 × ULN), occurring in six studies of six different NCEs ± piperaquine. Aspartate aminotransferase (AST) was elevated (> 2.5 × ULN) in 11.4% (13/114) of participants, across six of the 10 studies. Bilirubin was > 2 × ULN in one participant. Published data from other VIS models, using sporozoite inoculation by systemic administration or mosquito feeding, also showed moderate/severe liver enzyme elevations. In conclusion, liver enzyme elevations in IBSM studies are most likely multifactorial and could be caused by the model conditions, that is, malaria infection/parasite density and/or effective parasite clearance, or by participant-specific risk factors, acetaminophen administration, or direct hepatotoxicity of the test drug. We make recommendations that may mitigate the risk of liver enzyme elevations in future VISs and propose measures to assist their interpretation, should they occur.
Collapse
Affiliation(s)
| | | | - Anand Odedra
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | | | | | | | - Aditya H Gaur
- St. Jude Children's Research Hospital, Memphis, Tennessee
| | - G Dennis Shanks
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - James McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | |
Collapse
|
34
|
Kusi KA, Aggor FE, Amoah LE, Anum D, Nartey Y, Amoako-Sakyi D, Obiri-Yeboah D, Hollingdale M, Ganeshan H, Belmonte M, Peters B, Kim Y, Tetteh J, Kyei-Baafour E, Dodoo D, Villasante E, Sedegah M. Identification of Plasmodium falciparum circumsporozoite protein-specific CD8+ T cell epitopes in a malaria exposed population. PLoS One 2020; 15:e0228177. [PMID: 32040522 PMCID: PMC7010280 DOI: 10.1371/journal.pone.0228177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/08/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Sterile protection against malaria, most likely mediated by parasite-specific CD8+ T cells, has been achieved by attenuated sporozoite vaccination of animals as well as malaria-naïve and malaria-exposed subjects. The circumsporozoite protein (CSP)-based vaccine, RTS,S, shows low efficacy partly due to limited CD8+ T cell induction, and inclusion of such epitopes could improve RTS,S. This study assessed 8-10mer CSP peptide epitopes, present in predicted or previously positive P. falciparum 3D7 CSP 15mer overlapping peptide pools, for their ability to induce CD8+ T cell IFN-γ responses in natural malaria-exposed subjects. METHODS Cryopreserved PBMCs from nine HLA-typed subjects were stimulated with 23 8-10mer CSP peptides from the 3D7 parasite in IFN-ɣ ELISpot assays. The CD8+ T cell specificity of IFN-γ responses was confirmed in ELISpot assays using CD8+ T cell-enriched PBMC fractions after CD4+ cell depletion. RESULTS Ten of 23 peptide epitopes elicited responses in whole PBMCs from five of the nine subjects. Four peptides tested positive in CD8+ T cell-enriched PBMCs from two previously positive responders and one new subject. All four immunodominant peptides are restricted by globally common HLA supertypes (A02, A03, B07) and mapped to regions of the CSP antigen with limited or no reported polymorphism. Association of these peptide-specific responses with anti-malarial protection remains to be confirmed. CONCLUSIONS The relatively conserved nature of the four identified epitopes and their binding to globally common HLA supertypes makes them good candidates for inclusion in potential multi-epitope malaria vaccines.
Collapse
Affiliation(s)
- Kwadwo A. Kusi
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- * E-mail:
| | - Felix E. Aggor
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Linda E. Amoah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Dorothy Anum
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Yvonne Nartey
- Department of Microbiology and Immunology, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Daniel Amoako-Sakyi
- Department of Microbiology and Immunology, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Dorcas Obiri-Yeboah
- Department of Microbiology and Immunology, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Michael Hollingdale
- Malaria Department, Naval Medical Research Center, Silver Springs, MD, United States of America
| | - Harini Ganeshan
- Malaria Department, Naval Medical Research Center, Silver Springs, MD, United States of America
| | - Maria Belmonte
- Malaria Department, Naval Medical Research Center, Silver Springs, MD, United States of America
| | - Bjoern Peters
- La Jolla Institute for Immunology, La Jolla, CA, United States of America
| | - Yohan Kim
- La Jolla Institute for Immunology, La Jolla, CA, United States of America
| | - John Tetteh
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Eric Kyei-Baafour
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Daniel Dodoo
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Eileen Villasante
- Malaria Department, Naval Medical Research Center, Silver Springs, MD, United States of America
| | - Martha Sedegah
- Malaria Department, Naval Medical Research Center, Silver Springs, MD, United States of America
| |
Collapse
|
35
|
