1
|
Unger HW, Acharya S, Arnold L, Wu C, van Eijk AM, Gore-Langton GR, Ter Kuile FO, Lufele E, Chico RM, Price RN, Moore BR, Thriemer K, Rogerson SJ. The effect and control of malaria in pregnancy and lactating women in the Asia-Pacific region. Lancet Glob Health 2023; 11:e1805-e1818. [PMID: 37858590 DOI: 10.1016/s2214-109x(23)00415-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/10/2023] [Accepted: 08/23/2023] [Indexed: 10/21/2023]
Abstract
Half of all pregnancies at risk of malaria worldwide occur in the Asia-Pacific region, where Plasmodium falciparum and Plasmodium vivax co-exist. Despite substantial reductions in transmission, malaria remains an important cause of adverse health outcomes for mothers and offspring, including pre-eclampsia. Malaria transmission is heterogeneous, and infections are commonly subpatent and asymptomatic. High-grade antimalarial resistance poses a formidable challenge to malaria control in pregnancy in the region. Intermittent preventive treatment in pregnancy reduces infection risk in meso-endemic New Guinea, whereas screen-and-treat strategies will require more sensitive point-of-care tests to control malaria in pregnancy. In the first trimester, artemether-lumefantrine is approved, and safety data are accumulating for other artemisinin-based combinations. Safety of novel antimalarials to treat artemisinin-resistant P falciparum during pregnancy, and of 8-aminoquinolines during lactation, needs to be established. A more systematic approach to the prevention of malaria in pregnancy in the Asia-Pacific is required.
Collapse
Affiliation(s)
- Holger W Unger
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Department of Obstetrics and Gynaecology, Royal Darwin Hospital, Tiwi, NT, Australia; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Sanjaya Acharya
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Lachlan Arnold
- Royal Melbourne Hospital Clinical School, The University of Melbourne, Parkville, VIC, Australia
| | - Connie Wu
- Royal Melbourne Hospital Clinical School, The University of Melbourne, Parkville, VIC, Australia
| | - Anna Maria van Eijk
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Georgia R Gore-Langton
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Elvin Lufele
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Vector-Borne Diseases Unit, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - R Matthew Chico
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Ric N Price
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Brioni R Moore
- Curtin Medical School, Curtin University, Bentley, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia; Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, Australia
| | - Kamala Thriemer
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Stephen J Rogerson
- Department of Infectious Diseases, University of Melbourne, The Doherty Institute, Melbourne, VIC, Australia; Department of Medicine, University of Melbourne, The Doherty Institute, Melbourne, VIC, Australia
| |
Collapse
|
2
|
Saito M, Phyo AP, Chu C, Proux S, Rijken MJ, Beau C, Win HH, Archasuksan L, Wiladphaingern J, Phu NH, Hien TT, Day NP, Dondorp AM, White NJ, Nosten F, McGready R. Severe falciparum malaria in pregnancy in Southeast Asia: a multi-centre retrospective cohort study. BMC Med 2023; 21:320. [PMID: 37620809 PMCID: PMC10464355 DOI: 10.1186/s12916-023-02991-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Severe malaria in pregnancy causes maternal mortality, morbidity, and adverse foetal outcomes. The factors contributing to adverse maternal and foetal outcomes are not well defined. We aimed to identify the factors predicting higher maternal mortality and to describe the foetal mortality and morbidity associated with severe falciparum malaria in pregnancy. METHODS A retrospective cohort study was conducted of severe falciparum malaria in pregnancy, as defined by the World Health Organization severe malaria criteria. The patients were managed prospectively by the Shoklo Malaria Research Unit (SMRU) on the Thailand-Myanmar border or were included in hospital-based clinical trials in six Southeast Asian countries. Fixed-effects multivariable penalised logistic regression was used for analysing maternal mortality. RESULTS We included 213 (123 SMRU and 90 hospital-based) episodes of severe falciparum malaria in pregnancy managed between 1980 and 2020. The mean maternal age was 25.7 (SD 6.8) years, and the mean gestational age was 25.6 (SD 8.9) weeks. The overall maternal mortality was 12.2% (26/213). Coma (adjusted odds ratio [aOR], 7.18, 95% CI 2.01-25.57, p = 0.0002), hypotension (aOR 11.21, 95%CI 1.27-98.92, p = 0.03) and respiratory failure (aOR 4.98, 95%CI 1.13-22.01, p = 0.03) were associated with maternal mortality. Pregnant women with one or more of these three criteria had a mortality of 29.1% (25/86) (95%CI 19.5 to 38.7%) whereas there were no deaths in 88 pregnant women with hyperparasitaemia (> 10% parasitised erythrocytes) only or severe anaemia (haematocrit < 20%) only. In the SMRU prospective cohort, in which the pregnant women were followed up until delivery, the risks of foetal loss (23.3% by Kaplan-Meier estimator, 25/117) and small-for-gestational-age (38.3%, 23/60) after severe malaria were high. Maternal death, foetal loss and preterm birth occurred commonly within a week of diagnosis of severe malaria. CONCLUSIONS Vital organ dysfunction in pregnant women with severe malaria was associated with a very high maternal and foetal mortality whereas severe anaemia or hyperparasitaemia alone were not associated with poor prognosis, which may explain the variation of reported mortality from severe malaria in pregnancy. Access to antenatal care must be promoted to reduce barriers to early diagnosis and treatment of both malaria and anaemia.
Collapse
Affiliation(s)
- Makoto Saito
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
| | - Aung Pyae Phyo
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Cindy Chu
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Stephane Proux
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Marcus J Rijken
- Julius Global Health, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Obstetrics and Gynecology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Candy Beau
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Htun Htun Win
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Laypaw Archasuksan
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Jacher Wiladphaingern
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Nguyen H Phu
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Tran T Hien
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Nick P Day
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Arjen M Dondorp
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nicholas J White
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Akinola O, Ategbero E, Amusan AI, Gbotosho GO. Comparative efficacy of sulphadoxine-pyrimethamine and dihydroartemisinin-piperaquine against malaria infection during late-stage pregnancy in mice. Exp Parasitol 2023; 248:108500. [PMID: 36893971 DOI: 10.1016/j.exppara.2023.108500] [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: 10/05/2022] [Revised: 03/06/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023]
Abstract
The introduction of artemisinin combination therapies (ACTs) against malaria infections opened up a window of possibilities to combat malaria in pregnancy. However, the usefulness of ACTs in all stages of pregnancy must be critically assessed. This study was designed to evaluate dihydroartemisinin-piperaquine (DHAP) as a suitable alternative to sulphadoxine-pyrimethamine (SP) in the treatment of malaria during third-trimester pregnancy in mice. Experimental animals were inoculated with a parasitic dose of 1x106Plasmodium berghei (ANKA strain) infected erythrocytes and randomly allocated into treatment groups. The animals received standard doses of chloroquine alone (CQ)[10 mg/kg], SP [25 mg/kg] and [1.25 mg/kg] and DHAP [4 mg/kg] and [18 mg/kg] combinations. Maternal and pupil survival, litter sizes, pup weight and still-births were recorded, while the effect of the drug combinations on parasite suppression, recrudescence and parasite clearance time were evaluated. The day 4 chemo-suppression of parasitemia by DHAP in infected animals was comparable to SP, and CQ treatment (P > 0.05). The mean recrudescence time was significantly delayed (P = 0.031) in the DHAP treatment group compared to the CQ treatment group, while, there was no recrudescence in animals treated with SP. The birth rate in the SP group was significantly higher than in the DHAP group (P < 0.05). There was 100% maternal and pup survival in both combination treatments comparable with the uninfected gravid controls. The overall parasitological activity of SP against Plasmodium berghei in late-stage pregnancy appeared better than DHAP. In addition, SP treatment resulted in better birth outcomes assessed compared to DHAP treatment.
