1
|
Sanna A, Lambert Y, Jimeno Maroto I, Galindo MS, Plessis L, Bardon T, Carboni C, Bordalo J, Hiwat H, Cairo H, Musset L, Lazrek Y, Pelleau S, White M, Suárez Mutis M, Vreden S, Douine M. CUREMA project: a further step towards malaria elimination among hard-to-reach and mobile populations. Malar J 2024; 23:271. [PMID: 39256842 PMCID: PMC11385508 DOI: 10.1186/s12936-024-05040-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/09/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND In most countries engaged on the last mile towards malaria elimination, residual transmission mainly persists among vulnerable populations represented by isolated and mobile (often cross-border) communities. These populations are sometimes involved in informal or even illegal activities. In regions with Plasmodium vivax transmission, the specific biology of this parasite poses additional difficulties related to the need for a radical treatment against hypnozoites to prevent relapses. Among hard-to-reach communities, case management, a pillar of elimination strategy, is deficient: acute malaria attacks often occur in remote areas, where there is limited access to care, and drugs acquired outside formal healthcare are often inadequately used for treatment, which typically does not include radical treatment against P. vivax. For these reasons, P. vivax circulation among these communities represents one of the main challenges for malaria elimination in many non-African countries. The objective of this article is to describe the protocol of the CUREMA study, which aims to meet the challenge of targeting malaria in hard-to-reach populations with a focus on P. vivax. RESULTS CUREMA is a multi-centre, international public health intervention research project. The study population is represented by persons involved in artisanal and small-scale gold mining who are active and mobile in the Guiana Shield, deep inside the Amazon Forest. The CUREMA project includes a complex intervention composed of a package of actions: (1) health education activities; (2) targeted administration of treatment against P. vivax after screening against G6PD deficiency to asymptomatic persons considered at risk of silently carrying the parasite; (3) distribution of a self-testing and self-treatment kit (malakit) associated with user training for self-management of malaria symptoms occurring while in extreme isolation. These actions are offered by community health workers at settlements and neighbourhoods (often cross-border) that represent transit and logistic bases of gold miners. The study relies on hybrid design, aiming to evaluate both the effectiveness of the intervention on malaria transmission with a pre/post quasi-experimental design, and its implementation with a mixed methods approach. CONCLUSIONS The purpose of this study is to experiment an intervention that addresses both Plasmodium falciparum and P. vivax malaria elimination in a mobile and isolated population and to produce results that can be transferred to many contexts facing the same challenges around the world.
Collapse
Affiliation(s)
- Alice Sanna
- French West Indies-French Guiana Center for Clinical Investigation (CIC Inserm 1424), Department of Research, Innovation, and Public Health, Cayenne Hospital, Cayenne, French Guiana, France.
| | - Yann Lambert
- French West Indies-French Guiana Center for Clinical Investigation (CIC Inserm 1424), Department of Research, Innovation, and Public Health, Cayenne Hospital, Cayenne, French Guiana, France
| | - Irene Jimeno Maroto
- French West Indies-French Guiana Center for Clinical Investigation (CIC Inserm 1424), Department of Research, Innovation, and Public Health, Cayenne Hospital, Cayenne, French Guiana, France
| | - Muriel Suzanne Galindo
- French West Indies-French Guiana Center for Clinical Investigation (CIC Inserm 1424), Department of Research, Innovation, and Public Health, Cayenne Hospital, Cayenne, French Guiana, France
| | - Lorraine Plessis
- French West Indies-French Guiana Center for Clinical Investigation (CIC Inserm 1424), Department of Research, Innovation, and Public Health, Cayenne Hospital, Cayenne, French Guiana, France
| | - Teddy Bardon
- French West Indies-French Guiana Center for Clinical Investigation (CIC Inserm 1424), Department of Research, Innovation, and Public Health, Cayenne Hospital, Cayenne, French Guiana, France