Jongo SA, Church LWP, Mtoro AT, Chakravarty S, Ruben AJ, Swanson PA, Kassim KR, Mpina M, Tumbo AM, Milando FA, Qassim M, Juma OA, Bakari BM, Simon B, James ER, Abebe Y, Kc N, Saverino E, Gondwe L, Studer F, Fink M, Cosi G, El-Khorazaty J, Styers D, Seder RA, Schindler T, Billingsley PF, Daubenberger C, Sim BKL, Tanner M, Richie TL, Abdulla S, Hoffman SL. Safety and Differential Antibody and T-Cell Responses to the Plasmodium falciparum Sporozoite Malaria Vaccine, PfSPZ Vaccine, by Age in Tanzanian Adults, Adolescents, Children, and Infants. Am J Trop Med Hyg 2020; 100:1433-1444. [PMID: 30994090 PMCID: PMC6553883 DOI: 10.4269/ajtmh.18-0835] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In 2016, there were more cases and deaths caused by malaria globally than in 2015. An effective vaccine would be an ideal additional tool for reducing malaria's impact. Sanaria® PfSPZ Vaccine, composed of radiation-attenuated, aseptic, purified, cryopreserved Plasmodium falciparum (Pf) sporozoites (SPZ) has been well tolerated and safe in malaria-naïve and experienced adults in the United States and Mali and protective against controlled human malaria infection with Pf in the United States and field transmission of Pf in Mali, but had not been assessed in younger age groups. We, therefore, evaluated PfSPZ Vaccine in 93 Tanzanians aged 45 years to 6 months in a randomized, double-blind, normal saline placebo-controlled trial. There were no significant differences in adverse events between vaccinees and controls or between dosage regimens. Because all age groups received three doses of 9.0 × 105 PfSPZ of PfSPZ Vaccine, immune responses were compared at this dosage. Median antibody responses against Pf circumsporozoite protein and PfSPZ were highest in infants and lowest in adults. T-cell responses were highest in 6-10-year olds after one dose and 1-5-year olds after three doses; infants had no significant positive T-cell responses. The safety data were used to support initiation of trials in > 300 infants in Kenya and Equatorial Guinea. Because PfSPZ Vaccine-induced protection is thought to be mediated by T cells, the T-cell data suggest PfSPZ Vaccine may be more protective in children than in adults, whereas infants may not be immunologically mature enough to respond to the PfSPZ Vaccine immunization regimen assessed.
Collapse
Affiliation(s)
- Said A Jongo
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | - Ali T Mtoro
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | | | - Phillip A Swanson
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Kamaka R Kassim
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Maximillian Mpina
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Anneth-Mwasi Tumbo
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Florence A Milando
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Munira Qassim
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Omar A Juma
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Bakari M Bakari
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Beatus Simon
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | | | | | | | - Linda Gondwe
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Fabian Studer
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Martina Fink
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Glenda Cosi
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | | | - Robert A Seder
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Tobias Schindler
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | - Claudia Daubenberger
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - B Kim Lee Sim
- Protein Potential LLC, Rockville, Maryland.,Sanaria, Inc., Rockville, Maryland
| | - Marcel Tanner
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | - Salim Abdulla
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | |
Collapse
|
36
|
Seilie AM, Chang M, Hanron AE, Billman ZP, Stone BC, Zhou K, Olsen TM, Daza G, Ortega J, Cruz KR, Smith N, Healy SA, Neal J, Wallis CK, Shelton L, Mankowski TV, Wong-Madden S, Mikolajczak SA, Vaughan AM, Kappe SHI, Fishbaugher M, Betz W, Kennedy M, Hume JCC, Talley AK, Hoffman SL, Chakravarty S, Sim BKL, Richie TL, Wald A, Plowe CV, Lyke KE, Adams M, Fahle GA, Cowan EP, Duffy PE, Kublin JG, Murphy SC. Beyond Blood Smears: Qualification of Plasmodium 18S rRNA as a Biomarker for Controlled Human Malaria Infections. Am J Trop Med Hyg 2020; 100:1466-1476. [PMID: 31017084 PMCID: PMC6553913 DOI: 10.4269/ajtmh.19-0094] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
18S rRNA is a biomarker that provides an alternative to thick blood smears in controlled human malaria infection (CHMI) trials. We reviewed data from CHMI trials at non-endemic sites that used blood smears and Plasmodium 18S rRNA/rDNA biomarker nucleic acid tests (NATs) for time to positivity. We validated a multiplex quantitative reverse transcription–polymerase chain reaction (qRT-PCR) for Plasmodium 18S rRNA, prospectively compared blood smears and qRT-PCR for three trials, and modeled treatment effects at different biomarker-defined parasite densities to assess the impact on infection detection, symptom reduction, and measured intervention efficacy. Literature review demonstrated accelerated NAT-based infection detection compared with blood smears (mean acceleration: 3.2–3.6 days). For prospectively tested trials, the validated Plasmodium 18S rRNA qRT-PCR positivity was earlier (7.6 days; 95% CI: 7.1–8.1 days) than blood smears (11.0 days; 95% CI: 10.3–11.8 days) and significantly preceded the onset of grade 2 malaria-related symptoms (12.2 days; 95% CI: 10.6–13.3 days). Discrepant analysis showed that the risk of a blood smear–positive, biomarker-negative result was negligible. Data modeling predicted that treatment triggered by specific biomarker-defined thresholds can differentiate complete, partial, and non-protective outcomes and eliminate many grade 2 and most grade 3 malaria-related symptoms post-CHMI. Plasmodium 18S rRNA is a sensitive and specific biomarker that can justifiably replace blood smears for infection detection in CHMI trials in non-endemic settings. This study led to biomarker qualification through the U.S. Food and Drug Administration for use in CHMI studies at non-endemic sites, which will facilitate biomarker use for the qualified context of use in drug and vaccine trials.
