1
|
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 DOI: 10.1016/j.ebiom.2024.105419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
2
|
Wang LT, Idris AH, Kisalu NK, Crompton PD, Seder RA. Monoclonal antibodies to the circumsporozoite proteins as an emerging tool for malaria prevention. Nat Immunol 2024; 25:1530-1545. [PMID: 39198635 DOI: 10.1038/s41590-024-01938-2] [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: 05/06/2024] [Accepted: 07/17/2024] [Indexed: 09/01/2024]
Abstract
Despite various public health strategies, malaria caused by Plasmodium falciparum parasites remains a major global health challenge that requires development of new interventions. Extended half-life human monoclonal antibodies targeting the P. falciparum circumsporozoite protein on sporozoites, the infective form of malaria parasites, prevent malaria in rodents and humans and have been advanced into clinical development. The protective epitopes on the circumsporozoite protein targeted by monoclonal antibodies have been defined. Cryogenic electron and multiphoton microscopy have enabled mechanistic structural and functional investigations of how antibodies bind to the circumsporozoite protein and neutralize sporozoites. Moreover, innovations in bioinformatics and antibody engineering have facilitated enhancement of antibody potency and durability. Here, we summarize the latest scientific advances in understanding how monoclonal antibodies to the circumsporozoite protein prevent malaria and highlight existing clinical data and future plans for how this emerging intervention can be used alone or alongside existing antimalarial interventions to control malaria across at-risk populations.
Collapse
Affiliation(s)
- Lawrence T Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Azza H Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| | - Neville K Kisalu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- PATH's Center for Vaccine Innovation and Access, Washington, DC, USA
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Duffy PE, Gorres JP, Healy SA, Fried M. Malaria vaccines: a new era of prevention and control. Nat Rev Microbiol 2024:10.1038/s41579-024-01065-7. [PMID: 39025972 DOI: 10.1038/s41579-024-01065-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2024] [Indexed: 07/20/2024]
Abstract
Malaria killed over 600,000 people in 2022, a death toll that has not improved since 2015. Additionally, parasites and mosquitoes resistant to existing interventions are spreading across Africa and other regions. Vaccines offer hope to reduce the mortality burden: the first licensed malaria vaccines, RTS,S and R21, will be widely deployed in 2024 and should substantially reduce childhood deaths. In this Review, we provide an overview of the malaria problem and the Plasmodium parasite, then describe the RTS,S and R21 vaccines (the first vaccines for any human parasitic disease), summarizing their benefits and limitations. We explore next-generation vaccines designed using new knowledge of malaria pathogenesis and protective immunity, which incorporate antigens and platforms to elicit effective immune responses against different parasite stages in human or mosquito hosts. We describe a decision-making process that prioritizes malaria vaccine candidates for development in a resource-constrained environment. Future vaccines might improve upon the protective efficacy of RTS,S or R21 for children, or address the wider malaria scourge by preventing pregnancy malaria, reducing the burden of Plasmodium vivax or accelerating malaria elimination.
Collapse
Affiliation(s)
- Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - J Patrick Gorres
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Blanken SL, Barry A, Lanke K, Guelbeogo M, Ouedraogo A, Soulama I, Coulibaly SA, Teelen K, Graumans W, Dumont E, Stone W, Ramjith J, Marti M, Andrade CM, Drakeley C, Collins K, Tiono A, Bousema T. Plasmodium falciparum gametocyte production correlates with genetic markers of parasite replication but is not influenced by experimental exposure to mosquito biting. EBioMedicine 2024; 105:105190. [PMID: 38901148 PMCID: PMC11239461 DOI: 10.1016/j.ebiom.2024.105190] [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: 11/17/2023] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Plasmodium blood-stage parasites balance asexual multiplication with gametocyte development. Few studies link these dynamics with parasite genetic markers in vivo; even fewer in longitudinally monitored infections. Environmental influences on gametocyte formation, such as mosquito exposure, may influence the parasite's investment in gametocyte production. METHODS We investigated gametocyte production and asexual multiplication in two Plasmodium falciparum infected populations; a controlled human malaria infection (CHMI) study and a 28-day observational study in naturally infected individuals in Burkina Faso with controlled mosquito exposure. We measured gene transcript levels previously related to gametocyte formation (ap2-g, surfin1.2, surfin13.1, gexp-2) or inhibition of asexual multiplication (sir2a) and compared transcript levels to ring-stage parasite and mature gametocyte densities. FINDINGS Three of the five markers (ap2-g, surfin1.2, surfin13.1) predicted peak gametocytaemia in the CHMI study. An increase in all five markers in natural infections was associated with an increase in mature gametocytes 14 days later; the effect of sir2a on future gametocytes was strongest (fold change = 1.65, IQR = 1.22-2.24, P = 0.004). Mosquito exposure was not associated with markers of gametocyte formation (ap2-g P = 0.277; sir2a P = 0.499) or carriage of mature gametocytes (P = 0.379). INTERPRETATION All five parasite genetic markers predicted gametocyte formation over a single cycle of gametocyte formation and maturation in vivo; sir2a and ap2-g were most closely associated with gametocyte growth dynamics. We observed no evidence to support the hypothesis that exposure to Anopheles mosquito bites stimulates gametocyte formation. FUNDING This work was funded by the Bill & Melinda Gates Foundation (INDIE OPP1173572), the European Research Council fellowship (ERC-CoG 864180) and UKRI Medical Research Council (MR/T016272/1) and Wellcome Center (218676/Z/19/Z).
Collapse
Affiliation(s)
- Sara Lynn Blanken
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Aissata Barry
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Kjerstin Lanke
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Moussa Guelbeogo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Alphonse Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Issiaka Soulama
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Sam Aboubacar Coulibaly
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Karina Teelen
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Wouter Graumans
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Elin Dumont
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Will Stone
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Jordache Ramjith
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, Institute of Infection and Immunity, University of Glasgow, Glasgow, Scotland, UK
| | - Carolina M Andrade
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Chris Drakeley
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Katharine Collins
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Alfred Tiono
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
5
|
Hofer LM, Kweyamba PA, Sayi RM, Chabo MS, Maitra SL, Moore SJ, Tambwe MM. Malaria rapid diagnostic tests reliably detect asymptomatic Plasmodium falciparum infections in school-aged children that are infectious to mosquitoes. Parasit Vectors 2023; 16:217. [PMID: 37391770 DOI: 10.1186/s13071-023-05761-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/29/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Asymptomatic malaria infections (Plasmodium falciparum) are common in school-aged children and represent a disease transmission reservoir as they are potentially infectious to mosquitoes. To detect and treat such infections, convenient, rapid and reliable diagnostic tools are needed. In this study, malaria rapid diagnostic tests (mRDT), light microscopy (LM) and quantitative polymerase chain reaction (qPCR) were used to evaluate their performance detecting asymptomatic malaria infections that are infectious to mosquitoes. METHODS One hundred seventy asymptomatic school-aged children (6-14 years old) from the Bagamoyo district in Tanzania were screened for Plasmodium spp. infections using mRDT (SD BIOLINE), LM and qPCR. In addition, gametocytes were detected using reverse transcription quantitative polymerase chain reaction (RT-qPCR) for all qPCR-positive children. Venous blood from all P. falciparum positive children was fed to female Anopheles gambiae sensu stricto mosquitoes via direct membrane feeding assays (DMFAs) after serum replacement. Mosquitoes were dissected for oocyst infections on day 8 post-infection. RESULTS The P. falciparum prevalence in study participants was 31.7% by qPCR, 18.2% by mRDT and 9.4% by LM. Approximately one-third (31.2%) of asymptomatic malaria infections were infectious to mosquitoes in DMFAs. In total, 297 infected mosquitoes were recorded after dissections, from which 94.9% (282/297) were derived from infections detected by mRDT and 5.1% (15/297) from subpatent mRDT infections. CONCLUSION The mRDT can be used reliably to detect children carrying gametocyte densities sufficient to infect high numbers of mosquitoes. Subpatent mRDT infections contributed marginally to the pool of oocyts-infected mosquitoes.
Collapse
Affiliation(s)
- Lorenz M Hofer
- Vector Biology Unit, Department of Epidemiology and Public Health, Swiss Tropical and Public Health, Institute, Kreuzstrasse 2, 4123, Allschwil, Basel, Switzerland
- University of Basel, Petersplatz 1, 4001, Basel, Switzerland
- Vector Control Product Testing Unit (VCPTU) Ifakara Health Institute, Environmental Health, and Ecological Sciences, 74, Bagamoyo, Tanzania
| | - Prisca A Kweyamba
- Vector Control Product Testing Unit (VCPTU) Ifakara Health Institute, Environmental Health, and Ecological Sciences, 74, Bagamoyo, Tanzania
| | - Rajabu M Sayi
- Vector Control Product Testing Unit (VCPTU) Ifakara Health Institute, Environmental Health, and Ecological Sciences, 74, Bagamoyo, Tanzania
| | - Mohamed S Chabo
- Vector Control Product Testing Unit (VCPTU) Ifakara Health Institute, Environmental Health, and Ecological Sciences, 74, Bagamoyo, Tanzania
| | - Sonali L Maitra
- Vector Biology Unit, Department of Epidemiology and Public Health, Swiss Tropical and Public Health, Institute, Kreuzstrasse 2, 4123, Allschwil, Basel, Switzerland
- University of Basel, Petersplatz 1, 4001, Basel, Switzerland
- Vector Control Product Testing Unit (VCPTU) Ifakara Health Institute, Environmental Health, and Ecological Sciences, 74, Bagamoyo, Tanzania
| | - Sarah J Moore
- Vector Biology Unit, Department of Epidemiology and Public Health, Swiss Tropical and Public Health, Institute, Kreuzstrasse 2, 4123, Allschwil, Basel, Switzerland
- University of Basel, Petersplatz 1, 4001, Basel, Switzerland
- Vector Control Product Testing Unit (VCPTU) Ifakara Health Institute, Environmental Health, and Ecological Sciences, 74, Bagamoyo, Tanzania
- The Nelson Mandela African Institution of Science and Technology (NM-AIST), 447, Tengeru, Arusha, Tanzania
| | - Mgeni M Tambwe
- Vector Control Product Testing Unit (VCPTU) Ifakara Health Institute, Environmental Health, and Ecological Sciences, 74, Bagamoyo, Tanzania.
| |
Collapse
|
6
|
Umumararungu T, Nkuranga JB, Habarurema G, Nyandwi JB, Mukazayire MJ, Mukiza J, Muganga R, Hahirwa I, Mpenda M, Katembezi AN, Olawode EO, Kayitare E, Kayumba PC. Recent developments in antimalarial drug discovery. Bioorg Med Chem 2023; 88-89:117339. [PMID: 37236020 DOI: 10.1016/j.bmc.2023.117339] [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: 03/01/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023]
Abstract
Although malaria remains a big burden to many countries that it threatens their socio-economic stability, particularly in the countries where malaria is endemic, there have been great efforts to eradicate this disease with both successes and failures. For example, there has been a great improvement in malaria prevention and treatment methods with a net reduction in infection and mortality rates. However, the disease remains a global threat in terms of the number of people affected because it is one of the infectious diseases that has the highest prevalence rate, especially in Africa where the deadly Plasmodium falciparum is still widely spread. Methods to fight malaria are being diversified, including the use of mosquito nets, the target candidate profiles (TCPs) and target product profiles (TPPs) of medicine for malarial venture (MMV) strategy, the search for newer and potent drugs that could reverse chloroquine resistance, and the use of adjuvants such as rosiglitazone and sevuparin. Although these adjuvants have no antiplasmodial activity, they can help to alleviate the effects which result from plasmodium invasion such as cytoadherence. The list of new antimalarial drugs under development is long, including the out of ordinary new drugs MMV048, CDRI-97/78 and INE963 from South Africa, India and Novartis, respectively.
Collapse
Affiliation(s)
- Théoneste Umumararungu
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda.
| | - Jean Bosco Nkuranga
- Department of Chemistry, School of Science, College of Science and Technology, University of Rwanda, Rwanda
| | - Gratien Habarurema
- Department of Chemistry, School of Science, College of Science and Technology, University of Rwanda, Rwanda
| | - Jean Baptiste Nyandwi
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Marie Jeanne Mukazayire
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Janvier Mukiza
- Department of Mathematical Science and Physical Education, School of Education, College of Education, University of Rwanda, Rwanda; Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda
| | - Raymond Muganga
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda; Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda
| | - Innocent Hahirwa
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Matabishi Mpenda
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Alain Nyirimigabo Katembezi
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda; Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda
| | - Emmanuel Oladayo Olawode
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, 18301 N Miami Ave #1, Miami, FL 33169, USA
| | - Egide Kayitare
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Pierre Claver Kayumba
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| |
Collapse
|
7
|
Webster R, Mitchell H, Peters JM, Heunis J, O'Neill B, Gower J, Lynch S, Jennings H, Amante FH, Llewellyn S, Marquart L, Potter AJ, Birrell GW, Edstein MD, Shanks GD, McCarthy JS, Barber BE. Transmission Blocking Activity of Low-dose Tafenoquine in Healthy Volunteers Experimentally Infected With Plasmodium falciparum. Clin Infect Dis 2023; 76:506-512. [PMID: 35731843 DOI: 10.1093/cid/ciac503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/13/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Blocking the transmission of parasites from humans to mosquitoes is a key component of malaria control. Tafenoquine exhibits activity against all stages of the malaria parasite and may have utility as a transmission blocking agent. We aimed to characterize the transmission blocking activity of low-dose tafenoquine. METHODS Healthy adults were inoculated with Plasmodium falciparum 3D7-infected erythrocytes on day 0. Piperaquine was administered on days 9 and 11 to clear asexual parasitemia while allowing gametocyte development. A single 50-mg oral dose of tafenoquine was administered on day 25. Transmission was determined by enriched membrane feeding assays predose and at 1, 4, and 7 days postdose. Artemether-lumefantrine was administered following the final assay. Outcomes were the reduction in mosquito infection and gametocytemia after tafenoquine and safety parameters. RESULTS Six participants were enrolled, and all were infective to mosquitoes before tafenoquine, with a median 86% (range, 22-98) of mosquitoes positive for oocysts and 57% (range, 4-92) positive for sporozoites. By day 4 after tafenoquine, the oocyst and sporozoite positivity rate had reduced by a median 35% (interquartile range [IQR]: 16-46) and 52% (IQR: 40-62), respectively, and by day 7, 81% (IQR 36-92) and 77% (IQR 52-98), respectively. The decline in gametocyte density after tafenoquine was not significant. No significant participant safety concerns were identified. CONCLUSIONS Low-dose tafenoquine (50 mg) reduces P. falciparum transmission to mosquitoes, with a delay in effect.