| | - Carlotta Carboni
- French West Indies-French Guiana Center for Clinical Investigation (CIC Inserm 1424), Department of Research, Innovation, and Public Health, Cayenne Hospital, Cayenne, French Guiana, France
| | - Jane Bordalo
- Associação Desenvolvimento, Prevenção, Acompanhamento e Cooperação de Fronteiras (DPAC), Oiapoque, Brazil
| | - Helene Hiwat
- National Malaria Programme, Ministry of Health, Paramaribo, Suriname
| | - Hedley Cairo
- National Malaria Programme, Ministry of Health, Paramaribo, Suriname
| | - Lise Musset
- Laboratoire de Parasitologie, Institut Pasteur de la Guyane, Centre National de Référence du Paludisme, Cayenne, French Guiana, France
| | - Yassamine Lazrek
- Laboratoire de Parasitologie, Institut Pasteur de la Guyane, Centre National de Référence du Paludisme, Cayenne, French Guiana, France
| | - Stéphane Pelleau
- Infectious Disease Epidemiology and Analytics, Institut Pasteur, Université Paris Cité, Paris, France
| | - Michael White
- Infectious Disease Epidemiology and Analytics, Institut Pasteur, Université Paris Cité, Paris, France
| | - Martha Suárez Mutis
- Laboratory of Parasitic Diseases, Graduate Program in Tropical Medicine, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Stephen Vreden
- Foundation for the Advancement of Scientific Research in Suriname (SWOS), Paramaribo, Suriname
| | - Maylis Douine
- French West Indies-French Guiana Center for Clinical Investigation (CIC Inserm 1424), Department of Research, Innovation, and Public Health, Cayenne Hospital, Cayenne, French Guiana, France
| |
Collapse
|
2
|
Adissu W, Brito M, Garbin E, Macedo M, Monteiro W, Mukherjee SK, Myburg J, Alam MS, Bancone G, Bansil P, Pal S, Sharma A, Zobrist S, Bryan A, Chu CS, Das S, Domingo GJ, Hann A, Kublin J, Lacerda MVG, Layton M, Ley B, Murphy SC, Nosten F, Pereira D, Price RN, Talukdar A, Yilma D, Gerth-Guyette E. Clinical performance validation of the STANDARD G6PD test: A multi-country pooled analysis. PLoS Negl Trop Dis 2023; 17:e0011652. [PMID: 37824592 PMCID: PMC10597494 DOI: 10.1371/journal.pntd.0011652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 10/24/2023] [Accepted: 09/08/2023] [Indexed: 10/14/2023] Open
Abstract
INTRODUCTION Screening for G6PD deficiency can inform disease management including malaria. Treatment with the antimalarial drugs primaquine and tafenoquine can be guided by point-of-care testing for G6PD deficiency. METHODS AND FINDINGS Data from similar clinical studies evaluating the performance of the STANDARD G6PD Test (SD Biosensor, South Korea) conducted in Bangladesh, Brazil, Ethiopia, India, Thailand, the United Kingdom, and the United States were pooled. Test performance was assessed in a retrospective analysis on capillary and venous specimens. All study sites used spectrophotometry for reference G6PD testing, and either the HemoCue or complete blood count for reference hemoglobin measurement. The sensitivity of the STANDARD G6PD Test using the manufacturer thresholds for G6PD deficient and intermediate cases in capillary specimens from 4212 study participants was 100% (95% Confidence Interval (CI): 97.5%-100%) for G6PD deficient cases with <30% activity and 77% (95% CI 66.8%-85.4%) for females with intermediate activity between 30%-70%. Specificity was 98.1% (95% CI 97.6%-98.5%) and 92.8% (95% CI 91.6%-93.9%) for G6PD deficient individuals and intermediate females, respectively. Out of 20 G6PD intermediate females with false normal results, 12 had activity levels >60% on the reference assay. The negative predictive value for females with G6PD activity >60% was 99.6% (95% CI 99.1%-99.8%) on capillary specimens. Sensitivity among 396 P. vivax malaria cases was 100% (69.2%-100.0%) for both deficient and intermediate cases. Across the full dataset, 37% of those classified as G6PD deficient or intermediate resulted from true normal cases. Despite this, over 95% of cases would receive correct treatment with primaquine, over 87% of cases would receive correct treatment with tafenoquine, and no true G6PD deficient cases would be treated inappropriately based on the result of the STANDARD G6PD Test. CONCLUSIONS The STANDARD G6PD Test enables safe access to drugs which are contraindicated for individuals with G6PD deficiency. Operational considerations will inform test uptake in specific settings.
