1
|
Berry AA, Richie TL, Church LWP, Laurens MB, Boyce C, Kc N, Joshi S, Koudjra AR, Butler L, Chen MC, Abebe Y, Murshedkar T, James ER, Billingsley PF, Sim BKL, Hoffman SL, Lyke KE. Safety, tolerability and immunogenicity of a condensed, multi-dose prime regimen of PfSPZ Vaccine for the prevention of Plasmodium falciparum malaria infection. Malar J 2025; 24:88. [PMID: 40098097 PMCID: PMC11916963 DOI: 10.1186/s12936-025-05299-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND The World Health Organization (WHO) has called for new malaria vaccines with > 90% efficacy against Plasmodium falciparum infection to expand the anti-disease benefit provided by the RTS,S/AS01 and R21/Matrix M subunit vaccines currently administered to infants and young children in sub-Saharan Africa. Attenuated P. falciparum sporozoites (PfSPZ) are being developed as a traveller's vaccine and to fulfill WHO's call for high-level efficacy in endemic countries to support malaria elimination. METHODS PfSPZ Vaccine, comprised of radiation-attenuated PfSPZ, was compared with normal saline placebo in a randomized, double-blind trial targeting 60 malaria-naive US adults to assess safety, tolerability, immunogenicity, and efficacy against heterologous controlled human malaria infection three and twelve weeks after immunization. Pharmacists provided syringes to blinded clinicians using 3:1 (vaccine:placebo) blocked randomization, for administration by direct venous inoculation on days 1 and 8 (multidose prime) and day 29 (boost), a condensed regimen with superior efficacy. Primary outcomes included adverse events and antibody responses to the P. falciparum circumsporozoite protein (PfCSP). RESULTS 31 participants were screened, randomized and immunized twice (V1, V2) 5-7 days apart, with one withdrawal after an intercurrent adverse event. A vial issue, later traced to the vial manufacturer, halted further immunizations. Solicited local and systemic adverse events recorded for 2 and 7 days after immunizations, respectively, occurred with equal frequency and severity in the 23 vaccinees and 7 controls receiving two immunizations, as did unsolicited adverse events recorded for 28 days and laboratory abnormalities 1 and 5 weeks after V2. Four of 23 vaccinees and one of 7 controls (p = 1.00) developed grade 2 adverse events including subjective fever, headache, malaise, fatigue, rigors, arthralgia and myalgia after V2 but not V1, these symptoms generally resolving within 24 h. Twenty-two of 23 (96%) vaccinees developed IgG (median 99-fold increase over baseline) and IgM (median 1,110-fold increase) antibodies to PfCSP one week after V2. Antibody responses were not associated with reactogenicity. CONCLUSIONS The two-dose priming immunization regimen was safe, well tolerated and highly immunogenic. Larger studies may better define the adverse event profile of condensed regimens of PfSPZ Vaccine in malaria-naive adults. TRIAL REGISTRATION NUMBER clinicaltrial.gov NCT05604521.
Collapse
Affiliation(s)
- Andrea A Berry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | | | - L W Preston Church
- Sanaria Inc, Rockville, MD, 20850, USA
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Matthew B Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Colleen Boyce
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | | | - Sudhaunshu Joshi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Abra Rachida Koudjra
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lauryn Butler
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | | | | | | | | | - Peter F Billingsley
- Sanaria Inc, Rockville, MD, 20850, USA
- The Vital Narrative, Frederick, MD, 21701, USA
| | | | | | - Kirsten E Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
2
|
Kessy EJ, Olotu AI. Controlled human malaria infection: overview and potential application in the evaluation of transmission-blocking interventions in malaria-endemic areas. Malar J 2025; 24:33. [PMID: 39893367 PMCID: PMC11786456 DOI: 10.1186/s12936-025-05277-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 01/29/2025] [Indexed: 02/04/2025] Open
Abstract
Controlled human malaria infection (CHMI) involves the intentional infection of healthy individuals with malaria parasites, close observation of the volunteers, and clearance of the parasite at a predetermined endpoint. Depending on the need, CHMI can be initiated by either sporozoites or the administration of parasite-infected erythrocytes, with each of the two systems offering different advantages and caveats. Among other uses, CHMI has proven to be a useful tool for the evaluation of new malaria interventions, particularly vaccines and drugs. The majority of CHMI studies have been conducted in Europe, the USA and Australia, with only a handful of studies conducted in malaria-endemic countries. The slow adoption of CHMI in malaria-endemic countries may be attributed to a lack of infrastructure and expertise to conduct studies in malaria-endemic countries and the risk of undue influence and coercion as a result of volunteers' vulnerability due to a lack of education and financial situation. With the need to generate results relevant to the target populations, there has recently been an increase in CHMI studies that are being conducted in malaria-endemic countries. The use of CHMI models for the evaluation of preerythrocytic and blood-stage malaria interventions has been attempted in malaria-endemic countries with great success. There is a need for the adoption of a CHMI model for the evaluation of transmission-blocking interventions in malaria-endemic countries. The establishment of such a model in malaria-endemic countries will facilitate the selection of potential transmission-blocking intervention (TBI) candidates and accelerate their development. Here is an overview of CHMI, key challenges and ethical considerations in adopting CHMI for the evaluation of malaria transmission-blocking interventions in malaria-endemic countries.
Collapse
Affiliation(s)
- Enock J Kessy
- Ifakara Health Institute, P.O. Box 78 373, Dar Es Salaam, Tanzania.
- Nelson Mandela African Institution of Science and Technology, 404 Nganana, 2331 Kikwe, Arumeru, P.O.Box 447, Arusha, Tanzania.
| | - Ally I Olotu
- Ifakara Health Institute, P.O. Box 78 373, Dar Es Salaam, Tanzania
| |
Collapse
|
3
|
Hamisi MA, Asri NAM, Yassim ASM, Suppian R. A systematic review on malaria and Tuberculosis (TB) vaccine challenges in sub-Saharan African clinical trials. PLoS One 2025; 20:e0317233. [PMID: 39854359 PMCID: PMC11760592 DOI: 10.1371/journal.pone.0317233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 12/25/2024] [Indexed: 01/26/2025] Open
Abstract
OBJECTIVE For more than a century, developing novel and effective vaccines against malaria and Tuberculosis (TB) infections has been a challenge. This review sought to investigate the reasons for the slow progress of malaria and TB vaccine candidates in sub-Saharan African clinical trials. METHODS The systematic review protocol was registered on PROSPERO on July 26, 2023 (CRD42023445166). The research articles related to the immunogenicity, efficacy, or safety of malaria or TB vaccines that were published between January 1, 2012, and August 31, 2023, were searched on three databases: Web of Science (WoS), PubMed, and ClinicalTrials.gov. RESULTS A total of 2342 articles were obtained, 50 of which met the inclusion criteria. 28 (56%) articles reported on malaria vaccine attributes, while 22 (44%) articles reported on TB vaccines. In both cases, the major challenges in sub-Saharan African clinical trials were immunogenicity and efficacy, rather than safety. CONCLUSION Factors such as population characteristics, pathogen genetic diversity, vaccine nature, strategy, and formulation were associated with slow progress of the malaria and TB vaccine candidates in sub-Saharan African clinical trials.
Collapse
Affiliation(s)
- Maonezi Abas Hamisi
- School of Science and Technical Education (CoSTE), Mbeya University of Science and Technology, Mbeya, Tanzania
| | - Nur Ain Mohd Asri
- School of Health Sciences, Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| | | | - Rapeah Suppian
- School of Health Sciences, Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
4
|
Lamers OAC, Franke-Fayard BMD, Koopman JPR, Roozen GVT, Janse JJ, Chevalley-Maurel SC, Geurten FJA, de Bes-Roeleveld HM, Iliopoulou E, Colstrup E, Wessels E, van Gemert GJ, van de Vegte-Bolmer M, Graumans W, Stoter TR, Mordmüller BG, Houlder EL, Bousema T, Murugan R, McCall MBB, Janse CJ, Roestenberg M. Safety and Efficacy of Immunization with a Late-Liver-Stage Attenuated Malaria Parasite. N Engl J Med 2024; 391:1913-1923. [PMID: 39565990 DOI: 10.1056/nejmoa2313892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
BACKGROUND Currently licensed and approved malaria subunit vaccines provide modest, short-lived protection against malaria. Immunization with live-attenuated Plasmodium falciparum malaria parasites is an alternative vaccination strategy that has potential to improve protection. METHODS We conducted a double-blind, controlled clinical trial to evaluate the safety, side-effect profile, and efficacy of immunization, by means of mosquito bites, with a second-generation genetically attenuated parasite (GA2) - a mei2 single knockout P. falciparum NF54 parasite (sporozoite form) with extended development into the liver stage. After an open-label dose-escalation safety phase in which participants were exposed to the bites of 15 or 50 infected mosquitoes (stage A), healthy adults who had not had malaria were randomly assigned to be exposed to 50 mosquito bites per immunization of GA2, an early-arresting parasite (GA1), or placebo (bites from uninfected mosquitoes) (stage B). After the completion of three immunization sessions with 50 mosquito bites per session, we compared the protective efficacy of GA2 against homologous P. falciparum controlled human malaria infection with that of GA1 and placebo. The primary end points were the number and severity of adverse events (in stages A and B) and blood-stage parasitemia greater than 100 P. falciparum parasites per milliliter after bites from GA2-infected mosquitoes (in stage A) and after controlled human malaria infection (in stage B). RESULTS Adverse events were similar across the trial groups. Protective efficacy against subsequent controlled human malaria infection was observed in 8 of 9 participants (89%) in the GA2 group, in 1 of 8 participants (13%) in the GA1 group, and in 0 of 3 participants in the placebo group. A significantly higher frequency of P. falciparum-specific polyfunctional CD4+ and Vδ2+ γδ T cells were observed among participants who received GA2 than among those who received GA1, whereas GA2 and GA1 induced similar antibody titers targeting the P. falciparum circumsporozoite protein. CONCLUSIONS In this small trial, GA2 was associated with a favorable immune induction profile and protective efficacy, findings that warrant further evaluation. (Funded by the Bontius Foundation; ClinicalTrials.gov number, NCT04577066.).
Collapse
Affiliation(s)
- Olivia A C Lamers
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Blandine M D Franke-Fayard
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Jan Pieter R Koopman
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Geert V T Roozen
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Jacqueline J Janse
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Severine C Chevalley-Maurel
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Fiona J A Geurten
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Helena M de Bes-Roeleveld
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Eva Iliopoulou
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Emil Colstrup
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Els Wessels
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Geert-Jan van Gemert
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Marga van de Vegte-Bolmer
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Wouter Graumans
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Thabitha R Stoter
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Benjamin G Mordmüller
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Emma L Houlder
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Teun Bousema
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Rajagopal Murugan
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Matthew B B McCall
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Chris J Janse
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| | - Meta Roestenberg
- From Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden (O.A.C.L., B.M.D.F-.F., J.P.R.K., G.V.T.R., J.J.J., S.C.C.-M., F.J.A.G., H.M.B.-R., E.I., E.C., E.W., E.L.H., R.M., C.J.J., M.R.), and the Department of Medical Microbiology, Radboud University Medical Center, Nijmegen (G.-J.G., M.V.-B., W.G., T.R.S., B.G.M., T.B., M.B.B.M.) - both in the Netherlands
| |
Collapse
|
5
|
Pyuza JJ, van Dorst MM, Stam K, Wammes L, König M, Kullaya VI, Kruize Y, Huisman W, Andongolile N, Ngowi A, Shao ER, Mremi A, Hogendoorn PC, Msuya SE, Jochems SP, de Steenhuijsen Piters WA, Yazdanbakhsh M. Lifestyle score is associated with cellular immune profiles in healthy Tanzanian adults. Brain Behav Immun Health 2024; 41:100863. [PMID: 39398291 PMCID: PMC11470418 DOI: 10.1016/j.bbih.2024.100863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/31/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Immune system and vaccine responses vary across geographical locations worldwide, not only between high and low-middle income countries (LMICs), but also between rural and urban populations within the same country. Lifestyle factors such as housing conditions, exposure to microorganisms and parasites and diet are associated with rural-and urban-living. However, the relationships between these lifestyle factors and immune profiles have not been mapped in detail. Here, we profiled the immune system of 100 healthy Tanzanians living across four rural/urban areas using mass cytometry. We developed a lifestyle score based on an individual's household assets, housing condition and recent dietary history and studied the association with cellular immune profiles. Seventeen out of 80 immune cell clusters were associated with living location or lifestyle score, with eight identifiable only using lifestyle score. Individuals with low lifestyle score, most of whom live in rural settings, showed higher frequencies of NK cells, plasmablasts, atypical memory B cells, T helper 2 cells, regulatory T cells and activated CD4+ T effector memory cells expressing CD38, HLA-DR and CTLA-4. In contrast, those with high lifestyle score, most of whom live in urban areas, showed a less activated state of the immune system illustrated by higher frequencies of naïve CD8+ T cells. Using an elastic net machine learning model, we identified cellular immune signatures most associated with lifestyle score. Assuming a link between these immune profiles and vaccine responses, these signatures may inform us on the cellular mechanisms underlying poor responses to vaccines, but also reduced autoimmunity and allergies in low- and middle-income countries.