Collapse
Affiliation(s)
- Rebecca Webster
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Hayley Mitchell
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jenny M Peters
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Juanita Heunis
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Brighid O'Neill
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jeremy Gower
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Sean Lynch
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Helen Jennings
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Fiona H Amante
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | | | - Adam J Potter
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Geoffrey W Birrell
- Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - Michael D Edstein
- Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - G Dennis Shanks
- Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia.,The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | | |
Collapse
|
8
|
Choy RKM, Bourgeois AL, Ockenhouse CF, Walker RI, Sheets RL, Flores J. Controlled Human Infection Models To Accelerate Vaccine Development. Clin Microbiol Rev 2022; 35:e0000821. [PMID: 35862754 PMCID: PMC9491212 DOI: 10.1128/cmr.00008-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model's capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
Collapse
Affiliation(s)
- Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | - A. Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Richard I. Walker
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Jorge Flores
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| |
Collapse
|
9
|
Roberds A, Kifude C, Oyieko J, Ocholla S, Mutunga J, Oullo D, Waga C, Li Z, Luckhart S, Stewart VA. Longitudinal impact of asymptomatic malaria/HIV-1 co-infection on Plasmodium falciparum gametocyte transcript expression and transmission to Anopheles mosquitoes. Front Cell Infect Microbiol 2022; 12:934641. [PMID: 36189366 PMCID: PMC9523792 DOI: 10.3389/fcimb.2022.934641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/25/2022] [Indexed: 11/28/2022] Open
Abstract
Despite significant developments towards malaria reduction, parasite transmission in the common context of HIV-1 co-infection and treatment for one or both infections has not been fully characterized. This is particularly important given that HIV-1 and malaria chemotherapies have the potential to alter gametocyte burden and mosquito infectivity. In this study, we examined 782 blood samples collected from a longitudinal cohort of 300 volunteers with asymptomatic parasitemia seeking HIV testing or treatment in the endemic region of Kisumu, Kenya, to define the impacts of HIV-1-malaria co-infection, antiretroviral therapy (ART) plus trimethoprim-sulfamethoxazole (TS) and the antimalarials artemether/lumefantrine (AL) on Plasmodium falciparum gametocyte transcript prevalence and parasite transmission to the African malaria mosquito Anopheles gambiae. Volunteers were assigned to three distinct HIV-1 groups: HIV-1 positive on treatment, HIV-1 positive newly diagnosed, and HIV-1 negative. Volunteers were monitored monthly over the course of six months. Using our highly sensitive digital droplet PCR (ddPCR) assay of three gametocyte specific transcript markers, we detected gametocyte transcripts in 51.1% of 18S positive volunteers across all study groups and time points. After correcting for multiple comparisons, the factors of HIV-1 status, time, CD4+ T-cell levels and hematocrit were not predictive of gametocyte prevalence or transmission. However, among those volunteers who were newly diagnosed with HIV-1 and malaria positive by rapid diagnostic test (RDT) at enrollment, the initiation of ART/TS and AL treatment was associated with a significant reduction in gametocyte transcript prevalence in the subsequent month when compared to HIV-1 negative volunteers treated with AL. To assess gametocyte transmissibility, volunteer blood samples were used in standard membrane feeding assays (SFMA) with laboratory-reared A. gambiae, with evidence of transmission confirmed by at least one of 25 dissected mosquitoes per sample positive for at least one midgut oocyst. HIV-1 status, CD4+ T-cell levels and hematocrit were not significantly associated with successful transmission to A. gambiae. Analysis of SMFA blood samples revealed that 50% of transmission-positive blood samples failed to test positive by Plasmodium-specific 18S ribosomal RNA quantitative PCR (qPCR) and 35% failed to test positive for any gametocyte specific transcript marker by droplet digital (ddPCR), documenting that transmission occurred in the absence of molecular parasite/gametocyte detection. Overall, these findings highlight the complexity of HIV-1 malaria co-infection and the need to further define the unpredictable role of asymptomatic parasitemia in transmission to mosquitoes.
Collapse
Affiliation(s)
- Ashleigh Roberds
- Department of Preventive Medicine and Biostatistics, Division of Global Public Health, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Carolyne Kifude
- Kombewa Clinical Research Center, Kenya Medical Research Institute - United States Army Medical Research Directorate - Africa, Kisumu, Kenya
| | - Janet Oyieko
- Kombewa Clinical Research Center, Kenya Medical Research Institute - United States Army Medical Research Directorate - Africa, Kisumu, Kenya
| | - Stephen Ocholla
- Kombewa Clinical Research Center, Kenya Medical Research Institute - United States Army Medical Research Directorate - Africa, Kisumu, Kenya
| | - James Mutunga
- Department of Entomology and Vector Biology, United States Army Medical Research Directorate - Africa/Kenya Medical Research Institute, Kisumu, Kenya
- Department of Biological Sciences, Mount Kenya University, Thika, Kenya
| | - David Oullo
- Department of Entomology and Vector Biology, United States Army Medical Research Directorate - Africa/Kenya Medical Research Institute, Kisumu, Kenya
| | - Charles Waga
- Department of Entomology and Vector Biology, United States Army Medical Research Directorate - Africa/Kenya Medical Research Institute, Kisumu, Kenya
| | - Zhaozhang Li
- Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Shirley Luckhart
- Department of Entomology, Plant Pathology and Nematology and Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - V. Ann Stewart
- Department of Preventive Medicine and Biostatistics, Division of Global Public Health, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
10
|
Marquart L, Webb L, O'Rourke P, Gatton ML, Hsiang MS, Kalnoky M, Jang IK, Ntuku H, Mumbengegwi DR, Domingo GJ, McCarthy JS, Britton S. The in-vivo dynamics of Plasmodium falciparum HRP2: implications for the use of rapid diagnostic tests in malaria elimination. Malar J 2022; 21:233. [PMID: 35922803 PMCID: PMC9351188 DOI: 10.1186/s12936-022-04245-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 07/13/2022] [Indexed: 11/24/2022] Open
Abstract
Background Rapid diagnostic tests (RDTs) that rely on the detection of Plasmodium falciparum histidine-rich protein 2 (PfHRP2) have become key tools for diagnosing P. falciparum infection. The utility of RDTs can be limited by PfHRP2 persistence, however it can be a potential benefit in low transmission settings where detection of persistent PfHRP2 using newer ultra-sensitive PfHRP2 based RDTs can serve as a surveillance tool to identify recent exposure. Better understanding of the dynamics of PfHRP2 over the course of a malaria infection can inform optimal use of RDTs. Methods A previously published mathematical model was refined to mimic the production and decay of PfHRP2 during a malaria infection. Data from 15 individuals from volunteer infection studies were used to update the original model and estimate key model parameters. The refined model was applied to a cohort of patients from Namibia who received treatment for clinical malaria infection for whom longitudinal PfHRP2 concentrations were measured. Results The refinement of the PfHRP2 dynamic model indicated that in malaria naïve hosts, P. falciparum parasites of the 3D7 strain produce 33.6 × 10−15 g (95% CI 25.0–42.1 × 10−15 g) of PfHRP2 in vivo per parasite replication cycle, with an elimination half-life of 1.67 days (95% CI 1.11–3.40 days). The refined model included these updated parameters and incorporated individualized body fluid volume calculations, which improved predictive accuracy when compared to the original model. The performance of the model in predicting clearance of PfHRP2 post treatment in clinical samples from six adults with P. falciparum infection in Namibia improved when using a longer elimination half-life of 4.5 days, with 14% to 67% of observations for each individual within the predicted range. Conclusions The updated mathematical model can predict the growth and clearance of PfHRP2 during the production and decay of a mono-infection with P. falciparum, increasing the understanding of PfHRP2 antigen dynamics. This model can guide the optimal use of PfHRP2-based RDTs for reliable diagnosis of P. falciparum infection and re-infection in endemic settings, but also for malaria surveillance and elimination programmes in low transmission areas. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04245-z.
Collapse
Affiliation(s)
- Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia. .,University of Queensland, Brisbane, QLD, Australia.
| | - Lachlan Webb
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Peter O'Rourke
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | - Michelle S Hsiang
- Department of Pediatrics, University of Texas, Southwestern, Dallas, TX, USA.,Malaria Elimination Initiative, Institute for Global Health Services, University of California, San Francisco, CA, USA.,Department of Pediatrics, University of California, San Francisco, CA, USA
| | | | | | - Henry Ntuku
- Malaria Elimination Initiative, Institute for Global Health Services, University of California, San Francisco, CA, USA
| | | | | | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,University of Queensland, Brisbane, QLD, Australia
| | - Sumudu Britton
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
11
|
de Jong RM, Alkema M, Oulton T, Dumont E, Teelen K, Nakajima R, de Assis RR, Press KWD, Ngotho P, Tetteh KK, Felgner P, Marti M, Collins KA, Drakeley C, Bousema T, Stone WJ. The acquisition of humoral immune responses targeting Plasmodium falciparum sexual stages in controlled human malaria infections. Front Immunol 2022; 13:930956. [PMID: 35924245 PMCID: PMC9339717 DOI: 10.3389/fimmu.2022.930956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Individuals infected with P. falciparum develop antibody responses to intra-erythrocytic gametocyte proteins and exported gametocyte proteins present on the surface of infected erythrocytes. However, there is currently limited knowledge on the immunogenicity of gametocyte antigens and the specificity of gametocyte-induced antibody responses. In this study, we assessed antibody responses in participants of two controlled human malaria infection (CHMI) studies by ELISA, multiplexed bead-based antibody assays and protein microarray. By comparing antibody responses in participants with and without gametocyte exposure, we aimed to disentangle the antibody response induced by asexual and sexual stage parasites. We showed that after a single malaria infection, a significant anti-sexual stage humoral response is induced in malaria-naïve individuals, even after exposure to relatively low gametocyte densities (up to ~1,600 gametocytes/mL). In contrast to antibody responses to well-characterised asexual blood stage antigens that were detectable by day 21 after infection, responses to sexual stage antigens (including transmission blocking vaccine candidates Pfs48/45 and Pfs230) were only apparent at 51 days after infection. We found antigens previously associated with early gametocyte or anti-gamete immunity were highly represented among responses linked with gametocyte exposure. Our data provide detailed insights on the induction and kinetics of antibody responses to gametocytes and identify novel antigens that elicit antibody responses exclusively in individuals with gametocyte exposure. Our findings provide target identification for serological assays for surveillance of the malaria infectious reservoir, and support vaccine development by describing the antibody response to leading vaccine antigens after primary infection.
Collapse
Affiliation(s)
- Roos M. de Jong
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Manon Alkema
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Tate Oulton
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Elin Dumont
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Karina Teelen
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Rie Nakajima
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States
| | - Rafael Ramiro de Assis
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States
| | | | - Priscilla Ngotho
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Kevin K.A. Tetteh
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Phil Felgner
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Katharine A. Collins
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Chris Drakeley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Teun Bousema
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands,Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Will J.R. Stone
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom,*Correspondence: Will J.R. Stone,
| |
Collapse
|
12
|
Dash M, Sachdeva S, Bansal A, Sinha A. Gametogenesis in Plasmodium: Delving Deeper to Connect the Dots. Front Cell Infect Microbiol 2022; 12:877907. [PMID: 35782151 PMCID: PMC9241518 DOI: 10.3389/fcimb.2022.877907] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/09/2022] [Indexed: 11/16/2022] Open
Abstract
In the coming decades, eliminating malaria is the foremost goal of many tropical countries. Transmission control, along with an accurate and timely diagnosis of malaria, effective treatment and prevention are the different aspects that need to be met synchronously to accomplish the goal. The current review is focused on one of these aspects i.e., transmission control, by looking deeper into the event called gametogenesis. In the Plasmodium life cycle, gametocytes are the first life forms of the sexual phase. The transmission of the parasite and the disease is critically dependent on the number, viability and sex ratio of mature gametocytes and their further development inside mosquito vectors. Gametogenesis, the process of conversion of gametocytes into viable gametes, takes place inside the mosquito midgut, and is a tightly regulated event with fast and multiple rounds of DNA replication and diverse cellular changes going on within a short period. Interrupting the gametocyte-gamete transition is ought to restrict the successful transmission and progression of the disease and hence an area worth exploring for designing transmission-blocking strategies. This review summarizes an in-depth and up-to-date understanding of the biochemical and physiological mechanism of gametogenesis in Plasmodium, which could be targeted to control parasite and malaria transmission. This review also raises certain key questions regarding gametogenesis biology in Plasmodium and brings out gaps that still accompany in understanding the spectacular process of gametogenesis.
Collapse
Affiliation(s)
- Manoswini Dash
- Parasite Host Biology, Indian Council of Medical Research (ICMR)-National Institute of Malaria Research, New Delhi, India
- Central Molecular Laboratory, Govind Ballabh (GB) Pant Institute of Postgraduate Medical Education and Research, New Delhi, India
| | - Sherry Sachdeva
- Parasite Host Biology, Indian Council of Medical Research (ICMR)-National Institute of Malaria Research, New Delhi, India
| | - Abhisheka Bansal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Abhinav Sinha
- Parasite Host Biology, Indian Council of Medical Research (ICMR)-National Institute of Malaria Research, New Delhi, India
- *Correspondence: Abhinav Sinha,
| |
Collapse
|
13
|
Abstract
"The Primate Malarias" book has been a uniquely important resource for multiple generations of scientists, since its debut in 1971, and remains pertinent to the present day. Indeed, nonhuman primates (NHPs) have been instrumental for major breakthroughs in basic and pre-clinical research on malaria for over 50 years. Research involving NHPs have provided critical insights and data that have been essential for malaria research on many parasite species, drugs, vaccines, pathogenesis, and transmission, leading to improved clinical care and advancing research goals for malaria control, elimination, and eradication. Whilst most malaria scientists over the decades have been studying Plasmodium falciparum, with NHP infections, in clinical studies with humans, or using in vitro culture or rodent model systems, others have been dedicated to advancing research on Plasmodium vivax, as well as on phylogenetically related simian species, including Plasmodium cynomolgi, Plasmodium coatneyi, and Plasmodium knowlesi. In-depth study of these four phylogenetically related species over the years has spawned the design of NHP longitudinal infection strategies for gathering information about ongoing infections, which can be related to human infections. These Plasmodium-NHP infection model systems are reviewed here, with emphasis on modern systems biological approaches to studying longitudinal infections, pathogenesis, immunity, and vaccines. Recent discoveries capitalizing on NHP longitudinal infections include an advanced understanding of chronic infections, relapses, anaemia, and immune memory. With quickly emerging new technological advances, more in-depth research and mechanistic discoveries can be anticipated on these and additional critical topics, including hypnozoite biology, antigenic variation, gametocyte transmission, bone marrow dysfunction, and loss of uninfected RBCs. New strategies and insights published by the Malaria Host-Pathogen Interaction Center (MaHPIC) are recapped here along with a vision that stresses the importance of educating future experts well trained in utilizing NHP infection model systems for the pursuit of innovative, effective interventions against malaria.