Collapse
Affiliation(s)
- Wondimagegn Adissu
- School of Medical Laboratory Sciences, Jimma University, Jimma, Ethiopia
- Clinical Trial Unit, Jimma University, Jimma, Ethiopia
| | - Marcelo Brito
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil
- Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Eduardo Garbin
- Centro de Pesquisa Em Medicina Tropical (CEPEM), Porto Velho, Rondônia, Brazil
| | - Marcela Macedo
- Centro de Pesquisa Em Medicina Tropical (CEPEM), Porto Velho, Rondônia, Brazil
| | - Wuelton Monteiro
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil
- Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | | | - Jane Myburg
- Special Haematology Laboratory, Hammersmith Hospital, London, United Kingdom
| | - Mohammad Shafiul Alam
- Infectious Diseases Division, International Centre for Diarrheal Diseases Research, Bangladesh (icddr,b), Mohakhali, Dhaka, Bangladesh
| | - Germana Bancone
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Pooja Bansil
- Diagnostics, PATH, Seattle, Washington, United States of America
| | - Sampa Pal
- Diagnostics, PATH, Seattle, Washington, United States of America
| | - Abhijit Sharma
- Diagnostics, PATH, Seattle, Washington, United States of America
| | | | - Andrew Bryan
- Departments of Laboratory Medicine and Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Cindy S. Chu
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Santasabuj Das
- National Institute of Cholera and Enteric Diseases, Kolkata, India
| | | | - Amanda Hann
- Special Haematology Laboratory, Hammersmith Hospital, London, United Kingdom
| | - James Kublin
- Departments of Laboratory Medicine and Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Marcus V. G. Lacerda
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil
- Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
- Instituto Leônidas & Maria Deane (ILMD), Fiocruz, Manaus, Amazonas, Brazil
| | - Mark Layton
- Special Haematology Laboratory, Hammersmith Hospital, London, United Kingdom
| | - Benedikt Ley
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
- Center for Emerging and Reemerging Infectious Diseases, University of Washington, Seattle, Washington, United States of America
| | - Francois Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Dhélio Pereira
- Centro de Pesquisa Em Medicina Tropical (CEPEM), Porto Velho, Rondônia, Brazil
- Universidade Federal de Rondônia (UNIR), Porto Velho, Rondônia, Brazil
| | - Ric N. Price
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| | | | - Daniel Yilma
- Clinical Trial Unit, Jimma University, Jimma, Ethiopia
- Department of Internal Medicine, Jimma University, Jimma, Ethiopia
| | | |
Collapse
|
3
|
Taylor WRJ, Meagher N, Ley B, Thriemer K, Bancone G, Satyagraha A, Assefa A, Chand K, Chau NH, Dhorda M, Degaga TS, Ekawati LL, Hailu A, Hasanzai MA, Naddim MN, Pasaribu AP, Rahim AG, Sutanto I, Thanh NV, Tuyet-Trinh NT, Waithira N, Woyessa A, Dondorp A, von Seidlein L, Simpson JA, White NJ, Baird JK, Day NP, Price RN. Weekly primaquine for radical cure of patients with Plasmodium vivax malaria and glucose-6-phosphate dehydrogenase deficiency. PLoS Negl Trop Dis 2023; 17:e0011522. [PMID: 37672548 PMCID: PMC10482257 DOI: 10.1371/journal.pntd.0011522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 07/10/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND The World Health Organization recommends that primaquine should be given once weekly for 8-weeks to patients with Plasmodium vivax malaria and glucose-6-phosphate dehydrogenase (G6PD) deficiency, but data on its antirelapse efficacy and safety are limited. METHODS Within the context of a multicentre, randomised clinical trial of two primaquine regimens in P. vivax malaria, patients with G6PD deficiency were excluded and enrolled into a separate 12-month observational study. They were treated with a weekly dose of 0.75 mg/kg primaquine for 8 weeks (PQ8W) plus dihydroartemisinin piperaquine (Indonesia) or chloroquine (Afghanistan, Ethiopia, Vietnam). G6PD status was diagnosed using the fluorescent spot test and confirmed by genotyping for locally prevalent G6PD variants. The risk of P. vivax recurrence following PQ8W and the consequent haematological recovery were characterized in all patients and in patients with genotypically confirmed G6PD variants, and compared with the patients enrolled in the main randomised control trial. RESULTS Between July 2014 and November 2017, 42 male and 8 female patients were enrolled in Afghanistan (6), Ethiopia (5), Indonesia (19), and Vietnam (20). G6PD deficiency was confirmed by genotyping in 31 patients: Viangchan (14), Mediterranean (4), 357A-G (3), Canton (2), Kaiping (2), and one each for A-, Chatham, Gaohe, Ludhiana, Orissa, and Vanua Lava. Two patients had recurrent P. vivax parasitaemia (days 68 and 207). The overall 12-month cumulative risk of recurrent P. vivax malaria was 5.1% (95% CI: 1.3-18.9) and the incidence rate of recurrence was 46.8 per 1000 person-years (95% CI: 11.7-187.1). The risk of P. vivax recurrence was lower in G6PD deficient patients treated with PQ8W compared to G6PD normal patients in all treatment arms of the randomised controlled trial. Two of the 26 confirmed hemizygous males had a significant fall in haemoglobin (>5g/dl) after the first dose but were able to complete their 8 week regimen. CONCLUSIONS PQ8W was highly effective in preventing P. vivax recurrences. Whilst PQ8W was well tolerated in most patients across a range of different G6PD variants, significant falls in haemoglobin may occur after the first dose and require clinical monitoring. TRIAL REGISTRATION This trial is registered at ClinicalTrials.gov (NCT01814683).