Collapse
Affiliation(s)
- Olugbenga Akinola
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Ibadan, Ibadan, Oyo State, Nigeria; Malaria Research Laboratories, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Elizabeth Ategbero
- Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Abiodun I Amusan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Ibadan, Ibadan, Oyo State, Nigeria; Malaria Research Laboratories, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Grace O Gbotosho
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Ibadan, Ibadan, Oyo State, Nigeria; Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Nigeria; Malaria Research Laboratories, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| |
Collapse
|
4
|
D'Alessandro S, Menegola E, Parapini S, Taramelli D, Basilico N. Safety of Artemisinin Derivatives in the First Trimester of Pregnancy: A Controversial Story. Molecules 2020; 25:molecules25153505. [PMID: 32752056 PMCID: PMC7435965 DOI: 10.3390/molecules25153505] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 12/31/2022] Open
Abstract
Artemisinin combination therapy (ACT) is recommended by the World Health Organization (WHO) as first line treatment for uncomplicated malaria both in adults and children. During pregnancy, ACT is considered safe only in the second and third trimester, since animal studies have demonstrated that artemisinin derivatives can cause foetal death and congenital malformation within a narrow time window in early embryogenesis. During this period, artemisinin derivatives induce defective embryonic erythropoiesis and vasculogenesis/angiogenesis in experimental models. However, clinical data on the safety profile of ACT in pregnant women have not shown an increased risk of miscarriage, stillbirth, or congenital malformation, nor low birth weight, associated with exposure to artemisinins in the first trimester. Although further studies are needed, the evidence collected up to now is prompting the WHO towards a change in the guidelines for the treatment of uncomplicated malaria, allowing the use of ACT also in the first trimester of pregnancy.
Collapse
Affiliation(s)
- Sarah D'Alessandro
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy
| | - Elena Menegola
- Dipartimento di Scienze e Politiche Ambientali, Università degli Studi di Milano, 20133 Milan, Italy
| | - Silvia Parapini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
| | - Donatella Taramelli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy
| | - Nicoletta Basilico
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
5
|
Nambozi M, Tinto H, Mwapasa V, Tagbor H, Kabuya JBB, Hachizovu S, Traoré M, Valea I, Tahita MC, Ampofo G, Buyze J, Ravinetto R, Arango D, Thriemer K, Mulenga M, van Geertruyden JP, D'Alessandro U. Artemisinin-based combination therapy during pregnancy: outcome of pregnancy and infant mortality: a cohort study. Malar J 2019; 18:105. [PMID: 30922317 PMCID: PMC6437904 DOI: 10.1186/s12936-019-2737-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 03/20/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The World Health Organization (WHO) recommendation of treating uncomplicated malaria during the second and third trimester of pregnancy with an artemisinin-based combination therapy (ACT) has already been implemented by all sub-Saharan African countries. However, there is limited knowledge on the effect of ACT on pregnancy outcomes, and on newborn and infant's health. METHODS Pregnant women with malaria in four countries (Burkina Faso, Ghana, Malawi and Zambia) were treated with either artemether-lumefantrine (AL), amodiaquine-artesunate (ASAQ), mefloquine-artesunate (MQAS), or dihydroartemisinin-piperaquine (DHA-PQ); 3127 live new-borns (822 in the AL, 775 in the ASAQ, 765 in the MQAS and 765 in the DHAPQ arms) were followed-up until their first birthday. RESULTS Prevalence of placental malaria and low birth weight were 28.0% (738/2646) and 16.0% (480/2999), respectively, with no significant differences between treatment arms. No differences in congenital malformations (p = 0.35), perinatal mortality (p = 0.77), neonatal mortality (p = 0.21), and infant mortality (p = 0.96) were found. CONCLUSIONS Outcome of pregnancy and infant survival were similar between treatment arms indicating that any of the four artemisinin-based combinations could be safely used during the second and third trimester of pregnancy without any adverse effect on the baby. Nevertheless, smaller safety differences between artemisinin-based combinations cannot be excluded; country-wide post-marketing surveillance would be very helpful to confirm such findings. Trial registration ClinicalTrials.gov, NCT00852423, Registered on 27 February 2009, https://clinicaltrials.gov/ct2/show/NCT00852423.
Collapse
Affiliation(s)
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé-Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | | | - Harry Tagbor
- University of Health and Allied Science, Ho, Ghana
| | | | | | - Maminata Traoré
- Institut de Recherche en Sciences de la Santé-Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Innocent Valea
- Institut de Recherche en Sciences de la Santé-Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Marc Christian Tahita
- Institut de Recherche en Sciences de la Santé-Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Gifty Ampofo
- University of Health and Allied Science, Ho, Ghana
| | - Jozefien Buyze
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Raffaella Ravinetto
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | - Diana Arango
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Kamala Thriemer
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium.,Menzies School of Health Research, Darwin, Australia
| | | | | | - Umberto D'Alessandro
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia.
| |
Collapse
|
6
|
Sridharan K, Sivaramakrishnan G, Kanters S. Adverse pregnancy outcomes between the anti-malarial drugs: Is there a difference between the drugs recommended by World Health Organization? Results of a mixed treatment comparison analysis of randomized clinical trials and cohort studies. INTERNATIONAL JOURNAL OF RISK & SAFETY IN MEDICINE 2019; 30:73-89. [PMID: 30714973 DOI: 10.3233/jrs-180022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Data regarding the relative safety profile of anti-malarial drugs in pregnancy is sparse mainly limited by the absence of head-to-head clinical trials. The present study is a network meta-analysis of safety of anti-malarial drugs used to treat malaria in pregnant women. METHODS A thorough literature search using the search strategy "Malaria [tiab] AND (Pregnant [tiab] OR Pregnancy [tiab])" was carried out for either randomized controlled trials or prospective cohort studies in pregnant malarial women prescribed any of the recommended anti-malarial drugs by World Health Organization (WHO) and that have reported adverse pregnancy outcomes such as miscarriage, still birth, and neonatal deaths. Odds ratio with 95% confidence interval was used as the effect estimate. Random-effects model and Markov Chain Monte Carlo simulation method was used to generate pooled estimates. Sensitivity analysis was performed excluding data from first trimester and GRADE approach was used to categorize the quality of evidence. RESULTS A total of 1242 papers were obtained with the search strategy, of which seven evaluating 10 treatment arms in a total of 5510 participants were included in the present meta-analysis. The pooled estimates revealed significantly lower risks of abortion with quinine and artemisinin-lumefantrine compared to dihydroartemisinin-piperaquine, artesunate with mefloquine and artesunate with amodiaquine. But when a cohort study that was conducted in the first trimester of pregnancy was excluded, no significant differences were observed in the risk of abortion between the anti-malarial drugs. No significant differences in the risk of either stillbirths or neonatal deaths were observed with any of the drugs. The quality of evidence was found to be very low due to serious limitations in both the precision and indirectness. CONCLUSION WHO recommended anti-malarials in pregnancy have similar risk profiles with regard to abortion, stillbirth and neonatal deaths.
Collapse
Affiliation(s)
- Kannan Sridharan
- Department of Pharmacology and Therapeutics, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Gowri Sivaramakrishnan
- Department of Oral Health, College of Medicine, Nursing and Health Sciences, Fiji National University, Suva, Fiji
| | - Steve Kanters
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
| |
Collapse
|
7
|
Saito M, Gilder ME, McGready R, Nosten F. Antimalarial drugs for treating and preventing malaria in pregnant and lactating women. Expert Opin Drug Saf 2018; 17:1129-1144. [PMID: 30351243 DOI: 10.1080/14740338.2018.1535593] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Malaria in pregnancy and postpartum cause maternal mortality and adverse fetal outcomes. Efficacious and safe antimalarials are needed to treat and prevent such serious consequences. However, because of the lack of evidence on fetal safety, quinine, an old and less efficacious drug has long been recommended for pregnant women. Uncertainty about safety in relation to breastfeeding leads to withholding of efficacious treatments postpartum or cessation of breastfeeding. Areas covered: A search identified literature on humans in three databases (MEDLINE, Embase and Global health) using pregnancy or lactation, and the names of antimalarial drugs as search terms. Adverse reactions to the mother, fetus or breastfed infant were summarized together with efficacies. Expert opinion: Artemisinins are more efficacious and well-tolerated than quinine in pregnancy. Furthermore, the risks of miscarriage, stillbirth or congenital abnormality were not higher in pregnancies exposed to artemisinin derivatives for treatment of malaria than in pregnancies exposed to quinine or in the comparable background population unexposed to any antimalarials, and this was true for treatment in any trimester. Assessment of safety and efficacy of antimalarials including dose optimization for pregnant women is incomplete. Resistance to sulfadoxine-pyrimethamine in Plasmodium falciparum and long unprotected intervals between intermittent treatment doses begs reconsideration of current preventative recommendations in pregnancy. Data remain limited on antimalarials during breastfeeding; while most first-line drugs appear safe, further research is needed.