Collapse
Affiliation(s)
- Jeremia J. Pyuza
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Institute of Public Health, Kilimanjaro Christian University Medical College (KCMUCo), Moshi, Tanzania
- Kilimanjaro Clinical Research Institute (KCRI), Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Marloes M.A.R. van Dorst
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Koen Stam
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Linda Wammes
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Marion König
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Vesla I. Kullaya
- Kilimanjaro Clinical Research Institute (KCRI), Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical College (KCMUCo), Moshi, Tanzania
| | - Yvonne Kruize
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Wesley Huisman
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Nikuntufya Andongolile
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Anastazia Ngowi
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Elichilia R. Shao
- Department of Internal Medicine, Kilimanjaro Christian Medical University College (KCMUCo), Moshi, Tanzania
- Department of Internal Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Alex Mremi
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | | | - Sia E. Msuya
- Institute of Public Health, Kilimanjaro Christian University Medical College (KCMUCo), Moshi, Tanzania
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Simon P. Jochems
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | | | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| |
Collapse
|
6
|
Mukhiya R, Fleischmann WA, Loughland JR, Chan JA, de Labastida Rivera F, Andrew D, Beeson JG, McCarthy JS, Barber BE, Lopez JA, Engwerda C, Thomson-Luque R, Boyle MJ. Heterogeneity of the human immune response to malaria infection and vaccination driven by latent cytomegalovirus infection. EBioMedicine 2024; 109:105419. [PMID: 39490199 PMCID: PMC11576503 DOI: 10.1016/j.ebiom.2024.105419] [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: 08/05/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Human immune responses to infection and vaccination are heterogenous, driven by multiple factors including genetics, environmental exposures and personal infection histories. For malaria caused by Plasmodium falciparum parasites, host factors that impact on humoral immunity are poorly understood. METHODS We investigated the role of latent cytomegalovirus (CMV) on the host immune response to malaria using samples obtained from individuals in previously conducted Phase 1 trials of blood stage P. falciparum Controlled Human Malaria Infection (CHMI) and in a MSP1 vaccine clinical trial. Induced antibody and functions of antibodies, as well as CD4 T cell responses were quantified. FINDINGS CMV seropositivity was associated with reduced induction of parasite specific antibodies following malaria infection and vaccination. During infection, reduced antibody induction was associated with modifications to the T -follicular helper (Tfh) cell compartment. CMV seropositivity was associated with a skew towards Tfh1 cell subsets before and after malaria infection, and reduced activation of Tfh2 cells. Protective Tfh2 cell activation was only associated with antibody development in individuals who were CMV seronegative, and a higher proportion of Tfh1 cells was associated with lower antibody development in individuals who were CMV seropositive. During MSP1 vaccination, reduced antibody induction in individuals who were CMV seropositive was associated with CD4 T cell expression of terminal differentiation marker CD57. INTERPRETATION These findings suggest that CMV seropositivity may be negatively associated with malaria antibody development. Further studies in larger cohorts, particularly in malaria endemic regions are required to investigate whether CMV infection may modify immunity to malaria gained during infection or vaccination in children. FUNDING Work was funded by National Health and Medical Research Council of Australia, CSL Australia and Snow Medical Foundation. Funders had no role in data generation, writing of manuscript of decision to submit for publication.
Collapse
Affiliation(s)
- Reena Mukhiya
- Burnet Institute, Melbourne, Australia; School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Wim A Fleischmann
- Center for Infectious Diseases, Virology, Heidelberg University, Medical Faculty, University Hospital Heidelberg, Germany
| | - Jessica R Loughland
- Burnet Institute, Melbourne, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jo-Anne Chan
- Burnet Institute, Melbourne, Australia; Department of Infectious Diseases, University of Melbourne, Australia; Department of Microbiology and School of Translational Medicine, Monash University, Australia
| | | | - Dean Andrew
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - James G Beeson
- Burnet Institute, Melbourne, Australia; Department of Infectious Diseases, University of Melbourne, Australia; Department of Microbiology and School of Translational Medicine, Monash University, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia; Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | | | - J Alejandro Lopez
- School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Christian Engwerda
- School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Richard Thomson-Luque
- Sumaya-Biotech GmbH & Co. KG, Germany; Centre for Infectious Diseases, Parasitology, Heidelberg University, Medical Faculty, University Hospital Heidelberg, Germany
| | - Michelle J Boyle
- Burnet Institute, Melbourne, Australia; School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia; Department of Infectious Diseases, University of Melbourne, Australia; Department of Microbiology and School of Translational Medicine, Monash University, Australia.
| |
Collapse
|
7
|
Mishra A, Paul P, Srivastava M, Mishra S. A Plasmodium late liver stage arresting GAP provides superior protection in mice. NPJ Vaccines 2024; 9:193. [PMID: 39424860 PMCID: PMC11489731 DOI: 10.1038/s41541-024-00975-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/19/2024] [Indexed: 10/21/2024] Open
Abstract
Liver-stage genetically attenuated malaria parasites (GAPs) are powerful immunogens that provide protection against sporozoite challenge. We previously generated two late liver-stage-arresting GAPs by deleting the stearoyl-CoA desaturase (Scd) or sporozoite conserved orthologous transcript 1 (Scot1) genes in Plasmodium berghei. Immunization with Scd or Scot1 GAP conferred complete protection against a sporozoite challenge. In a safety study, we observed rare breakthrough blood-stage infections in mice inoculated with high doses of sporozoites, indicating that both GAPs were incompletely attenuated. In this study, we generated a Scd/Scot1 GAP by dual gene deletion. This resulted in complete attenuation of the parasites in the liver and did not transition to blood-stage infection despite a high-dose sporozoite challenge. The Scd/Scot1 KO and WT GFP parasites were indistinguishable during blood, mosquito and early liver stage development. Moreover, Scd/Scot1 KO liver-stage schizonts exhibited an abnormal apicoplast biogenesis and nuclear division phenotype, failed to form hepatic merozoites, and exhibited late liver-stage arrest. Compared with early-arresting Speld KO immunization, late-stage liver-arresting Scd/Scot1 KO induces greater and broader CD8+ T-cell responses and elicits stage-transcending immunity that provides superior protection in C57BL/6 mice. These data prove that multiple gene deletions lead to complete attenuation of the parasite and support the development of late liver stage-arresting P. falciparum GAP.
Collapse
Affiliation(s)
- Akancha Mishra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Plabita Paul
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Mrigank Srivastava
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Satish Mishra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
8
|
Chutiyami M, Saravanakumar P, Bello UM, Salihu D, Adeleye K, Kolo MA, Dawa KK, Hamina D, Bhandari P, Sulaiman SK, Sim J. Malaria vaccine efficacy, safety, and community perception in Africa: a scoping review of recent empirical studies. Infection 2024; 52:2007-2028. [PMID: 38441731 PMCID: PMC11499420 DOI: 10.1007/s15010-024-02196-y] [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: 09/21/2023] [Accepted: 01/22/2024] [Indexed: 10/24/2024]
Abstract
AIM The review summarizes the recent empirical evidence on the efficacy, safety, and community perception of malaria vaccines in Africa. METHODS Academic Search Complete, African Journals Online, CINAHL, Medline, PsychInfo, and two gray literature sources were searched in January 2023, and updated in June 2023. Relevant studies published from 2012 were included. Studies were screened, appraised, and synthesized in line with the review aim. Statistical results are presented as 95% Confidence Intervals and proportions/percentages. RESULTS Sixty-six (N = 66) studies met the inclusion criteria. Of the vaccines identified, overall efficacy at 12 months was highest for the R21 vaccine (N = 3) at 77.0%, compared to the RTS,S vaccine (N = 15) at 55%. The efficacy of other vaccines was BK-SE36 (11.0-50.0%, N = 1), ChAd63/MVA ME-TRAP (- 4.7-19.4%, N = 2), FMP2.1/AS02A (7.6-9.9%, N = 1), GMZ2 (0.6-60.0%, N = 5), PfPZ (20.0-100.0%, N = 5), and PfSPZ-CVac (24.8-33.6%, N = 1). Injection site pain and fever were the most common adverse events (N = 26), while febrile convulsion (N = 8) was the most reported, vaccine-related Serious Adverse Event. Mixed perceptions of malaria vaccines were found in African communities (N = 17); awareness was generally low, ranging from 11% in Tanzania to 60% in Nigeria (N = 9), compared to willingness to accept the vaccines, which varied from 32.3% in Ethiopia to 96% in Sierra Leone (N = 15). Other issues include availability, logistics, and misconceptions. CONCLUSION Malaria vaccines protect against malaria infection in varying degrees, with severe side effects rarely occurring. Further research is required to improve vaccine efficacy and community involvement is needed to ensure successful widespread use in African communities.
Collapse
Affiliation(s)
- Muhammad Chutiyami
- School of Nursing and Midwifery, University of Technology Sydney, Sydney, Australia.
| | - Priya Saravanakumar
- School of Nursing and Midwifery, University of Technology Sydney, Sydney, Australia
| | - Umar Muhammad Bello
- Department of Physiotherapy and Paramedicine, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Dauda Salihu
- College of Nursing, Jouf University, Sakaka, Saudi Arabia
| | - Khadijat Adeleye
- College of Nursing, University of Massachusetts, Amherst, MA, 01003, USA
| | | | - Kabiru Kasamu Dawa
- School of Nursing, Midwifery and Health Education, University of Bedfordshire, Luton, UK
| | - Dathini Hamina
- Department of Nursing Science, University of Maiduguri, Maiduguri, Nigeria
| | - Pratibha Bhandari
- School of Nursing and Midwifery, University of Technology Sydney, Sydney, Australia
| | | | - Jenny Sim
- WHO Collaborating Centre for Nursing, Midwifery and Health Development, University of Technology Sydney, Sydney, Australia
- School of Nursing, Midwifery and Paramedicine, Australian Catholic University, Sydney, Australia
| |
Collapse
|
9
|
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
|
10
|
Abuelazm MT, Elzeftawy MA, Kamal MA, Badr H, Gamal M, Aboulgheit M, Abdelazeem B, Abd-Elsalam S, Abouzid M. Protective efficacy and safety of radiation-attenuated and chemo-attenuated Plasmodium Falciparum sporozoite vaccines against controlled and natural malaria infection: a systematic review and meta-analysis of randomized controlled trials. Infection 2024; 52:707-722. [PMID: 38319556 DOI: 10.1007/s15010-024-02174-4] [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: 08/28/2023] [Accepted: 01/01/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND AND OBJECTIVE Despite the significant burden of Plasmodium falciparum (Pf) malaria and the licensure of two vaccines for use in infants and young children that are partially effective in preventing clinical malaria caused by Pf, a highly effective vaccine against Pf infection is still lacking. Live attenuated vaccines using Pf sporozoites as the immunogen (PfSPZ Vaccines) hold promise for addressing this gap. Here we review the safety and efficacy of two of the most promising PfSPZ approaches: PfSPZ Vaccine (radiation attenuated PfSPZ) and PfSPZ-CVac (chemo-attenuated PfSPZ). METHODS We conducted a systematic review and meta-analysis by searching PubMed, EMBASE, SCOPUS, CENTRAL, and WOS until 22nd December 2021. We included randomized controlled trials (RCTs) of these two vaccine approaches that measured protection against parasitaemia following controlled human malaria infection (CHMI) in malaria-naive and malaria-exposed adults or following exposure to naturally transmitted Pf malaria in African adults and children (primary outcome) and that also measured the incidence of solicited and unsolicited adverse events as indicators of safety and tolerability after vaccination (secondary outcome). We included randomized controlled trials (RCTs) that measured the detected parasitaemia after vaccination (primary outcome) and the incidence of various solicited and unsolicited adverse events (secondary outcome). The quality of the included RCTs using the Cochrane ROB 1 tool and the quality of evidence using the GRADE system were evaluated. We pooled dichotomous data using the risk ratio (RR) for development of parasitemia in vaccinees relative to controls as a measure of vaccine efficacy (VE), including the corresponding confidence interval (CI). This study was registered with PROSPERO (CRD42022308057). RESULTS We included 19 RCTs. Pooled RR favoured PfSPZ Vaccine (RR: 0.65 with 95% CI [0.53, 0.79], P = 0.0001) and PfSPZ-table (RR: 0.42 with 95% CI [0.27, 0.67], P = 0.0002) for preventing parasitaemia, relative to normal saline placebo. Pooled RR showed no difference between PfSPZ Vaccine and the control in the occurrence of any solicited adverse event (RR: 1.00 with 95% CI [0.82, 1.23], P = 0.98), any local solicited adverse events (RR: 0.73 with 95% CI [0.49, 1.08], P = 0.11), any systemic solicited adverse events (RR: 0.94 with 95% CI [0.75, 1.17], P = 0.58), and any unsolicited adverse event (RR: 0.93 with 95% CI [0.78, 1.10], P = 0.37). CONCLUSION PfSPZ and PfSPZ-CVacs showed comparable efficacy. Therefore, they can introduce a promising strategy for malaria prophylaxis, but more large-scale field trials are required to sustain efficacy and yield clinically applicable findings.
Collapse
Affiliation(s)
| | | | | | - Helmy Badr
- Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | | | - Basel Abdelazeem
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| | | | - Mohamed Abouzid
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznan, Poland.