Collapse
Affiliation(s)
- Walter R. J. Taylor
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Niamh Meagher
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
- Department of Infectious Diseases University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Benedikt Ley
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| | - Kamala Thriemer
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| | - Germana Bancone
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Ari Satyagraha
- Eijkman Institute of Molecular Biology, Jakarta, Indonesia.8. Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | | | - Krisin Chand
- Oxford University Clinical Research Unit, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Nguyen Hoang Chau
- Oxford University Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Mehul Dhorda
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tamiru S. Degaga
- College of Medicine & Health Sciences, Arbaminch University, Arbaminch, Ethiopia
| | - Lenny L. Ekawati
- Oxford University Clinical Research Unit, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Asrat Hailu
- College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | | | | | | | - Awab Ghulam Rahim
- Nangarhar Medical Faculty, Nangarhar University, Ministry of Higher Education, Jalalabad, Afghanistan
- Health and Social Development Organization, Kabul, Afghanistan
| | - Inge Sutanto
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Ngo Viet Thanh
- Oxford University Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Tuyet-Trinh
- Oxford University Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Naomi Waithira
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Adugna Woyessa
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Arjen Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Lorenz von Seidlein
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Julie A. Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Nicholas J. White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - J. Kevin Baird
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Oxford University Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Nicholas P. Day
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ric N. Price
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| |
Collapse
|
4
|
Mordue DG, Hale SJ, Dennis WE, Vuong CV, Li XM, Yang N, Wormser GP. Plasma Blood Levels of Tafenoquine following a Single Oral Dosage in BALBc Mice with Acute Babesia microti Infection That Resulted in Rapid Clearance of Microscopically Detectable Parasitemia. Pathogens 2023; 12:1113. [PMID: 37764921 PMCID: PMC10534932 DOI: 10.3390/pathogens12091113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/26/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Previous studies of mice infected with Babesia microti have shown that a single dose of tafenoquine administered orally is extremely effective at decreasing microscopically detectable parasitemia. However, a critical limitation of studies to date is the lack of data concerning the plasma levels of tafenoquine that are needed to treat babesiosis. In the current study, we begin to address this gap by examining the plasma levels of tafenoquine associated with the rapid reduction of B. microti patent parasitemia in a mouse model of babesiosis. In the current study, we infected BALB/c mice with 1 × 107B. microti-infected red blood cells. Two days post-infection, mice were treated with 20 mg/kg of tafenoquine succinate or vehicle control administered orally by gavage. Parasitemia and plasma levels of tafenoquine were evaluated every 24 h post-treatment for 96 h. This allowed us to correlate blood plasma levels of tafenoquine with reductions in parasitemia in treated mice. Consistent with previous studies, a single oral dose of 20 mg/kg tafenoquine resulted in a rapid reduction in parasitemia. Plasma levels of tafenoquine 24 h post-administration ranged from 347 to 503 ng/mL and declined thereafter. This blood plasma tafenoquine level is similar to that achieved in humans using the current FDA-approved dose for the prevention of malaria.
Collapse
Affiliation(s)
- Dana G. Mordue
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10520, USA
| | - Synthia J. Hale
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10520, USA
| | - William E. Dennis
- Walter Reed Army Institute of Research, Experimental Therapeutics Branch, Department of Drug Development, Silver Spring, MD 20910, USA; (W.E.D.)
| | - Chau V. Vuong
- Walter Reed Army Institute of Research, Experimental Therapeutics Branch, Department of Drug Development, Silver Spring, MD 20910, USA; (W.E.D.)
| | - Xiu-Min Li
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10520, USA
| | - Nan Yang
- General Nutraceutical Technology, LLC, Elmsford, NY 10523, USA;
| | - Gary P. Wormser
- Division of Infectious Diseases, New York Medical College, Valhalla, NY 10520, USA;
| |
Collapse
|
5
|
Llanos-Cuentas A, Manrrique P, Rosas-Aguirre A, Herrera S, Hsiang MS. Tafenoquine for the treatment of Plasmodium vivax malaria. Expert Opin Pharmacother 2022; 23:759-768. [PMID: 35379070 DOI: 10.1080/14656566.2022.2058394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Plasmodium vivax malaria causes significant disease burden worldwide, especially in Latin America, Southeast Asia, and Oceania. P. vivax is characterized by the production of liver hypnozoites that cause clinical relapses upon periodic activation. Primaquine, an 8-aminoquinoline drug, has been the standard of care for decades to treat liver-stage P. vivax malaria; however, it requires long treatment regimens (one to two weeks) that lead to poor adherence and thus clinical relapses. Tafenoquine (TFQ), a newly available and efficacious single-dose 8-aminoquinoline, aims to address this challenge. Safe administration is possible when paired with the use of glucose-6-phosphate dehydrogenase (G6PD) diagnostics to prevent 8-aminoquinoline-induced hemolysis in patients with underlying G6PD deficiency (G6PDd). AREAS COVERED In this review, the authors present the recent literature regarding the pharmacology, efficacy, safety, and tolerability of TFQ and highlight regional differences in these areas. The authors also discuss the potential for TFQ, complemented with primaquine PQ and effective screening for G6PDd, to improve P. vivax clinical management and facilitate targeted mass drug administration in communities to decrease transmission. EXPERT OPINION Clinical studies show therapeutic efficacy of TFQ as well as a good performance in terms of safety and tolerability. Additional research regarding the effectiveness and safety TFQ in malaria elimination strategies such as targeted or mass drug administration are needed.