Collapse
Affiliation(s)
- Annette M Seilie
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Ming Chang
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Amelia E Hanron
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Zachary P Billman
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Brad C Stone
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Kevin Zhou
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Tayla M Olsen
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Glenda Daza
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Jose Ortega
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Kurtis R Cruz
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Nahum Smith
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Washington, Seattle, Washington.,Center for Global Infectious Disease Research, Seattle Children's Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington
| | - Jillian Neal
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Carolyn K Wallis
- Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| | - Lisa Shelton
- Center for Global Infectious Disease Research, Seattle Children's Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington
| | - Tracie VonGoedert Mankowski
- Center for Global Infectious Disease Research, Seattle Children's Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington
| | - Sharon Wong-Madden
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Sebastian A Mikolajczak
- Center for Global Infectious Disease Research, Seattle Children's Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington
| | - Matt Fishbaugher
- Center for Global Infectious Disease Research, Seattle Children's Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington
| | - Will Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington
| | - Mark Kennedy
- Center for Global Infectious Disease Research, Seattle Children's Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington
| | - Jen C C Hume
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Angela K Talley
- Center for Global Infectious Disease Research, Seattle Children's Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington
| | | | | | | | | | - Anna Wald
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington
| | | | - Kirsten E Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Matthew Adams
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Gary A Fahle
- Microbiology Service, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | | | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - James G Kublin
- Seattle Malaria Clinical Trials Center, Fred Hutch Cancer Research Center, Seattle, Washington.,Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sean C Murphy
- Department of Microbiology, University of Washington, Seattle, Washington.,Seattle Malaria Clinical Trials Center, Fred Hutch Cancer Research Center, Seattle, Washington.,Center for Global Infectious Disease Research, Seattle Children's Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington.,Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington
| |
Collapse
|
37
|
Atmar RL, Keitel WA. Searching for Improved Flu Vaccines-The Time Is Now. J Infect Dis 2020; 221:1-4. [PMID: 31665360 DOI: 10.1093/infdis/jiz545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Robert L Atmar
- Departments of Medicine and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Wendy A Keitel
- Departments of Medicine and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
38
|
Kapulu MC, Njuguna P, Hamaluba MM. Controlled Human Malaria Infection in Semi-Immune Kenyan Adults (CHMI-SIKA): a study protocol to investigate in vivo Plasmodium falciparum malaria parasite growth in the context of pre-existing immunity. Wellcome Open Res 2019; 3:155. [PMID: 31803847 PMCID: PMC6871356 DOI: 10.12688/wellcomeopenres.14909.2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2019] [Indexed: 01/09/2023] Open
Abstract
Malaria remains a major public health burden despite approval for implementation of a partially effective pre-erythrocytic malaria vaccine. There is an urgent need to accelerate development of a more effective multi-stage vaccine. Adults in malaria endemic areas may have substantial immunity provided by responses to the blood stages of malaria parasites, but field trials conducted on several blood-stage vaccines have not shown high levels of efficacy. We will use the controlled human malaria infection (CHMI) models with malaria-exposed volunteers to identify correlations between immune responses and parasite growth rates in vivo. Immune responses more strongly associated with control of parasite growth should be prioritized to accelerate malaria vaccine development. We aim to recruit up to 200 healthy adult volunteers from areas of differing malaria transmission in Kenya, and after confirming their health status through clinical examination and routine haematology and biochemistry, we will comprehensively characterize immunity to malaria using >100 blood-stage antigens. We will administer 3,200 aseptic, purified, cryopreserved Plasmodium falciparum sporozoites (PfSPZ Challenge) by direct venous inoculation. Serial quantitative polymerase chain reaction to measure parasite growth rate in vivo will be undertaken. Clinical and laboratory monitoring will be undertaken to ensure volunteer safety. In addition, we will also explore the perceptions and experiences of volunteers and other stakeholders in participating in a malaria volunteer infection study. Serum, plasma, peripheral blood mononuclear cells and whole blood will be stored to allow a comprehensive assessment of adaptive and innate host immunity. We will use CHMI in semi-immune adult volunteers to relate parasite growth outcomes with antibody responses and other markers of host immunity. Registration: ClinicalTrials.gov identifier NCT02739763.