Collapse
Affiliation(s)
- Makoto Saito
- a Shoklo Malaria Research Unit (SMRU), Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine , Mahidol University , Tak , Thailand.,b Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine , University of Oxford , Oxford , UK.,c WorldWide Antimalarial Resistance Network (WWARN) , Oxford , UK
| | - Mary Ellen Gilder
- a Shoklo Malaria Research Unit (SMRU), Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine , Mahidol University , Tak , Thailand
| | - Rose McGready
- a Shoklo Malaria Research Unit (SMRU), Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine , Mahidol University , Tak , Thailand.,b Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine , University of Oxford , Oxford , UK
| | - François Nosten
- a Shoklo Malaria Research Unit (SMRU), Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine , Mahidol University , Tak , Thailand.,b Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine , University of Oxford , Oxford , UK
| |
Collapse
|
8
|
Moore BR, Davis TME. Pharmacotherapy for the prevention of malaria in pregnant women: currently available drugs and challenges. Expert Opin Pharmacother 2018; 19:1779-1796. [PMID: 30289730 DOI: 10.1080/14656566.2018.1526923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Malaria in pregnancy continues to be a significant public health burden globally, with over 100 million women at risk each year. Sulfadoxine-pyrimethamine (SP) is the only antimalarial recommended for intermittent preventive therapy in pregnancy (IPTp) but increasing parasite resistance threatens its viability. There are few other available antimalarial therapies that currently have sufficient evidence of tolerability, safety, and efficacy to replace SP. AREAS COVERED Novel antimalarial combinations are under investigation for potential use as chemoprophylaxis and in IPTp regimens. The present review summarizes currently available therapies, emerging candidate combination therapies, and the potential challenges to integrating these into mainstream policy. EXPERT OPINION Alternative drugs or combination therapies to SP for IPTp are desperately required. Dihydroartemisinin-piperaquine and azithromycin-based combinations are showing great promise as potential candidates for IPTp but pharmacokinetic data suggest that dose modification may be required to ensure adequate prophylactic efficacy. If a suitable candidate regimen is not identified in the near future, the success of chemopreventive strategies such as IPTp may be in jeopardy.
Collapse
Affiliation(s)
- Brioni R Moore
- a School of Pharmacy and Biomedical Sciences , Curtin University , Bentley , Western Australia , Australia.,b Medical School , University of Western Australia , Crawley , Western Australia , Australia
| | - Timothy M E Davis
- b Medical School , University of Western Australia , Crawley , Western Australia , Australia
| |
Collapse
|
9
|
Chan XHS, Win YN, Mawer LJ, Tan JY, Brugada J, White NJ. Risk of sudden unexplained death after use of dihydroartemisinin-piperaquine for malaria: a systematic review and Bayesian meta-analysis. THE LANCET. INFECTIOUS DISEASES 2018; 18:913-923. [PMID: 29887371 PMCID: PMC6060085 DOI: 10.1016/s1473-3099(18)30297-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/01/2018] [Accepted: 05/04/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Dihydroartemisinin-piperaquine is an effective and well tolerated artemisinin-based combination therapy that has been assessed extensively for the prevention and treatment of malaria. Piperaquine, similar to several structurally related antimalarials currently used, can prolong cardiac ventricular repolarisation duration and the electrocardiographic QT interval, leading to concerns about its proarrhythmic potential. We aimed to assess the risk of potentially lethal iatrogenic ventricular arrhythmias in individuals receiving dihydroartemisinin-piperaquine. METHODS We did a systematic review and Bayesian meta-analysis. We searched clinical bibliographic databases (last on May 24, 2017) for studies of dihydroartemisinin-piperaquine in human beings. Further unpublished studies were identified with the WHO Evidence Review Group on the Cardiotoxicity of Antimalarials. We searched for articles containing "dihydroartemisinin-piperaquine" as title, abstract, or subject heading keywords, with synonyms and variant spellings as additional search terms. We excluded animal studies, but did not apply limits on language or publication date. Eligible studies were prospective, randomised, controlled trials or cohort studies in which individuals received at least one 3-day treatment course of dihydroartemisinin-piperaquine for mass drug administration, preventive therapy, or case management of uncomplicated malaria, with follow-up over at least 3 days. At least two independent reviewers screened titles, abstracts, and full texts, agreed study eligibility, and extracted information about study and participant characteristics, adverse event surveillance methodology, dihydroartemisinin-piperaquine exposures, loss-to-follow up, and any deaths after dihydroartemisinin-piperaquine treatment into a standardised database. The risk of sudden unexplained death after dihydroartemisinin-piperaquine with 95% credible intervals (CI) generated by Bayesian meta-analysis was compared with the baseline rate of sudden cardiac death. FINDINGS Our search identified 94 eligible primary studies including data for 197 867 individuals who had received dihydroartemisinin-piperaquine: 154 505 in mass drug administration programmes; 15 188 in 14 studies of repeated courses in preventive therapies and case management of uncomplicated malaria; and 28 174 as single-course treatments of uncomplicated malaria in 76 case-management studies. There was one potentially drug-related sudden unexplained death: a healthy woman aged 16 in Mozambique who developed heart palpitations several hours after the second dose of dihydroartemisinin-piperaquine and collapsed and died on the way to hospital (no autopsy or ECG was done). The median pooled risk estimate of sudden unexplained death after dihydroartemisinin-piperaquine was 1 in 757 950 (95% CI 1 in 2 854 490 to 1 in 209 114). This risk estimate was not higher than the baseline rate of sudden cardiac death (0·7-11·9 per 100 000 person-years or 1 in 1 714 280 to 1 in 100 835 over a 30-day risk period). The risk of bias was low in most studies and unclear in a few. INTERPRETATION Dihydroartemisinin-piperaquine was associated with a low risk of sudden unexplained death that was not higher than the baseline rate of sudden cardiac death. Concerns about repolarisation-related cardiotoxicity need not limit its current use for the prevention and treatment of malaria. FUNDING Wellcome Trust, UK Medical Research Council, WHO, Bill & Melinda Gates Foundation, and University of Oxford.
Collapse
Affiliation(s)
- Xin Hui S Chan
- 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, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Yan Naung Win
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Defence Services Medical Research Centre & Health and Disease Control Unit, Naypyidaw, Myanmar
| | - Laura J Mawer
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Royal Free London NHS Foundation Trust, London, UK
| | - Jireh Y Tan
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Josep Brugada
- Arrhythmia Section, Cardiology Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), University of Barcelona, Spain
| | - 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, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
10
|
D'Alessandro U, Hill J, Tarning J, Pell C, Webster J, Gutman J, Sevene E. Treatment of uncomplicated and severe malaria during pregnancy. THE LANCET. INFECTIOUS DISEASES 2018; 18:e133-e146. [PMID: 29395998 DOI: 10.1016/s1473-3099(18)30065-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 07/19/2017] [Accepted: 10/13/2017] [Indexed: 12/13/2022]
Abstract
Over the past 10 years, the available evidence on the treatment of malaria during pregnancy has increased substantially. Owing to their relative ease of use, good sensitivity and specificity, histidine rich protein 2 based rapid diagnostic tests are appropriate for symptomatic pregnant women; however, such tests are less appropriate for systematic screening because they will not detect an important proportion of infections among asymptomatic women. The effect of pregnancy on the pharmacokinetics of antimalarial drugs varies greatly between studies and class of antimalarial drugs, emphasising the need for prospective studies in pregnant and non-pregnant women. For the treatment of malaria during the first trimester, international guidelines are being reviewed by WHO. For the second and third trimester of pregnancy, results from several trials have confirmed that artemisinin-based combination treatments are safe and efficacious, although tolerability and efficacy might vary by treatment. It is now essential to translate such evidence into policies and clinical practice that benefit pregnant women in countries where malaria is endemic. Access to parasitological diagnosis or appropriate antimalarial treatment remains low in many countries and regions. Therefore, there is a pressing need for research to identify quality improvement interventions targeting pregnant women and health providers. In addition, efficient and practical systems for pharmacovigilance are needed to further expand knowledge on the safety of antimalarial drugs, particularly in the first trimester of pregnancy.