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland.
| |
Collapse
|
11
|
Senkpeil L, Bhardwaj J, Little MR, Holla P, Upadhye A, Fusco EM, Swanson PA, Wiegand RE, Macklin MD, Bi K, Flynn BJ, Yamamoto A, Gaskin EL, Sather DN, Oblak AL, Simpson E, Gao H, Haining WN, Yates KB, Liu X, Murshedkar T, Richie TL, Sim BKL, Otieno K, Kariuki S, Xuei X, Liu Y, Polidoro RB, Hoffman SL, Oneko M, Steinhardt LC, Schmidt NW, Seder RA, Tran TM. Innate immune activation restricts priming and protective efficacy of the radiation-attenuated PfSPZ malaria vaccine. JCI Insight 2024; 9:e167408. [PMID: 38687615 PMCID: PMC11382880 DOI: 10.1172/jci.insight.167408] [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: 12/05/2022] [Accepted: 04/24/2024] [Indexed: 05/02/2024] Open
Abstract
A systems analysis was conducted to determine the potential molecular mechanisms underlying differential immunogenicity and protective efficacy results of a clinical trial of the radiation-attenuated whole-sporozoite PfSPZ vaccine in African infants. Innate immune activation and myeloid signatures at prevaccination baseline correlated with protection from P. falciparum parasitemia in placebo controls. These same signatures were associated with susceptibility to parasitemia among infants who received the highest and most protective PfSPZ vaccine dose. Machine learning identified spliceosome, proteosome, and resting DC signatures as prevaccination features predictive of protection after highest-dose PfSPZ vaccination, whereas baseline circumsporozoite protein-specific (CSP-specific) IgG predicted nonprotection. Prevaccination innate inflammatory and myeloid signatures were associated with higher sporozoite-specific IgG Ab response but undetectable PfSPZ-specific CD8+ T cell responses after vaccination. Consistent with these human data, innate stimulation in vivo conferred protection against infection by sporozoite injection in malaria-naive mice while diminishing the CD8+ T cell response to radiation-attenuated sporozoites. These data suggest a dichotomous role of innate stimulation for malaria protection and induction of protective immunity by whole-sporozoite malaria vaccines. The uncoupling of vaccine-induced protective immunity achieved by Abs from more protective CD8+ T cell responses suggests that PfSPZ vaccine efficacy in malaria-endemic settings may be constrained by opposing antigen presentation pathways.
Collapse
Affiliation(s)
- Leetah Senkpeil
- Division of Infectious Diseases, Department of Medicine
- Department of Microbiology and Immunology, and
| | | | - Morgan R Little
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Prasida Holla
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Aditi Upadhye
- Division of Infectious Diseases, Department of Medicine
| | | | - Phillip A Swanson
- Cellular Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Ryan E Wiegand
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Kevin Bi
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Barbara J Flynn
- Cellular Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Ayako Yamamoto
- Cellular Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Erik L Gaskin
- Division of Infectious Diseases, Department of Medicine
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | | | - Edward Simpson
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hongyu Gao
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kathleen B Yates
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Xiaowen Liu
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | | | | | | | - Kephas Otieno
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Simon Kariuki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Xiaoling Xuei
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yunlong Liu
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rafael B Polidoro
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Martina Oneko
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Laura C Steinhardt
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nathan W Schmidt
- Department of Microbiology and Immunology, and
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert A Seder
- Cellular Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Tuan M Tran
- Division of Infectious Diseases, Department of Medicine
- Department of Microbiology and Immunology, and
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
12
|
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: 16] [Impact Index Per Article: 16.0] [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
|
13
|
van Dorst MMAR, Pyuza JJ, Nkurunungi G, Kullaya VI, Smits HH, Hogendoorn PCW, Wammes LJ, Everts B, Elliott AM, Jochems SP, Yazdanbakhsh M. Immunological factors linked to geographical variation in vaccine responses. Nat Rev Immunol 2024; 24:250-263. [PMID: 37770632 DOI: 10.1038/s41577-023-00941-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/30/2023]
Abstract
Vaccination is one of medicine's greatest achievements; however, its full potential is hampered by considerable variation in efficacy across populations and geographical regions. For example, attenuated malaria vaccines in high-income countries confer almost 100% protection, whereas in low-income regions these same vaccines achieve only 20-50% protection. This trend is also observed for other vaccines, such as bacillus Calmette-Guérin (BCG), rotavirus and yellow fever vaccines, in terms of either immunogenicity or efficacy. Multiple environmental factors affect vaccine responses, including pathogen exposure, microbiota composition and dietary nutrients. However, there has been variable success with interventions that target these individual factors, highlighting the need for a better understanding of their downstream immunological mechanisms to develop new ways of modulating vaccine responses. Here, we review the immunological factors that underlie geographical variation in vaccine responses. Through the identification of causal pathways that link environmental influences to vaccine responsiveness, it might become possible to devise modulatory compounds that can complement vaccines for better outcomes in regions where they are needed most.
Collapse
Affiliation(s)
- Marloes M A R van Dorst
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Jeremia J Pyuza
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Gyaviira Nkurunungi
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Vesla I Kullaya
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Linda J Wammes
- Department of Medical Microbiology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Alison M Elliott
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Simon P Jochems
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
14
|
Watson FN, Shears MJ, Kalata AC, Duncombe CJ, Seilie AM, Chavtur C, Conrad E, Cruz Talavera I, Raappana A, Sather DN, Chakravarty S, Sim BKL, Hoffman SL, Tsuji M, Murphy SC. Ultra-low volume intradermal administration of radiation-attenuated sporozoites with the glycolipid adjuvant 7DW8-5 completely protects mice against malaria. Sci Rep 2024; 14:2881. [PMID: 38311678 PMCID: PMC10838921 DOI: 10.1038/s41598-024-53118-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 01/28/2024] [Indexed: 02/06/2024] Open
Abstract
Radiation-attenuated sporozoite (RAS) vaccines can completely prevent blood stage Plasmodium infection by inducing liver-resident memory CD8+ T cells to target parasites in the liver. Such T cells can be induced by 'Prime-and-trap' vaccination, which here combines DNA priming against the P. yoelii circumsporozoite protein (CSP) with a subsequent intravenous (IV) dose of liver-homing RAS to "trap" the activated and expanding T cells in the liver. Prime-and-trap confers durable protection in mice, and efforts are underway to translate this vaccine strategy to the clinic. However, it is unclear whether the RAS trapping dose must be strictly administered by the IV route. Here we show that intradermal (ID) RAS administration can be as effective as IV administration if RAS are co-administrated with the glycolipid adjuvant 7DW8-5 in an ultra-low inoculation volume. In mice, the co-administration of RAS and 7DW8-5 in ultra-low ID volumes (2.5 µL) was completely protective and dose sparing compared to standard volumes (10-50 µL) and induced protective levels of CSP-specific CD8+ T cells in the liver. Our finding that adjuvants and ultra-low volumes are required for ID RAS efficacy may explain why prior reports about higher volumes of unadjuvanted ID RAS proved less effective than IV RAS. The ID route may offer significant translational advantages over the IV route and could improve sporozoite vaccine development.
Collapse
Affiliation(s)
- Felicia N Watson
- Graduate Program in Pathobiology, Department of Global Health, University of Washington, Seattle, WA, 98109, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Melanie J Shears
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Anya C Kalata
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Caroline J Duncombe
- Graduate Program in Pathobiology, Department of Global Health, University of Washington, Seattle, WA, 98109, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - A Mariko Seilie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Chris Chavtur
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Ethan Conrad
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Irene Cruz Talavera
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA
| | - Andrew Raappana
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA
| | - Sumana Chakravarty
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - B Kim Lee Sim
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Stephen L Hoffman
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sean C Murphy
- Graduate Program in Pathobiology, Department of Global Health, University of Washington, Seattle, WA, 98109, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, WA, 98109, USA.
- Department of Microbiology, University of Washington, Seattle, WA, 98109, USA.
- Washington National Primate Research Center, University of Washington, Seattle, WA, 98109, USA.
- Department of Laboratories, Seattle Children's Hospital, Seattle, WA, 98105, USA.
| |
Collapse
|
15
|
MacMillen Z, Hatzakis K, Simpson A, Shears MJ, Watson F, Erasmus JH, Khandhar AP, Wilder B, Murphy SC, Reed SG, Davie JW, Avril M. Accelerated prime-and-trap vaccine regimen in mice using repRNA-based CSP malaria vaccine. NPJ Vaccines 2024; 9:12. [PMID: 38200025 PMCID: PMC10781674 DOI: 10.1038/s41541-023-00799-4] [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: 06/09/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Malaria, caused by Plasmodium parasites, remains one of the most devastating infectious diseases worldwide, despite control efforts to lower morbidity and mortality. Both advanced candidate vaccines, RTS,S and R21, are subunit (SU) vaccines that target a single Plasmodium falciparum (Pf) pre-erythrocytic (PE) sporozoite (spz) surface protein known as circumsporozoite (CS). These vaccines induce humoral immunity but fail to elicit CD8 + T-cell responses sufficient for long-term protection. In contrast, whole-organism (WO) vaccines, such as Radiation Attenuated Sporozoites (RAS), achieved sterile protection but require a series of intravenous doses administered in multiple clinic visits. Moreover, these WO vaccines must be produced in mosquitos, a burdensome process that severely limits their availability. To reduce reliance on WO while maintaining protection via both antibodies and Trm responses, we have developed an accelerated vaccination regimen that combines two distinct agents in a prime-and-trap strategy. The priming dose is a single dose of self-replicating RNA encoding the full-length P. yoelii CS protein, delivered via an advanced cationic nanocarrier (LIONTM). The trapping dose consists of one dose of WO RAS. Our vaccine induces a strong immune response when administered in an accelerated regimen, i.e., either 5-day or same-day immunization. Additionally, mice after same-day immunization showed a 2-day delay of blood patency with 90% sterile protection against a 3-week spz challenge. The same-day regimen also induced durable 70% sterile protection against a 2-month spz challenge. Our approach presents a clear path to late-stage preclinical and clinical testing of dose-sparing, same-day regimens that can confer sterilizing protection against malaria.
Collapse
Affiliation(s)
- Zachary MacMillen
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle, WA, 98102, USA
| | - Kiara Hatzakis
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle, WA, 98102, USA
| | - Adrian Simpson
- HDT Bio, 1150 Eastlake Ave E, Suite 200A, Seattle, WA, 98109, USA
| | - Melanie J Shears
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA, 98109, USA
| | - Felicia Watson
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA, 98109, USA
| | - Jesse H Erasmus
- HDT Bio, 1150 Eastlake Ave E, Suite 200A, Seattle, WA, 98109, USA
| | - Amit P Khandhar
- HDT Bio, 1150 Eastlake Ave E, Suite 200A, Seattle, WA, 98109, USA
| | - Brandon Wilder
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Building 1, Room 2220, 505 NW 185th Ave, Beaverton, OR, 97006, USA
| | - Sean C Murphy
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA, 98109, USA
| | - Steven G Reed
- HDT Bio, 1150 Eastlake Ave E, Suite 200A, Seattle, WA, 98109, USA
| | - James W Davie
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle, WA, 98102, USA
| | - Marion Avril
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle, WA, 98102, USA.
| |
Collapse
|
16
|
Jongo S, Church LP, Milando F, Qassim M, Schindler T, Rashid M, Tumbo A, Nyaulingo G, Bakari BM, Athuman Mbaga T, Mohamed L, Kassimu K, Simon BS, Mpina M, Zaidi I, Duffy PE, Swanson PA, Seder R, Herman JD, Mendu M, Zur Y, Alter G, KC N, Riyahi P, Abebe Y, Murshedkar T, James ER, Billingsley PF, Sim BKL, Richie TL, Daubenberger C, Abdulla S, Hoffman SL. Safety and protective efficacy of PfSPZ Vaccine administered to HIV-negative and -positive Tanzanian adults. J Clin Invest 2024; 134:e169060. [PMID: 38194272 PMCID: PMC10940097 DOI: 10.1172/jci169060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 12/20/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUNDSanaria PfSPZ Vaccine, composed of attenuated Plasmodium falciparum (Pf) sporozoites (SPZ), protects against malaria. We conducted this clinical trial to assess the safety and efficacy of PfSPZ Vaccine in HIV-positive (HIV+) individuals, since the HIV-infection status of participants in mass vaccination programs may be unknown.METHODSThis randomized, double-blind, placebo-controlled trial enrolled 18- to 45-year-old HIV-negative (HIV-) and well-controlled HIV+ Tanzanians (HIV viral load <40 copies/mL, CD4 counts >500 cells/μL). Participants received 5 doses of PfSPZ Vaccine or normal saline (NS) over 28 days, followed by controlled human malaria infection (CHMI) 3 weeks later.RESULTSThere were no solicited adverse events in the 9 HIV- and 12 HIV+ participants. After CHMI, 6 of 6 NS controls, 1 of 5 HIV- vaccinees, and 4 of 4 HIV+ vaccinees were Pf positive by quantitative PCR (qPCR). After immunization, anti-Pf circumsporozoite protein (anti-PfCSP) (isotype and IgG subclass) and anti-PfSPZ antibodies, anti-PfSPZ CD4+ T cell responses, and Vδ2+ γδ CD3+ T cells were nonsignificantly higher in HIV- than in HIV+ vaccinees. Sera from HIV- vaccinees had significantly higher inhibition of PfSPZ invasion of hepatocytes in vitro and antibody-dependent complement deposition (ADCD) and Fcγ3B binding by anti-PfCSP and ADCD by anti-cell-traversal protein for ookinetes and SPZ (anti-PfCelTOS) antibodies.CONCLUSIONSPfSPZ Vaccine was safe and well tolerated in HIV+ vaccinees, but not protective. Vaccine efficacy was 80% in HIV- vaccinees (P = 0.012), whose sera had significantly higher inhibition of PfSPZ invasion of hepatocytes and enrichment of multifunctional PfCSP antibodies. A more potent PfSPZ vaccine or regimen is needed to protect those living with HIV against Pf infection in Africa.TRIAL REGISTRATIONClinicalTrials.gov NCT03420053.FUNDINGEquatorial Guinea Malaria Vaccine Initiative (EGMVI), made up of the Government of Equatorial Guinea Ministries of Mines and Hydrocarbons, and Health and Social Welfare, Marathon Equatorial Guinea Production Limited, Noble Energy, Atlantic Methanol Production Company, and EG LNG; Swiss government, through ESKAS scholarship grant no. 2016.0056; Intramural Research Program of the National Institute of Allergy and Infectious Diseases, NIH; NIH grant 1U01AI155354-01.