Collapse
Affiliation(s)
- Mary R Galinski
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Emory Vaccine Center, Emory University, Atlanta, GA, USA.
- Emory National Primate Research Center (Yerkes National Primate Research Center), Emory University, Atlanta, GA, USA.
| |
Collapse
|
14
|
Ramjith J, Alkema M, Bradley J, Dicko A, Drakeley C, Stone W, Bousema T. Quantifying Reductions in Plasmodium falciparum Infectivity to Mosquitos: A Sample Size Calculator to Inform Clinical Trials on Transmission-Reducing Interventions. Front Immunol 2022; 13:899615. [PMID: 35720362 PMCID: PMC9205189 DOI: 10.3389/fimmu.2022.899615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Malaria transmission depends on the presence of mature Plasmodium transmission stages (gametocytes) that may render blood-feeding Anopheles mosquitos infectious. Transmission-blocking antimalarial drugs and vaccines can prevent transmission by reducing gametocyte densities or infectivity to mosquitos. Mosquito infection outcomes are thereby informative biological endpoints of clinical trials with transmission blocking interventions. Nevertheless, trials are often primarily designed to determine intervention safety; transmission blocking efficacy is difficult to incorporate in sample size considerations due to variation in infection outcomes and considerable inter-study variation. Here, we use clinical trial data from studies in malaria naive and naturally exposed study participants to present an online sample size calculator tool. This sample size calculator allows studies to be powered to detect reductions in the proportion of infected mosquitos or infection burden (oocyst density) in mosquitos. The utility of this online tool is illustrated using trial data with transmission blocking malaria drugs.
Collapse
Affiliation(s)
- Jordache Ramjith
- Radboud Institute for Molecular Life Sciences, Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department for Health Evidence, Biostatistics Research Group, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Manon Alkema
- Radboud Institute for Molecular Life Sciences, Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - John Bradley
- Medical Research Council (MRC) International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Alassane Dicko
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Chris Drakeley
- Medical Research Council (MRC) International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Will Stone
- Medical Research Council (MRC) International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Teun Bousema
- Radboud Institute for Molecular Life Sciences, Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
15
|
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: 11] [Impact Index Per Article: 5.5] [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
|
16
|
Barbieri D, Gomez L, Royer L, Dupuy F, Franetich JF, Tefit M, N’Dri ME, Mazier D, Silvie O, Moreno-Sabater A, Lavazec C. The Phosphodiesterase Inhibitor Tadalafil Promotes Splenic Retention of Plasmodium falciparum Gametocytes in Humanized Mice. Front Cell Infect Microbiol 2022; 12:883759. [PMID: 35694548 PMCID: PMC9174641 DOI: 10.3389/fcimb.2022.883759] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
The persistence of erythrocytes infected with Plasmodium falciparum gametocytes in the bloodstream is closely related to the modulation of their mechanical properties. New drugs that increase the stiffness of infected erythrocytes may thus represent a novel approach to block malaria parasite transmission. The phosphodiesterase inhibitor tadalafil has been shown to impair the ability of infected erythrocytes to circulate in an in vitro model for splenic retention. Here, we used a humanized mouse model to address in vivo the effect of tadalafil on the circulation kinetics of mature gametocyte-infected erythrocytes. We show that stiff immature gametocyte-infected erythrocytes are retained in the spleen of humanized mice at rates comparable to that of the in vitro model. Accordingly, tadalafil-induced stiffening of mature gametocyte-infected erythrocytes impairs their circulation in the bloodstream and triggers their retention by the spleen. These in vivo results validate that tadalafil is a novel drug lead potentially capable of blocking malaria parasite transmission by targeting GIE mechanical properties.
Collapse
Affiliation(s)
- Daniela Barbieri
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
| | - Lina Gomez
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
| | - Ludivine Royer
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
| | - Florian Dupuy
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
| | - Jean-François Franetich
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
| | - Maurel Tefit
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
| | - Marie-Esther N’Dri
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
| | - Dominique Mazier
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
| | - Olivier Silvie
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
| | - Alicia Moreno-Sabater
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
- Service de Parasitologie-Mycologie Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Paris, France
| | - Catherine Lavazec
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
- *Correspondence: Catherine Lavazec,
| |
Collapse
|
17
|
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: 4.5] [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
|
18
|
Alkema M, Yap XZ, de Jong GM, Reuling IJ, de Mast Q, van Crevel R, Ockenhouse CF, Collins KA, Bousema T, McCall MBB, Sauerwein RW. Controlled human malaria infections by mosquito bites induce more severe clinical symptoms than asexual blood-stage challenge infections. EBioMedicine 2022; 77:103919. [PMID: 35278741 PMCID: PMC8917304 DOI: 10.1016/j.ebiom.2022.103919] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Fever and inflammation are a hallmark of clinical Plasmodium falciparum (Pf) malaria induced by circulating asexual parasites. Although clinical manifestations of inflammation are associated with parasite density, this relationship is influenced by a complex network of immune-modulating factors of both human and parasite origin. METHODS In the Controlled Human Malaria infection (CHMI) model, we compared clinical inflammation in healthy malaria-naïve volunteers infected by either Pf-infected mosquito bites (MB, n=12) or intravenous administration of Pf-infected red blood cells (BS, n=12). FINDINGS All volunteers developed patent parasitaemia, but both the incidence and duration of severe adverse events were significantly higher after MB infection. Similarly, clinical laboratory markers of inflammation were significantly increased in the MB-group, as well as serum pro-inflammatory cytokine concentrations including IFN-γ, IL-6, MCP1 and IL-8. Parasite load, as reflected by maximum parasite density and area under the curve, was similar, but median duration of parasitaemia until treatment was longer in the BS-group compared to the MB-group (8 days [range 8 - 8 days] versus 5·5 days [range 3·5 - 12·5 days]). The in vitro response of subsets of peripheral blood mononuclear cells showed attenuated Pf-specific IFNγ production by γδ T-cells in the BS-arm. INTERPRETATION In conclusion, irrespective the parasite load, Pf-infections by MB induce stronger signs and symptoms of inflammation compared to CHMI by BS infection. The pathophysiological basis remains speculative but may relate to induced immune tolerance. FUNDING The trial was supported by PATH's Malaria Vaccine Initiative; the current analyses were supported by the AMMODO Science Award 2019 (TB).
Collapse
Affiliation(s)
- Manon Alkema
- Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud university medical center, 6500 HB Nijmegen, The Netherlands
| | - X Zen Yap
- Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud university medical center, 6500 HB Nijmegen, The Netherlands
| | - Gerdie M de Jong
- Department of Medical Microbiology and Infectious Diseases, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| | - Isaie J Reuling
- Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud university medical center, 6500 HB Nijmegen, The Netherlands
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud university medical center, 6500 HB Nijmegen, The Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud university medical center, 6500 HB Nijmegen, The Netherlands
| | | | - Katharine A Collins
- Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud university medical center, 6500 HB Nijmegen, The Netherlands
| | - Teun Bousema
- Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud university medical center, 6500 HB Nijmegen, The Netherlands
| | - Matthew B B McCall
- Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud university medical center, 6500 HB Nijmegen, The Netherlands.
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud university medical center, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
19
|
Birkholtz LM, Alano P, Leroy D. Transmission-blocking drugs for malaria elimination. Trends Parasitol 2022; 38:390-403. [DOI: 10.1016/j.pt.2022.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 12/24/2022]
|
20
|
Alahakoon P, McCaw JM, Taylor PG. Estimation of the probability of epidemic fade-out from multiple outbreak data. Epidemics 2022; 38:100539. [DOI: 10.1016/j.epidem.2022.100539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/13/2021] [Accepted: 01/05/2022] [Indexed: 11/03/2022] Open
|
21
|
Woolley SD, Marquart L, Woodford J, Chalon S, Moehrle JJ, McCarthy JS, Barber BE. Haematological response in experimental human Plasmodium falciparum and Plasmodium vivax malaria. Malar J 2021; 20:470. [PMID: 34930260 PMCID: PMC8685492 DOI: 10.1186/s12936-021-04003-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/01/2021] [Indexed: 11/25/2022] Open
Abstract
Background Malaria-associated anaemia, arising from symptomatic, asymptomatic and submicroscopic infections, is a significant cause of morbidity worldwide. Induced blood stage malaria volunteer infection studies (IBSM-VIS) provide a unique opportunity to evaluate the haematological response to early Plasmodium falciparum and Plasmodium vivax infection. Methods This study was an analysis of the haemoglobin, red cell counts, and parasitaemia data from 315 participants enrolled in IBSM-VIS between 2012 and 2019, including 269 participants inoculated with the 3D7 strain of P. falciparum (Pf3D7), 15 with an artemisinin-resistant P. falciparum strain (PfK13) and 46 with P. vivax. Factors associated with the fractional fall in haemoglobin (Hb-FF) were evaluated, and the malaria-attributable erythrocyte loss after accounting for phlebotomy-related losses was estimated. The relative contribution of parasitized erythrocytes to the malaria-attributable erythrocyte loss was also estimated. Results The median peak parasitaemia prior to treatment was 10,277 parasites/ml (IQR 3566–27,815), 71,427 parasites/ml [IQR 33,236–180,213], and 34,840 parasites/ml (IQR 13,302–77,064) in participants inoculated with Pf3D7, PfK13, and P. vivax, respectively. The median Hb-FF was 10.3% (IQR 7.8–13.3), 14.8% (IQR 11.8–15.9) and 11.7% (IQR 8.9–14.5) in those inoculated with Pf3D7, PfK13 and P. vivax, respectively, with the haemoglobin nadir occurring a median 12 (IQR 5–21), 15 (IQR 7–22), and 8 (IQR 7–15) days following inoculation. In participants inoculated with P. falciparum, recrudescence was associated with a greater Hb-FF, while in those with P. vivax, the Hb-FF was associated with a higher pre-treatment parasitaemia and later day of anti-malarial treatment. After accounting for phlebotomy-related blood losses, the estimated Hb-FF was 4.1% (IQR 3.1–5.3), 7.2% (IQR 5.8–7.8), and 4.9% (IQR 3.7–6.1) in participants inoculated with Pf3D7, PfK13, and P. vivax, respectively. Parasitized erythrocytes were estimated to account for 0.015% (IQR 0.006–0.06), 0.128% (IQR 0.068–0.616) and 0.022% (IQR 0.008–0.082) of the malaria-attributable erythrocyte loss in participants inoculated with Pf3D7, PfK13, and P. vivax, respectively. Conclusion Early experimental P. falciparum and P. vivax infection resulted in a small but significant fall in haemoglobin despite parasitaemia only just at the level of microscopic detection. Loss of parasitized erythrocytes accounted for < 0.2% of the total malaria-attributable haemoglobin loss. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-04003-7.
Collapse
|
22
|
Doolan DL. Malaria research in Australia: looking through the lens of the past towards the future. Int J Parasitol 2021; 51:1255-1263. [PMID: 34780720 DOI: 10.1016/j.ijpara.2021.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
Abstract
Malaria remains a global health priority, with substantial resources devoted to control and intervention since the causative parasite was first identified in 1880. Major advances have been made in discovery and translational research activities aimed at prevention, treatment and control. Laboratory-based, clinical, and field-based studies have complemented public health approaches. Australian scientists have played important roles, developing and applying innovative approaches, novel research tools and cutting-edge technologies in animal and human models of disease, as well as in disease-endemic settings. This article will provide an insight into 50 years of Australian efforts to discover mechanisms and targets of immunity and pathogenesis; develop new diagnostics, drugs, vaccines, and therapeutics; and assess new public health interventions and control measures in malaria-endemic settings.
Collapse
Affiliation(s)
- Denise L Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns QLD 4878, Australia.
| |
Collapse
|
23
|
Ayanful-Torgby R, Sarpong E, Abagna HB, Donu D, Obboh E, Mensah BA, Adjah J, Williamson KC, Amoah LE. Persistent Plasmodium falciparum infections enhance transmission-reducing immunity development. Sci Rep 2021; 11:21380. [PMID: 34725428 PMCID: PMC8560775 DOI: 10.1038/s41598-021-00973-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 10/21/2021] [Indexed: 11/09/2022] Open
Abstract
Subclinical infections that serve as reservoir populations to drive transmission remain a hurdle to malaria control. Data on infection dynamics in a geographical area is required to strategically design and implement malaria interventions. In a longitudinal cohort, we monitored Plasmodium falciparum infection prevalence and persistence, and anti-parasite immunity to gametocyte and asexual antigens for 10 weeks. Of the 100 participants, only 11 were never infected, whilst 16 had persistent infections detected by reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR), and one participant had microscopic parasites at all visits. Over 70% of the participants were infected three or more times, and submicroscopic gametocyte prevalence was high, ≥ 48% of the parasite carriers. Naturally induced responses against recombinant Pfs48/45.6C, Pfs230proC, and EBA175RIII-V antigens were not associated with either infection status or gametocyte carriage, but the antigen-specific IgG titers inversely correlated with parasite and gametocyte densities consistent with partial immunity. Longitudinal analysis of gametocyte diversity indicated at least four distinct clones circulated throughout the study period. The high prevalence of children infected with distinct gametocyte clones coupled with marked variation in infection status at the individual level suggests ongoing transmission and should be targeted in malaria control programs.