Collapse
Affiliation(s)
- Umberto D'Alessandro
- Medical Research Council Unit, Banjul, The Gambia; London School of Hygiene & Tropical Medicine, London, UK.
| | - Jenny Hill
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Joel Tarning
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Christopher Pell
- Centre for Social Science and Global Health, University of Amsterdam, Amsterdam, Netherlands; Amsterdam Institute for Global Health and Development, Amsterdam, Netherlands
| | - Jayne Webster
- London School of Hygiene & Tropical Medicine, London, UK
| | - Julie Gutman
- Malaria Branch, US Centers for Diseases Control and Prevention, Atlanta, GA, USA
| | - Esperanca Sevene
- Manhiça Health Research Center (CISM), Manhiça, Mozambique; Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| |
Collapse
|
11
|
Olafuyi O, Coleman M, Badhan RK. The application of physiologically based pharmacokinetic modelling to assess the impact of antiretroviral-mediated drug-drug interactions on piperaquine antimalarial therapy during pregnancy. Biopharm Drug Dispos 2017; 38:464-478. [DOI: 10.1002/bdd.2087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/22/2017] [Accepted: 07/06/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Olusola Olafuyi
- Aston Healthy Research Group, Aston Pharmacy School; Aston University; Birmingham B4 7ET UK
| | - Michael Coleman
- Aston Pharmacy School; Aston University; Birmingham B4 7ET UK
| | - Raj K.S. Badhan
- Aston Healthy Research Group, Aston Pharmacy School; Aston University; Birmingham B4 7ET UK
- Aston Pharmacy School; Aston University; Birmingham B4 7ET UK
| |
Collapse
|
12
|
Natureeba P, Kakuru A, Muhindo M, Ochieng T, Ategeka J, Koss CA, Plenty A, Charlebois ED, Clark TD, Nzarubara B, Nakalembe M, Cohan D, Rizzuto G, Muehlenbachs A, Ruel T, Jagannathan P, Havlir DV, Kamya MR, Dorsey G. Intermittent Preventive Treatment With Dihydroartemisinin-Piperaquine for the Prevention of Malaria Among HIV-Infected Pregnant Women. J Infect Dis 2017; 216:29-35. [PMID: 28329368 DOI: 10.1093/infdis/jix110] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 02/21/2017] [Indexed: 11/13/2022] Open
Abstract
Background Daily trimethoprim-sulfamethoxazole (TMP-SMX) and insecticide-treated nets remain the main interventions for prevention of malaria in human immunodeficiency virus (HIV)-infected pregnant women in Africa. However, antifolate and pyrethroid resistance threaten the effectiveness of these interventions, and new ones are needed. Methods We conducted a double-blinded, randomized, placebo-controlled trial comparing daily TMP-SMX plus monthly dihydroartemisinin-piperaquine (DP) to daily TMP-SMX alone in HIV-infected pregnant women in an area of Uganda where indoor residual spraying of insecticide had recently been implemented. Participants were enrolled between gestation weeks 12 and 28 and given an insecticide-treated net. The primary outcome was detection of active or past placental malarial infection by histopathologic analysis. Secondary outcomes included incidence of malaria, parasite prevalence, and adverse birth outcomes. Result All 200 women enrolled were followed through delivery, and the primary outcome was assessed in 194. There was no statistically significant difference in the risk of histopathologically detected placental malarial infection between the daily TMP-SMX plus DP arm and the daily TMP-SMX alone arm (6.1% vs. 3.1%; relative risk, 1.96; 95% confidence interval, .50-7.61; P = .50). Similarly, there were no differences in secondary outcomes. Conclusions Among HIV-infected pregnant women in the setting of indoor residual spraying of insecticide, adding monthly DP to daily TMP-SMX did not reduce the risk of placental or maternal malaria or improve birth outcomes. Clinical Trials Registration NCT02282293.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Atis Muehlenbachs
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | | | | - Moses R Kamya
- School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | | |
Collapse
|
13
|
Osarfo J, Tagbor H, Cairns M, Alifrangis M, Magnussen P. Dihydroartemisinin-piperaquine versus artesunate-amodiaquine for treatment of malaria infection in pregnancy in Ghana: an open-label, randomised, non-inferiority trial. Trop Med Int Health 2017; 22:1043-1052. [PMID: 28556586 DOI: 10.1111/tmi.12905] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To determine whether dihydroartemisinin-piperaquine (DHA-PPQ) is non-inferior to artesunate-amodiaquine (ASAQ) for treating uncomplicated malaria infection in pregnancy. METHODS A total of 417 second/ third trimester pregnant women with confirmed asymptomatic Plasmodium falciparum parasitaemia were randomised to receive DHA-PPQ or ASAQ over 3 days. Women were followed up on days 1, 2, 3, 7, 14, 28 and 42 after treatment start and at delivery for parasitological, haematological, birth outcomes and at 6-week post-partum to ascertain the health status of the babies. Parasitological efficacy (PE) by days 28 and 42 were co-primary outcomes. Analysis was per-protocol (PP) and modified intention-to-treat (ITT). Non-inferiority was declared if the two-sided 95% confidence interval for PE at the endpoints excluded 5% lower efficacy for DHA-PPQ. Secondary outcomes were assessed for superiority. RESULTS In PP analysis, PE was 91.6% for DHA-PPQ and 89.3% for ASAQ by day 28 and 89.0% and 86.5%, respectively, by day 42. DHA-PPQ was non-inferior to ASAQ with respect to uncorrected PE [adjusted difference by day 28 (DHA-PPQ-ASAQ); 3.5% (95%CI: -1.5, 8.5); and day 42: 3.9% (95%CI: -2.7, 10.4)]. ITT analysis gave similar results. PCR to distinguish recrudescence and reinfection was unsuccessful. DHA-PPQ recipients had fewer adverse events of vomiting, dizziness, and general weakness compared to ASAQ. Both drugs were well-tolerated, and there was no excess of adverse birth outcomes. CONCLUSION DHA-PPQ was non-inferior to ASAQ for treatment of malaria infection during pregnancy. No safety concerns were identified. Our findings contribute to growing evidence that DHA-PPQ is useful for control of malaria in pregnancy.