Collapse
Affiliation(s)
- Said Jongo
- Ifakara Health Institute (IHI), Bagamoyo, Tanzania
| | | | | | | | - Tobias Schindler
- Swiss Tropical Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Anneth Tumbo
- Ifakara Health Institute (IHI), Bagamoyo, Tanzania
- Swiss Tropical Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | | | | | | | | | | | - Maxmillian Mpina
- Ifakara Health Institute (IHI), Bagamoyo, Tanzania
- Swiss Tropical Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Irfan Zaidi
- Laboratory of Malaria Immunology and Vaccinology and
| | | | | | - Robert Seder
- Vaccine Research Center, NIH, Bethesda, Maryland, USA
| | - Jonathan D. Herman
- Division of Infectious Disease, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Maanasa Mendu
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yonatan Zur
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Galit Alter
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Natasha KC
- Sanaria Inc., Rockville, Maryland, USA
- Protein Potential LLC, Rockville, Maryland, USA
| | | | | | | | | | | | - B. Kim Lee Sim
- Sanaria Inc., Rockville, Maryland, USA
- Protein Potential LLC, Rockville, Maryland, USA
| | | | - Claudia Daubenberger
- Swiss Tropical Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | | |
Collapse
|
17
|
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
|
18
|
Abstract
Malaria is a mosquito-borne disease caused by protozoan parasites of the genus Plasmodium. Despite significant declines in malaria-attributable morbidity and mortality over the last two decades, it remains a major public health burden in many countries. This underscores the critical need for improved strategies to prevent, treat and control malaria if we are to ultimately progress towards the eradication of this disease. Ideally, this will include the development and deployment of a highly effective malaria vaccine that is able to induce long-lasting protective immunity. There are many malaria vaccine candidates in development, with more than a dozen of these in clinical development. RTS,S/AS01 (also known as Mosquirix) is the most advanced malaria vaccine and was shown to have modest efficacy against clinical malaria in phase III trials in 5- to 17-month-old infants. Following pilot implementation trials, the World Health Organisation has recommended it for use in Africa in young children who are most at risk of infection with P. falciparum, the deadliest of the human malaria parasites. It is well recognised that more effective malaria vaccines are needed. In this review, we discuss malaria vaccine candidates that have progressed into clinical evaluation and highlight the most advanced candidates: Sanaria's irradiated sporozoite vaccine (PfSPZ Vaccine), the chemoattenuated sporozoite vaccine (PfSPZ-CVac), RTS,S/AS01 and the novel malaria vaccine candidate, R21, which displayed promising, high-level efficacy in a recent small phase IIb trial in Africa.
Collapse
Affiliation(s)
- Danielle I Stanisic
- 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.
| |
Collapse
|
19
|
Nyandele JP, Kibondo UA, Issa F, Van Geertruyden JP, Warimwe G, Jongo S, Abdulla S, Olotu A. Pre-vaccination monocyte-to-lymphocyte ratio as a biomarker for the efficacy of malaria candidate vaccines: A subgroup analysis of pooled clinical trial data. PLoS One 2023; 18:e0291244. [PMID: 37708143 PMCID: PMC10501550 DOI: 10.1371/journal.pone.0291244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/24/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Pre-vaccination monocyte-to-lymphocyte ratio was previously suggested as a marker for malaria vaccine effectiveness. We investigated the potential of this cell ratio as a marker for malaria vaccine efficacy and effectiveness. Effectiveness was investigated by using clinical malaria endpoint, and efficacy was investigated by using surrogate endpoints of Plasmodium falciparum prepatent period, parasite density, and multiplication rates in a controlled human malaria infection trial (CHMI). METHODS We evaluated the correlation between monocyte-to-lymphocyte ratio and RTS,S vaccine effectiveness using Cox regression modeling with clinical malaria as the primary endpoint. Of the 1704 participants in the RTS,S field trial, data on monocyte-to-lymphocyte ratio was available for 842 participants, of whom our analyses were restricted. We further used Spearman Correlations and Cox regression modeling to evaluate the correlation between monocyte-to-lymphocyte ratio and Whole Sporozoite malaria vaccine efficacy using the surrogate endpoints. Of the 97 participants in the controlled human malaria infection vaccine trials, hematology and parasitology information were available for 82 participants, of whom our analyses were restricted. RESULTS The unadjusted efficacy of RTS,S malaria vaccine was 54% (95% CI: 37%-66%, p <0.001). No correlation was observed between monocyte-to-lymphocyte ratio and RTS,S vaccine efficacy (Hazard Rate (HR):0.90, 95%CI:0.45-1.80; p = 0.77). The unadjusted efficacy of Whole Sporozoite malaria vaccine in the appended dataset was 17.6% (95%CI:10%-28.5%, p<0.001). No association between monocyte-to-lymphocyte ratio and the Whole Sporozoite malaria vaccine was found against either the prepatent period (HR = 1.16; 95%CI:0.51-2.62, p = 0.72), parasite density (rho = 0.004, p = 0.97) or multiplication rates (rho = 0.031, p = 0.80). CONCLUSION Monocyte-to-lymphocyte ratio alone may not be an adequate marker for malaria vaccine efficacy. Further investigations on immune correlates and underlying mechanisms of immune protection against malaria could provide a clearer explanation of the differences between those protected in comparison with those not protected against malaria by vaccination.
Collapse
Affiliation(s)
- Jane Paula Nyandele
- Global Health Institute, University of Antwerp, Antwerp, Belgium
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Ummi Abdul Kibondo
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Fatuma Issa
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | | | | | - Said Jongo
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Salim Abdulla
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Ally Olotu
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| |
Collapse
|
20
|
Watson FN, Shears MJ, Kalata AC, Duncombe CJ, Seilie AM, Chavtur C, Conrad E, Talavera IC, Raappana A, Sather DN, Chakravarty S, Sim BKL, Hoffman SL, Tsuji M, Murphy SC. Ultra-low volume intradermal administration of radiation-attenuated sporozoites with the glycolipid adjuvant 7DW8-5 completely protects mice against malaria. RESEARCH SQUARE 2023:rs.3.rs-3243319. [PMID: 37609210 PMCID: PMC10441511 DOI: 10.21203/rs.3.rs-3243319/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Malaria is caused by Plasmodium parasites and was responsible for over 247 million infections and 619,000 deaths in 2021. Radiation-attenuated sporozoite (RAS) vaccines can completely prevent blood stage infection by inducing protective liver-resident memory CD8+ T cells. Such T cells can be induced by 'prime-and-trap' vaccination, which here combines DNA priming against the P. yoelii circumsporozoite protein (CSP) with a subsequent intravenous (IV) dose of liver-homing RAS to "trap" the activated and expanding T cells in the liver. Prime-and-trap confers durable protection in mice, and efforts are underway to translate this vaccine strategy to the clinic. However, it is unclear whether the RAS trapping dose must be strictly administered by the IV route. Here we show that intradermal (ID) RAS administration can be as effective as IV administration if RAS are co-administrated with the glycolipid adjuvant 7DW8-5 in an ultra-low inoculation volume. In mice, the co-administration of RAS and 7DW8-5 in ultra-low ID volumes (2.5 μL) was completely protective and dose sparing compared to standard volumes (10-50 μL) and induced protective levels of CSP-specific CD8+ T cells in the liver. Our finding that adjuvants and ultra-low volumes are required for ID RAS efficacy may explain why prior reports about higher volumes of unadjuvanted ID RAS proved less effective. The ID route may offer significant translational advantages over the IV route and could improve sporozoite vaccine development.
Collapse
|
21
|
MacMillen Z, Hatzakis K, Simpson A, Shears M, Watson F, Erasmus J, Khandhar A, Wilder B, Murphy S, Reed S, Davie J, Avril M. Accelerated prime-and-trap vaccine regimen in mice using repRNA-based CSP malaria vaccine. RESEARCH SQUARE 2023:rs.3.rs-3045076. [PMID: 37461621 PMCID: PMC10350175 DOI: 10.21203/rs.3.rs-3045076/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Malaria, caused by Plasmodium parasites, remains one of the most devastating infectious diseases worldwide, despite control efforts that have lowered morbidity and mortality. The only P. falciparum vaccine candidates to show field efficacy are those targeting the asymptomatic pre-erythrocytic (PE) stages of infection. The subunit (SU) RTS,S/AS01 vaccine, the only licensed malaria vaccine to date, is only modestly effective against clinical malaria. Both RTS,S/AS01 and the SU R21 vaccine candidate target the PE sporozoite (spz) circumsporozoite (CS) protein. These candidates elicit high-titer antibodies that provide short-term protection from disease, but do not induce the liver-resident memory CD8+ T cells (Trm) that confer strong PE immunity and long-term protection. In contrast, whole-organism (WO) vaccines, employing for example radiation-attenuated spz (RAS), elicit both high antibody titers and Trm, and have achieved high levels of sterilizing protection. However, they require multiple intravenous (IV) doses, which must be administered at intervals of several weeks, complicating mass administration in the field. Moreover, the quantities of spz required present production difficulties. To reduce reliance on WO while maintaining protection via both antibodies and Trm responses, we have developed an accelerated vaccination regimen that combines two distinct agents in a prime-and-trap strategy. While the priming dose is a self-replicating RNA encoding P. yoelii CS protein, delivered via an advanced cationic nanocarrier (LION™), the trapping dose consists of WO RAS. This accelerated regime confers sterile protection in the P. yoelii mouse model of malaria. Our approach presents a clear path to late-stage preclinical and clinical testing of dose-sparing, same-day regimens that can confer sterilizing protection against malaria.
Collapse
|
22
|
MacMillen Z, Hatzakis K, Simpson A, Shears MJ, Watson F, Erasmus JH, Khandhar AP, Wilder B, Murphy SC, Reed SG, Davie JW, Avril M. Accelerated prime-and-trap vaccine regimen in mice using repRNA-based CSP malaria vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541932. [PMID: 37292739 PMCID: PMC10245832 DOI: 10.1101/2023.05.23.541932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Malaria, caused by Plasmodium parasites, remains one of the most devastating infectious diseases worldwide, despite control efforts that have lowered morbidity and mortality. The only P. falciparum vaccine candidates to show field efficacy are those targeting the asymptomatic pre-erythrocytic (PE) stages of infection. The subunit (SU) RTS,S/AS01 vaccine, the only licensed malaria vaccine to date, is only modestly effective against clinical malaria. Both RTS,S/AS01 and the SU R21 vaccine candidate target the PE sporozoite (spz) circumsporozoite (CS) protein. These candidates elicit high-titer antibodies that provide short-term protection from disease, but do not induce the liver-resident memory CD8+ T cells (Trm) that confer strong PE immunity and long-term protection. In contrast, whole-organism (WO) vaccines, employing for example radiation-attenuated spz (RAS), elicit both high antibody titers and Trm, and have achieved high levels of sterilizing protection. However, they require multiple intravenous (IV) doses, which must be administered at intervals of several weeks, complicating mass administration in the field. Moreover, the quantities of spz required present production difficulties. To reduce reliance on WO while maintaining protection via both antibodies and Trm responses, we have developed an accelerated vaccination regimen that combines two distinct agents in a prime-and-trap strategy. While the priming dose is a self-replicating RNA encoding P. yoelii CS protein, delivered via an advanced cationic nanocarrier (LION™), the trapping dose consists of WO RAS. This accelerated regime confers sterile protection in the P. yoelii mouse model of malaria. Our approach presents a clear path to late-stage preclinical and clinical testing of dose-sparing, same-day regimens that can confer sterilizing protection against malaria.
Collapse
Affiliation(s)
| | - Kiara Hatzakis
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle WA 98102
| | - Adrian Simpson
- HDT Bio, 1616 Eastlake Ave E, Suite 280, Seattle WA 98102
| | - Melanie J. Shears
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA 98109
| | - Felicia Watson
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA 98109
| | | | | | - Brandon Wilder
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Building 1, Room 2220, 505 NW 185th Ave, Beaverton, OR 97006
| | - Sean C. Murphy
- University of Washington, Department of Laboratory Medicine and Pathology, 750 Republican St., F870, Seattle, WA 98109
| | - Steven G. Reed
- HDT Bio, 1616 Eastlake Ave E, Suite 280, Seattle WA 98102
| | - James W. Davie
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle WA 98102
| | - Marion Avril
- MalarVx, Inc 1551 Eastlake Ave E, Suite 100, Seattle WA 98102
| |
Collapse
|
23
|
El-Moamly AA, El-Sweify MA. Malaria vaccines: the 60-year journey of hope and final success-lessons learned and future prospects. Trop Med Health 2023; 51:29. [PMID: 37198702 DOI: 10.1186/s41182-023-00516-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/18/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND The world has made great strides towards beating malaria, although about half of the world population is still exposed to the risk of contracting malaria. Developing an effective malaria vaccine was a huge challenge for medical science. In 2021 the World Health Organization (WHO) approved the first malaria vaccine, RTS,S/AS01 vaccine (Mosquirix™), for widespread use. This review highlights the history of development, and the different approaches and types of malaria vaccines, and the literature to date. It covers the developmental stages of RTS,S/AS01 and recommends steps for its deployment. The review explores other potential vaccine candidates and their status, and suggests options for their further development. It also recommends future roles for vaccines in eradicating malaria. Questions remain on how RTS,S vaccine will work in widespread use and how it can best be utilized to benefit vulnerable communities. CONCLUSION Malaria vaccines have been in development for almost 60 years. The RTS,S/AS01 vaccine has now been approved, but cannot be a stand-alone solution. Development should continue on promising candidates such as R21, PfSPZ and P. vivax vaccines. Multi-component vaccines may be a useful addition to other malaria control techniques in achieving eradication of malaria.