Collapse
Affiliation(s)
- Ruth Ayanful-Torgby
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.
| | | | - Hamza B Abagna
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Dickson Donu
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | | | - Benedicta A Mensah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Joshua Adjah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Kim C Williamson
- Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Linda E Amoah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.
| |
Collapse
|
24
|
Minassian AM, Themistocleous Y, Silk SE, Barrett JR, Kemp A, Quinkert D, Nielsen CM, Edwards NJ, Rawlinson TA, Ramos Lopez F, Roobsoong W, Ellis KJ, Cho JS, Aunin E, Otto TD, Reid AJ, Bach FA, Labbé GM, Poulton ID, Marini A, Zaric M, Mulatier M, Lopez Ramon R, Baker M, Mitton CH, Sousa JC, Rachaphaew N, Kumpitak C, Maneechai N, Suansomjit C, Piteekan T, Hou MM, Khozoee B, McHugh K, Roberts DJ, Lawrie AM, Blagborough AM, Nugent FL, Taylor IJ, Johnson KJ, Spence PJ, Sattabongkot J, Biswas S, Rayner JC, Draper SJ. Controlled human malaria infection with a clone of Plasmodium vivax with high quality genome assembly. JCI Insight 2021; 6:152465. [PMID: 34609964 PMCID: PMC8675201 DOI: 10.1172/jci.insight.152465] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Controlled human malaria infection (CHMI) provides a highly informative means to investigate host-pathogen interactions and enable in vivo proof-of-concept efficacy testing of new drugs and vaccines. However, unlike Plasmodium falciparum, well-characterized P. vivax parasites that are safe and suitable for use in modern CHMI models are limited. Here, two healthy malaria-naïve UK adults with universal donor blood group were safely infected with a clone of P. vivax from Thailand by mosquito-bite CHMI. Parasitemia developed in both volunteers and, prior to treatment, each volunteer donated blood to produce a cryopreserved stabilate of infected red blood cells. Following stringent safety screening, the parasite stabilate from one of these donors ("PvW1") was thawed and used to inoculate six healthy malaria-naïve UK adults by blood-stage CHMI, at three different dilutions. Parasitemia developed in all volunteers, who were then successfully drug treated. PvW1 parasite DNA was isolated and sequenced to produce a high quality genome assembly by using a hybrid assembly method. We analysed leading vaccine candidate antigens and multigene families, including the Vivax interspersed repeat (VIR) genes of which we identified 1145 in the PvW1 genome. Our genomic analysis will guide future assessment of candidate vaccines and drugs, as well as experimental medicine studies.
Collapse
Affiliation(s)
| | | | - Sarah E Silk
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jordan R Barrett
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Alison Kemp
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Doris Quinkert
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Nick J Edwards
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | - Jee-Sun Cho
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Eerik Aunin
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Thomas D Otto
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Adam J Reid
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Florian A Bach
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Ian D Poulton
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Arianna Marini
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Marija Zaric
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Margaux Mulatier
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Megan Baker
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Celia H Mitton
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jason C Sousa
- Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Maryland, United States of America
| | | | | | | | | | - Tianrat Piteekan
- Mahidol Vivax Research Unit, Mahidol University, Bangkok, Thailand
| | - Mimi M Hou
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Baktash Khozoee
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Kirsty McHugh
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - David J Roberts
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, United Kingdom
| | - Alison M Lawrie
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Fay L Nugent
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Iona J Taylor
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Philip J Spence
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Sumi Biswas
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Julian C Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
25
|
Kapulu MC, Njuguna P, Hamaluba M, Kimani D, Ngoi JM, Musembi J, Ngoto O, Otieno E, Billingsley PF. Safety and PCR monitoring in 161 semi-immune Kenyan adults following controlled human malaria infection. JCI Insight 2021; 6:e146443. [PMID: 34264864 PMCID: PMC8492329 DOI: 10.1172/jci.insight.146443] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 07/14/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUNDNaturally acquired immunity to malaria is incompletely understood. We used controlled human malaria infection (CHMI) to study the impact of past exposure on malaria in Kenyan adults in relation to infection with a non-Kenyan parasite strain.METHODSWe administered 3.2 × 103 aseptic, purified, cryopreserved Plasmodium falciparum sporozoites (Sanaria PfSPZ Challenge, NF54 West African strain) by direct venous inoculation and undertook clinical monitoring and serial quantitative PCR (qPCR) of the 18S ribosomal RNA gene. The study endpoint was met when parasitemia reached 500 or more parasites per μL blood, clinically important symptoms were seen, or at 21 days after inoculation. All volunteers received antimalarial drug treatment upon meeting the endpoint.RESULTSOne hundred and sixty-one volunteers underwent CHMI between August 4, 2016, and February 14, 2018. CHMI was well tolerated, with no severe or serious adverse events. Nineteen volunteers (11.8%) were excluded from the analysis based on detection of antimalarial drugs above the minimal inhibitory concentration or parasites genotyped as non-NF54. Of the 142 volunteers who were eligible for analysis, 26 (18.3%) had febrile symptoms and were treated; 30 (21.1%) reached 500 or more parasites per μL and were treated; 53 (37.3%) had parasitemia without meeting thresholds for treatment; and 33 (23.2%) remained qPCR negative.CONCLUSIONWe found that past exposure to malaria, as evidenced by location of residence, in some Kenyan adults can completely suppress in vivo growth of a parasite strain originating from outside Kenya.TRIAL REGISTRATIONClinicalTrials.gov NCT02739763.FUNDINGWellcome Trust.
Collapse
Affiliation(s)
- Melissa C. Kapulu
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Patricia Njuguna
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Mainga Hamaluba
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Domtila Kimani
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Joyce M. Ngoi
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Janet Musembi
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Omar Ngoto
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Edward Otieno
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | | |
Collapse
|
26
|
Koepfli C, Nguitragool W, de Almeida ACG, Kuehn A, Waltmann A, Kattenberg E, Ome-Kaius M, Rarau P, Obadia T, Kazura J, Monteiro W, Darcy AW, Wini L, Bassat Q, Felger I, Sattabongkot J, Robinson LJ, Lacerda M, Mueller I. Identification of the asymptomatic Plasmodium falciparum and Plasmodium vivax gametocyte reservoir under different transmission intensities. PLoS Negl Trop Dis 2021; 15:e0009672. [PMID: 34449764 PMCID: PMC8428688 DOI: 10.1371/journal.pntd.0009672] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 09/09/2021] [Accepted: 07/21/2021] [Indexed: 11/25/2022] Open
Abstract
Background Understanding epidemiological variables affecting gametocyte carriage and density is essential to design interventions that most effectively reduce malaria human-to-mosquito transmission. Methodology/Principal findings Plasmodium falciparum and P. vivax parasites and gametocytes were quantified by qPCR and RT-qPCR assays using the same methodologies in 5 cross-sectional surveys involving 16,493 individuals in Brazil, Thailand, Papua New Guinea, and Solomon Islands. The proportion of infections with detectable gametocytes per survey ranged from 44–94% for P. falciparum and from 23–72% for P. vivax. Blood-stage parasite density was the most important predictor of the probability to detect gametocytes. In moderate transmission settings (prevalence by qPCR>5%), parasite density decreased with age and the majority of gametocyte carriers were children. In low transmission settings (prevalence<5%), >65% of gametocyte carriers were adults. Per survey, 37–100% of all individuals positive for gametocytes by RT-qPCR were positive by light microscopy for asexual stages or gametocytes (overall: P. falciparum 178/348, P. vivax 235/398). Conclusions/Significance Interventions to reduce human-to-mosquito malaria transmission in moderate-high endemicity settings will have the greatest impact when children are targeted. In contrast, all age groups need to be included in control activities in low endemicity settings to achieve elimination. Detection of infections by light microscopy is a valuable tool to identify asymptomatic blood stage infections that likely contribute most to ongoing transmission at the time of sampling. Plasmodium vivax and Plasmodium falciparum cause the vast majority of all human malaria cases. Across all transmission settings, a large proportion of infections of the two species remain asymptomatic. These infections are not diagnosed and treated by control programs focusing on clinical cases. They can carry gametocytes, the sexual stage of the parasite that establishes infections in mosquitos, thus asymptomatic infections contribute to transmission. In order to determine who is likely to contribute to transmission, gametocyte densities were measured by sensitive molecular methods in afebrile individuals in four countries. The proportion of infections with gametocytes varied greatly among surveys, and was higher in regions that had experienced low transmission for extended periods of time. In moderate-high transmission settings, gametocyte densities were particularly high in children below six years, highlighting the importance that interventions to reduce transmission include this age group. The majority of gametocyte carriers was positive by light microscopy. The comprehensive data on gametocyte carriage presented here lays the foundation for the development of more effective screen and treat activities to reduce malaria transmission.
Collapse
Affiliation(s)
- Cristian Koepfli
- Population Health & Immunity Division, Walter & Eliza Hall Institute, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- University of Notre Dame, Eck Institute for Global Health, Department of Biological Sciences, Notre Dame, Indiana, United States of America
- * E-mail:
| | - Wang Nguitragool
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Anne Cristine Gomes de Almeida
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
- Universidade do Estado do Amazonas, Manaus, Brazil
| | - Andrea Kuehn
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Spain
| | - Andreea Waltmann
- Population Health & Immunity Division, Walter & Eliza Hall Institute, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Eline Kattenberg
- Population Health & Immunity Division, Walter & Eliza Hall Institute, Parkville, Australia
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Maria Ome-Kaius
- Population Health & Immunity Division, Walter & Eliza Hall Institute, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Patricia Rarau
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Thomas Obadia
- Hub de Bioinformatique et Biostatistique, Département Biologie Computationnelle, Institut Pasteur, Paris, France
- Unité Malaria: parasites et Hôtes, Département Parasites et Insectes Vecteurs, Institut Pasteur, Paris, France
| | - James Kazura
- Centre for Global Health & Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Wuelton Monteiro
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
- Universidade do Estado do Amazonas, Manaus, Brazil
| | - Andrew W. Darcy
- National Health Training and Research Institute, Ministry of Health, Honiara, Solomon Islands
| | - Lyndes Wini
- Vector Borne Diseases Program, Ministry of Health, Honiara, Solomon Islands
| | - Quique Bassat
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- ICREA, Barcelona, Spain
- Pediatric Infectious Diseases Unit, Pediatrics Department, Hospital Sant Joan de Déu (University of Barcelona), Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Ingrid Felger
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Leanne J. Robinson
- Population Health & Immunity Division, Walter & Eliza Hall Institute, Parkville, Australia
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Marcus Lacerda
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Ivo Mueller
- Population Health & Immunity Division, Walter & Eliza Hall Institute, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Unité Malaria: parasites et Hôtes, Département Parasites et Insectes Vecteurs, Institut Pasteur, Paris, France
| |
Collapse
|
27
|
Schneider P, Reece SE. The private life of malaria parasites: Strategies for sexual reproduction. Mol Biochem Parasitol 2021; 244:111375. [PMID: 34023299 PMCID: PMC8346949 DOI: 10.1016/j.molbiopara.2021.111375] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 12/22/2022]
Abstract
Malaria parasites exhibit a complex lifecycle, requiring extensive asexual replication in the liver and blood of the vertebrate host, and in the haemocoel of the insect vector. Yet, they must also undergo a single round of sexual reproduction, which occurs in the vector's midgut upon uptake of a blood meal. Sexual reproduction is obligate for infection of the vector and thus, is essential for onwards transmission to new hosts. Sex in malaria parasites involves several bottlenecks in parasite number, making the stages involved attractive targets for blocking disease transmission. Malaria parasites have evolved a suite of adaptations ("strategies") to maximise the success of sexual reproduction and transmission, which could undermine transmission-blocking interventions. Yet, understanding parasite strategies may also reveal novel opportunities for such interventions. Here, we outline how evolutionary and ecological theories, developed to explain reproductive strategies in multicellular taxa, can be applied to explain two reproductive strategies (conversion rate and sex ratio) expressed by malaria parasites within the vertebrate host.
Collapse
Affiliation(s)
- Petra Schneider
- Institute of Evolutionary Biology, Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK.
| | - Sarah E Reece
- Institute of Evolutionary Biology, Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
28
|
Graumans W, Lanke K, van Gemert GJ, Alkema M, van de Vegte-Bolmer M, Bousema T, Collins KA. The effect of anticoagulants in blood collection tubes on Plasmodium falciparum transmission in direct membrane feeding assays. Trans R Soc Trop Med Hyg 2021; 116:187-189. [PMID: 34182573 PMCID: PMC8804877 DOI: 10.1093/trstmh/trab095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 11/13/2022] Open
Abstract
Background Direct membrane feeding assays assess the transmission potential of malaria-infected individuals using whole blood collected in anticoagulant vacutainers. Methods The potential inhibitory effect of four commonly used anticoagulants on gametocyte infectivity to mosquitoes was assessed in standard membrane feeding assays with cultured Plasmodium falciparum. Results Infection burden in mosquitoes was significantly reduced when blood was collected in sodium citrate and EDTA. Transmission was highest when blood was collected in lithium heparin and sodium heparin, although a concentration-dependent inhibition of mosquito infection was also observed. Conclusions Although anticoagulants can reduce transmission efficiency, lithium heparin and sodium heparin are the best anticoagulants for evaluating malaria transmission.
Collapse
Affiliation(s)
- Wouter Graumans
- Radboud University Medical Center , Radboud Institute for Health Sciences , Department of Medical Microbiology, 268, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Kjerstin Lanke
- Radboud University Medical Center , Radboud Institute for Health Sciences , Department of Medical Microbiology, 268, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Geert-Jan van Gemert
- Radboud University Medical Center , Radboud Institute for Health Sciences , Department of Medical Microbiology, 268, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Manon Alkema
- Radboud University Medical Center , Radboud Institute for Health Sciences , Department of Medical Microbiology, 268, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Marga van de Vegte-Bolmer
- Radboud University Medical Center , Radboud Institute for Health Sciences , Department of Medical Microbiology, 268, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Teun Bousema
- Radboud University Medical Center , Radboud Institute for Health Sciences , Department of Medical Microbiology, 268, PO Box 9101, 6500 HB Nijmegen, The Netherlands.,Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, Keppel St, London WC1E 7HT, UK
| | - Katharine A Collins
- Radboud University Medical Center , Radboud Institute for Health Sciences , Department of Medical Microbiology, 268, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
29
|
Meibalan E, Barry A, Gibbins MP, Awandu S, Meerstein-Kessel L, Achcar F, Bopp S, Moxon C, Diarra A, Debe S, Ouédraogo N, Barry-Some I, Badoum ES, Fagnima T, Lanke K, Gonçalves BP, Bradley J, Wirth D, Drakeley C, Guelbeogo WM, Tiono AB, Marti M, Bousema T. Plasmodium falciparum Gametocyte Density and Infectivity in Peripheral Blood and Skin Tissue of Naturally Infected Parasite Carriers in Burkina Faso. J Infect Dis 2021; 223:1822-1830. [PMID: 31875909 PMCID: PMC8161640 DOI: 10.1093/infdis/jiz680] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/23/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Plasmodium falciparum transmission depends on mature gametocytes that can be ingested by mosquitoes taking a blood meal on human skin. Although gametocyte skin sequestration has long been hypothesized as important contributor to efficient malaria transmission, this has never been formally tested. METHODS In naturally infected gametocyte carriers from Burkina Faso, we assessed infectivity to mosquitoes by direct skin feeding and membrane feeding. We directly quantified male and female gametocytes and asexual parasites in finger-prick and venous blood samples, skin biopsy samples, and in of mosquitoes that fed on venous blood or directly on skin. Gametocytes were visualized in skin tissue with confocal microscopy. RESULTS Although more mosquitoes became infected when feeding directly on skin then when feeding on venous blood (odds ratio, 2.01; 95% confidence interval, 1.21-3.33; P = .007), concentrations of gametocytes were not higher in the subdermal skin vasculature than in other blood compartments; only sparse gametocytes were observed in skin tissue. DISCUSSION Our data strongly suggest that there is no significant skin sequestration of P. falciparum gametocytes. Gametocyte densities in peripheral blood are thus informative for predicting onward transmission potential to mosquitoes and can be used to target and monitor malaria elimination initiatives.