Collapse
Affiliation(s)
| | - Paulo Manrrique
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, PA, USA
| | - Angel Rosas-Aguirre
- Public Health and Administration, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Sonia Herrera
- Department of Epidemiology, Division of Infectious Diseases and Global Health, Department of Pediatrics, Division of Pediatric Infectious Diseases, University of California San Francisco, San Francisco, CA, United States
| | - Michelle S Hsiang
- Department of Epidemiology, Division of Infectious Diseases and Global Health, Department of Pediatrics, Division of Pediatric Infectious Diseases, University of California San Francisco, San Francisco, CA, United States.,Department of Epidemiology and Biostatistics, University of California San Francisco (UCSF), San Francisco, CA, USA.,Department of PediatricsUniversity of California San Francisco (UCSF), San Francisco, CA, USA
| |
Collapse
|
6
|
Fasinu PS, Chaurasiya ND, Dhammika Nanayakkara NP, Wang YH, Bandara Herath HMT, Avula B, McChesney JD, Jollow D, Walker LA, Tekwani BL. Comparative pharmacokinetics and tissue distribution of primaquine enantiomers in mice. Malar J 2022; 21:33. [PMID: 35123453 PMCID: PMC8817607 DOI: 10.1186/s12936-022-04054-4] [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: 11/26/2021] [Accepted: 01/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Primaquine (PQ) has been used for the radical cure of relapsing Plasmodium vivax malaria for more than 60 years. PQ is also recommended for prophylaxis and prevention of transmission of Plasmodium falciparum. However, clinical utility of PQ has been limited due to toxicity in individuals with genetic deficiencies in glucose 6-phosphate dehydrogenase (G6PD). PQ is currently approved for clinical use as a racemic mixture. Recent studies in animals as well as humans have established differential pharmacological and toxicological properties of the two enantiomers of PQ. This has been attributed to differential metabolism and pharmacokinetics of individual PQ enantiomers. The aim of the current study is to evaluate the comparative pharmacokinetics (PK), tissue distribution and metabolic profiles of the individual enantiomers in mice. METHODS Two groups of 21 male Albino ND4 Swiss mice were dosed orally with 45 mg/kg of S-(+)-PQ and R-(-)PQ respectively. Each of the enantiomers was comprised of a 50:50 mixture of 12C- and 13C- stable isotope labelled species (at 6 carbons on the benzene ring of the quinoline core). Three mice were euthanized from each group at different time points (at 0, 0.5, 1, 2, 4, 8, 24 h) and blood was collected by terminal cardiac bleed. Liver, spleen, lungs, kidneys and brain were removed, extracted and analysed using UPLC/MS. The metabolites were profiled by tandem mass (MS/MS) fragmentation profile and fragments with 12C-13C twin peaks. Non-compartmental analysis was performed using the Phoenix WinNonLin PK software module. RESULTS The plasma AUC0-last (µg h/mL) (1.6 vs. 0.6), T1/2 (h) (1.9 vs. 0.45), and Tmax (h) (1 vs. 0.5) were greater for SPQ as compared to RPQ. Generally, the concentration of SPQ was higher in all tissues. At Tmax, (0.5-1 h in all tissues), the level of SPQ was 3 times that of RPQ in the liver. Measured Cmax of SPQ and RPQ in the liver were about 100 and 40 times the Cmax values in plasma, respectively. Similar observations were recorded in other tissues where the concentration of SPQ was higher compared to RPQ (2× in the spleen, 6× in the kidneys, and 49× in the lungs) than in the plasma. CPQ, the major metabolite, was preferentially generated from RPQ, with higher levels in all tissues (> 10× in the liver, and 3.5× in the plasma) than from SPQ. The PQ-o-quinone was preferentially formed from the SPQ (> 4× compared to RPQ), with higher concentrations in the liver. CONCLUSION These studies show that in mice, PQ enantiomers are differentially biodistributed and metabolized, which may contribute to differential pharmacologic and toxicity profiles of PQ enantiomers. The findings on higher levels of PQ-o-quinone in liver and RBCs compared to plasma and preferential generation of this metabolite from SPQ are consistent with the higher anti-malarial efficacy of SPQ observed in the mouse causal prophylaxis test, and higher haemolytic toxicity in the humanized mouse model of G6PD deficiency. Potential relevance of these findings to clinical use of racemic PQ and other 8-aminoquinolines vis-à-vis need for further clinical evaluation of individual enantiomers are discussed.