Collapse
Affiliation(s)
- Amal A El-Moamly
- Department of Medical Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Mohamed A El-Sweify
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
24
|
Oyong DA, Duffy FJ, Neal ML, Du Y, Carnes J, Schwedhelm KV, Hertoghs N, Jun SH, Miller H, Aitchison JD, De Rosa SC, Newell EW, McElrath MJ, McDermott SM, Stuart KD. Distinct immune responses associated with vaccination status and protection outcomes after malaria challenge. PLoS Pathog 2023; 19:e1011051. [PMID: 37195999 PMCID: PMC10228810 DOI: 10.1371/journal.ppat.1011051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/30/2023] [Accepted: 04/26/2023] [Indexed: 05/19/2023] Open
Abstract
Understanding immune mechanisms that mediate malaria protection is critical for improving vaccine development. Vaccination with radiation-attenuated Plasmodium falciparum sporozoites (PfRAS) induces high level of sterilizing immunity against malaria and serves as a valuable tool for the study of protective mechanisms. To identify vaccine-induced and protection-associated responses during malarial infection, we performed transcriptome profiling of whole blood and in-depth cellular profiling of PBMCs from volunteers who received either PfRAS or noninfectious mosquito bites, followed by controlled human malaria infection (CHMI) challenge. In-depth single-cell profiling of cell subsets that respond to CHMI in mock-vaccinated individuals showed a predominantly inflammatory transcriptome response. Whole blood transcriptome analysis revealed that gene sets associated with type I and II interferon and NK cell responses were increased in prior to CHMI while T and B cell signatures were decreased as early as one day following CHMI in protected vaccinees. In contrast, non-protected vaccinees and mock-vaccinated individuals exhibited shared transcriptome changes after CHMI characterized by decreased innate cell signatures and inflammatory responses. Additionally, immunophenotyping data showed different induction profiles of vδ2+ γδ T cells, CD56+ CD8+ T effector memory (Tem) cells, and non-classical monocytes between protected vaccinees and individuals developing blood-stage parasitemia, following treatment and resolution of infection. Our data provide key insights in understanding immune mechanistic pathways of PfRAS-induced protection and infective CHMI. We demonstrate that vaccine-induced immune response is heterogenous between protected and non-protected vaccinees and that inducted-malaria protection by PfRAS is associated with early and rapid changes in interferon, NK cell and adaptive immune responses. Trial Registration: ClinicalTrials.gov NCT01994525.
Collapse
Affiliation(s)
- Damian A. Oyong
- Center for Global Infectious Disease Research (CGIDR), Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Fergal J. Duffy
- Center for Global Infectious Disease Research (CGIDR), Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Maxwell L. Neal
- Center for Global Infectious Disease Research (CGIDR), Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Ying Du
- Center for Global Infectious Disease Research (CGIDR), Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Jason Carnes
- Center for Global Infectious Disease Research (CGIDR), Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Katharine V. Schwedhelm
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Nina Hertoghs
- Center for Global Infectious Disease Research (CGIDR), Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Seong-Hwan Jun
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Helen Miller
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - John D. Aitchison
- Center for Global Infectious Disease Research (CGIDR), Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Evan W. Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Suzanne M. McDermott
- Center for Global Infectious Disease Research (CGIDR), Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Kenneth D. Stuart
- Center for Global Infectious Disease Research (CGIDR), Seattle Children’s Research Institute, Seattle, Washington, United States of America
| |
Collapse
|
25
|
Edwards CL, Ng SS, de Labastida Rivera F, Corvino D, Engel JA, Montes de Oca M, Bukali L, Frame TC, Bunn PT, Chauhan SB, Singh SS, Wang Y, Na J, Amante FH, Loughland JR, Soon MS, Waddell N, Mukhopadhay P, Koufariotis LT, Johnston RL, Lee JS, Kuns R, Zhang P, Boyle MJ, Hill GR, McCarthy JS, Kumar R, Engwerda CR. IL-10-producing Th1 cells possess a distinct molecular signature in malaria. J Clin Invest 2023; 133:e153733. [PMID: 36594463 PMCID: PMC9797345 DOI: 10.1172/jci153733] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/18/2022] [Indexed: 01/04/2023] Open
Abstract
Control of intracellular parasites responsible for malaria requires host IFN-γ+T-bet+CD4+ T cells (Th1 cells) with IL-10 produced by Th1 cells to mitigate the pathology induced by this inflammatory response. However, these IL-10-producing Th1 (induced type I regulatory [Tr1]) cells can also promote parasite persistence or impair immunity to reinfection or vaccination. Here, we identified molecular and phenotypic signatures that distinguished IL-10-Th1 cells from IL-10+Tr1 cells in Plasmodium falciparum-infected people who participated in controlled human malaria infection studies, as well as C57BL/6 mice with experimental malaria caused by P. berghei ANKA. We also identified a conserved Tr1 cell molecular signature shared between patients with malaria, dengue, and graft-versus-host disease. Genetic manipulation of primary human CD4+ T cells showed that the transcription factor cMAF played an important role in the induction of IL-10, while BLIMP-1 promoted the development of human CD4+ T cells expressing multiple coinhibitory receptors. We also describe heterogeneity of Tr1 cell coinhibitory receptor expression that has implications for targeting these molecules for clinical advantage during infection. Overall, this work provides insights into CD4+ T cell development during malaria that offer opportunities for creation of strategies to modulate CD4+ T cell functions and improve antiparasitic immunity.
Collapse
Affiliation(s)
- Chelsea L. Edwards
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | - Susanna S. Ng
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Griffith University, School of Natural Sciences, Nathan, Australia
- Institute of Experimental Oncology, University of Bonn, Bonn, Germany
| | | | - Dillon Corvino
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Institute of Experimental Oncology, University of Bonn, Bonn, Germany
| | | | - Marcela Montes de Oca
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Luzia Bukali
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | - Teija C.M. Frame
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | - Patrick T. Bunn
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Shashi Bhushan Chauhan
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Siddharth Sankar Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Yulin Wang
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Griffith University, School of Natural Sciences, Nathan, Australia
| | - Jinrui Na
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | - Fiona H. Amante
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Megan S.F. Soon
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | | | | | - Jason S. Lee
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Rachel Kuns
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Ping Zhang
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Clinical Research Division, Fred Hutchinson Cancer Research Centre, Seattle, Washington, USA
| | | | - Geoffrey R. Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Centre, Seattle, Washington, USA
| | - James S. McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Victorian Infectious Diseases Services, Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
26
|
Richie TL, Church LWP, Murshedkar T, Billingsley PF, James ER, Chen MC, Abebe Y, KC N, Chakravarty S, Dolberg D, Healy SA, Diawara H, Sissoko MS, Sagara I, Cook DM, Epstein JE, Mordmüller B, Kapulu M, Kreidenweiss A, Franke-Fayard B, Agnandji ST, López Mikue MSA, McCall MBB, Steinhardt L, Oneko M, Olotu A, Vaughan AM, Kublin JG, Murphy SC, Jongo S, Tanner M, Sirima SB, Laurens MB, Daubenberger C, Silva JC, Lyke KE, Janse CJ, Roestenberg M, Sauerwein RW, Abdulla S, Dicko A, Kappe SHI, Lee Sim BK, Duffy PE, Kremsner PG, Hoffman SL. Sporozoite immunization: innovative translational science to support the fight against malaria. Expert Rev Vaccines 2023; 22:964-1007. [PMID: 37571809 PMCID: PMC10949369 DOI: 10.1080/14760584.2023.2245890] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
INTRODUCTION Malaria, a devastating febrile illness caused by protozoan parasites, sickened 247,000,000 people in 2021 and killed 619,000, mostly children and pregnant women in sub-Saharan Africa. A highly effective vaccine is urgently needed, especially for Plasmodium falciparum (Pf), the deadliest human malaria parasite. AREAS COVERED Sporozoites (SPZ), the parasite stage transmitted by Anopheles mosquitoes to humans, are the only vaccine immunogen achieving >90% efficacy against Pf infection. This review describes >30 clinical trials of PfSPZ vaccines in the U.S.A., Europe, Africa, and Asia, based on first-hand knowledge of the trials and PubMed searches of 'sporozoites,' 'malaria,' and 'vaccines.' EXPERT OPINION First generation (radiation-attenuated) PfSPZ vaccines are safe, well tolerated, 80-100% efficacious against homologous controlled human malaria infection (CHMI) and provide 18-19 months protection without boosting in Africa. Second generation chemo-attenuated PfSPZ are more potent, 100% efficacious against stringent heterologous (variant strain) CHMI, but require a co-administered drug, raising safety concerns. Third generation, late liver stage-arresting, replication competent (LARC), genetically-attenuated PfSPZ are expected to be both safe and highly efficacious. Overall, PfSPZ vaccines meet safety, tolerability, and efficacy requirements for protecting pregnant women and travelers exposed to Pf in Africa, with licensure for these populations possible within 5 years. Protecting children and mass vaccination programs to block transmission and eliminate malaria are long-term objectives.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sara A. Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Halimatou Diawara
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Mahamadou S. Sissoko
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Issaka Sagara
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - David M. Cook
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Judith E. Epstein
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Mordmüller
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Melissa Kapulu
- Biosciences Department, Kenya Medical Research Institute KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrea Kreidenweiss
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | | | - Selidji T. Agnandji
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | | - Matthew B. B. McCall
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Laura Steinhardt
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Martina Oneko
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Ally Olotu
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Ashley M. Vaughan
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - James G. Kublin
- Department of Global Health, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases and Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Said Jongo
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Marcel Tanner
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Matthew B. Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Claudia Daubenberger
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Joana C. Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kirsten E. Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chris J. Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert W. Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Salim Abdulla
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Alassane Dicko
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Stefan H. I. Kappe
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter G. Kremsner
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | |
Collapse
|
27
|
Duffy FJ, Hertoghs N, Du Y, Neal ML, Oyong D, McDermott S, Minkah N, Carnes J, Schwedhelm KV, McElrath MJ, De Rosa SC, Newell E, Aitchison JD, Stuart K. Longitudinal immune profiling after radiation-attenuated sporozoite vaccination reveals coordinated immune processes correlated with malaria protection. Front Immunol 2022; 13:1042741. [PMID: 36591224 PMCID: PMC9798120 DOI: 10.3389/fimmu.2022.1042741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Background Identifying immune processes required for liver-stage sterilizing immunity to malaria remains an open problem. The IMRAS trial comprised 5x immunizations with radiation-attenuated sporozoites resulting in 55% protection from subsequent challenge. Methods To identify correlates of vaccination and protection, we performed detailed systems immunology longitudinal profiling of the entire trial time course including whole blood transcriptomics, detailed PBMC cell phenotyping and serum antigen array profiling of 11 IMRAS radiation-attenuated sporozoite (RAS) vaccinees at up to 21 timepoints each. Results RAS vaccination induced serum antibody responses to CSP, TRAP, and AMA1 in all vaccinees. We observed large numbers of differentially expressed genes associated with vaccination response and protection, with distinctly differing transcriptome responses elicited after each immunization. These included inflammatory and proliferative responses, as well as increased abundance of monocyte and DC subsets after each immunization. Increases in Vδ2 γδ; T cells and MAIT cells were observed in response to immunization over the course of study, and CD1c+ CD40+ DC abundance was significantly associated with protection. Interferon responses strongly differed between protected and non-protected individuals with high interferon responses after the 1st immunization, but not the 2nd-5th. Blood transcriptional interferon responses were correlated with abundances of different circulating classical and non-classical monocyte populations. Conclusions This study has revealed multiple coordinated immunological processes induced by vaccination and associated with protection. Our work represents the most detailed immunological profiling of a RAS vaccine trial performed to date and will guide the design and interpretation of future malaria vaccine trials.