Collapse
Affiliation(s)
- Elamaran Meibalan
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Aissata Barry
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
- Radboud Institute for Health Sciences, Radboud University Medical Center, the Netherlands
| | - Matthew P Gibbins
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Shehu Awandu
- Radboud Institute for Health Sciences, Radboud University Medical Center, the Netherlands
| | | | - Fiona Achcar
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Selina Bopp
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Christopher Moxon
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Amidou Diarra
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Siaka Debe
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Nicolas Ouédraogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Ines Barry-Some
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Emilie S Badoum
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Traoré Fagnima
- Centre Hospitalier Universitaire Régional de Ouahigoua, Université de Ouahigouya, Burkina Faso
| | - Kjerstin Lanke
- Radboud Institute for Health Sciences, Radboud University Medical Center, the Netherlands
| | - Bronner P Gonçalves
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John Bradley
- MRC Tropical Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Dyann Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Chris Drakeley
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Alfred B Tiono
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Matthias Marti
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Teun Bousema
- Radboud Institute for Health Sciences, Radboud University Medical Center, the Netherlands
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
30
|
McCarthy JS, Donini C, Chalon S, Woodford J, Marquart L, Collins KA, Rozenberg FD, Fidock DA, Cherkaoui-Rbati MH, Gobeau N, Möhrle JJ. A Phase 1, Placebo-controlled, Randomized, Single Ascending Dose Study and a Volunteer Infection Study to Characterize the Safety, Pharmacokinetics, and Antimalarial Activity of the Plasmodium Phosphatidylinositol 4-Kinase Inhibitor MMV390048. Clin Infect Dis 2021; 71:e657-e664. [PMID: 32239164 PMCID: PMC7744986 DOI: 10.1093/cid/ciaa368] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/01/2020] [Indexed: 02/01/2023] Open
Abstract
Background MMV390048 is the first Plasmodium phosphatidylinositol 4-kinase inhibitor to reach clinical development as a new antimalarial. We aimed to characterize the safety, pharmacokinetics, and antimalarial activity of a tablet formulation of MMV390048. Methods A 2-part, phase 1 trial was conducted in healthy adults. Part 1 was a double-blind, randomized, placebo-controlled, single ascending dose study consisting of 3 cohorts (40, 80, 120 mg MMV390048). Part 2 was an open-label volunteer infection study using the Plasmodium falciparum induced blood-stage malaria model consisting of 2 cohorts (40 mg and 80 mg MMV390048). Results Twenty four subjects were enrolled in part 1 (n = 8 per cohort, randomized 3:1 MMV390048:placebo) and 15 subjects were enrolled in part 2 (40 mg [n = 7] and 80 mg [n = 8] cohorts). One subject was withdrawn from part 2 (80 mg cohort) before dosing and was not included in analyses. No serious or severe adverse events were attributed to MMV390048. The rate of parasite clearance was greater in subjects administered 80 mg compared to those administered 40 mg (clearance half-life 5.5 hours [95% confidence interval {CI}, 5.2–6.0 hours] vs 6.4 hours [95% CI, 6.0–6.9 hours]; P = .005). Pharmacokinetic/pharmacodynamic modeling estimated a minimum inhibitory concentration of 83 ng/mL and a minimal parasiticidal concentration that would achieve 90% of the maximum effect of 238 ng/mL, and predicted that a single 120-mg dose would achieve an adequate clinical and parasitological response with 92% certainty. Conclusions The safety, pharmacokinetics, and pharmacodynamics of MMV390048 support its further development as a partner drug of a single-dose combination therapy for malaria. Clinical Trials Registration NCT02783820 (part 1); NCT02783833 (part 2).
Collapse
Affiliation(s)
- James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | | | - John Woodford
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Felix D Rozenberg
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA.,Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | | | | | | |
Collapse
|
31
|
Vekemans J, Schellenberg D, Benns S, O'Brien K, Alonso P. Meeting report: WHO consultation on malaria vaccine development, Geneva, 15-16 July 2019. Vaccine 2021; 39:2907-2916. [PMID: 33931251 DOI: 10.1016/j.vaccine.2021.03.093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 01/25/2023]
Abstract
Considerable progress has been made in malaria control in the last two decades, but progress has stalled in the last few years. New tools are needed to achieve public health goals in malaria control and elimination. A first generation vaccine, RTS,S/AS01, is currently being evaluated as it undergoes pilot implementation through routine health systems in parts of three African countries. The development of this vaccine took over 30 years and has been full of uncertainties. Even now, important unknowns remain as to its future role in public health. Lessons need to be learnt for second generation and future vaccines, including how to facilitate early planning of investments, streamlining of development, regulatory and policy pathways. A number of candidate vaccines populate the current development pipeline, some of which have the potential to contribute to burden reduction if efficacy is confirmed in conditions of natural exposure, and if they are amenable to affordable supply and programmatic implementation. New, innovative technologies will be needed if future malaria vaccines are to overcome important scientific hurdles and induce durable, high level protection. WHO convened a stakeholder consultation on the status of malaria vaccine research and development to inform the recently reconstituted Malaria Vaccine Advisory Committee (MALVAC) which will assist WHO in updating its current guidance and recommendations about priorities and product preferences for malaria vaccines.
Collapse
Affiliation(s)
- Johan Vekemans
- World Health Organization, 20 Av Appia, 1211 Geneva 27, Switzerland
| | | | | | - Kate O'Brien
- World Health Organization, 20 Av Appia, 1211 Geneva 27, Switzerland
| | - Pedro Alonso
- World Health Organization, 20 Av Appia, 1211 Geneva 27, Switzerland
| |
Collapse
|
32
|
Barry A, Bradley J, Stone W, Guelbeogo MW, Lanke K, Ouedraogo A, Soulama I, Nébié I, Serme SS, Grignard L, Patterson C, Wu L, Briggs JJ, Janson O, Awandu SS, Ouedraogo M, Tarama CW, Kargougou D, Zongo S, Sirima SB, Marti M, Drakeley C, Tiono AB, Bousema T. Higher gametocyte production and mosquito infectivity in chronic compared to incident Plasmodium falciparum infections. Nat Commun 2021; 12:2443. [PMID: 33903595 PMCID: PMC8076179 DOI: 10.1038/s41467-021-22573-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 03/09/2021] [Indexed: 11/09/2022] Open
Abstract
Plasmodium falciparum gametocyte kinetics and infectivity may differ between chronic and incident infections. In the current study, we assess parasite kinetics and infectivity to mosquitoes among children (aged 5-10 years) from Burkina Faso with (a) incident infections following parasite clearance (n = 48) and (b) chronic asymptomatic infections (n = 60). In the incident infection cohort, 92% (44/48) of children develop symptoms within 35 days, compared to 23% (14/60) in the chronic cohort. All individuals with chronic infection carried gametocytes or developed them during follow-up, whereas only 35% (17/48) in the incident cohort produce gametocytes before becoming symptomatic and receiving treatment. Parasite multiplication rate (PMR) and the relative abundance of ap2-g and gexp-5 transcripts are positively associated with gametocyte production. Antibody responses are higher and PMR lower in chronic infections. The presence of symptoms and sexual stage immune responses are associated with reductions in gametocyte infectivity to mosquitoes. We observe that most incident infections require treatment before the density of mature gametocytes is sufficient to infect mosquitoes. In contrast, chronic, asymptomatic infections represent a significant source of mosquito infections. Our observations support the notion that malaria transmission reduction may be expedited by enhanced case management, involving both symptom-screening and infection detection.
Collapse
Affiliation(s)
- Aissata Barry
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
- Radboud Institute for Health Sciences and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - John Bradley
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Will Stone
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Moussa W Guelbeogo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Kjerstin Lanke
- Radboud Institute for Health Sciences and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Alphonse Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Issiaka Soulama
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Issa Nébié
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Samuel S Serme
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Lynn Grignard
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Catriona Patterson
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Lindsey Wu
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Jessica J Briggs
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Owen Janson
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Shehu S Awandu
- Radboud Institute for Health Sciences and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mireille Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Casimire W Tarama
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Désiré Kargougou
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Soumanaba Zongo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Sodiomon B Sirima
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Chris Drakeley
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Alfred B Tiono
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Teun Bousema
- Radboud Institute for Health Sciences and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands.
| |
Collapse
|
33
|
Wang CYT, Ballard EL, Pava Z, Marquart L, Gaydon J, Murphy SC, Whiley D, O'Rourke P, McCarthy JS. Analytical validation of a real-time hydrolysis probe PCR assay for quantifying Plasmodium falciparum parasites in experimentally infected human adults. Malar J 2021; 20:181. [PMID: 33838672 PMCID: PMC8035755 DOI: 10.1186/s12936-021-03717-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 03/29/2021] [Indexed: 11/10/2022] Open
Abstract
Background Volunteer infection studies have become a standard model for evaluating drug efficacy against Plasmodium infections. Molecular techniques such as qPCR are used in these studies due to their ability to provide robust and accurate estimates of parasitaemia at increased sensitivity compared to microscopy. The validity and reliability of assays need to be ensured when used to evaluate the efficacy of candidate drugs in clinical trials. Methods A previously described 18S rRNA gene qPCR assay for quantifying Plasmodium falciparum in blood samples was evaluated. Assay performance characteristics including analytical sensitivity, reportable range, precision, accuracy and specificity were assessed using experimental data and data compiled from phase 1 volunteer infection studies conducted between 2013 and 2019. Guidelines for validation of laboratory-developed molecular assays were followed. Results The reportable range was 1.50 to 6.50 log10 parasites/mL with a limit of detection of 2.045 log10 parasites/mL of whole blood based on a parasite diluted standard series over this range. The assay was highly reproducible with minimal intra-assay (SD = 0.456 quantification cycle (Cq) units [0.137 log10 parasites/mL] over 21 replicates) and inter-assay (SD = 0.604 Cq units [0.182 log10 parasites/mL] over 786 qPCR runs) variability. Through an external quality assurance program, the QIMR assay was shown to generate accurate results (quantitative bias + 0.019 log10 parasites/mL against nominal values). Specificity was 100% after assessing 164 parasite-free human blood samples. Conclusions The 18S rRNA gene qPCR assay is specific and highly reproducible and can provide reliable and accurate parasite quantification. The assay is considered fit for use in evaluating drug efficacy in malaria clinical trials. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03717-y.
Collapse
Affiliation(s)
- Claire Y T Wang
- Centre for Children's Health Research, Children's Health Queensland, Brisbane, Australia. .,Child Health Research Centre, The University of Queensland, Brisbane, Australia.
| | - Emma L Ballard
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Zuleima Pava
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jane Gaydon
- Centre for Children's Health Research, Children's Health Queensland, Brisbane, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, Australia
| | - Sean C Murphy
- Departments of Laboratory Medicine and Microbiology, University of Washington, Seattle, WA, USA.,Center for Emerging and Re-Emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - David Whiley
- Centre for Children's Health Research, Children's Health Queensland, Brisbane, Australia.,UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Peter O'Rourke
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia.,School of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
34
|
Omondi BR, Muthui MK, Muasya WI, Orindi B, Mwakubambanya RS, Bousema T, Drakeley C, Marsh K, Bejon P, Kapulu MC. Antibody Responses to Crude Gametocyte Extract Predict Plasmodium falciparum Gametocyte Carriage in Kenya. Front Immunol 2021; 11:609474. [PMID: 33633729 PMCID: PMC7902058 DOI: 10.3389/fimmu.2020.609474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/17/2020] [Indexed: 11/18/2022] Open
Abstract
Background Malaria caused by Plasmodium falciparum remains a serious global public health challenge especially in Africa. Interventions that aim to reduce malaria transmission by targeting the gametocyte reservoir are key to malaria elimination and/or eradication. However, factors that are associated with gametocyte carriage have not been fully explored. Consequently, identifying predictors of the infectious reservoir is fundamental in the elimination campaign. Methods We cultured P. falciparum NF54 gametocytes (to stage V) and prepared crude gametocyte extract. Samples from a total of 687 participants (aged 6 months to 67 years) representing two cross-sectional study cohorts in Kilifi, Kenya were used to assess IgG antibody responses by ELISA. We also analyzed IgG antibody responses to the blood-stage antigen AMA1 as a marker of asexual parasite exposure. Gametocytemia and asexual parasitemia data quantified by microscopy and molecular detection (QT-NASBA) were used to determine the relationship with antibody responses, season, age, and transmission setting. Multivariable logistic regression models were used to study the association between antibody responses and gametocyte carriage. The predictive power of the models was tested using the receiver operating characteristic (ROC) curve. Results Multivariable logistic regression analysis showed that IgG antibody response to crude gametocyte extract predicted both microscopic (OR=1.81 95% CI: 1.06-3.07, p=0.028) and molecular (OR=1.91, 95% CI: 1.11-3.29, p=0.019) P. falciparum gametocyte carriage. Antibody responses to AMA1 were also associated with both microscopic (OR=1.61 95% CI: 1.08-2.42, p=0.020) and molecular (OR=3.73 95% CI: 2.03-6.74, p<0.001) gametocytemia. ROC analysis showed that molecular (AUC=0.897, 95% CI: 0.868-0.926) and microscopic (AUC=0.812, 95% CI: 0.758-0.865) multivariable models adjusted for gametocyte extract showed very high predictive power. Molecular (AUC=0.917, 95% CI: 0.891-0.943) and microscopic (AUC=0.806, 95% CI: 0.755-0.858) multivariable models adjusted for AMA1 were equally highly predictive. Conclusion In our study, it appears that IgG responses to crude gametocyte extract are not an independent predictor of gametocyte carriage after adjusting for AMA1 responses but may predict gametocyte carriage as a proxy marker of exposure to parasites. Serological responses to AMA1 or to gametocyte extract may facilitate identification of individuals within populations who contribute to malaria transmission and support implementation of transmission-blocking interventions.