Collapse
Affiliation(s)
- Melissa C. Kapulu
- KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, UK
| | | | | | - CHMI-SIKA Study Team
- KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, UK
| |
Collapse
|
39
|
Laurens MB, Berry AA, Travassos MA, Strauss K, Adams M, Shrestha B, Li T, Eappen A, Manoj A, Abebe Y, Murshedkar T, Gunasekera A, Richie TL, Lyke KE, Plowe CV, Kennedy JK, Potter GE, Deye GA, Sim BKL, Hoffman SL. Dose-Dependent Infectivity of Aseptic, Purified, Cryopreserved Plasmodium falciparum 7G8 Sporozoites in Malaria-Naive Adults. J Infect Dis 2019; 220:1962-1966. [PMID: 31419294 PMCID: PMC6834064 DOI: 10.1093/infdis/jiz410] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/13/2019] [Indexed: 12/11/2022] Open
Abstract
Direct venous inoculation of 3.2 × 103 aseptic, purified, cryopreserved, vialed Plasmodium falciparum (Pf) strain NF54 sporozoites, PfSPZ Challenge (NF54), has been used for controlled human malaria infection (CHMI) in the United States, 4 European countries, and 6 African countries. In nonimmune adults, this results in 100% infection rates. We conducted a double-blind, randomized, dose-escalation study to assess the infectivity of the 7G8 clone of Pf (PfSPZ Challenge [7G8]). Results showed dose-dependent infectivity from 43% for 8 × 102 PfSPZ to 100% for 4.8 × 103 PfSPZ. PfSPZ Challenge (7G8) will allow for more complete assessment by CHMI of antimalarial vaccines and drugs.
Collapse
Affiliation(s)
- Matthew B Laurens
- Malaria Research Group, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - Andrea A Berry
- Malaria Research Group, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - Mark A Travassos
- Malaria Research Group, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - Kathy Strauss
- Malaria Research Group, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - Matthew Adams
- Malaria Research Group, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - Biraj Shrestha
- Malaria Research Group, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - Tao Li
- Sanaria, Inc, Rockville, Maryland
| | | | | | | | | | | | | | - Kirsten E Lyke
- Malaria Research Group, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | | | | | | | - Gregory A Deye
- Parasitology and International Programs Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
40
|
Goswami D, Minkah NK, Kappe SHI. Designer Parasites: Genetically Engineered Plasmodium as Vaccines To Prevent Malaria Infection. THE JOURNAL OF IMMUNOLOGY 2019; 202:20-28. [PMID: 30587570 DOI: 10.4049/jimmunol.1800727] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/21/2018] [Indexed: 12/20/2022]
Abstract
A highly efficacious malaria vaccine that prevents disease and breaks the cycle of infection remains an aspirational goal of medicine. Whole parasite vaccines based on the sporozoite forms of the parasite that target the clinically silent pre-erythrocytic stages of infection have emerged as one of the leading candidates. In animal models of malaria, these vaccines elicit potent neutralizing Ab responses against the sporozoite stage and cytotoxic T cells that eliminate parasite-infected hepatocytes. Among whole-sporozoite vaccines, immunization with live, replication-competent whole parasites engenders superior immunity and protection when compared with live replication-deficient sporozoites. As such, the genetic design of replication-competent vaccine strains holds the promise for a potent, broadly protective malaria vaccine. In this report, we will review the advances in whole-sporozoite vaccine development with a particular focus on genetically attenuated parasites both as malaria vaccine candidates and also as valuable tools to interrogate protective immunity against Plasmodium infection.
Collapse
Affiliation(s)
- Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109; and
| | - Nana K Minkah
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109; and
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109; and .,Department of Global Health, University of Washington, Seattle, WA 98195
| |
Collapse
|
41
|
Loiseau C, Cooper MM, Doolan DL. Deciphering host immunity to malaria using systems immunology. Immunol Rev 2019; 293:115-143. [PMID: 31608461 DOI: 10.1111/imr.12814] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 09/18/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
A century of conceptual and technological advances in infectious disease research has changed the face of medicine. However, there remains a lack of effective interventions and a poor understanding of host immunity to the most significant and complex pathogens, including malaria. The development of successful interventions against such intractable diseases requires a comprehensive understanding of host-pathogen immune responses. A major advance of the past decade has been a paradigm switch in thinking from the contemporary reductionist (gene-by-gene or protein-by-protein) view to a more holistic (whole organism) view. Also, a recognition that host-pathogen immunity is composed of complex, dynamic interactions of cellular and molecular components and networks that cannot be represented by any individual component in isolation. Systems immunology integrates the field of immunology with omics technologies and computational sciences to comprehensively interrogate the immune response at a systems level. Herein, we describe the system immunology toolkit and report recent studies deploying systems-level approaches in the context of natural exposure to malaria or controlled human malaria infection. We contribute our perspective on the potential of systems immunity for the rational design and development of effective interventions to improve global public health.