Collapse
Affiliation(s)
- Joseph Osarfo
- Ghana Health Service, Effiduase District Hospital, Effiduase, Ashanti Region, Ghana
| | - Harry Tagbor
- School of Public Health, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.,School of Medicine, University of Health and Allied Sciences, Ho, Ghana
| | - Matthew Cairns
- MRC Tropical Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Michael Alifrangis
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Pascal Magnussen
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,Department of Veterinary Disease Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Clark RL. Animal Embryotoxicity Studies of Key Non-Artemisinin Antimalarials and Use in Women in the First Trimester. Birth Defects Res 2017. [DOI: 10.1002/bdr2.1035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Dellicour S, Sevene E, McGready R, Tinto H, Mosha D, Manyando C, Rulisa S, Desai M, Ouma P, Oneko M, Vala A, Rupérez M, Macete E, Menéndez C, Nakanabo-Diallo S, Kazienga A, Valéa I, Calip G, Augusto O, Genton B, Njunju EM, Moore KA, d’Alessandro U, Nosten F, ter Kuile F, Stergachis A. First-trimester artemisinin derivatives and quinine treatments and the risk of adverse pregnancy outcomes in Africa and Asia: A meta-analysis of observational studies. PLoS Med 2017; 14:e1002290. [PMID: 28463996 PMCID: PMC5412992 DOI: 10.1371/journal.pmed.1002290] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 03/23/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Animal embryotoxicity data, and the scarcity of safety data in human pregnancies, have prevented artemisinin derivatives from being recommended for malaria treatment in the first trimester except in lifesaving circumstances. We conducted a meta-analysis of prospective observational studies comparing the risk of miscarriage, stillbirth, and major congenital anomaly (primary outcomes) among first-trimester pregnancies treated with artemisinin derivatives versus quinine or no antimalarial treatment. METHODS AND FINDINGS Electronic databases including Medline, Embase, and Malaria in Pregnancy Library were searched, and investigators contacted. Five studies involving 30,618 pregnancies were included; four from sub-Saharan Africa (n = 6,666 pregnancies, six sites) and one from Thailand (n = 23,952). Antimalarial exposures were ascertained by self-report or active detection and confirmed by prescriptions, clinic cards, and outpatient registers. Cox proportional hazards models, accounting for time under observation and gestational age at enrollment, were used to calculate hazard ratios. Individual participant data (IPD) meta-analysis was used to combine the African studies, and the results were then combined with those from Thailand using aggregated data meta-analysis with a random effects model. There was no difference in the risk of miscarriage associated with the use of artemisinins anytime during the first trimester (n = 37/671) compared with quinine (n = 96/945; adjusted hazard ratio [aHR] = 0.73 [95% CI 0.44, 1.21], I2 = 0%, p = 0.228), in the risk of stillbirth (artemisinins, n = 10/654; quinine, n = 11/615; aHR = 0.29 [95% CI 0.08-1.02], p = 0.053), or in the risk of miscarriage and stillbirth combined (pregnancy loss) (aHR = 0.58 [95% CI 0.36-1.02], p = 0.099). The corresponding risks of miscarriage, stillbirth, and pregnancy loss in a sensitivity analysis restricted to artemisinin exposures during the embryo sensitive period (6-12 wk gestation) were as follows: aHR = 1.04 (95% CI 0.54-2.01), I2 = 0%, p = 0.910; aHR = 0.73 (95% CI 0.26-2.06), p = 0.551; and aHR = 0.98 (95% CI 0.52-2.04), p = 0.603. The prevalence of major congenital anomalies was similar for first-trimester artemisinin (1.5% [95% CI 0.6%-3.5%]) and quinine exposures (1.2% [95% CI 0.6%-2.4%]). Key limitations of the study include the inability to control for confounding by indication in the African studies, the paucity of data on potential confounders, the limited statistical power to detect differences in congenital anomalies, and the lack of assessment of cardiovascular defects in newborns. CONCLUSIONS Compared to quinine, artemisinin treatment in the first trimester was not associated with an increased risk of miscarriage or stillbirth. While the data are limited, they indicate no difference in the prevalence of major congenital anomalies between treatment groups. The benefits of 3-d artemisinin combination therapy regimens to treat malaria in early pregnancy are likely to outweigh the adverse outcomes of partially treated malaria, which can occur with oral quinine because of the known poor adherence to 7-d regimens. REVIEW REGISTRATION PROSPERO CRD42015032371.
Collapse
Affiliation(s)
- Stephanie Dellicour
- Malaria Epidemiology Unit, Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- * E-mail: (SD); (AS)
| | - Esperança Sevene
- Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
- Centro de Investigação em Saúde da Manhiça, Manhiça, Mozambique
| | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | | | | | - Stephen Rulisa
- University Teaching Hospital of Kigali, University of Rwanda, Kigali, Rwanda
| | - Meghna Desai
- Malaria Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Peter Ouma
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Martina Oneko
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Anifa Vala
- Centro de Investigação em Saúde da Manhiça, Manhiça, Mozambique
| | - Maria Rupérez
- Centro de Investigação em Saúde da Manhiça, Manhiça, Mozambique
- Instituto de Salud Global de Barcelona, Barcelona, Spain
| | - Eusébio Macete
- Centro de Investigação em Saúde da Manhiça, Manhiça, Mozambique
| | - Clara Menéndez
- Centro de Investigação em Saúde da Manhiça, Manhiça, Mozambique
- Instituto de Salud Global de Barcelona, Barcelona, Spain
| | - Seydou Nakanabo-Diallo
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Adama Kazienga
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Innocent Valéa
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Gregory Calip
- Department of Pharmacy Systems, Outcomes and Policy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Orvalho Augusto
- Centro de Investigação em Saúde da Manhiça, Manhiça, Mozambique
| | - Blaise Genton
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- Infectious Diseases Service, Lausanne University Hospital, Lausanne, Switzerland
| | - Eric M. Njunju
- School of Medicine, Copperbelt University, Ndola, Zambia
| | - Kerryn A. Moore
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - Umberto d’Alessandro
- Medical Research Council, Fajara, The Gambia
- Institute of Tropical Medicine, Antwerp, Belgium
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Francois Nosten
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Feiko ter Kuile
- Malaria Epidemiology Unit, Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Andy Stergachis
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, United States of America
- Department of Global Health, School of Public Health, University of Washington, Seattle, Washington, United States of America
- * E-mail: (SD); (AS)
| |
Collapse
|
16
|
Gutman J, Kovacs S, Dorsey G, Stergachis A, Ter Kuile FO. Safety, tolerability, and efficacy of repeated doses of dihydroartemisinin-piperaquine for prevention and treatment of malaria: a systematic review and meta-analysis. THE LANCET. INFECTIOUS DISEASES 2017; 17:184-193. [PMID: 27865890 PMCID: PMC5266794 DOI: 10.1016/s1473-3099(16)30378-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/14/2016] [Accepted: 09/16/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Intermittent preventive treatment (IPT) for malaria is used in infants, children, adults, and pregnant women. Dihydroartemisinin-piperaquine (DP) is an effective, well tolerated artemisinin-based combination therapy. The long half-life of piperaquine makes it attractive for IPT. We conducted a systematic review and meta-analysis to establish the efficacy and safety of repeated treatment with DP. METHODS Following PRISMA guidelines, we searched multiple databases on Sept 1, 2016, with the terms: "human" AND "dihydroartemisinin-piperaquine" OR "DHA-PPQ". Studies were eligible if they were randomised controlled trials (RCTs) or prospective cohort studies involving repeat exposures to standard 3-day courses of DP for either seasonal malaria chemoprevention, mass drug administration, or treatment of clinical malaria, conducted at any time and in any geographic location. Random-effects meta-analysis was used to generate pooled incidence rate ratios and relative risks, or risk differences. FINDINGS 11 studies were included: two repeat treatment studies (one in children younger than 5 years and one in pregnant women), and nine IPT trials (five in children younger than 5 years, one in schoolchildren, one in adults, two in pregnant women). Comparator interventions included placebo, artemether-lumefantrine, sulfadoxine-pyrimethamine (SP), SP+amodiaquine, SP+piperaquine, SP+chloroquine, and co-trimoxazole. Of 14 628 participants, 3935 received multiple DP courses (2-18). Monthly IPT-DP was associated with an 84% reduction in the incidence of malaria parasitaemia measured by microscopy compared with placebo. Monthly IPT-DP was associated with fewer serious adverse events than placebo, daily co-trimoxazole, or monthly SP. Among 56 IPT-DP recipients (26 children, 30 pregnant women) with cardiac parameters, all QTc intervals were within normal limits, with no significant increase in QTc prolongation with increasing courses of DP. INTERPRETATION Monthly DP appears well tolerated and effective for IPT. Additional data are needed in pregnancy and to further explore the cardiac safety with monthly dosing. FUNDING Bill & Melinda Gates Foundation and NIH.