Collapse
Affiliation(s)
- Brian R. Omondi
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Biochemistry and Molecular Biology, Egerton University, Nakuru, Kenya
| | - Michelle K. Muthui
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - William I. Muasya
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Benedict Orindi
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Chris Drakeley
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Kevin Marsh
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Philip Bejon
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Melissa C. Kapulu
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
35
|
Collins KA, Wang CY, Adams M, Mitchell H, Robinson GJ, Rampton M, Elliott S, Odedra A, Khoury D, Ballard E, Shelper TB, Lucantoni L, Avery VM, Chalon S, Moehrle JJ, McCarthy JS. A Plasmodium vivax experimental human infection model for evaluating efficacy of interventions. J Clin Invest 2021; 130:2920-2927. [PMID: 32045385 PMCID: PMC7259989 DOI: 10.1172/jci134923] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/04/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Interventions that interrupt Plasmodium vivax transmission or eliminate dormant P. vivax liver-stage parasites will be essential for malaria elimination. Development of these interventions has been hindered by the lack of P. vivax in vitro culture and could be accelerated by a safe and reproducible clinical model in malaria-naive individuals. METHODS Healthy, malaria-naive adults were enrolled in 2 studies to assess the safety, infectivity, and transmissibility of a new P. vivax isolate. Participants (Study 1, n = 2; Study 2, n = 24) were inoculated with P. vivax–infected red blood cells to initiate infection, and were treated with artemether-lumefantrine (Study 1) or chloroquine (Study 2). Primary endpoints were safety and infectivity of the new isolate. In Study 2, transmission to mosquitoes was also evaluated using mosquito feeding assays, and sporozoite viability was assessed using in vitro cultured hepatocytes. RESULTS Parasitemia and gametocytemia developed in all participants and was cleared by antimalarial treatment. Adverse events were mostly mild or moderate and none were serious. Sixty-nine percent of participants (11/16) were infectious to Anopheles mosquitoes at peak gametocytemia. Mosquito infection rates reached 97% following membrane feeding with gametocyte-enriched blood, and sporozoites developed into liver-stage schizonts in culture. CONCLUSION We have demonstrated the safe, reproducible, and efficient transmission of P. vivax gametocytes from humans to mosquitoes, and have established an experimental model that will accelerate the development of interventions targeting multiple stages of the P. vivax life cycle. TRIAL REGISTRATION ACTRN12614000930684 and ACTRN12616000174482. FUNDING (Australian) National Health and Medical Research Council Program Grant 1132975 (Study 1). Bill and Melinda Gates Foundation (OPP1111147) (Study 2).
Collapse
Affiliation(s)
- Katharine A Collins
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Claire Yt Wang
- Queensland Paediatric Infectious Diseases Laboratory, Centre for Children's Health Research, Brisbane, Queensland, Australia
| | - Matthew Adams
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Hayley Mitchell
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Greg J Robinson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Melanie Rampton
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Anand Odedra
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - David Khoury
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Emma Ballard
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Todd B Shelper
- Discovery Biology, Griffith University, Brisbane, Queensland, Australia
| | | | - Vicky M Avery
- Discovery Biology, Griffith University, Brisbane, Queensland, Australia
| | | | | | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
36
|
Abstract
Plasmodium vivax bench research greatly lags behind Plasmodium falciparum because of an inability to culture in vitro. A century ago, intentionally inducing a malaria infection was a strategy commonly used to cure late-stage syphilis. These controlled human malaria infections were used with expertise and persisted to the end of World War II. While controlled malaria liver-stage infection has been achieved for both P. vivax and P. falciparum, controlled human transmission to mosquitoes falls short for both species. In this issue of the JCI, Collins et al. present groundbreaking work that establishes a system to transmit P. vivax gametocytes from humans to mosquitoes. The authors injected a unique human isolate of P. vivax that reached high gametocyte density within weeks. This study provides a technical advance that will facilitate the study and eradication of the human parasite P. vivax.
Collapse
|
37
|
Defining the Antimalarial Activity of Cipargamin in Healthy Volunteers Experimentally Infected with Blood-Stage Plasmodium falciparum. Antimicrob Agents Chemother 2021; 65:AAC.01423-20. [PMID: 33199389 PMCID: PMC7849011 DOI: 10.1128/aac.01423-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022] Open
Abstract
The spiroindolone cipargamin, a new antimalarial compound that inhibits Plasmodium ATP4, is currently in clinical development. This study aimed to characterize the antimalarial activity of cipargamin in healthy volunteers experimentally infected with blood-stage Plasmodium falciparum. The spiroindolone cipargamin, a new antimalarial compound that inhibits Plasmodium ATP4, is currently in clinical development. This study aimed to characterize the antimalarial activity of cipargamin in healthy volunteers experimentally infected with blood-stage Plasmodium falciparum. Eight subjects were intravenously inoculated with parasite-infected erythrocytes and received a single oral dose of 10 mg cipargamin 7 days later. Blood samples were collected to monitor the development and clearance of parasitemia and plasma cipargamin concentrations. Parasite regrowth was treated with piperaquine monotherapy to clear asexual parasites, while allowing gametocyte transmissibility to mosquitoes to be investigated. An initial rapid decrease in parasitemia occurred in all participants following cipargamin dosing, with a parasite clearance half-life of 3.99 h. As anticipated from the dose selected, parasite regrowth occurred in all 8 subjects 3 to 8 days after dosing and allowed the pharmacokinetic/pharmacodynamic relationship to be determined. Based on the limited data from the single subtherapeutic dose cohort, a MIC of 11.6 ng/ml and minimum parasiticidal concentration that achieves 90% of maximum effect of 23.5 ng/ml were estimated, and a single 95-mg dose (95% confidence interval [CI], 50 to 270) was predicted to clear 109 parasites/ml. Low gametocyte densities were detected in all subjects following piperaquine treatment, which did not transmit to mosquitoes. Serious adverse liver function changes were observed in three subjects, which led to premature study termination. The antimalarial activity characterized in this study supports the further clinical development of cipargamin as a new treatment for P. falciparum malaria, although the hepatic safety profile of the compound warrants further evaluation. (This study has been registered at ClinicalTrials.gov under identifier NCT02543086.)
Collapse
|
38
|
Safety and feasibility of apheresis to harvest and concentrate parasites from subjects with induced blood stage Plasmodium vivax infection. Malar J 2021; 20:43. [PMID: 33446191 PMCID: PMC7807416 DOI: 10.1186/s12936-021-03581-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 01/04/2021] [Indexed: 11/27/2022] Open
Abstract
Background In the absence of a method to culture Plasmodium vivax, the only way to source parasites is ex vivo. This hampers many aspects of P. vivax research. This study aimed to assess the safety of apheresis, a method for selective removal of specific components of blood as a means of extracting and concentrating P. vivax parasites. Methods An iterative approach was employed across four non-immune healthy human subjects in single subject cohorts. All four subjects were inoculated with ~ 564 blood stage P. vivax (HMP013-Pv) and subjected to apheresis 10 to 11 days later. Blood samples collected during apheresis (haematocrit layers 0.5% to 11%) were tested for the presence and concentration of P. vivax by microscopy, flow cytometry, 18S rDNA qPCR for total parasites, and pvs25 qRT-PCR for female gametocyte transcripts. Safety was determined by monitoring adverse events. Malaria transmission to mosquitoes was assessed by membrane feeding assays. Results There were no serious adverse events and no significant safety concerns. Apheresis concentrated asexual parasites by up to 4.9-fold (range: 0.9–4.9-fold) and gametocytes by up to 1.45-fold (range: 0.38–1.45-fold) compared to pre-apheresis densities. No single haematocrit layer contained > 40% of all the recovered P. vivax asexual parasites. Ex vivo concentration of parasites by Percoll gradient centrifugation of whole blood achieved greater concentration of gametocytes than apheresis. Mosquito transmission was enhanced by up to fivefold in a single apheresis sample compared to pre-apheresis. Conclusion The modest level of parasite concentration suggests that the use of apheresis may not be an ideal method for harvesting P. vivax. Trial Registration Australia New Zealand Clinical Trials Registry (ANZCTR) Trial ID: ACTRN12617001502325 registered on 19th October 2017. https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=373812.
Collapse
|
39
|
Mahamar A, Lanke K, Graumans W, Diawara H, Sanogo K, Diarra K, Niambele SM, Gosling R, Drakeley C, Chen I, Dicko A, Bousema T, Roh ME. Persistence of mRNA indicative of Plasmodium falciparum ring-stage parasites 42 days after artemisinin and non-artemisinin combination therapy in naturally infected Malians. Malar J 2021; 20:34. [PMID: 33422068 PMCID: PMC7797096 DOI: 10.1186/s12936-020-03576-z] [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: 09/13/2020] [Accepted: 12/31/2020] [Indexed: 11/10/2022] Open
Abstract
Background Malaria control in sub-Saharan Africa relies upon prompt case management with artemisinin-based combination therapy (ACT). Ring-stage parasite mRNA, measured by sbp1 quantitative reverse-transcriptase PCR (qRT-PCR), was previously reported to persist after ACT treatment and hypothesized to reflect temporary arrest of the growth of ring-stage parasites (dormancy) following exposure to artemisinins. Here, the persistence of ring-stage parasitaemia following ACT and non-ACT treatment was examined. Methods Samples were used from naturally infected Malian gametocyte carriers who received dihydroartemisinin–piperaquine (DP) or sulfadoxine–pyrimethamine (SP–AQ) with or without gametocytocidal drugs. Gametocytes and ring-stage parasites were quantified by qRT-PCR during 42 days of follow-up. Results At baseline, 89% (64/73) of participants had measurable ring-stage parasite mRNA. Following treatment, the proportion of ring-stage parasite-positive individuals and estimated densities declined for all four treatment groups. Ring-stage parasite prevalence and density was generally lower in arms that received DP compared to SP–AQ. This finding was most apparent days 1, 2, and 42 of follow-up (p < 0.01). Gametocytocidal drugs did not influence ring-stage parasite persistence. Ring-stage parasite density estimates on days 14 and 28 after initiation of treatment were higher among individuals who subsequently experienced recurrent parasitaemia compared to those who remained free of parasites until day 42 after initiation of treatment (pday 14 = 0.011 and pday 28 = 0.068). No association of ring-stage persistence with gametocyte carriage was observed. Conclusions The current findings of lower ring-stage persistence after ACT without an effect of gametocytocidal partner drugs affirms the use of sbp1 as ring-stage marker. Lower persistence of ring-stage mRNA after ACT treatment suggests the marker may not reflect dormant parasites whilst it was predictive of re-appearance of parasitaemia.
Collapse
Affiliation(s)
- Almahamoudou Mahamar
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Kjerstin Lanke
- Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, PO Box 9101, 6525GA, Nijmegen, The Netherlands
| | - Wouter Graumans
- Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, PO Box 9101, 6525GA, Nijmegen, The Netherlands
| | - Halimatou Diawara
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Koualy Sanogo
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Kalifa Diarra
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Sidi Mohamed Niambele
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Roly Gosling
- Global Health Group, Malaria Elimination Initiative, University of California, San Francisco, CA, USA
| | - Chris Drakeley
- Department of Infection & Immunity, London School of Hygiene & Tropical Medicine, London, UK
| | - Ingrid Chen
- Global Health Group, Malaria Elimination Initiative, University of California, San Francisco, CA, USA
| | - Alassane Dicko
- Malaria Research and Training Centre, Faculty of Pharmacy, Medicine, and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Teun Bousema
- Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 26-28, PO Box 9101, 6525GA, Nijmegen, The Netherlands. .,Department of Infection & Immunity, London School of Hygiene & Tropical Medicine, London, UK.
| | - Michelle E Roh
- Global Health Group, Malaria Elimination Initiative, University of California, San Francisco, CA, USA
| |
Collapse
|
40
|
Th2-like T Follicular Helper Cells Promote Functional Antibody Production during Plasmodium falciparum Infection. CELL REPORTS MEDICINE 2020; 1:100157. [PMID: 33377128 PMCID: PMC7762767 DOI: 10.1016/j.xcrm.2020.100157] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/08/2020] [Accepted: 11/19/2020] [Indexed: 01/10/2023]
Abstract
CD4+ T follicular helper cells (Tfh) are key drivers of antibody development. During Plasmodium falciparum malaria in children, the activation of Tfh is restricted to the Th1 subset and not associated with antibody levels. To identify Tfh subsets that are associated with antibody development in malaria, we assess Tfh and antibodies longitudinally in human volunteers with experimental P. falciparum infection. Tfh cells activate during infection, with distinct dynamics in different Tfh subsets. Th2-Tfh cells activate early, during peak infection, while Th1-Tfh cells activate 1 week after peak infection and treatment. Th2-Tfh cell activation is associated with the functional breadth and magnitude of parasite antibodies. In contrast, Th1-Tfh activation is not associated with antibody development but instead with plasma cells, which have previously been shown to play a detrimental role in the development of long-lived immunity. Thus, our study identifies the contrasting roles of Th2 and Th1-Tfh cells during experimental P. falciparum malaria.
Collapse
|
41
|
Prajapati SK, Ayanful-Torgby R, Pava Z, Barbeau MC, Acquah FK, Cudjoe E, Kakaney C, Amponsah JA, Obboh E, Ahmed AE, Abuaku BK, McCarthy JS, Amoah LE, Williamson KC. The transcriptome of circulating sexually committed Plasmodium falciparum ring stage parasites forecasts malaria transmission potential. Nat Commun 2020; 11:6159. [PMID: 33268801 PMCID: PMC7710746 DOI: 10.1038/s41467-020-19988-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 10/06/2020] [Indexed: 11/30/2022] Open
Abstract
Malaria is spread by the transmission of sexual stage parasites, called gametocytes. However, with Plasmodium falciparum, gametocytes can only be detected in peripheral blood when they are mature and transmissible to a mosquito, which complicates control efforts. Here, we identify the set of genes overexpressed in patient blood samples with high levels of gametocyte-committed ring stage parasites. Expression of all 18 genes is regulated by transcription factor AP2-G, which is required for gametocytogenesis. We select three genes, not expressed in mature gametocytes, to develop as biomarkers. All three biomarkers we validate in vitro using 6 different parasite lines and develop an algorithm that predicts gametocyte production in ex vivo samples and volunteer infection studies. The biomarkers are also sensitive enough to monitor gametocyte production in asymptomatic P. falciparum carriers allowing early detection and treatment of infectious reservoirs, as well as the in vivo analysis of factors that modulate sexual conversion. Malaria gametocytes are sexual-stage parasites transmitted from mammalian host’s blood back to their insect vector. Here, Prajapati et al. identify gametocyte-committed ring-stage biomarkers allowing to forecast malaria transmission potential.