Collapse
Affiliation(s)
- Claire Loiseau
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Qld, Australia
| | - Martha M Cooper
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Qld, Australia
| | - Denise L Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Qld, Australia
| |
Collapse
|
42
|
Reporter lines based on the gexp02 promoter enable early quantification of sexual conversion rates in the malaria parasite Plasmodium falciparum. Sci Rep 2019; 9:14595. [PMID: 31601834 PMCID: PMC6787211 DOI: 10.1038/s41598-019-50768-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/16/2019] [Indexed: 12/19/2022] Open
Abstract
Transmission of malaria parasites from humans to mosquito vectors requires that some asexual parasites differentiate into sexual forms termed gametocytes. The balance between proliferation in the same host and conversion into transmission forms can be altered by the conditions of the environment. The ability to accurately measure the rate of sexual conversion under different conditions is essential for research addressing the mechanisms underlying sexual conversion, and to assess the impact of environmental factors. Here we describe new Plasmodium falciparum transgenic lines with genome-integrated constructs in which a fluorescent reporter is expressed under the control of the promoter of the gexp02 gene. Using these parasite lines, we developed a sexual conversion assay that shortens considerably the time needed for an accurate determination of sexual conversion rates, and dispenses the need to add chemicals to inhibit parasite replication. Furthermore, we demonstrate that gexp02 is expressed specifically in sexual parasites, with expression starting as early as the sexual ring stage, which makes it a candidate marker for circulating sexual rings in epidemiological studies.
Collapse
|
43
|
Hoo R, Bruske E, Dimonte S, Zhu L, Mordmüller B, Sim BKL, Kremsner PG, Hoffman SL, Bozdech Z, Frank M, Preiser PR. Transcriptome profiling reveals functional variation in Plasmodium falciparum parasites from controlled human malaria infection studies. EBioMedicine 2019; 48:442-452. [PMID: 31521613 PMCID: PMC6838377 DOI: 10.1016/j.ebiom.2019.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/29/2019] [Accepted: 09/01/2019] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The transcriptome of Plasmodium falciparum clinical isolates varies according to strain, mosquito bites, disease severity and clinical history. Therefore, it remains a challenge to directly interpret the parasite's transcriptomic information into a more general biological signature in a natural human malaria infection. These confounding variations can be potentially overcome with parasites derived from controlled-human malaria infection (CHMI) studies. METHODS We performed CHMI studies in healthy and immunologically naïve volunteers receiving the same P. falciparum strain ((Sanaria® PfSPZ Challenge (NF54)), but with different sporozoite dosage and route of infection. Parasites isolated from these volunteers at the day of patency were subjected to in vitro culture for several generations and synchronized ring-stage parasites were subjected to transcriptome profiling. FINDINGS We observed clear deviations between CHMI-derived parasites from volunteer groups receiving different PfSPZ dose and route. CHMI-derived parasites and the pre-mosquito strain used for PfSPZ generation showed significant transcriptional variability for gene clusters associated with malaria pathogenesis, immune evasion and transmission. These transcriptional variation signature clusters were also observed in the transcriptome of P. falciparum isolates from acute clinical infections. INTERPRETATION Our work identifies a previously unrecognized transcriptional pattern in malaria infections in a non-immune background. Significant transcriptome heterogeneity exits between parasites derived from human infections and the pre-mosquito strain, implying that the malaria parasites undergo a change in functional state to adapt to its host environment. Our work also highlights the potential use of transcriptomics data from CHMI study advance our understanding of malaria parasite adaptation and transmission in humans. FUND: This work is supported by German Israeli Foundation, German ministry for education and research, MOE Tier 1 from the Singapore Ministry of Education Academic Research Fund, Singapore Ministry of Health's National Medical Research Council, National Institute of Allergy and Infectious Diseases, National Institutes of Health, USA and the German Centre for Infection Research (Deutsches Zentrum für Infektionsforschung-DZIF).