Collapse
Affiliation(s)
- Pius S Fasinu
- Department of Pharmacology & Toxicology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Narayan D Chaurasiya
- Department of Infectious Diseases, Division of Scientific Platforms, Southern Research, Birmingham, AL, 35205, USA
| | - N P Dhammika Nanayakkara
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, 38677, USA
| | - Yan-Hong Wang
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, 38677, USA
| | - H M T Bandara Herath
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, 38677, USA
| | - Bharathi Avula
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, 38677, USA
| | | | - David Jollow
- Department of Pharmacology, Medical University of South Carolina, Charleston, SC, USA
| | - Larry A Walker
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, 38677, USA.
| | - Babu L Tekwani
- Department of Infectious Diseases, Division of Scientific Platforms, Southern Research, Birmingham, AL, 35205, USA.
| |
Collapse
|
7
|
Stewart AGA, Zimmerman PA, McCarthy JS. Genetic Variation of G6PD and CYP2D6: Clinical Implications on the Use of Primaquine for Elimination of Plasmodium vivax. Front Pharmacol 2021; 12:784909. [PMID: 34899347 PMCID: PMC8661410 DOI: 10.3389/fphar.2021.784909] [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: 09/28/2021] [Accepted: 11/05/2021] [Indexed: 12/03/2022] Open
Abstract
Primaquine, an 8-aminoquinoline, is the only medication approved by the World Health Organization to treat the hypnozoite stage of Plasmodium vivax and P. ovale malaria. Relapse, triggered by activation of dormant hypnozoites in the liver, can occur weeks to years after primary infection, and provides the predominant source of transmission in endemic settings. Hence, primaquine is essential for individual treatment and P. vivax elimination efforts. However, primaquine use is limited by the risk of life-threatening acute hemolytic anemia in glucose-6-phosphate dehydrogenase (G6PD) deficient individuals. More recently, studies have demonstrated decreased efficacy of primaquine due to cytochrome P450 2D6 (CYP2D6) polymorphisms conferring an impaired metabolizer phenotype. Failure of standard primaquine therapy has occurred in individuals with decreased or absent CYP2D6 activity. Both G6PD and CYP2D6 are highly polymorphic genes, with considerable geographic and interethnic variability, adding complexity to primaquine use. Innovative strategies are required to overcome the dual challenge of G6PD deficiency and impaired primaquine metabolism. Further understanding of the pharmacogenetics of primaquine is key to utilizing its full potential. Accurate CYP2D6 genotype-phenotype translation may optimize primaquine dosing strategies for impaired metabolizers and expand its use in a safe, efficacious manner. At an individual level the current challenges with G6PD diagnostics and CYP2D6 testing limit clinical implementation of pharmacogenetics. However, further characterisation of the overlap and spectrum of G6PD and CYP2D6 activity may optimize primaquine use at a population level and facilitate region-specific dosing strategies for mass drug administration. This precision public health approach merits further investigation for P. vivax elimination.
Collapse
Affiliation(s)
| | - Peter A Zimmerman
- The Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, United States
| | - James S McCarthy
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, VIC, Australia.,Peter Doherty Institute of Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
8
|
Angrisano F, Robinson LJ. Plasmodium vivax - How hidden reservoirs hinder global malaria elimination. Parasitol Int 2021; 87:102526. [PMID: 34896312 DOI: 10.1016/j.parint.2021.102526] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 01/23/2023]
Abstract
Plasmodium vivax is the most geographically widespread human malaria parasite. Global malaria efforts have been less successful at reducing the burden of P. vivax compared to P. falciparum, owing to the unique biology and related treatment complexity of P. vivax. As a result, P. vivax is now the dominant malaria parasite throughout the Asia-Pacific and South America causing up to 14 million clinical cases every year and is considered a major obstacle to malaria elimination. Key features circumventing existing malaria control tools are the transmissibility of asymptomatic, low-density circulating infections and reservoirs of persistent dormant liver stages (hypnozoites) that are undetectable but reactivate to cause relapsing infections and sustain transmission. In this review we summarise the new knowledge shaping our understanding of the global epidemiology of P. vivax infections, highlighting the challenges for elimination and the tools that will be required achieve this.
Collapse
|
9
|
Elexpe A, Nieto N, Fernández-Cuétara C, Domínguez-Fernández C, Morera-Herreras T, Torrecilla M, Miguélez C, Laso A, Ochoa E, Bailen M, González-Coloma A, Angulo-Barturen I, Astigarraga E, Barreda-Gómez G. Study of Tissue-Specific Reactive Oxygen Species Formation by Cell Membrane Microarrays for the Characterization of Bioactive Compounds. MEMBRANES 2021; 11:membranes11120943. [PMID: 34940444 PMCID: PMC8705675 DOI: 10.3390/membranes11120943] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 01/26/2023]
Abstract
The production of reactive oxygen species (ROS) increases considerably in situations of cellular stress, inducing lipid peroxidation and multiple alterations in proteins and nucleic acids. However, sensitivity to oxidative damage varies between organs and tissues depending on the triggering process. Certain drugs used in the treatment of diverse diseases such as malaria have side effects similar to those produced by oxidative damage, although no specific study has been conducted. For this purpose, cell membrane microarrays were developed and the superoxide production evoked by the mitochondrial activity was assayed in the presence of specific inhibitors: rotenone, antimycin A and azide. Once the protocol was set up on cell membrane isolated from rat brain areas, the effect of six antimalarial drugs (atovaquone, quinidine, doxycycline, mefloquine, artemisinin, and tafenoquine) and two essential oils (Rosmarinus officinalis and Origanum majoricum) were evaluated in multiple human samples. The basal activity was different depending on the type of tissue, the liver, jejunum and adrenal gland being the ones with the highest amount of superoxide. The antimalarial drugs studied showed specific behavior according to the type of human tissue analyzed, with atovaquone and quinidine producing the highest percentage of superoxide formation, and doxycycline the lowest. In conclusion, the analysis of superoxide production evaluated in cell membranes of a collection of human tissues allowed for the characterization of the safety profile of these antimalarial drugs against toxicity mediated by oxidative stress.