Collapse
Affiliation(s)
- Surendra K Prajapati
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Ruth Ayanful-Torgby
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Zuleima Pava
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Michelle C Barbeau
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,University of Virginia, Charlottesville, VA, USA
| | - Festus K Acquah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Elizabeth Cudjoe
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Courage Kakaney
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Jones A Amponsah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | | | - Anwar E Ahmed
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Benjamin K Abuaku
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Linda E Amoah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Kim C Williamson
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
42
|
Woodford J, Collins KA, Odedra A, Wang C, Jang IK, Domingo GJ, Watts R, Marquart L, Berriman M, Otto TD, McCarthy JS. An Experimental Human Blood-Stage Model for Studying Plasmodium malariae Infection. J Infect Dis 2020; 221:948-955. [PMID: 30852586 DOI: 10.1093/infdis/jiz102] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/06/2019] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Plasmodium malariae is considered a minor malaria parasite, although its global disease burden is underappreciated. The aim of this study was to develop an induced blood-stage malaria (IBSM) model of P. malariae to study parasite biology, diagnostic assays, and treatment. METHODS This clinical trial involved 2 healthy subjects who were intravenously inoculated with cryopreserved P. malariae-infected erythrocytes. Subjects were treated with artemether-lumefantrine after development of clinical symptoms. Prior to antimalarial therapy, mosquito-feeding assays were performed to investigate transmission, and blood samples were collected for rapid diagnostic testing and parasite transcription profiling. Serial blood samples were collected for biomarker analysis. RESULTS Both subjects experienced symptoms and signs typical of early malaria. Parasitemia was detected 7 days after inoculation, and parasite concentrations increased until antimalarial treatment was initiated 25 and 21 days after inoculation for subjects 1 and 2 respectively (peak parasitemia levels, 174 182 and 50 291 parasites/mL, respectively). The parasite clearance half-life following artemether-lumefantrine treatment was 6.7 hours. Mosquito transmission was observed for 1 subject, while in vivo parasite transcription and biomarkers were successfully profiled. CONCLUSIONS An IBSM model of P. malariae has been successfully developed and may be used to study the biology of, diagnostic testing for, and treatment of this neglected malaria species. CLINICAL TRIALS REGISTRATION ACTRN12617000048381.
Collapse
Affiliation(s)
- John Woodford
- QIMR Berghofer Medical Research Institute
- The University of Queensland
| | | | | | - Claire Wang
- Queensland Paediatric Infectious Diseases Laboratory, Brisbane, Australia
| | | | | | | | | | | | - Thomas D Otto
- Wellcome Sanger Institute, Hinxton
- Centre of Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute
- The University of Queensland
| |
Collapse
|
43
|
Wockner LF, Hoffmann I, Webb L, Mordmüller B, Murphy SC, Kublin JG, O'Rourke P, McCarthy JS, Marquart L. Growth Rate of Plasmodium falciparum: Analysis of Parasite Growth Data From Malaria Volunteer Infection Studies. J Infect Dis 2020; 221:963-972. [PMID: 31679015 PMCID: PMC7198127 DOI: 10.1093/infdis/jiz557] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/29/2019] [Indexed: 12/16/2022] Open
Abstract
Background Growth rate of malaria parasites in the blood of infected subjects is an important
measure of efficacy of drugs and vaccines. Methods We used log-linear and sine-wave models to estimate the parasite growth rate of the 3D7
strain of Plasmodium falciparum using data from 177 subjects from 14
induced blood stage malaria (IBSM) studies conducted at QIMR Berghofer. We estimated
parasite multiplication rate per 48 hour (PMR48), PMR per life-cycle
(PMRLC), and parasite life-cycle duration. We compared these parameters to
those from studies conducted elsewhere with infections induced by IBSM (n=66),
sporozoites via mosquito bite (n=336) or injection (n=51). Results The parasite growth rate of 3D7 in QIMR Berghofer studies was 0.75/day (95% CI:
0.73–0.77/day), PMR48 was 31.9 (95% CI: 28.7–35.4),
PMRLC was 16.4 (95% CI: 15.1–17.8) and parasite life-cycle was 38.8
hour (95% CI: 38.3–39.2 hour). These parameters were similar to estimates from
IBSM studies elsewhere (0.71/day, 95% CI: 0.67–0.75/day; PMR48 26.6,
95% CI: 22.2–31.8), but significantly higher (P < 0.001)
than in sporozoite studies (0.47/day, 95% CI: 0.43–0.50/day; PMR48
8.6, 95% CI: 7.3–10.1). Conclusions Parasite growth rates were similar across different IBSM studies and higher than
infections induced by sporozoite.
Collapse
Affiliation(s)
- Leesa F Wockner
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Isabell Hoffmann
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Lachlan Webb
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Sean C Murphy
- Departments of Laboratory Medicine and Microbiology, University of Washington, Seattle, Washington, USA
| | - James G Kublin
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Peter O'Rourke
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
44
|
Gruenberg M, Hofmann NE, Nate E, Karl S, Robinson LJ, Lanke K, Smith TA, Bousema T, Felger I. qRT-PCR versus IFA-based Quantification of Male and Female Gametocytes in Low-Density Plasmodium falciparum Infections and Their Relevance for Transmission. J Infect Dis 2020; 221:598-607. [PMID: 31437280 PMCID: PMC7325619 DOI: 10.1093/infdis/jiz420] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/14/2019] [Indexed: 11/22/2022] Open
Abstract
Background Accurate quantification of female and male gametocytes and sex ratios in asymptomatic low-density malaria infections are important for assessing their transmission potential. Gametocytes often escape detection even by molecular methods, therefore ultralow gametocyte densities were quantified in large blood volumes. Methods Female and male gametocytes were quantified in 161 PCR-positive Plasmodium falciparum infections from a cross-sectional survey in Papua New Guinea. Ten-fold concentrated RNA from 800 µL blood was analyzed using female-specific pfs25 and male-specific pfmget or mssp qRT-PCR. Gametocyte sex ratios from qRT-PCR were compared with those from immunofluorescence assays (IFA). Results Gametocytes were identified in 58% (93/161) P. falciparum-positive individuals. Mean gametocyte densities were frequently below 1 female and 1 male gametocyte/µL by qRT-PCR. The mean proportion of males was 0.39 (95% confidence interval, 0.33–0.44) by pfs25/pfmget qRT-PCR; this correlated well with IFA results (Pearsons r2 = 0.91; P < .001). A Poisson model fitted to our data predicted 16% P. falciparum-positive individuals that are likely to transmit, assuming at least 1 female and 1 male gametocyte per 2.5 µL mosquito bloodmeal. Conclusions Based on model estimates of female and male gametocytes per 2.5 µL blood, P. falciparum-positive individuals detected exclusively by ultrasensitive diagnostics are negligible for human-to-mosquito transmission. Estimating the transmission potential of ultralow-density malaria infections informs interventions. Almost all infections with ≥1 female and male gametocyte per 2.5 µL mosquito bloodmeal, and thus with highest likelihood of contributing to human-to-mosquito transmission, were detectable by standard molecular diagnostics.
Collapse
Affiliation(s)
- Maria Gruenberg
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Natalie E Hofmann
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Elma Nate
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Stephan Karl
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Leanne J Robinson
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Kjerstin Lanke
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas A Smith
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Teun Bousema
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ingrid Felger
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| |
Collapse
|
45
|
Gruenberg M, Moniz CA, Hofmann NE, Koepfli C, Robinson LJ, Nate E, Monteiro WM, de Melo GC, Kuehn A, Siqueira AM, Nguitragool W, Bassat Q, Lacerda M, Sattabongkot J, Mueller I, Felger I. Utility of ultra-sensitive qPCR to detect Plasmodium falciparum and Plasmodium vivax infections under different transmission intensities. Malar J 2020; 19:319. [PMID: 32883308 PMCID: PMC7469345 DOI: 10.1186/s12936-020-03374-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/13/2020] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The use of molecular diagnostics has revealed an unexpectedly large number of asymptomatic low-density malaria infections in many malaria endemic areas. This study compared the gains in parasite prevalence obtained by the use of ultra-sensitive (us)-qPCR as compared to standard qPCR in cross-sectional surveys conducted in Thailand, Brazil and Papua New Guinea (PNG). The compared assays differed in the copy number of qPCR targets in the parasite genome. METHODS Plasmodium falciparum (Pf) and Plasmodium vivax (Pv) parasites were quantified by qPCR amplifying the low-copy Pf_ and Pv_18S rRNA genes or the multi-copy targets Pf_varATS and Pv_mtCOX1. Cross-sectional surveys at the three study sites included 2252 participants of all ages and represented different transmission intensities. RESULTS In the two low-transmission areas, P. falciparum positivity was 1.3% (10/773) (Thailand) and 0.8% (5/651) (Brazil) using standard Pf_18S rRNA qPCR. In these two countries, P. falciparum positivity by Pf_varATS us-qPCR increased to 1.9% (15/773) and 1.7% (11/651). In PNG, an area with moderate transmission intensity, P. falciparum positivity significantly increased from 8.6% (71/828) by standard qPCR to 12.2% (101/828) by us-qPCR. The proportions of P. falciparum infections not detected by standard qPCR were 33%, 55% and 30% in Thailand, Brazil and PNG. Plasmodium vivax was the predominating species in Thailand and Brazil, with 3.9% (30/773) and 4.9% (32/651) positivity by Pv_18S rRNA qPCR. In PNG, P. vivax positivity was similar to P. falciparum, at 8.0% (66/828). Use of Pv_mtCOX1 us-qPCR led to a significant increase in positivity to 5.1% (39/773), 6.4% (42/651) and 11.5% (95/828) in Thailand, Brazil, and PNG. The proportions of P. vivax infections missed by standard qPCR were similar at all three sites, with 23%, 24% and 31% in Thailand, Brazil and PNG. CONCLUSION The proportional gains in the detection of P. falciparum and P. vivax infections by ultra-sensitive diagnostic assays were substantial at all three study sites. Thus, us-qPCR yields more precise prevalence estimates for both P. falciparum and P. vivax at all studied levels of endemicity and represents a significant diagnostic improvement. Improving sensitivity in P. vivax surveillance by us-qPCR is of particular benefit, because the additionally detected P. vivax infections signal the potential presence of hypnozoites and subsequent risk of relapse and further transmission.
Collapse
Affiliation(s)
- Maria Gruenberg
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Clara Antunes Moniz
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Natalie E Hofmann
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Cristian Koepfli
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA
| | - Leanne J Robinson
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea.,Burnet Institute, Melbourne, Australia
| | - Elma Nate
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | | | | | - Andrea Kuehn
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil.,ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Andre M Siqueira
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil.,Instituto Nacional de Infectologia Evandro Chagas, Fiocruz, Rio de Janeiro, Brazil
| | - Wang Nguitragool
- Department of Molecular Tropical Medicine & Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Quique Bassat
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Marcus Lacerda
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil.,Universidade Do Estado Do Amazonas, Manaus, Brazil
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Ivo Mueller
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia.,Malaria Parasite & Hosts Unit, Institut Pasteur, Paris, France
| | - Ingrid Felger
- Swiss Tropical and Public Health Institute, Basel, Switzerland. .,University of Basel, Basel, Switzerland.
| |
Collapse
|
46
|
Stepniewska K, Humphreys GS, Gonçalves BP, Craig E, Gosling R, Guerin PJ, Price RN, Barnes KI, Raman J, Smit MR, D’Alessandro U, Stone WJR, Bjorkman A, Samuels AM, Arroyo-Arroyo MI, Bastiaens GJH, Brown JM, Dicko A, El-Sayed BB, Elzaki SEG, Eziefula AC, Kariuki S, Kwambai TK, Maestre AE, Martensson A, Mosha D, Mwaiswelo RO, Ngasala BE, Okebe J, Roh ME, Sawa P, Tiono AB, Chen I, Drakeley CJ, Bousema T. Efficacy of Single-Dose Primaquine With Artemisinin Combination Therapy on Plasmodium falciparum Gametocytes and Transmission: An Individual Patient Meta-Analysis. J Infect Dis 2020; 225:1215-1226. [PMID: 32778875 PMCID: PMC8974839 DOI: 10.1093/infdis/jiaa498] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 08/06/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Since the World Health Organization recommended single low-dose (0.25 mg/kg) primaquine (PQ) in combination with artemisinin-based combination therapies (ACTs) in areas of low transmission or artemisinin-resistant Plasmodium falciparum, several single-site studies have been conducted to assess efficacy. METHODS An individual patient meta-analysis to assess gametocytocidal and transmission-blocking efficacy of PQ in combination with different ACTs was conducted. Random effects logistic regression was used to quantify PQ effect on (1) gametocyte carriage in the first 2 weeks post treatment; and (2) the probability of infecting at least 1 mosquito or of a mosquito becoming infected. RESULTS In 2574 participants from 14 studies, PQ reduced PCR-determined gametocyte carriage on days 7 and 14, most apparently in patients presenting with gametocytemia on day 0 (odds ratio [OR], 0.22; 95% confidence interval [CI], .17-.28 and OR, 0.12; 95% CI, .08-.16, respectively). Rate of decline in gametocyte carriage was faster when PQ was combined with artemether-lumefantrine (AL) compared to dihydroartemisinin-piperaquine (DP) (P = .010 for day 7). Addition of 0.25 mg/kg PQ was associated with near complete prevention of transmission to mosquitoes. CONCLUSIONS Transmission blocking is achieved with 0.25 mg/kg PQ. Gametocyte persistence and infectivity are lower when PQ is combined with AL compared to DP.