Collapse
Affiliation(s)
- Fergal J. Duffy
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States,*Correspondence: Fergal J. Duffy, ; Ken Stuart,
| | - Nina Hertoghs
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Ying Du
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Maxwell L. Neal
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Damian Oyong
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Suzanne McDermott
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Nana Minkah
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Jason Carnes
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Katharine V. Schwedhelm
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Evan Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - John D. Aitchison
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Ken Stuart
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States,*Correspondence: Fergal J. Duffy, ; Ken Stuart,
| |
Collapse
|
28
|
Sirima SB, Ouédraogo A, Tiono AB, Kaboré JM, Bougouma EC, Ouattara MS, Kargougou D, Diarra A, Henry N, Ouédraogo IN, Billingsley PF, Manoj A, Abebe Y, Kc N, Ruben A, Richie TL, James ER, Joshi S, Shrestha B, Strauss K, Lyke KE, Plowe CV, Potter GE, Cox C, Jones W, Sim BKL, Hoffman SL, Laurens MB. A randomized controlled trial showing safety and efficacy of a whole sporozoite vaccine against endemic malaria. Sci Transl Med 2022; 14:eabj3776. [PMID: 36475905 PMCID: PMC10041996 DOI: 10.1126/scitranslmed.abj3776] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A highly effective malaria vaccine remains elusive despite decades of research. Plasmodium falciparum sporozoite vaccine (PfSPZ Vaccine), a metabolically active, nonreplicating, whole parasite vaccine demonstrated safety and vaccine efficacy (VE) against endemic P. falciparum for 6 months in Malian adults receiving a five-dose regimen. Safety, immunogenicity, and VE of a three-dose regimen were assessed in adults in Balonghin, Burkina Faso in a two-component study: an open-label dose escalation trial with 32 participants followed by a double-blind, randomized, placebo-controlled trial (RCT) with 80 participants randomized to receive three doses of 2.7 × 106 PfSPZ (N = 39) or normal saline (N = 41) just before malaria season. To clear parasitemia, artesunate monotherapy was administered before first and last vaccinations. Thick blood smear microscopy was performed on samples collected during illness and every 4 weeks for 72 weeks after last vaccinations, including two 6-month malaria transmission seasons. Safety outcomes were assessed in all 80 participants who received at least one dose and VE for 79 participants who received three vaccinations. Myalgia was the only symptom that differed between groups. VE (1 - risk ratio; primary VE endpoint) was 38% at 6 months (P = 0.017) and 15% at 18 months (0.078). VE (1 - hazard ratio) was 48% and 46% at 6 and 18 months (P = 0.061 and 0.018). Two weeks after the last dose, antibodies to P. falciparum circumsporozoite protein and PfSPZ were higher in protected versus unprotected vaccinees. A three-dose regimen of PfSPZ Vaccine demonstrated safety and efficacy against malaria infection in malaria-experienced adults.
Collapse
Affiliation(s)
- Sodiomon B Sirima
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso.,Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Alphonse Ouédraogo
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso.,Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Alfred B Tiono
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso.,Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Jean M Kaboré
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso.,Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Edith C Bougouma
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso.,Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Maurice S Ouattara
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso.,Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Désiré Kargougou
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Amidou Diarra
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso.,Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Noelie Henry
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso.,Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Issa N Ouédraogo
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso.,Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | | | | | | | | | | | | | | | - Sudhaunshu Joshi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Biraj Shrestha
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kathy Strauss
- Center for Vaccine Development and Global Health, 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
| | - Christopher V Plowe
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | - Walter Jones
- Parasitic and International Programs Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Matthew B Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Creation and preclinical evaluation of genetically attenuated malaria parasites arresting growth late in the liver. NPJ Vaccines 2022; 7:139. [PMCID: PMC9636417 DOI: 10.1038/s41541-022-00558-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
AbstractWhole-sporozoite (WSp) malaria vaccines induce protective immune responses in animal malaria models and in humans. A recent clinical trial with a WSp vaccine comprising genetically attenuated parasites (GAP) which arrest growth early in the liver (PfSPZ-GA1), showed that GAPs can be safely administered to humans and immunogenicity is comparable to radiation-attenuated PfSPZ Vaccine. GAPs that arrest late in the liver stage (LA-GAP) have potential for increased potency as shown in rodent malaria models. Here we describe the generation of four putative P. falciparum LA-GAPs, generated by CRISPR/Cas9-mediated gene deletion. One out of four gene-deletion mutants produced sporozoites in sufficient numbers for further preclinical evaluation. This mutant, PfΔmei2, lacking the mei2-like RNA gene, showed late liver growth arrest in human liver-chimeric mice with human erythrocytes, absence of unwanted genetic alterations and sensitivity to antimalarial drugs. These features of PfΔmei2 make it a promising vaccine candidate, supporting further clinical evaluation. PfΔmei2 (GA2) has passed regulatory approval for safety and efficacy testing in humans based on the findings reported in this study.
Collapse
|
30
|
KC N, Church LWP, Riyahi P, Chakravarty S, Seder RA, Epstein JE, Lyke KE, Mordmüller B, Kremsner PG, Sissoko MS, Healy S, Duffy PE, Jongo SA, Nchama VUNN, Abdulla S, Mpina M, Sirima SB, Laurens MB, Steinhardt LC, Oneko M, Li M, Murshedkar T, Billingsley PF, Sim BKL, Richie TL, Hoffman SL. Increased levels of anti-PfCSP antibodies in post-pubertal females versus males immunized with PfSPZ Vaccine does not translate into increased protective efficacy. Front Immunol 2022; 13:1006716. [PMID: 36389797 PMCID: PMC9641621 DOI: 10.3389/fimmu.2022.1006716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/30/2022] [Indexed: 11/25/2022] Open
Abstract
Background While prior research has shown differences in the risk of malaria infection and sickness between males and females, little is known about sex differences in vaccine-induced immunity to malaria. Identifying such differences could elucidate important aspects of malaria biology and facilitate development of improved approaches to malaria vaccination. Methods Using a standardized enzyme-linked immunosorbent assay, IgG antibodies to the major surface protein on Plasmodium falciparum (Pf) sporozoites (SPZ), the Pf circumsporozoite protein (PfCSP), were measured before and two weeks after administration of a PfSPZ-based malaria vaccine (PfSPZ Vaccine) to 5-month to 61-year-olds in 11 clinical trials in Germany, the US and five countries in Africa, to determine if there were differences in vaccine elicited antibody response between males and females and if these differences were associated with differential protection against naturally transmitted Pf malaria (Africa) or controlled human malaria infection (Germany, the US and Africa). Results Females ≥ 11 years of age made significantly higher levels of antibodies to PfCSP than did males in most trials, while there was no indication of such differences in infants or children. Although adult females had higher levels of antibodies, there was no evidence of improved protection compared to males. In 2 of the 7 trials with sufficient data, protected males had significantly higher levels of antibodies than unprotected males, and in 3 other trials protected females had higher levels of antibodies than did unprotected females. Conclusion Immunization with PfSPZ Vaccine induced higher levels of antibodies in post-pubertal females but showed equivalent protection in males and females. We conclude that the increased antibody levels in post-pubertal females did not contribute substantially to improved protection. We hypothesize that while antibodies to PfCSP (and PfSPZ) may potentially contribute directly to protection, they primarily correlate with other, potentially protective immune mechanisms, such as antibody dependent and antibody independent cellular responses in the liver.
Collapse
Affiliation(s)
- Natasha KC
- Sanaria Inc., Rockville, MD, United States
| | | | | | | | - Robert A. Seder
- Vaccine Research Center, National Institute of Heath, Bethesda, MD, United States
| | - Judith E. Epstein
- Naval Medical Research Center (NMRC), Silver Spring, MD, United States
| | - Kirsten E. Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Benjamin Mordmüller
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen and German Center for Infection Research, Tübingen, Germany
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter G. Kremsner
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen and German Center for Infection Research, Tübingen, Germany
- Centre de Recherches Medicales de Lambaréné, Lambaréné, Gabon
| | - Mahamadou S. Sissoko
- Malaria Research and Training Center (MRTC), Mali National Institute of Allergy and Infectious Diseases International Centers for Excellence in Research, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Sara Healy
- Laboratory of Malaria Immunology and Parasitology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health (LMIV/NIAID/NIH), Rockville, MD, United States
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Parasitology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health (LMIV/NIAID/NIH), Rockville, MD, United States
| | - Said A. Jongo
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | | | - Salim Abdulla
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Maxmillian Mpina
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
- Swiss Tropical Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Sodiomon B. Sirima
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Matthew B. Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Laura C. Steinhardt
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Martina Oneko
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - MingLin Li
- Sanaria Inc., Rockville, MD, United States
| | | | | | | | | | - Stephen L. Hoffman
- Sanaria Inc., Rockville, MD, United States
- *Correspondence: Stephen L. Hoffman,
| |
Collapse
|
31
|
Coulibaly D, Kone AK, Traore K, Niangaly A, Kouriba B, Arama C, Zeguime A, Dolo A, Lyke KE, Plowe CV, Abebe Y, Potter GE, Kennedy JK, Galbiati SM, Nomicos E, Deye GA, Richie TL, James ER, KC N, Sim BKL, Hoffman SL, Doumbo OK, Thera MA, Laurens MB. PfSPZ-CVac malaria vaccine demonstrates safety among malaria-experienced adults: A randomized, controlled phase 1 trial. EClinicalMedicine 2022; 52:101579. [PMID: 35928033 PMCID: PMC9343417 DOI: 10.1016/j.eclinm.2022.101579] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Plasmodium falciparum (Pf) Sporozoite (SPZ) Chemoprophylaxis Vaccine (PfSPZ-CVac) involves concurrently administering infectious PfSPZ and malaria drug, often chloroquine (CQ), to kill liver-emerging parasites. PfSPZ-CVac (CQ) protected 100% of malaria-naïve participants against controlled human malaria infection. We investigated the hypothesis that PfSPZ-CVac (CQ) is safe and efficacious against seasonal, endemic Pf in malaria-exposed adults. METHODS Healthy 18-45 year olds were enrolled in a double-blind, placebo-controlled trial in Bougoula-Hameau, Mali, randomized 1:1 to 2.048 × 105 PfSPZ (PfSPZ Challenge) or normal saline administered by direct venous inoculation at 0, 4, 8 weeks. Syringes were prepared by pharmacy staff using online computer-based enrolment that randomized allocations. Clinical team and participant masking was assured by identical appearance of vaccine and placebo. Participants received chloroquine 600mg before first vaccination, 10 weekly 300mg doses during vaccination, then seven daily doses of artesunate 200mg before 24-week surveillance during the rainy season. Safety outcomes were solicited adverse events (AEs) and related unsolicited AEs within 12 days of injections, and all serious AEs. Pf infection was detected by thick blood smears performed every four weeks and during febrile illness over 48 weeks. Primary vaccine efficacy (VE) endpoint was time to infection at 24 weeks. NCT02996695. FINDINGS 62 participants were enrolled in April/May 2017. Proportions of participants experiencing at least one solicited systemic AE were similar between treatment arms: 6/31 (19.4%, 95%CI 9.2-36.3) of PfSPZ-CVac recipients versus 7/31 (22.6%, 95%CI 29.2-62.2) of controls (p value = 1.000). Two/31 (6%) in each group reported related, unsolicited AEs. One unrelated death occurred. Of 59 receiving 3 immunizations per protocol, fewer vaccinees (16/29, 55.2%) became infected than controls (22/30, 73.3%). VE was 33.6% by hazard ratio (p = 0.21, 95%CI -27·9, 65·5) and 24.8% by risk ratio (p = 0.10, 95%CI -4·8, 54·3). Antibody responses to PfCSP were poor; 28% of vaccinees sero-converted. INTERPRETATION PfSPZ-CVac (CQ) was well-tolerated. The tested dosing regimen failed to significantly protect against Pf infection in this very high transmission setting. FUNDING U.S. National Institutes of Health, Sanaria. REGISTRATION NUMBER ClinicalTrials.gov identifier (NCT number): NCT02996695.
Collapse
Key Words
- ALT, alanine aminotransferase
- CHMI, Controlled Human Malaria Infection
- CQ, chloroquine
- CSP, circumsporozoite protein
- DOT, directly observed therapy
- DVI, direct venous inoculation
- ELISA, enzyme linked immunosorbent assay
- HR, hazard ratio
- Malaria vaccine
- PCR, polymerase chain reaction
- Pf, Plasmodium falciparum
- PfSPZ Vaccine
- PfSPZ-CVac
- PfSPZ-CVac, Plasmodium falciparum Sporozoite Chemoprophylaxis Vaccine
- Plasmodium falciparum
- SMC, safety monitoring committee
- SPZ, sporozoite
- Sporozoite
- TBS, thick blood smear
- VE, vaccine efficacy
Collapse
Affiliation(s)
- Drissa Coulibaly
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
- Corresponding author.
| | - Abdoulaye K. Kone
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Karim Traore
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Amadou Niangaly
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Bourema Kouriba
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Charles Arama
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Amatigue Zeguime
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Amagana Dolo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Kirsten E. Lyke
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Christopher V. Plowe
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | - Effie Nomicos
- Parasitic and International Programs Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, U. S. National Institutes of Health, Bethesda, MD, United States
| | - Gregory A. Deye
- Parasitic and International Programs Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, U. S. National Institutes of Health, Bethesda, MD, United States
| | | | | | | | | | | | - Ogobara K. Doumbo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Mahamadou A. Thera
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Matthew B. Laurens
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
32
|
Owalla TJ, Hergott DEB, Seilie AM, Staubus W, Chavtur C, Chang M, Kublin JG, Egwang TG, Murphy SC. Rethinking detection of pre-existing and intervening Plasmodium infections in malaria clinical trials. Front Immunol 2022; 13:1003452. [PMID: 36203582 PMCID: PMC9531235 DOI: 10.3389/fimmu.2022.1003452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/22/2022] [Indexed: 02/02/2023] Open
Abstract
Pre-existing and intervening low-density Plasmodium infections complicate the conduct of malaria clinical trials. These infections confound infection detection endpoints, and their immunological effects may detract from intended vaccine-induced immune responses. Historically, these infections were often unrecognized since infrequent and often analytically insensitive parasitological testing was performed before and during trials. Molecular diagnostics now permits their detection, but investigators must weigh the cost, complexity, and personnel demands on the study and the laboratory when scheduling such tests. This paper discusses the effect of pre-existing and intervening, low-density Plasmodium infections on malaria vaccine trial endpoints and the current methods employed for their infection detection. We review detection techniques, that until recently, provided a dearth of cost-effective strategies for detecting low density infections. A recently deployed, field-tested, simple, and cost-effective molecular diagnostic strategy for detecting pre-existing and intervening Plasmodium infections from dried blood spots (DBS) in malaria-endemic settings is discussed to inform new clinical trial designs. Strategies that combine sensitive molecular diagnostic techniques with convenient DBS collections and cost-effective pooling strategies may enable more thorough and informative infection monitoring in upcoming malaria clinical trials and epidemiological studies.