Collapse
Affiliation(s)
- Julie Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Stephanie Kovacs
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Grant Dorsey
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA, USA
| | - Andy Stergachis
- Department of Global Health, University of Washington, Seattle, WA, USA; Department of Pharmacy, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
17
|
Kovacs SD, van Eijk AM, Sevene E, Dellicour S, Weiss NS, Emerson S, Steketee R, ter Kuile FO, Stergachis A. The Safety of Artemisinin Derivatives for the Treatment of Malaria in the 2nd or 3rd Trimester of Pregnancy: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0164963. [PMID: 27824884 PMCID: PMC5100961 DOI: 10.1371/journal.pone.0164963] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 10/04/2016] [Indexed: 11/26/2022] Open
Abstract
Given the high morbidity for mother and fetus associated with malaria in pregnancy, safe and efficacious drugs are needed for treatment. Artemisinin derivatives are the most effective antimalarials, but are associated with teratogenic and embryotoxic effects in animal models when used in early pregnancy. However, several organ systems are still under development later in pregnancy. We conducted a systematic review and meta-analysis of the occurrence of adverse pregnancy outcomes among women treated with artemisinins monotherapy or as artemisinin-based combination therapy during the 2nd or 3rd trimesters relative to pregnant women who received non-artemisinin antimalarials or none at all. Pooled odds ratio (POR) were calculated using Mantel-Haenszel fixed effects model with a 0.5 continuity correction for zero events. Eligible studies were identified through Medline, Embase, and the Malaria in Pregnancy Consortium Library. Twenty studies (11 cohort studies and 9 randomized controlled trials) contributed to the analysis, with 3,707 women receiving an artemisinin, 1,951 a non-artemisinin antimalarial, and 13,714 no antimalarial. The PORs (95% confidence interval (CI)) for stillbirth, fetal loss, and congenital anomalies when comparing artemisinin versus quinine were 0.49 (95% CI 0.24-0.97, I2 = 0%, 3 studies); 0.58 (95% CI 0.31-1.16, I2 = 0%, 6 studies); and 1.00 (95% CI 0.27-3.75, I2 = 0%, 3 studies), respectively. The PORs comparing artemisinin users to pregnant women who received no antimalarial were 1.13 (95% CI 0.77-1.66, I2 = 86.7%, 3 studies); 1.10 (95% CI 0.79-1.54, I2 = 0%, 4 studies); and 0.79 (95% CI 0.37-1.67, I2 = 0%, 3 studies) for miscarriage, stillbirth and congenital anomalies respectively. Treatment with artemisinin in 2nd and 3rd trimester was not associated with increased risks of congenital malformations or miscarriage and may be was associated with a reduced risk of stillbirths compared to quinine. This study updates the reviews conducted by the WHO in 2002 and 2006 and supports the current WHO malaria treatment guidelines malaria in pregnancy.
Collapse
Affiliation(s)
- Stephanie D. Kovacs
- Department of Epidemiology, University of Washington, Seattle, WA, United States of America
| | | | - Esperanca Sevene
- Manhiça Health Research Centre, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| | | | - Noel S. Weiss
- Department of Epidemiology, University of Washington, Seattle, WA, United States of America
| | - Scott Emerson
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | | | - Feiko O. ter Kuile
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Kenya Medical Research Institute (KEMRI) Centre for Global Health, Kisumu, Kenya
| | - Andy Stergachis
- Department of Global Health, University of Washington, Seattle, WA, United States of America
- Department of Pharmacy, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
18
|
Surjadjaja C, Surya A, Baird JK. Epidemiology of Plasmodium vivax in Indonesia. Am J Trop Med Hyg 2016; 95:121-132. [PMID: 27708185 PMCID: PMC5201218 DOI: 10.4269/ajtmh.16-0093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 03/07/2016] [Indexed: 11/07/2022] Open
Abstract
Endemic malaria occurs across much of the vast Indonesian archipelago. All five species of Plasmodium known to naturally infect humans occur here, along with 20 species of Anopheles mosquitoes confirmed as carriers of malaria. Two species of plasmodia cause the overwhelming majority and virtually equal shares of malaria infections in Indonesia: Plasmodium falciparum and Plasmodium vivax. The challenge posed by P. vivax is especially steep in Indonesia because chloroquine-resistant strains predominate, along with Chesson-like strains that relapse quickly and multiple times at short intervals in almost all patients. Indonesia's hugely diverse human population carries many variants of glucose-6-phosphate dehydrogenase (G6PD) deficiency, most of them exhibiting severely impaired enzyme activity. Therefore, the patients most likely to benefit from primaquine therapy by preventing aggressive relapse, may also be most likely to suffer harm without G6PD deficiency screening. Indonesia faces the challenge of controlling and eventually eliminating malaria across > 13,500 islands stretching > 5,000 km and an enormous diversity of ecological, ethnographic, and socioeconomic settings, and extensive human migrations. This article describes the occurrence of P. vivax in Indonesia and the obstacles faced in eliminating its transmission.
Collapse
Affiliation(s)
| | - Asik Surya
- Sub-Directorate for Malaria Control, Ministry of Health, Jakarta, Indonesia
| | - J Kevin Baird
- Eijkman-Oxford Clinical Research Unit, Jakarta, Indonesia.,Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Abstract
Introduction: Relapses are important contributors to illness and morbidity in Plasmodium vivax and P. ovale infections. Relapse prevention (radical cure) with primaquine is required for optimal management, control and ultimately elimination of Plasmodium vivax malaria. A review was conducted with publications in English, French, Portuguese and Spanish using the search terms ‘P. vivax’ and ‘relapse’. Areas covered: Hypnozoites causing relapses may be activated weeks or months after initial infection. Incidence and temporal patterns of relapse varies geographically. Relapses derive from parasites either genetically similar or different from the primary infection indicating that some derive from previous infections. Malaria illness itself may activate relapse. Primaquine is the only widely available treatment for radical cure. However, it is often not given because of uncertainty over the risks of primaquine induced haemolysis when G6PD deficiency testing is unavailable. Recommended dosing of primaquine for radical cure in East Asia and Oceania is 0.5 mg base/kg/day and elsewhere is 0.25 mg base/kg/day. Alternative treatments are under investigation. Expert commentary: Geographic heterogeneity in relapse patterns and chloroquine susceptibility of P. vivax, and G6PD deficiency epidemiology mean that radical treatment should be given much more than it is today. G6PD testing should be made widely available so primaquine can be given more safely.
Collapse
Affiliation(s)
- Cindy S Chu
- a Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine , Mahidol University , Mae Sot , Thailand.,b Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine , Mahidol University , Bangkok , Thailand
| | - Nicholas J White
- b Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine , Mahidol University , Bangkok , Thailand.,c Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine , University of Oxford , Oxford , UK
| |
Collapse
|
20
|
Briand V, Le Hesran JY, Mayxay M, Newton PN, Bertin G, Houzé S, Keomany S, Inthavong Y, Vannavong N, Chindavongsa K, Hongvanthong B, Fievet N. Prevalence of malaria in pregnancy in southern Laos: a cross-sectional survey. Malar J 2016; 15:436. [PMID: 27566274 PMCID: PMC5002160 DOI: 10.1186/s12936-016-1492-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/16/2016] [Indexed: 11/28/2022] Open
Abstract
Background There are no data on the burden of malaria in pregnancy (MiP) in Laos, where malaria still remains prevalent in the south. Methods Two cross-sectional surveys were conducted in 2014 to assess the prevalence of MiP in Vapi District, Salavan Province, southern Laos: the first consisted of screening 204 pregnant women during pregnancies [mean (95 % CI) gestational age: 23 (22–25) weeks] living in 30 randomly selected villages in Vapi District; the second was conducted among 331 pregnant women, who delivered during the study period in Vapi and Toumlane District Hospitals and in Salavan Provincial Hospital. Peripheral and placental malaria was detected using rapid diagnostic tests (RDT), thick blood smears (TBS) and real-time quantitative polymerase chain reactions (RT-qPCR). Factors associated with low birth weight (LBW) and maternal anaemia were assessed. Results In the villages, 12/204 women (5.9 %; 95 % CI 3.1–10.0) were infected with malaria as determined by RT-qPCR: 11 were Plasmodium vivax infections and 1 was mixed Plasmodium vivax/Plasmodium falciparum infection, among which 9 were sub-microscopic (as not detected by TBS). History of malaria during current pregnancy tended to be associated with a higher risk of MiP (aIRR 3.05; 95 % CI 0.94–9.88). At delivery, two Plasmodium falciparum sub-microscopic infections (one peripheral and one placental) were detected (4.5 %; 0.6–15.5) in Vapi District. In both surveys, all infected women stated they had slept under a bed net the night before the survey, and 86 % went to the forest for food-finding 1 week before the survey in median. The majority of infections (94 %) were asymptomatic and half of them were associated with anaemia. Overall, 24 % of women had LBW newborns. Factors associated with a higher risk of LBW were tobacco use (aIRR 2.43; 95 % CI 1.64–3.60) and pre-term delivery (aIRR 3.17; 95 % CI 2.19–4.57). Factors associated with a higher risk of maternal anaemia were no iron supplementation during pregnancy, Lao Theung ethnicity and place of living. Conclusions The prevalence of MiP in this population was noticeable. Most infections were asymptomatic and sub-microscopic vivax malaria, which raises the question of reliability of recommended national strategies for the screening and prevention of MiP in Laos. Electronic supplementary material The online version of this article (doi:10.1186/s12936-016-1492-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Valérie Briand
- Faculté de Pharmacie, Institut de Recherche pour le Développement (IRD), Mère et enfant face aux infections tropicales (UMR216), 4 avenue de l'Observatoire, 75006, Paris, France. .,COMUE Sorbonne Paris Cité, Faculté de Pharmacie, Université Paris Descartes, Paris, France.