Collapse
Affiliation(s)
- Ane Elexpe
- Research and Development Division, IMG Pharma Biotech, 48160 Derio, Spain; (A.E.); (N.N.); (C.D.-F.); (E.A.)
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (T.M.-H.); (M.T.); (C.M.)
| | - Nerea Nieto
- Research and Development Division, IMG Pharma Biotech, 48160 Derio, Spain; (A.E.); (N.N.); (C.D.-F.); (E.A.)
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (T.M.-H.); (M.T.); (C.M.)
| | - Claudia Fernández-Cuétara
- Department of Preventive Medicine and Public Health and Microbiology, Faculty of Medicine, Autonomus University of Madrid UAM, 28029 Madrid, Spain; (C.F.-C.); (M.B.)
| | - Celtia Domínguez-Fernández
- Research and Development Division, IMG Pharma Biotech, 48160 Derio, Spain; (A.E.); (N.N.); (C.D.-F.); (E.A.)
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (T.M.-H.); (M.T.); (C.M.)
| | - Teresa Morera-Herreras
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (T.M.-H.); (M.T.); (C.M.)
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Instiute, 48903 Barakaldo, Spain
| | - María Torrecilla
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (T.M.-H.); (M.T.); (C.M.)
| | - Cristina Miguélez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (T.M.-H.); (M.T.); (C.M.)
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Instiute, 48903 Barakaldo, Spain
| | - Antonio Laso
- Research and Development Division, AleoVitro, 48160 Derio, Spain; (A.L.); (E.O.)
| | - Eneko Ochoa
- Research and Development Division, AleoVitro, 48160 Derio, Spain; (A.L.); (E.O.)
| | - María Bailen
- Department of Preventive Medicine and Public Health and Microbiology, Faculty of Medicine, Autonomus University of Madrid UAM, 28029 Madrid, Spain; (C.F.-C.); (M.B.)
| | - Azucena González-Coloma
- Institute of Agricultural Sciences (ICA), Spanish Research Council (CSIC), 28006 Madrid, Spain;
| | | | - Egoitz Astigarraga
- Research and Development Division, IMG Pharma Biotech, 48160 Derio, Spain; (A.E.); (N.N.); (C.D.-F.); (E.A.)
| | - Gabriel Barreda-Gómez
- Research and Development Division, IMG Pharma Biotech, 48160 Derio, Spain; (A.E.); (N.N.); (C.D.-F.); (E.A.)
- Correspondence: ; Tel.: +34-94-4316-577; Fax: +34-94-6013-455
| |
Collapse
|
10
|
Long-term safety of the tafenoquine antimalarial chemoprophylaxis regimen: A 12-month, randomized, double-blind, placebo-controlled trial. Travel Med Infect Dis 2021; 45:102211. [PMID: 34801714 DOI: 10.1016/j.tmaid.2021.102211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/09/2021] [Accepted: 11/15/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Tafenoquine is a long-acting 8-aminoquinoline approved for antimalarial prophylaxis for ≤6 months. Additional data is needed to establish the drug's longer-term safety profile, including potential ophthalmic or neuropsychiatric effects. METHOD This was a randomized, double-blind, placebo-controlled trial in 600 healthy adults. Eligible subjects were randomized 1:1 to receive tafenoquine 200 mg weekly (antimalarial prophylactic regimen) or placebo for 52 weeks. Scheduled safety visits occurred at Weeks 4, 12, 24, 52 (dosing completed), and 64 (final follow-up). Safety assessments included ophthalmic changes, general and neuropsychiatric adverse events (AEs), and laboratory value changes. RESULTS The percentage of subjects with a protocol-defined Serious Ophthalmic Safety Event was lower in the Tafenoquine Group (18.2%) versus the Placebo Group (19%, p = 0.308). There was no significant difference between the percentages of subjects with at least one AE in the Tafenoquine Group (91.0%) versus Placebo (89.9%, p = 0.65). Common AEs seen at a significantly higher incidence for tafenoquine included reversible cornea verticillata (54.5%) and nausea (13.0%), leading to 0.0% and 0.7% discontinuations. Psychiatric AEs occurred at similar percentages in both study groups. Reversible changes in hemoglobin, methemoglobin, creatinine, and blood urea nitrogen (BUN) were noted. CONCLUSIONS This study supports the safety of extended 52-week tafenoquine prophylaxis. CLINICAL TRIAL REGISTRATION NUMBER/CLINICALTRIALS. GOV IDENTIFIER NCT03320174.