Collapse
Affiliation(s)
- Tonny J. Owalla
- Department of Immunology and Parasitology, Med Biotech Laboratories, Kampala, Uganda
| | - Dianna E. B. Hergott
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States
| | - Annette M. Seilie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Weston Staubus
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Chris Chavtur
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Ming Chang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - James G. Kublin
- Department of Global Health, University of Washington, Seattle, WA, United States,Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Thomas G. Egwang
- Department of Immunology and Parasitology, Med Biotech Laboratories, Kampala, Uganda
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States,Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States,Department of Microbiology, University of Washington, Seattle, WA, United States,*Correspondence: Sean C. Murphy,
| |
Collapse
|
33
|
Nunes-Cabaço H, Moita D, Prudêncio M. Five decades of clinical assessment of whole-sporozoite malaria vaccines. Front Immunol 2022; 13:977472. [PMID: 36159849 PMCID: PMC9493004 DOI: 10.3389/fimmu.2022.977472] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
In 1967, pioneering work by Ruth Nussenzweig demonstrated for the first time that irradiated sporozoites of the rodent malaria parasite Plasmodium berghei protected mice against a challenge with infectious parasites of the same species. This remarkable finding opened up entirely new prospects of effective vaccination against malaria using attenuated sporozoites as immunization agents. The potential for whole-sporozoite-based immunization in humans was established in a clinical study in 1973, when a volunteer exposed to X-irradiated P. falciparum sporozoites was found to be protected against malaria following challenge with a homologous strain of this parasite. Nearly five decades later, much has been achieved in the field of whole-sporozoite malaria vaccination, and multiple reports on the clinical evaluation of such candidates have emerged. However, this process has known different paces before and after the turn of the century. While only a few clinical studies were published in the 1970’s, 1980’s and 1990’s, remarkable progress was made in the 2000’s and beyond. This article reviews the history of the clinical assessment of whole-sporozoite malaria vaccines over the last forty-nine years, highlighting the impressive achievements made over the last few years, and discussing some of the challenges ahead.
Collapse
|
34
|
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: 1.3] [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
|
35
|
Mordmüller B, Sulyok Z, Sulyok M, Molnar Z, Lalremruata A, Calle CL, Bayon PG, Esen M, Gmeiner M, Held J, Heimann HL, Woldearegai TG, Ibáñez J, Flügge J, Fendel R, Kreidenweiss A, Kc N, Murshedkar T, Chakravarty S, Riyahi P, Billingsley PF, Church LWP, Richie TL, Sim BKL, Hoffman SL, Kremsner PG. 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: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [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
Affiliation(s)
- Benjamin Mordmüller
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany.
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany.
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Zita Sulyok
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Mihály Sulyok
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Zsofia Molnar
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Albert Lalremruata
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Carlos Lamsfus Calle
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Patricia Granados Bayon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Meral Esen
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Markus Gmeiner
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jana Held
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Henri-Lynn Heimann
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Tamirat Gebru Woldearegai
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Javier Ibáñez
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Judith Flügge
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Rolf Fendel
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
| | - Andrea Kreidenweiss
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Natasha Kc
- Sanaria Inc., Rockville, MD, USA
- Protein Potential, LLC, Rockville, MD, USA
| | | | | | | | | | | | | | - B Kim Lee Sim
- Sanaria Inc., Rockville, MD, USA
- Protein Potential, LLC, Rockville, MD, USA
| | | | - Peter G Kremsner
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|
36
|
Visweswaran GRR, Vijayan K, Chandrasekaran R, Trakhimets O, Brown SL, Vigdorovich V, Yang A, Raappana A, Watson A, Selman W, Zuck M, Dambrauskas N, Kaushansky A, Sather DN. Germinal center activity and B cell maturation are associated with protective antibody responses against Plasmodium pre-erythrocytic infection. PLoS Pathog 2022; 18:e1010671. [PMID: 35793394 PMCID: PMC9292112 DOI: 10.1371/journal.ppat.1010671] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/18/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022] Open
Abstract
Blocking Plasmodium, the causative agent of malaria, at the asymptomatic pre-erythrocytic stage would abrogate disease pathology and prevent transmission. However, the lack of well-defined features within vaccine-elicited antibody responses that correlate with protection represents a major roadblock to improving on current generation vaccines. We vaccinated mice (BALB/cJ and C57BL/6J) with Py circumsporozoite protein (CSP), the major surface antigen on the sporozoite, and evaluated vaccine-elicited humoral immunity and identified immunological factors associated with protection after mosquito bite challenge. Vaccination achieved 60% sterile protection and otherwise delayed blood stage patency in BALB/cJ mice. In contrast, all C57BL/6J mice were infected similar to controls. Protection was mediated by antibodies and could be passively transferred from immunized BALB/cJ mice into naïve C57BL/6J. Dissection of the underlying immunological features of protection revealed early deficits in antibody titers and polyclonal avidity in C57BL/6J mice. Additionally, PyCSP-vaccination in BALB/cJ induced a significantly higher proportion of antigen-specific B-cells and class-switched memory B-cell (MBCs) populations than in C57BL/6J mice. Strikingly, C57BL/6J mice also had markedly fewer CSP-specific germinal center experienced B cells and class-switched MBCs compared to BALB/cJ mice. Analysis of the IgG γ chain repertoires by next generation sequencing in PyCSP-specific memory B-cell repertoires also revealed higher somatic hypermutation rates in BALB/cJ mice than in C57BL/6J mice. These findings indicate that the development of protective antibody responses in BALB/cJ mice in response to vaccination with PyCSP was associated with increased germinal center activity and somatic mutation compared to C57BL/6J mice, highlighting the key role B cell maturation may have in the development of vaccine-elicited protective antibodies against CSP. Identifying specific features of vaccine-elicited antibody responses that are associated with protection from malaria infection is a key step toward the development of a safe and effective vaccine. Here we compared antibody and B cell responses in two mouse strains that exhibited a differential ability to generate antibodies that protect from infection challenge. We found that protection was due to the presence of vaccine-elicited antibodies and could be transferred between strains, and that the ability of antibodies to neutralize the parasite was directly linked to the strength (affinity) with which it binds CSP. Thus, we sought to understand if there were differences in the two strains in the process of B cell maturation that leads to generation of high affinity, protective antibody responses after vaccination. Overall, our comparative analysis indicates that germinal center (GC) activity, a key process in B cell maturation, was significantly diminished in the non-protected strain. Further, we observed evidence of higher levels of somatic mutation, which is a result of germinal center activity, in protected mice. Thus, our results indicate that the ability to generate protective antibody responses was linked to enhanced B cell maturation in the protected strain, providing a key clue to the type of responses that should be generated by future vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ashton Yang
- Seattle Children’s Research Institute, Seattle, Washington
| | | | - Alex Watson
- Seattle Children’s Research Institute, Seattle, Washington
| | - William Selman
- Seattle Children’s Research Institute, Seattle, Washington
| | - Meghan Zuck
- Seattle Children’s Research Institute, Seattle, Washington
| | | | - Alexis Kaushansky
- Seattle Children’s Research Institute, Seattle, Washington
- Department of Pediatrics, University of Washington, Seattle, Washington
- Department of Global Health, University of Washington, Seattle, Washington
- Brotman Baty Research Institute, Seattle, Washington
- Institute for Stem Cell and Regenerative Medicine, Seattle, Washington
- * E-mail: (AK); (DNS)
| | - D. Noah Sather
- Seattle Children’s Research Institute, Seattle, Washington
- Department of Pediatrics, University of Washington, Seattle, Washington
- Department of Global Health, University of Washington, Seattle, Washington
- * E-mail: (AK); (DNS)
| |
Collapse
|
37
|
Silva JC, Dwivedi A, Moser KA, Sissoko MS, Epstein JE, Healy SA, Lyke KE, Mordmüller B, Kremsner PG, Duffy PE, Murshedkar T, Sim BKL, Richie TL, Hoffman SL. Plasmodium falciparum 7G8 challenge provides conservative prediction of efficacy of PfNF54-based PfSPZ Vaccine in Africa. Nat Commun 2022; 13:3390. [PMID: 35697668 PMCID: PMC9189790 DOI: 10.1038/s41467-022-30882-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/24/2022] [Indexed: 11/26/2022] Open
Abstract
Controlled human malaria infection (CHMI) has supported Plasmodium falciparum (Pf) malaria vaccine development by providing preliminary estimates of vaccine efficacy (VE). Because CHMIs generally use Pf strains similar to vaccine strains, VE against antigenically heterogeneous Pf in the field has been required to establish VE. We increased the stringency of CHMI by selecting a Brazilian isolate, Pf7G8, which is genetically distant from the West African parasite (PfNF54) in our PfSPZ vaccines. Using two regimens to identically immunize US and Malian adults, VE over 24 weeks in the field was as good as or better than VE against CHMI at 24 weeks in the US. To explain this finding, here we quantify differences in the genome, proteome, and predicted CD8 T cell epitopes of PfNF54 relative to 704 Pf isolates from Africa and Pf7G8. We show that Pf7G8 is more distant from PfNF54 than any African isolates tested. We propose VE against Pf7G8 CHMI for providing pivotal data for malaria vaccine licensure for travelers to Africa, and potentially for endemic populations, because the genetic distance of Pf7G8 from the Pf vaccine strain makes it a stringent surrogate for Pf parasites in Africa. Here the authors show that controlled human malaria infection with a Brazilian parasite highly divergent from vaccine and West African field strains can provide estimates of vaccine efficacy in Mali, and could replace field testing, streamlining vaccine development.
Collapse
Affiliation(s)
- Joana C Silva
- Institute for Genomic Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ankit Dwivedi
- Institute for Genomic Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kara A Moser
- Institute for Genomic Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mahamadou S Sissoko
- Malaria Research and Training Center, Mali National Institute of Allergy and Infectious Diseases International Centers for Excellence in Research, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Judith E Epstein
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, NIAID, NIH, Bethesda, MD, USA
| | - Kirsten E Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Benjamin Mordmüller
- Institute of Tropical Medicine, University of Tübingen and German Center for Infection Research, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter G Kremsner
- Institute of Tropical Medicine, University of Tübingen and German Center for Infection Research, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, NIAID, NIH, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
38
|
Mulamba C, Williams C, Kreppel K, Ouedraogo JB, Olotu AI. Evaluation of the Pfs25-IMX313/Matrix-M malaria transmission-blocking candidate vaccine in endemic settings. Malar J 2022; 21:159. [PMID: 35655174 PMCID: PMC9161629 DOI: 10.1186/s12936-022-04173-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/02/2022] [Indexed: 11/10/2022] Open
Abstract
Malaria control relies heavily on the use of anti-malarial drugs and insecticides against malaria parasites and mosquito vectors. Drug and insecticide resistance threatens the effectiveness of conventional malarial interventions; alternative control approaches are, therefore, needed. The development of malaria transmission-blocking vaccines that target the sexual stages in humans or mosquito vectors is among new approaches being pursued. Here, the immunological mechanisms underlying malaria transmission blocking, status of Pfs25-based vaccines are viewed, as well as approaches and capacity for first in-human evaluation of a transmission-blocking candidate vaccine Pfs25-IMX313/Matrix-M administered to semi-immune healthy individuals in endemic settings. It is concluded that institutions in low and middle income settings should be supported to conduct first-in human vaccine trials in order to stimulate innovative research and reduce the overdependence on developed countries for research and local interventions against many diseases of public health importance.
Collapse
Affiliation(s)
- Charles Mulamba
- Interventions & Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania.,Nelson Mandela African Institution of Science and Technology, Tengeru, P. O. Box 447, Arusha, Tanzania
| | - Chris Williams
- The Jenner Institute, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7DQ, UK
| | - Katharina Kreppel
- Nelson Mandela African Institution of Science and Technology, Tengeru, P. O. Box 447, Arusha, Tanzania
| | | | - Ally I Olotu
- Interventions & Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania.
| |
Collapse
|
39
|
Crabtree JN, Caffrey DR, de Souza Silva L, Kurt-Jones EA, Dobbs K, Dent A, Fitzgerald KA, Golenbock DT. Lymphocyte crosstalk is required for monocyte-intrinsic trained immunity to Plasmodium falciparum. J Clin Invest 2022; 132:e139298. [PMID: 35642634 PMCID: PMC9151696 DOI: 10.1172/jci139298] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 04/21/2022] [Indexed: 01/16/2023] Open
Abstract
Plasmodium falciparum (P. falciparum) induces trained innate immune responses in vitro, where initial stimulation of adherent PBMCs with P. falciparum-infected RBCs (iRBCs) results in hyperresponsiveness to subsequent ligation of TLR2. This response correlates with the presence of T and B lymphocytes in adherent PBMCs, suggesting that innate immune training is partially due to adaptive immunity. We found that T cell-depleted PBMCs and purified monocytes alone did not elicit hyperproduction of IL-6 and TNF-α under training conditions. Analysis of P. falciparum-trained PBMCs showed that DCs did not develop under control conditions, and IL-6 and TNF-α were primarily produced by monocytes and DCs. Transwell experiments isolating purified monocytes from either PBMCs or purified CD4+ T cells, but allowing diffusion of secreted proteins, enabled monocytes trained with iRBCs to hyperproduce IL-6 and TNF-α after TLR restimulation. Purified monocytes stimulated with IFN-γ hyperproduced IL-6 and TNF-α, whereas blockade of IFN-γ in P. falciparum-trained PBMCs inhibited trained responses. Assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-Seq) on monocytes from patients with malaria showed persistently open chromatin at genes that appeared to be trained in vitro. Together, these findings indicate that the trained immune response of monocytes to P. falciparum is not completely cell intrinsic but depends on soluble signals from lymphocytes.