Collapse
Affiliation(s)
- Kasia Stepniewska
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom,Infectious Diseases Data Observatory, Oxford, United Kingdom,Kasia Stepniewska, PhD, WorldWide Antimalarial Resistance Network (WWARN), Centre for Tropical Medicine and Global Health, Churchill Hospital, CCVTM, University of Oxford, Old Road, Oxford OX3 7LE, UK
| | - Georgina S Humphreys
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom,Infectious Diseases Data Observatory, Oxford, United Kingdom,Green Templeton College, University of Oxford, Oxford, United Kingdom
| | - Bronner P Gonçalves
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Elaine Craig
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom,Infectious Diseases Data Observatory, Oxford, United Kingdom
| | - Roly Gosling
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA,Global Health Group, Malaria Elimination Initiative, University of California, San Francisco, California, USA
| | - Philippe J Guerin
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom,Infectious Diseases Data Observatory, Oxford, United Kingdom
| | - Ric N Price
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom,Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Norther Territory, Australia,Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Karen I Barnes
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom,University of Cape Town/Medical Research Council Collaborating Centre for Optimising Antimalarial Therapy, University of Cape Town, Cape Town, South Africa,Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Jaishree Raman
- University of Cape Town/Medical Research Council Collaborating Centre for Optimising Antimalarial Therapy, University of Cape Town, Cape Town, South Africa,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, National Health Laboratory Services, Johannesburg, South Africa,Wits Research Institute for Malaria, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Menno R Smit
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Umberto D’Alessandro
- Medical Research Council Unit The Gambia, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Will J R Stone
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom,Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anders Bjorkman
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Aaron M Samuels
- Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA,Centers for Disease Control and Prevention, Kisumu, Kenya
| | - Maria I Arroyo-Arroyo
- Grupo Salud y Comunidad, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Guido J H Bastiaens
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands,Department of Microbiology and Immunology, Rijnstate Hospital, Arnhem, the Netherlands
| | - Joelle M Brown
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Alassane Dicko
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Badria B El-Sayed
- Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
| | - Salah-Eldin G Elzaki
- Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
| | - Alice C Eziefula
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom,Department of Global Health and Infection, Brighton and Sussex Medical School, Brighton, United Kingdom
| | | | - Titus K Kwambai
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom,Kenya Medical Research Institute, Kisian, Kenya
| | - Amanda E Maestre
- Grupo Salud y Comunidad, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Andreas Martensson
- Department of Women’s and Children’s Health, International Maternal and Child Health, Uppsala University, Uppsala, Sweden
| | - Dominic Mosha
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania,Africa Academy for Public Health, Dar es Salaam, Tanzania
| | - Richard O Mwaiswelo
- Department of Parasitology and Medical Entomology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Billy E Ngasala
- Department of Parasitology and Medical Entomology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Joseph Okebe
- Department of International Public Health, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Michelle E Roh
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA,Global Health Group, Malaria Elimination Initiative, University of California, San Francisco, California, USA
| | - Patrick Sawa
- Human Health Division, International Centre for Insect Physiology and Ecology, Mbita Point, Kenya
| | - Alfred B Tiono
- Department of Biomedical Sciences, Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Ingrid Chen
- Global Health Group, Malaria Elimination Initiative, University of California, San Francisco, California, USA
| | - Chris J Drakeley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Teun Bousema
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom,Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands,Correspondence: Teun Bousema, PhD, Department of Medical Microbiology, Radboud Institute for Health Science, Radboudumc, PO Box 9101, 6500 HB Nijmegen, The Netherlands ()
| |
Collapse
|
47
|
Graumans W, Heutink R, van Gemert GJ, van de Vegte-Bolmer M, Bousema T, Collins KA. A mosquito feeding assay to examine Plasmodium transmission to mosquitoes using small blood volumes in 3D printed nano-feeders. Parasit Vectors 2020; 13:401. [PMID: 32771047 PMCID: PMC7414548 DOI: 10.1186/s13071-020-04269-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/29/2020] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND To understand the dynamics of malaria transmission, membrane feeding assays with glass feeders are used to assess the transmission potential of malaria infected individuals to mosquitoes. However, in some circumstances, use of these assays is hindered by both the blood volume requirement and the availability of fragile, specially crafted glass feeders. 3D printed plastic feeders that require very small volumes of blood would thus expand the utility of membrane feeding assays. METHODS Using two 3D printing production methods, MultiJet (MJ) and Digital Light Processing (DLP), we developed a plastic version of the most commonly used standard glass feeder (the mini-feeder) with an improved design, and also a smaller feeder requiring only 60 µl of blood (the nano-feeder). Performance of the 3D printed feeders was compared to standard glass mini-feeders by assessing infectivity of gametocytes to mosquitoes in standard membrane feeding assays with laboratory reared Anopheles stephensi mosquitoes and cultured Plasmodium falciparum gametocytes. In addition, the optimum number of mosquitoes that can feed on the nano-feeder was determined by evaluating fully fed mosquitoes visually and by assessing blood- meal volume with a colorimetric haemoglobin assay. RESULTS The 3D printing methods allowed quick and inexpensive production of durable feeders. Infectivity of gametocytes to mosquitoes was comparable for MJ and DLP 3D printed feeders and glass feeders, and the performance of the 3D printed feeders was not influenced by repeated washing with bleach. There was no loss in transmission efficiency when the feeder size was reduced from mini-feeder to nano-feeder, and blood-meal volume assessment indicated ~10 An. stephensi mosquitoes can take a full blood-meal (median volume 3.44 µl) on a nano-feeder. CONCLUSIONS Here we present 3D printed mini- and nano-feeders with comparable performance to the currently used glass mini-feeders. These feeders do not require specialized glass craftsmanship, making them easily accessible. Moreover, the smaller nano-feeders will enable evaluation of smaller blood volumes that can be collected from finger prick, thus expanding the utility of membrane feeding assays and facilitating a more thorough evaluation of the human infectious reservoir for malaria.
Collapse
Affiliation(s)
- Wouter Graumans
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Roel Heutink
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Marga van de Vegte-Bolmer
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Katharine A. Collins
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| |
Collapse
|
48
|
Watts RE, Odedra A, Marquart L, Webb L, Abd-Rahman AN, Cascales L, Chalon S, Rebelo M, Pava Z, Collins KA, Pasay C, Chen N, Peatey CL, Möhrle JJ, McCarthy JS. Safety and parasite clearance of artemisinin-resistant Plasmodium falciparum infection: A pilot and a randomised volunteer infection study in Australia. PLoS Med 2020; 17:e1003203. [PMID: 32822347 PMCID: PMC7444516 DOI: 10.1371/journal.pmed.1003203] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 07/21/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Artemisinin resistance is threatening malaria control. We aimed to develop and test a human model of artemisinin-resistant (ART-R) Plasmodium falciparum to evaluate the efficacy of drugs against ART-R malaria. METHODS AND FINDINGS We conducted 2 sequential phase 1, single-centre, open-label clinical trials at Q-Pharm, Brisbane, Australia, using the induced blood-stage malaria (IBSM) model, whereby healthy participants are intravenously inoculated with blood-stage parasites. In a pilot study, participants were inoculated (Day 0) with approximately 2,800 viable P. falciparum ART-R parasites. In a comparative study, participants were randomised to receive approximately 2,800 viable P. falciparum ART-R (Day 0) or artemisinin-sensitive (ART-S) parasites (Day 1). In both studies, participants were administered a single approximately 2 mg/kg oral dose of artesunate (AS; Day 9). Primary outcomes were safety, ART-R parasite infectivity, and parasite clearance. In the pilot study, 2 participants were enrolled between April 27, 2017, and September 12, 2017, and included in final analyses (males n = 2 [100%], mean age = 26 years [range, 23-28 years]). In the comparative study, 25 participants were enrolled between October 26, 2017, and October 18, 2018, of whom 22 were inoculated and included in final analyses (ART-R infected participants: males n = 7 [53.8%], median age = 22 years [range, 18-40 years]; ART-S infected participants: males n = 5 [55.6%], median age = 28 years [range, 22-35 years]). In both studies, all participants inoculated with ART-R parasites became parasitaemic. A total of 36 adverse events were reported in the pilot study and 277 in the comparative study. Common adverse events in both studies included headache, pyrexia, myalgia, nausea, and chills; none were serious. Seven participants experienced transient severe falls in white cell counts and/or elevations in liver transaminase levels which were considered related to malaria. Additionally, 2 participants developed ventricular extrasystoles that were attributed to unmasking of a predisposition to benign fever-induced tachyarrhythmia. In the comparative study, parasite clearance half-life after AS was significantly longer for ART-R infected participants (n = 13, 6.5 hours; 95% confidence interval [CI] 6.3-6.7 hours) compared with ART-S infected participants (n = 9, 3.2 hours; 95% CI 3.0-3.3 hours; p < 0.001). The main limitation of this study was that the ART-R and ART-S parasite strains did not share the same genetic background. CONCLUSIONS We developed the first (to our knowledge) human model of ART-R malaria. The delayed clearance profile of ART-R parasites after AS aligns with field study observations. Although based on a relatively small sample size, results indicate that this model can be safely used to assess new drugs against ART-R P. falciparum. TRIAL REGISTRATION The studies were registered with the Australian New Zealand Clinical Trials Registry: ACTRN12617000244303 (https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=372357) and ACTRN12617001394336 (https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=373637).
Collapse
Affiliation(s)
| | - Anand Odedra
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Lachlan Webb
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Laura Cascales
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Maria Rebelo
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Zuleima Pava
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Cielo Pasay
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Nanhua Chen
- Australian Army Malaria Institute, Brisbane, Australia
| | | | | | - James S. McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- * E-mail:
| |
Collapse
|
49
|
Wang CYT, Ballard E, Llewellyn S, Marquart L, Bousema T, McCarthy JS, Collins KA. Assays for quantification of male and female gametocytes in human blood by qRT-PCR in the absence of pure sex-specific gametocyte standards. Malar J 2020; 19:218. [PMID: 32576184 PMCID: PMC7310411 DOI: 10.1186/s12936-020-03291-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/17/2020] [Indexed: 12/25/2022] Open
Abstract
Background Malaria transmission from humans to Anopheles mosquitoes requires the presence of gametocytes in human peripheral circulation, and the dynamics of transmission are determined largely by the density and sex ratio of the gametocytes. Molecular methods are thus employed to measure gametocyte densities, particularly when assessing transmission epidemiology and the efficacy of transmission-blocking interventions. However, accurate quantification of male and female gametocytes with molecular methods requires pure male and female gametocytes as reference standards, which are not widely available. Methods qRT-PCR assays were used to quantify levels of sex-specific mRNA transcripts in Plasmodium falciparum female and male gametocytes (pfs25 and pfMGET, respectively) using synthetic complimentary RNA standards and in vitro cultured gametocytes. Assays were validated and assay performance was investigated in blood samples of clinical trial participants using these standards and compared to absolute quantification by droplet digital PCR (ddPCR). Results The number of transcript copies per gametocyte were determined to be 279.3 (95% CI 253.5–307.6) for the female-specific transcript pfs25, and 12.5 (95% CI 10.6–14.9) for the male-specific transcript pfMGET. These numbers can be used to convert from transcript copies/mL to gametocyte/mL. The reportable range was determined to be 5.71 × 106 to 5.71 female gametocytes/mL for pfs25, and 1.73 × 107 to 1.73 × 101 male gametocytes/mL for pfMGET. The limit of detection was 3.9 (95% CI 2.5–8.2) female gametocytes/mL for pfs25, and 26.9 (95% CI 19.3–51.7) male gametocytes/mL for PfMGET. Both assays showed minimal intra-assay and inter-assay variability with coefficient of variation < 3%. No cross-reactivity was observed in both assays in uninfected human blood samples. Comparison of results from ddPCR to qRT-PCR assays on clinical blood samples indicated a high-level agreement (ICC = 0.998 for pfs25 and 0.995 for pfMGET). Conclusions This study reports the validation of qRT-PCR assays that are able to accurately quantify female and male P. falciparum gametocytes at sub-microscopic densities. The assays showed excellent reproducibility, sensitivity, precision, specificity, and accuracy. The methodology will enable the estimation of gametocyte density in the absence of pure female and male gametocyte standards, and will facilitate clinical trials and epidemiological studies.
Collapse
Affiliation(s)
- Claire Y T Wang
- QPID Laboratory, Centre for Children's Health Research, Brisbane, QLD, Australia
| | - Emma Ballard
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Stacey Llewellyn
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Teun Bousema
- Radboud Institute for Health Science, Radboud University Medical Center, Nijmegen, The Netherlands
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Katharine A Collins
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia. .,Radboud Institute for Health Science, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
50
|
Collins KA, Abd-Rahman AN, Marquart L, Ballard E, Gobeau N, Griffin P, Chalon S, Möhrle JJ, McCarthy JS. Antimalarial activity of artefenomel against asexual parasites and transmissible gametocytes during experimental blood-stage Plasmodium vivax infection. J Infect Dis 2020; 225:1062-1069. [PMID: 32479608 PMCID: PMC8922009 DOI: 10.1093/infdis/jiaa287] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022] Open
Abstract
Background Interventions that effectively target Plasmodium vivax are critical for the future control and elimination of malaria. We conducted a P. vivax volunteer infection study to characterize the antimalarial activity of artefenomel, a new drug candidate. Methods Eight healthy, malaria-naive participants were intravenously inoculated with blood-stage P. vivax and subsequently received a single oral 200-mg dose of artefenomel. Blood samples were collected to monitor the development and clearance of parasitemia, and plasma artefenomel concentration. Mosquito feeding assays were conducted before artefenomel dosing to investigate parasite transmissibility. Results Initial parasite clearance occurred in all participants after artefenomel administration (log10 parasite reduction ratio over 48 hours, 1.67; parasite clearance half-life, 8.67 hours). Recrudescence occurred in 7 participants 11–14 days after dosing. A minimum inhibitory concentration of 0.62 ng/mL and minimum parasiticidal concentration that achieves 90% of maximum effect of 0.83 ng/mL were estimated, and a single 300-mg dose was predicted to clear 109 parasites per milliliter with 95% certainty. Gametocytemia developed in all participants and was cleared 4–8 days after dosing. At peak gametocytemia, 75% of participants were infectious to mosquitoes. Conclusions The in vivo antimalarial activity of artefenomel supports its further clinical development as a treatment for P. vivax malaria. Clinical Trials Registration NCT02573857.
Collapse
Affiliation(s)
| | | | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Herston QLD, Australia
| | - Emma Ballard
- QIMR Berghofer Medical Research Institute, Herston QLD, Australia
| | - Nathalie Gobeau
- Medicine for Malaria Venture, Route de Pré-Bois, Meyrin, Switzerland
| | - Paul Griffin
- QIMR Berghofer Medical Research Institute, Herston QLD, Australia.,The University of Queensland, Brisbane QLD, Australia.,Department of Medicine and Infectious Diseases, Mater Hospital and Mater Research, Raymond Terrace, South Brisbane QLD, Australia
| | - Stephan Chalon
- Medicine for Malaria Venture, Route de Pré-Bois, Meyrin, Switzerland
| | - Jörg J Möhrle
- Medicine for Malaria Venture, Route de Pré-Bois, Meyrin, Switzerland
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Herston QLD, Australia.,The University of Queensland, Brisbane QLD, Australia
| |
Collapse
|