Collapse
Affiliation(s)
- Regina Hoo
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Ellen Bruske
- Institute of Tropical Medicine, Wilhelmstr. 27, University of Tübingen, 72074 Tübingen, Germany
| | - Sandra Dimonte
- Institute of Tropical Medicine, Wilhelmstr. 27, University of Tübingen, 72074 Tübingen, Germany
| | - Lei Zhu
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Benjamin Mordmüller
- Institute of Tropical Medicine, Wilhelmstr. 27, University of Tübingen, 72074 Tübingen, Germany; German Center for Infection Research, partner site Tübingen, Germany
| | - B Kim Lee Sim
- Sanaria Inc, 9800 Medical Center Dr A209, Rockville, MD 20850, USA
| | - Peter G Kremsner
- Institute of Tropical Medicine, Wilhelmstr. 27, University of Tübingen, 72074 Tübingen, Germany; Centre de Recherches Médicales de Lambaréné, BP 242 Lambaréné, Gabon
| | | | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Matthias Frank
- Institute of Tropical Medicine, Wilhelmstr. 27, University of Tübingen, 72074 Tübingen, Germany.
| | - Peter R Preiser
- School of Biological Sciences, Nanyang Technological University, Singapore.
| |
Collapse
|
44
|
Winkel BMF, de Korne CM, van Oosterom MN, Staphorst D, Meijhuis M, Baalbergen E, Ganesh MS, Dechering KJ, Vos MW, Chevalley-Maurel SC, Franke-Fayard B, van Leeuwen FWB, Roestenberg M. Quantification of wild-type and radiation attenuated Plasmodium falciparum sporozoite motility in human skin. Sci Rep 2019; 9:13436. [PMID: 31530862 PMCID: PMC6748968 DOI: 10.1038/s41598-019-49895-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/31/2019] [Indexed: 11/09/2022] Open
Abstract
Given the number of global malaria cases and deaths, the need for a vaccine against Plasmodium falciparum (Pf) remains pressing. Administration of live, radiation-attenuated Pf sporozoites can fully protect malaria-naïve individuals. Despite the fact that motility of these attenuated parasites is key to their infectivity and ultimately protective efficacy, sporozoite motility in human tissue (e.g. skin) remains wholly uncharacterized to date. We show that the ability to quantitatively address the complexity of sporozoite motility in human tissue provides an additional tool in the development of attenuated sporozoite vaccines. We imaged Pf movement in the skin of its natural host and compared wild-type and radiation-attenuated GFP-expressing Pf sporozoites. Using custom image analysis software and human skin explants we were able to quantitatively study their key motility features. This head-to-head comparison revealed that radiation attenuation impaired the capacity of sporozoites to vary their movement angle, velocity and direction, promoting less refined movement patterns. Understanding and overcoming these changes in motility will contribute to the development of an efficacious attenuated parasite malaria vaccine.
Collapse
Affiliation(s)
- Béatrice M F Winkel
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Clarize M de Korne
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Matthias N van Oosterom
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Diego Staphorst
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Mark Meijhuis
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Els Baalbergen
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Munisha S Ganesh
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Koen J Dechering
- TropIQ Health Sciences, Transistorweg 5, 6534, Nijmegen, The Netherlands
| | - Martijn W Vos
- TropIQ Health Sciences, Transistorweg 5, 6534, Nijmegen, The Netherlands
| | - Séverine C Chevalley-Maurel
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Blandine Franke-Fayard
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
45
|
Cooper MM, Loiseau C, McCarthy JS, Doolan DL. Human challenge models: tools to accelerate the development of malaria vaccines. Expert Rev Vaccines 2019; 18:241-251. [PMID: 30732492 DOI: 10.1080/14760584.2019.1580577] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Malaria challenge models, where healthy human volunteers are intentionally infected with Plasmodium species parasites under controlled conditions, can be undertaken in several well-defined ways. These challenge models enable evaluation of the kinetics of parasite growth and clearance, host-pathogen interactions and the host immune response. They can facilitate discovery of candidate diagnostic biomarkers and novel vaccine targets. As translational tools they can facilitate testing of candidate vaccines and drugs and evaluation of diagnostic tests. AREAS COVERED Until recently, malaria human challenge models have been limited to only a few Plasmodium falciparum strains and used exclusively in malaria-naïve volunteers in non-endemic regions. Several recent advances include the use of alternate P. falciparum strains and other species of Plasmodia, as well as strains attenuated by chemical, radiation or genetic modification, and the conduct of studies in pre-exposed individuals. Herein, we discuss how this diversification is enabling more thorough vaccine efficacy testing and informing rational vaccine development. EXPERT OPINION The ability to comprehensively evaluate vaccine efficacy in controlled settings will continue to accelerate the translation of candidate malaria vaccines to the clinic, and inform the development and optimisation of potential vaccines that would be effective against multiple strains in geographically and demographically diverse settings.