| | - Jean-Yves Le Hesran
- Faculté de Pharmacie, Institut de Recherche pour le Développement (IRD), Mère et enfant face aux infections tropicales (UMR216), 4 avenue de l'Observatoire, 75006, Paris, France.,COMUE Sorbonne Paris Cité, Faculté de Pharmacie, Université Paris Descartes, Paris, France
| | - Mayfong Mayxay
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Microbiology Laboratory, Vientiane, Lao People's Democratic Republic.,Faculty of Postgraduate Studies, University of Health Sciences, Vientiane, Lao People's Democratic Republic.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Paul N Newton
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Microbiology Laboratory, Vientiane, Lao People's Democratic Republic.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Gwladys Bertin
- Faculté de Pharmacie, Institut de Recherche pour le Développement (IRD), Mère et enfant face aux infections tropicales (UMR216), 4 avenue de l'Observatoire, 75006, Paris, France.,COMUE Sorbonne Paris Cité, Faculté de Pharmacie, Université Paris Descartes, Paris, France
| | - Sandrine Houzé
- Faculté de Pharmacie, Institut de Recherche pour le Développement (IRD), Mère et enfant face aux infections tropicales (UMR216), 4 avenue de l'Observatoire, 75006, Paris, France.,COMUE Sorbonne Paris Cité, Faculté de Pharmacie, Université Paris Descartes, Paris, France.,Laboratoire de Parasitologie, CNR du Paludisme, AP-HP, Hôpital Bichat, Paris, France
| | - Sommay Keomany
- Salavan Provincial Hospital, Salavan, Salavan Province, Lao People's Democratic Republic
| | - Yom Inthavong
- Vapi District Hospital, Vapi, Salavan Province, Lao People's Democratic Republic
| | - Nanthasane Vannavong
- Champasack Provincial Health Office, Champasack, Champasack Province, Lao People's Democratic Republic
| | | | - Bouasy Hongvanthong
- Center of Malariology, Parasitology and Entomology, Vientiane, Lao People's Democratic Republic
| | - Nadine Fievet
- Faculté de Pharmacie, Institut de Recherche pour le Développement (IRD), Mère et enfant face aux infections tropicales (UMR216), 4 avenue de l'Observatoire, 75006, Paris, France.,COMUE Sorbonne Paris Cité, Faculté de Pharmacie, Université Paris Descartes, Paris, France
| |
Collapse
|
21
|
Gomes C, Boareto AC, Dalsenter PR. Clinical and non-clinical safety of artemisinin derivatives in pregnancy. Reprod Toxicol 2016; 65:194-203. [PMID: 27506918 DOI: 10.1016/j.reprotox.2016.08.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/03/2016] [Accepted: 08/05/2016] [Indexed: 10/21/2022]
Abstract
Malaria in pregnancy is a clinically wasting infectious disease, where drug therapy has to be promptly initiated. Currently, the treatment of this infection depends on the use of artemisinin derivatives. The World Health Organization does not recommend the use of these drugs in the first trimester of pregnancy due to non-clinical findings that have shown embryolethality and teratogenic effects. Nevertheless, until now, this toxicity has not been proved in humans. Artemisinin derivatives mechanisms of embryotoxicity are related to depletion of circulating embryonic primitive erythroblasts. Species differences in this sensitive period for toxicity and the presence of malaria infection, which could reduce drug distribution to the fetus, are significant to the risk assessment of artemisinin derivatives treatment to pregnant women. In this review we aimed to assess the results of non-clinical and clinical studies with artemisinin derivatives, their mechanisms of embryotoxicity and discuss the safety of their use during pregnancy.
Collapse
Affiliation(s)
- Caroline Gomes
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil.
| | - Ana Cláudia Boareto
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil.
| | | |
Collapse
|
22
|
Moore BR, Salman S, Davis TME. Treatment regimens for pregnant women with falciparum malaria. Expert Rev Anti Infect Ther 2016; 14:691-704. [PMID: 27322015 DOI: 10.1080/14787210.2016.1202758] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION With increasing parasite drug resistance, the WHO has updated treatment recommendations for falciparum malaria including in pregnancy. This review assesses the evidence for choice of treatment for pregnant women. AREAS COVERED Relevant studies, primarily those published since 2010, were identified from reference databases and were used to identify secondary data sources. Expert commentary: WHO recommends use of intravenous artesunate for severe malaria, quinine-clindamycin for uncomplicated malaria in first trimester, and artemisinin combination therapy for uncomplicated malaria in second/third trimesters. Because fear of adverse outcomes has often excluded pregnant women from conventional drug development, available data for novel therapies are usually based on preclinical studies and cases of inadvertent exposure. Changes in antimalarial drug disposition in pregnancy have been observed but are yet to be translated into specific treatment recommendations. Such targeted regimens may become important as parasite resistance demands that drug exposure is optimized.
Collapse
Affiliation(s)
- Brioni R Moore
- a Fiona Stanley Hospital Unit, School of Medicine and Pharmacology , University of Western Australia , Perth , Australia.,b School of Pharmacy , Curtin University , Perth , Australia
| | - Sam Salman
- c Linear Clinical Research Limited, QEII Medical Centre , Nedlands , Australia.,d Fremantle Hospital Unit, School of Medicine and Pharmacology , University of Western Australia , Fremantle , Australia
| | - Timothy M E Davis
- d Fremantle Hospital Unit, School of Medicine and Pharmacology , University of Western Australia , Fremantle , Australia
| |
Collapse
|
23
|
Moore BR, Benjamin JM, Auyeung SO, Salman S, Yadi G, Griffin S, Page-Sharp M, Batty KT, Siba PM, Mueller I, Rogerson SJ, Davis TM. Safety, tolerability and pharmacokinetic properties of coadministered azithromycin and piperaquine in pregnant Papua New Guinean women. Br J Clin Pharmacol 2016; 82:199-212. [PMID: 26889763 PMCID: PMC4917786 DOI: 10.1111/bcp.12910] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/09/2016] [Accepted: 02/14/2016] [Indexed: 12/14/2022] Open
Abstract
AIMS The aim of the present study was to investigate the safety, tolerability and pharmacokinetics of coadministered azithromycin (AZI) and piperaquine (PQ) for treating malaria in pregnant Papua New Guinean women. METHODS Thirty pregnant women (median age 22 years; 16-32 weeks' gestation) were given three daily doses of 1 g AZI plus 960 mg PQ tetraphosphate with detailed monitoring/blood sampling over 42 days. Plasma AZI and PQ were assayed using liquid chromatography-mass spectrometry and high-performance liquid chromatography, respectively. Pharmacokinetic analysis was by population-based compartmental models. RESULTS The treatment was well tolerated. The median (interquartile range) increase in the rate-corrected electrocardiographic QT interval 4 h postdose [12 (6-26) ms(0) (.5) ] was similar to that found in previous studies of AZI given in pregnancy with other partner drugs. Six women with asymptomatic malaria cleared their parasitaemias within 72 h. Two apararasitaemic women developed late uncomplicated Plasmodium falciparum infections on Days 42 and 83. Compared with previous pregnancy studies, the area under the concentration-time curve (AUC0-∞ ) for PQ [38818 (24354-52299) μg h l(-1) ] was similar to published values but there was a 52% increase in relative bioavailability with each dose. The AUC0-∞ for AZI [46799 (43526-49462) μg h l(-1) ] was at least as high as reported for higher-dose regimens, suggesting saturable absorption and/or concentration-dependent tissue uptake and clearance from the central compartment. CONCLUSIONS AZI-PQ appears to be well tolerated and safe in pregnancy. Based on the present/other data, total AZI doses higher than 3 g for the treatment and prevention of malaria may be unnecessary in pregnant women, while clearance of parasitaemia could improve the relative bioavailability of PQ.