Collapse
|
11
|
Markus MB. Safety and Efficacy of Tafenoquine for Plasmodium vivax Malaria Prophylaxis and Radical Cure: Overview and Perspectives. Ther Clin Risk Manag 2021; 17:989-999. [PMID: 34526770 PMCID: PMC8435617 DOI: 10.2147/tcrm.s269336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/20/2021] [Indexed: 11/23/2022] Open
Abstract
This article is inter alia a brief, first-stop guide to possible adverse events (AEs) associated with tafenoquine (TQ) intake. Safety and efficacy findings for TQ in Plasmodium vivax malaria prophylaxis and radical cure are summarized and some of the latest TQ-related studies (published in 2020 and 2021) are highlighted. In addition, little-known biological and other matters concerning malaria parasites and 8-aminoquinoline (8-AQ) drug action are discussed and some correct terminology pertinent to malaria is explained.
Collapse
Affiliation(s)
- Miles B Markus
- School of Animal, Plant and Environmental Sciences, Faculty of Science, University of the Witwatersrand, Johannesburg, South Africa
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
12
|
Agudelo Higuita NI, White BP, Franco-Paredes C, McGhee MA. An update on prevention of malaria in travelers. Ther Adv Infect Dis 2021; 8:20499361211040690. [PMID: 34484736 PMCID: PMC8408895 DOI: 10.1177/20499361211040690] [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: 04/27/2021] [Accepted: 08/02/2021] [Indexed: 11/20/2022] Open
Abstract
Malaria, a parasitic disease caused by protozoa belonging to the genus Plasmodium, continues to represent a formidable public health challenge. Despite being a preventable disease, cases reported among travelers have continued to increase in recent decades. Protection of travelers against malaria, a potentially life-threatening disease, is of paramount importance, and it is therefore necessary for healthcare professionals to be up to date with the most recent recommendations. The present review provides an update of the existent measures for malaria prevention among travelers.
Collapse
Affiliation(s)
| | - Bryan Pinckney White
- Infectious Diseases Clinical Pharmacist, Oklahoma University Medical Center, Oklahoma City, OK, USA
| | - Carlos Franco-Paredes
- Department of Medicine, University of Colorado Denver School of Medicine, Aurora, CO, USA
| | - Miranda Ann McGhee
- Department of Medicine, Section of Infectious Diseases, University of Oklahoma Health Science Center, 800 Stanton L. Young Blvd., Suite 7300, Oklahoma City, OK 73104, USA
| |
Collapse
|
13
|
Renard I, Ben Mamoun C. Treatment of Human Babesiosis: Then and Now. Pathogens 2021; 10:pathogens10091120. [PMID: 34578153 PMCID: PMC8469882 DOI: 10.3390/pathogens10091120] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 12/26/2022] Open
Abstract
Babesiosis is an emerging tick-borne disease caused by apicomplexan parasites of the genus Babesia. With its increasing incidence worldwide and the risk of human-to-human transmission through blood transfusion, babesiosis is becoming a rising public health concern. The current arsenal for the treatment of human babesiosis is limited and consists of combinations of atovaquone and azithromycin or clindamycin and quinine. These combination therapies were not designed based on biological criteria unique to Babesia parasites, but were rather repurposed based on their well-established efficacy against other apicomplexan parasites. However, these compounds are associated with mild or severe adverse events and a rapid emergence of drug resistance, thus highlighting the need for new therapeutic strategies that are specifically tailored to Babesia parasites. Herein, we review ongoing babesiosis therapeutic and management strategies and their limitations, and further review current efforts to develop new, effective, and safer therapies for the treatment of this disease.
Collapse
|
14
|
White NJ, Nosten FH. SERCAP: is the perfect the enemy of the good? Malar J 2021; 20:281. [PMID: 34167536 PMCID: PMC8223304 DOI: 10.1186/s12936-021-03821-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 06/15/2021] [Indexed: 01/07/2023] Open
Abstract
Single Encounter Radical Cure and Prophylaxis (SERCAP) describes an ideal anti-malarial drug that cures all malaria in a single dose. This target product profile has dominated anti-malarial drug discovery and development over the past decade. The operational advantage of a single encounter has to be balanced against the need for a high dose, reliable absorption, little variability in pharmacokinetic properties, slow elimination (to ensure curative drug exposures in all patients) and a very low rate of vomiting. The demanding aspirational target may have hindered anti-malarial drug development. Aiming for three-day regimens, as in current anti-malarial treatments, would be better.
Collapse
Affiliation(s)
- Nicholas J White
- Mahidol Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand. .,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK.
| | - François H Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK.,Shoklo Malaria Research Unit, MORU, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak, Thailand
| |
Collapse
|
15
|
Abstract
Lorenz von Seidlein and Nicholas White introduce a Collection on Plasmodium vivax malaria.
Collapse
|