Collapse
Affiliation(s)
- Martha M Cooper
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - Claire Loiseau
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - James S McCarthy
- b Infectious Diseases Programme , QIMR Berghofer Medical Research Institute , Brisbane , Australia
| | - Denise L Doolan
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| |
Collapse
|
46
|
Walk J, Stok JE, Sauerwein RW. Can Patrolling Liver-Resident T Cells Control Human Malaria Parasite Development? Trends Immunol 2019; 40:186-196. [PMID: 30713008 DOI: 10.1016/j.it.2019.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/08/2019] [Accepted: 01/13/2019] [Indexed: 12/23/2022]
Abstract
Recently, a population of non-recirculating, tissue-resident memory CD8+ T cells has been identified; cells that seems to act as key sentinels for invading microorganisms with enhanced effector functions. In malaria, the liver represents the first site for parasite development before a definite infection is established in circulating red blood cells. Here, we discuss the evidence obtained from animal models on several diseases and hypothesize that liver-resident memory CD8+ T cells (hepatic TRM) play a critical role in providing protective liver-stage immunity against Plasmodium malaria parasites. Although observations in human malaria trials are limited to peripheral blood, we propose recommendations for the translation of some of these findings to human malaria research.
Collapse
Affiliation(s)
- Jona Walk
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Jorn E Stok
- University Medical Center Utrecht, PO Box 85500, Utrecht, The Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
47
|
O'Brochta DA, Alford R, Harrell R, Aluvihare C, Eappen AG, Li T, Chakravarty S, Sim BKL, Hoffman SL, Billingsley PF. Is Saglin a mosquito salivary gland receptor for Plasmodium falciparum? Malar J 2019; 18:2. [PMID: 30602380 PMCID: PMC6317240 DOI: 10.1186/s12936-018-2634-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/20/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Saglin, a 100 kDa protein composed of two 50 kDa homodimers, is present in the salivary glands of Anopheles gambiae and has been considered an essential receptor for sporozoites (SPZ) of Plasmodium berghei and Plasmodium falciparum (Pf), allowing SPZ to recognize, bind to, and infect mosquito salivary glands. Spatial and temporal patterns of Saglin expression reported here, however, suggest that this model does not fully describe the Saglin-SPZ interaction. RESULTS Saglin protein was detected by indirect immunofluorescence microscopy only in the medial and proximal-lateral lobes, but not in the distal-lateral lobes, of the salivary glands of An. gambiae; the pattern of expression was independent of mosquito age or physiological state. These results were confirmed by steady-state Saglin transcript and protein expression using qRT-PCR and Western-blot analysis, respectively. Saglin was localized to the basal surface of the cells of the medial lobes and was undetectable elsewhere (intracellularly, on the lateral or apical membranes, the cells' secretory vacuoles, or in the salivary duct). In the cells of the proximal lateral lobes of the salivary glands, Saglin was distinctly intracellular and was not localized to any of the cell surfaces. Transgenic Anopheles stephensi were produced that expressed An. gambiae Saglin in the distal lateral lobes of the salivary gland. Additional Saglin expression did not enhance infection by PfSPZ compared to non-transgenic siblings fed on the same gametocyte-containing blood meal. CONCLUSIONS The absence of Saglin in the distal lateral lobes of the salivary glands, a primary destination for SPZ, suggests Saglin is not an essential receptor for Plasmodium SPZ. The lack of any correlation between increased Saglin expression in the distal lateral lobes of the salivary glands of transgenic An. stephensi and PfSPZ infection is also consistent with Saglin not being an essential salivary gland receptor for Plasmodium SPZ.
Collapse
Affiliation(s)
- David A O'Brochta
- Department of Entomology and The Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, 9600 Gudelsky Drive, Rockville, MD, 20850, USA.,Foundation for the National Institutes of Health, 11400 Rockville Pike, Suite 600, North Bethesda, MD, 20852, USA
| | - Robert Alford
- Department of Entomology and The Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Robert Harrell
- University of Maryland Insect Transformation Facility, The Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Channa Aluvihare
- University of Maryland Insect Transformation Facility, The Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Abraham G Eappen
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Tao Li
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Sumana Chakravarty
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - B Kim Lee Sim
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Stephen L Hoffman
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Peter F Billingsley
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA.
| |
Collapse
|
48
|
Matuschewski K, Borrmann S. Controlled Human Malaria Infection (CHMI) Studies: Over 100 Years of Experience with Parasite Injections. Methods Mol Biol 2019; 2013:91-101. [PMID: 31267496 DOI: 10.1007/978-1-4939-9550-9_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Human experimentation by deliberate infection with malarial parasites seems unethical yet has a long history in infectious disease research. After rigorous screening, volunteers are inoculated with Plasmodium sporozoites or blood stages and monitored under strict clinical supervision until they are treated with a licensed malaria drug and the infection is completely resolved. Historically, experimental Plasmodium challenge infections were applied to confirm that Anopheles mosquitoes were the malaria vector and to treat neurosyphilis in Treponema pallidum-infected patients. The lifesaving treatment with reliable parasite inoculation, monitoring, and drug cure was awarded with a Nobel Prize in 1927 and paved the way for human trials for clinical tests of candidate drugs and vaccines. Importantly, controlled human malaria infection (CHMI) studies are indispensable to bridge the major gap between phase I safety and phase II field trials. Here, we describe the biological basis, historical experiences, applications, and ethical considerations for CHMI studies. Acceleration of antimalarial drug and vaccine development remains a priority in medical research and critically depends on capacity building for CHMI studies.