Collapse
Affiliation(s)
- Juliet N. Crabtree
- Program in Innate Immunity and
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Daniel R. Caffrey
- Program in Innate Immunity and
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Leandro de Souza Silva
- Program in Innate Immunity and
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Evelyn A. Kurt-Jones
- Program in Innate Immunity and
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | | - Arlene Dent
- Case Western University, Cleveland, Ohio, USA
| | - Katherine A. Fitzgerald
- Program in Innate Immunity and
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Douglas T. Golenbock
- Program in Innate Immunity and
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
40
|
Nunes-Cabaço H, Moita D, Rôla C, Mendes AM, Prudêncio M. Impact of Dietary Protein Restriction on the Immunogenicity and Efficacy of Whole-Sporozoite Malaria Vaccination. Front Immunol 2022; 13:869757. [PMID: 35529859 PMCID: PMC9070679 DOI: 10.3389/fimmu.2022.869757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/28/2022] [Indexed: 11/24/2022] Open
Abstract
Malaria remains one of the world’s most prevalent infectious diseases. Several vaccination strategies currently under investigation aim at hampering the development of the Plasmodium parasite during the clinically silent liver stage of its life cycle in the mammalian host, preventing the subsequent disease-associated blood stage of infection. Immunization with radiation-attenuated sporozoites (RAS), the liver-infecting parasite forms, can induce sterile protection against malaria. However, the efficacy of vaccine candidates in malaria-naïve individuals in high-income countries is frequently higher than that found in populations where malaria is endemic. Malnutrition has been associated with immune dysfunction and with a delay or impairment of the immune response to some vaccines. Since vaccine efficacy depends on the generation of competent immune responses, and malaria-endemic regions are often associated with malnutrition, we hypothesized that an inadequate host nutritional status, specifically resulting from a reduction in dietary protein, could impact on the establishment of an efficient anti-malarial immune response. We developed a model of RAS immunization under low protein diet to investigate the impact of a reduced host protein intake on the immunogenicity and protective efficacy of this vaccine. Our analysis of the circulating and tissue-associated immune compartments revealed that a reduction in dietary protein intake during immunization resulted in a decrease in the frequency of circulating CD4+ T cells and of hepatic NK cells. Nevertheless, the profile of CD8+ T cells in the blood, liver and spleen was robust and minimally affected by the dietary protein content during RAS immunization, as assessed by supervised and in-depth unsupervised X-shift clustering analysis. Although mice immunized under low protein diet presented higher parasite liver load upon challenge than those immunized under adequate protein intake, the two groups displayed similar levels of protection from disease. Overall, our data indicate that dietary protein reduction may have minimal impact on the immunogenicity and efficacy of RAS-based malaria vaccination. Importantly, this experimental model can be extended to assess the impact of other nutrient imbalances and immunization strategies, towards the refinement of future translational interventions that improve vaccine efficacy in malnourished individuals.
Collapse
|
41
|
Aleshnick M, Florez-Cuadros M, Martinson T, Wilder BK. Monoclonal antibodies for malaria prevention. Mol Ther 2022; 30:1810-1821. [PMID: 35395399 PMCID: PMC8979832 DOI: 10.1016/j.ymthe.2022.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/04/2022] [Accepted: 04/01/2022] [Indexed: 11/29/2022] Open
Abstract
Monoclonal antibodies are highly specific proteins that are cloned from a single B cell and bind to a single epitope on a pathogen. These laboratory-made molecules can serve as prophylactics or therapeutics for infectious diseases and have an impressive capacity to modulate the progression of disease, as demonstrated for the first time on a large scale during the COVID-19 pandemic. The high specificity and natural starting point of monoclonal antibodies afford an encouraging safety profile, yet the high cost of production remains a major limitation to their widespread use. While a monoclonal antibody approach to abrogating malaria infection is not yet available, the unique life cycle of the malaria parasite affords many opportunities for such proteins to act, and preliminary research into the efficacy of monoclonal antibodies in preventing malaria infection, disease, and transmission is encouraging. This review examines the current status and future outlook for monoclonal antibodies against malaria in the context of the complex life cycle and varied antigenic targets expressed in the human and mosquito hosts, and provides insight into the strengths and limitations of this approach to curtailing one of humanity’s oldest and deadliest diseases.
Collapse
Affiliation(s)
- Maya Aleshnick
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | - Thomas Martinson
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Brandon K Wilder
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA; Department of Parasitology, U.S. Naval Medical Research 6 (NAMRU-6), Lima, Peru
| |
Collapse
|
42
|
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: 0.3] [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
|
43
|
Daubenberger C. Assessment of experimental malaria vaccine induced protection in pre-exposed populations. THE LANCET. INFECTIOUS DISEASES 2022; 22:305-307. [PMID: 34801111 DOI: 10.1016/s1473-3099(21)00359-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 06/13/2023]
Affiliation(s)
- Claudia Daubenberger
- Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland; University of Basel, Basel, Switzerland.
| |
Collapse
|
44
|
Preimmunization correlates of protection shared across malaria vaccine trials in adults. NPJ Vaccines 2022; 7:5. [PMID: 35031601 PMCID: PMC8760258 DOI: 10.1038/s41541-021-00425-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/02/2021] [Indexed: 12/15/2022] Open
Abstract
Identifying preimmunization biological characteristics that promote an effective vaccine response offers opportunities for illuminating the critical immunological mechanisms that confer vaccine-induced protection, for developing adjuvant strategies, and for tailoring vaccination regimens to individuals or groups. In the context of malaria vaccine research, studying preimmunization correlates of protection can help address the need for a widely effective malaria vaccine, which remains elusive. In this study, common preimmunization correlates of protection were identified using transcriptomic data from four independent, heterogeneous malaria vaccine trials in adults. Systems-based analyses showed that a moderately elevated inflammatory state prior to immunization was associated with protection against malaria challenge. Functional profiling of protection-associated genes revealed the importance of several inflammatory pathways, including TLR signaling. These findings, which echo previous studies that associated enhanced preimmunization inflammation with protection, illuminate common baseline characteristics that set the stage for an effective vaccine response across diverse malaria vaccine strategies in adults.
Collapse
|
45
|
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: 0.5] [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
|
46
|
Lefebvre MN, Drewry LL, Pewe LL, Hancox LS, Reyes-Sandoval A, Harty JT. Cutting Edge: Subunit Booster Vaccination Confers Sterilizing Immunity against Liver-Stage Malaria in Mice Initially Primed with a Weight-Normalized Dose of Radiation-Attenuated Sporozoites. THE JOURNAL OF IMMUNOLOGY 2021; 207:2631-2635. [PMID: 34716185 DOI: 10.4049/jimmunol.2100818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/25/2021] [Indexed: 11/19/2022]
Abstract
Radiation-attenuated sporozoite (RAS) vaccination offers hope for global malaria control through induction of protective liver-stage-specific memory CD8 T cells. Effective RAS vaccination regimens exist; however, widespread implementation remains unfeasible. A key difficulty resides in the need to administer three or more doses i.v. to achieve sufficient immunity. Strategies to reduce the number of RAS doses are therefore desirable. Here we used mice to model human immune responses to a single, suboptimal weight-normalized RAS dose administered i.v. followed by subunit vaccination to amplify liver-stage-specific memory CD8 T cells. RAS+subunit prime-boost regimens increased the numbers of liver-stage-specific memory CD8 T cells to a level greater than is present after one RAS vaccination. Both i.v. and i.m. subunit vaccine delivery induced immunity in mice, and many vaccinated mice completely cleared liver infection. These findings are particularly relevant to human vaccine development because RAS+subunit prime-boost vaccination would reduce the logistical challenges of multiple RAS-only immunizations.
Collapse
Affiliation(s)
- Mitchell N Lefebvre
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA; and
| | - Lisa L Drewry
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Lecia L Pewe
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Lisa S Hancox
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | - John T Harty
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; .,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA; and
| |
Collapse
|
47
|
Lefebvre MN, Surette FA, Anthony SM, Vijay R, Jensen IJ, Pewe LL, Hancox LS, Van Braeckel-Budimir N, van de Wall S, Urban SL, Mix MR, Kurup SP, Badovinac VP, Butler NS, Harty JT. Expeditious recruitment of circulating memory CD8 T cells to the liver facilitates control of malaria. Cell Rep 2021; 37:109956. [PMID: 34731605 PMCID: PMC8628427 DOI: 10.1016/j.celrep.2021.109956] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/08/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Circulating memory CD8 T cell trafficking and protective capacity during liver-stage malaria infection remains undefined. We find that effector memory CD8 T cells (Tem) infiltrate the liver within 6 hours after malarial or bacterial infections and mediate pathogen clearance. Tem recruitment coincides with rapid transcriptional upregulation of inflammatory genes in Plasmodium-infected livers. Recruitment requires CD8 T cell-intrinsic LFA-1 expression and the presence of liver phagocytes. Rapid Tem liver infiltration is distinct from recruitment to other non-lymphoid tissues in that it occurs both in the absence of liver tissue resident memory "sensing-and-alarm" function and ∼42 hours earlier than in lung infection by influenza virus. These data demonstrate relevance for Tem in protection against malaria and provide generalizable mechanistic insights germane to control of liver infections.
Collapse
Affiliation(s)
- Mitchell N Lefebvre
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Medical Scientist Training Program, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA
| | - Fionna A Surette
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA; Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Scott M Anthony
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Rahul Vijay
- Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Isaac J Jensen
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA
| | - Lecia L Pewe
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Lisa S Hancox
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | | | - Stephanie van de Wall
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Stina L Urban
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Madison R Mix
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Medical Scientist Training Program, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA
| | - Samarchith P Kurup
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA; Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Noah S Butler
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA; Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - John T Harty
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA.
| |
Collapse
|
48
|
Immunosuppression in Malaria: Do Plasmodium falciparum Parasites Hijack the Host? Pathogens 2021; 10:pathogens10101277. [PMID: 34684226 PMCID: PMC8536967 DOI: 10.3390/pathogens10101277] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Malaria reflects not only a state of immune activation, but also a state of general immune defect or immunosuppression, of complex etiology that can last longer than the actual episode. Inhabitants of malaria-endemic regions with lifelong exposure to the parasite show an exhausted or immune regulatory profile compared to non- or minimally exposed subjects. Several studies and experiments to identify and characterize the cause of this malaria-related immunosuppression have shown that malaria suppresses humoral and cellular responses to both homologous (Plasmodium) and heterologous antigens (e.g., vaccines). However, neither the underlying mechanisms nor the relative involvement of different types of immune cells in immunosuppression during malaria is well understood. Moreover, the implication of the parasite during the different stages of the modulation of immunity has not been addressed in detail. There is growing evidence of a role of immune regulators and cellular components in malaria that may lead to immunosuppression that needs further research. In this review, we summarize the current evidence on how malaria parasites may directly and indirectly induce immunosuppression and investigate the potential role of specific cell types, effector molecules and other immunoregulatory factors.
Collapse
|
49
|
Safety, immunogenicity and efficacy of PfSPZ Vaccine against malaria in infants in western Kenya: a double-blind, randomized, placebo-controlled phase 2 trial. Nat Med 2021; 27:1636-1645. [PMID: 34518679 DOI: 10.1038/s41591-021-01470-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/16/2021] [Indexed: 01/13/2023]
Abstract
The radiation-attenuated Plasmodium falciparum sporozoite (PfSPZ) vaccine provides protection against P. falciparum infection in malaria-naïve adults. Preclinical studies show that T cell-mediated immunity is required for protection and is readily induced in humans after vaccination. However, previous malaria exposure can limit immune responses and vaccine efficacy (VE) in adults. We hypothesized that infants with less previous exposure to malaria would have improved immunity and protection. We conducted a multi-arm, randomized, double-blind, placebo-controlled trial in 336 infants aged 5-12 months to determine the safety, tolerability, immunogenicity and efficacy of the PfSPZ Vaccine in infants in a high-transmission malaria setting in western Kenya ( NCT02687373 ). Groups of 84 infants each received 4.5 × 105, 9.0 × 105 or 1.8 × 106 PfSPZ Vaccine or saline three times at 8-week intervals. The vaccine was well tolerated; 52 (20.6%) children in the vaccine groups and 20 (23.8%) in the placebo group experienced related solicited adverse events (AEs) within 28 d postvaccination and most were mild. There was 1 grade 3-related solicited AE in the vaccine group (0.4%) and 2 in the placebo group (2.4%). Seizures were more common in the highest-dose group (14.3%) compared to 6.0% of controls, with most being attributed to malaria. There was no significant protection against P. falciparum infection in any dose group at 6 months (VE in the 9.0 × 105 dose group = -6.5%, P = 0.598, the primary statistical end point of the study). VE against clinical malaria 3 months after the last dose in the highest-dose group was 45.8% (P = 0.027), an exploratory end point. There was a dose-dependent increase in antibody responses that correlated with VE at 6 months in the lowest- and highest-dose groups. T cell responses were undetectable across all dose groups. Detection of Vδ2+Vγ9+ T cells, which have been correlated with induction of PfSPZ Vaccine T cell immunity and protection in adults, were infrequent. These data suggest that PfSPZ Vaccine-induced T cell immunity is age-dependent and may be influenced by Vδ2+Vγ9+ T cell frequency. Since there was no significant VE at 6 months in these infants, these vaccine regimens will likely not be pursued further in this age group.
Collapse
|
50
|
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: 3.5] [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
|