Collapse
Affiliation(s)
- Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.
| | - Steffen Borrmann
- Institute for Tropical Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany.
| |
Collapse
|
49
|
Kapulu MC, Njuguna P, Hamaluba MM. Controlled Human Malaria Infection in Semi-Immune Kenyan Adults (CHMI-SIKA): a study protocol to investigate in vivo Plasmodium falciparum malaria parasite growth in the context of pre-existing immunity. Wellcome Open Res 2018; 3:155. [PMID: 31803847 PMCID: PMC6871356 DOI: 10.12688/wellcomeopenres.14909.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2018] [Indexed: 10/20/2023] Open
Abstract
Malaria remains a major public health burden despite approval for implementation of a partially effective pre-erythrocytic malaria vaccine. There is an urgent need to accelerate development of a more effective multi-stage vaccine. Adults in malaria endemic areas may have substantial immunity provided by responses to the blood stages of malaria parasites, but field trials conducted on several blood-stage vaccines have not shown high levels of efficacy. We will use controlled human malaria infection (CHMI) studies with malaria-exposed volunteers to identify correlations between immune responses and parasite growth rates in vivo. Immune responses more strongly associated with control of parasite growth should be prioritized to accelerate malaria vaccine development. We aim to recruit up to 200 healthy adult volunteers from areas of differing malaria transmission in Kenya, and after confirming their health status through clinical examination and routine haematology and biochemistry, we will comprehensively characterize immunity to malaria using >100 blood-stage antigens. We will administer 3,200 aseptic, purified, cryopreserved Plasmodium falciparum sporozoites (PfSPZ Challenge) by direct venous inoculation. Serial quantitative polymerase chain reaction to measure parasite growth rate in vivo will be undertaken. Clinical and laboratory monitoring will be undertaken to ensure volunteer safety. In addition, we will also explore the perceptions and experiences of volunteers and other stakeholders in participating in a malaria volunteer infection study. Serum, plasma, peripheral blood mononuclear cells and extracted DNA will be stored to allow a comprehensive assessment of adaptive and innate host immunity. We will use CHMI in semi-immune adult volunteers to relate parasite growth outcomes with antibody responses and other markers of host immunity. Registration: ClinicalTrials.gov identifier NCT02739763.
Collapse
Affiliation(s)
- Melissa C. Kapulu
- KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, UK
| | | | | | - CHMI-SIKA Study Team
- KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, UK
| |
Collapse
|
50
|
Artemisinin resistance phenotypes and K13 inheritance in a Plasmodium falciparum cross and Aotus model. Proc Natl Acad Sci U S A 2018; 115:12513-12518. [PMID: 30455312 DOI: 10.1073/pnas.1813386115] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Concerns about malaria parasite resistance to treatment with artemisinin drugs (ARTs) have grown with findings of prolonged parasite clearance t 1/2s (>5 h) and their association with mutations in Plasmodium falciparum Kelch-propeller protein K13. Here, we describe a P. falciparum laboratory cross of K13 C580Y mutant with C580 wild-type parasites to investigate ART response phenotypes in vitro and in vivo. After genotyping >400 isolated progeny, we evaluated 20 recombinants in vitro: IC50 measurements of dihydroartemisinin were at similar low nanomolar levels for C580Y- and C580-type progeny (mean ratio, 1.00; 95% CI, 0.62-1.61), whereas, in a ring-stage survival assay, the C580Y-type progeny had 19.6-fold (95% CI, 9.76-39.2) higher average counts. In splenectomized Aotus monkeys treated with three daily doses of i.v. artesunate, t 1/2 calculations by three different methods yielded mean differences of 0.01 h (95% CI, -3.66 to 3.67), 0.80 h (95% CI, -0.92 to 2.53), and 2.07 h (95% CI, 0.77-3.36) between C580Y and C580 infections. Incidences of recrudescence were 57% in C580Y (4 of 7) versus 70% in C580 (7 of 10) infections (-13% difference; 95% CI, -58% to 35%). Allelic substitution of C580 in a C580Y-containing progeny clone (76H10) yielded a transformant (76H10C580Rev) that, in an infected monkey, recrudesced regularly 13 times over 500 d. Frequent recrudescences of ART-treated P. falciparum infections occur with or without K13 mutations and emphasize the need for improved partner drugs to effectively eliminate the parasites that persist through the ART component of combination therapy.
Collapse
|