Collapse
Affiliation(s)
- Brioni R Moore
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia, Australia
- Papua New Guinea Institute of Medical Research, Madang, Madang Province, Papua New Guinea
| | - John M Benjamin
- Papua New Guinea Institute of Medical Research, Madang, Madang Province, Papua New Guinea
| | - Siu On Auyeung
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Sam Salman
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Gumul Yadi
- Papua New Guinea Institute of Medical Research, Madang, Madang Province, Papua New Guinea
| | - Suzanne Griffin
- Papua New Guinea Institute of Medical Research, Madang, Madang Province, Papua New Guinea
| | - Madhu Page-Sharp
- School of Pharmacy, Curtin University of Technology, Bentley, Western Australia, Australia
| | - Kevin T Batty
- School of Pharmacy, Curtin University of Technology, Bentley, Western Australia, Australia
| | - Peter M Siba
- Papua New Guinea Institute of Medical Research, Madang, Madang Province, Papua New Guinea
| | - Ivo Mueller
- Infection and Immunity, Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
- Barcelona Institute for Global Health (ISGLOBAL), Barcelona, Spain
| | - Stephen J Rogerson
- Department of Medicine (RMH), The University of Melbourne, Parkville, Victoria, Australia
| | - Timothy Me Davis
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
24
|
Pasay CJ, Rockett R, Sekuloski S, Griffin P, Marquart L, Peatey C, Wang CYT, O'Rourke P, Elliott S, Baker M, Möhrle JJ, McCarthy JS. Piperaquine Monotherapy of Drug-Susceptible Plasmodium falciparum Infection Results in Rapid Clearance of Parasitemia but Is Followed by the Appearance of Gametocytemia. J Infect Dis 2016; 214:105-13. [PMID: 27056954 PMCID: PMC4907420 DOI: 10.1093/infdis/jiw128] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/24/2016] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Piperaquine, coformulated with dihydroartemisinin, is a component of a widely used artemisinin combination therapy. There is a paucity of data on its antimalarial activity as a single agent. Such data, if available, would inform selection of new coformulations. METHODS We undertook a study in healthy subjects, using the induced blood stage malaria (IBSM) model to test the antimalarial activity of single doses of piperaquine (960, 640, and 480 mg) in 3 cohorts. In a pilot study in the third cohort, gametocyte clearance following administration of 15 mg, or 45 mg or no primaquine was investigated. RESULTS Parasite clearance over the 48-hour period after piperaquine administration was more rapid in the 960 mg cohort, compared with the 640 mg cohort (parasite reduction ratio, 2951 [95% confidence interval {CI}, 1520-5728] vs 586 [95% CI, 351-978]; P < .001). All 24 subjects developed gametocytemia as determined by pfs25 transcripts. Clearance of pfs25 was significantly faster in those receiving primaquine than in those not receiving primaquine (P < .001). CONCLUSIONS Piperaquine possesses rapid parasite-clearing activity, but monotherapy is followed by the appearance of gametocytemia, which could facilitate the spread of malaria. This new information should be taken into account when developing future antimalarial coformulations. CLINICAL TRIALS REGISTRATION ACTRN12613000565741.
Collapse
Affiliation(s)
| | - Rebecca Rockett
- School of Chemistry and Molecular Biosciences Queensland Pediatric Infectious Diseases Laboratory
| | | | - Paul Griffin
- QIMR Berghofer Medical Research Institute School of Medicine, University of Queensland Q-Pharm Department of Infectious Diseases, Mater Health Services and Mater Research Institute
| | | | | | | | | | | | | | | | - James S McCarthy
- QIMR Berghofer Medical Research Institute School of Medicine, University of Queensland Q-Pharm
| |
Collapse
|
25
|
Kakuru A, Jagannathan P, Muhindo MK, Natureeba P, Awori P, Nakalembe M, Opira B, Olwoch P, Ategeka J, Nayebare P, Clark TD, Feeney ME, Charlebois ED, Rizzuto G, Muehlenbachs A, Havlir DV, Kamya MR, Dorsey G. Dihydroartemisinin-Piperaquine for the Prevention of Malaria in Pregnancy. N Engl J Med 2016; 374:928-39. [PMID: 26962728 PMCID: PMC4847718 DOI: 10.1056/nejmoa1509150] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Intermittent treatment with sulfadoxine-pyrimethamine is widely recommended for the prevention of malaria in pregnant women in Africa. However, with the spread of resistance to sulfadoxine-pyrimethamine, new interventions are needed. METHODS We conducted a double-blind, randomized, controlled trial involving 300 human immunodeficiency virus (HIV)-uninfected pregnant adolescents or women in Uganda, where sulfadoxine-pyrimethamine resistance is widespread. We randomly assigned participants to a sulfadoxine-pyrimethamine regimen (106 participants), a three-dose dihydroartemisinin-piperaquine regimen (94 participants), or a monthly dihydroartemisinin-piperaquine regimen (100 participants). The primary outcome was the prevalence of histopathologically confirmed placental malaria. RESULTS The prevalence of histopathologically confirmed placental malaria was significantly higher in the sulfadoxine-pyrimethamine group (50.0%) than in the three-dose dihydroartemisinin-piperaquine group (34.1%, P=0.03) or the monthly dihydroartemisinin-piperaquine group (27.1%, P=0.001). The prevalence of a composite adverse birth outcome was lower in the monthly dihydroartemisinin-piperaquine group (9.2%) than in the sulfadoxine-pyrimethamine group (18.6%, P=0.05) or the three-dose dihydroartemisinin-piperaquine group (21.3%, P=0.02). During pregnancy, the incidence of symptomatic malaria was significantly higher in the sulfadoxine-pyrimethamine group (41 episodes over 43.0 person-years at risk) than in the three-dose dihydroartemisinin-piperaquine group (12 episodes over 38.2 person-years at risk, P=0.001) or the monthly dihydroartemisinin-piperaquine group (0 episodes over 42.3 person-years at risk, P<0.001), as was the prevalence of parasitemia (40.5% in the sulfadoxine-pyrimethamine group vs. 16.6% in the three-dose dihydroartemisinin-piperaquine group [P<0.001] and 5.2% in the monthly dihydroartemisinin-piperaquine group [P<0.001]). In each treatment group, the risk of vomiting after administration of any dose of the study agents was less than 0.4%, and there were no significant differences among the groups in the risk of adverse events. CONCLUSIONS The burden of malaria in pregnancy was significantly lower among adolescent girls or women who received intermittent preventive treatment with dihydroartemisinin-piperaquine than among those who received sulfadoxine-pyrimethamine, and monthly treatment with dihydroartemisinin-piperaquine was superior to three-dose dihydroartemisinin-piperaquine with regard to several outcomes. (Funded by the Eunice Kennedy Shriver National Institute of Child Health and Human Development; ClinicalTrials.gov number, NCT02163447.).
Collapse
Affiliation(s)
- Abel Kakuru
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Prasanna Jagannathan
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Mary K Muhindo
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Paul Natureeba
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Patricia Awori
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Miriam Nakalembe
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Bishop Opira
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Peter Olwoch
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - John Ategeka
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Patience Nayebare
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Tamara D Clark
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Margaret E Feeney
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Edwin D Charlebois
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Gabrielle Rizzuto
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Atis Muehlenbachs
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Diane V Havlir
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Moses R Kamya
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| | - Grant Dorsey
- From the Infectious Diseases Research Collaboration (A.K., M.K.M., P. Natureeba, P.A., B.O., P.O., J.A., P. Nayebare), the Department of Obstetrics and Gynecology, Makerere University College of Health Sciences (M.N.), and the School of Medicine, Makerere University College of Health Sciences (M.R.K.) - all in Kampala, Uganda; the Departments of Medicine (P.J., T.D.C., M.E.F., D.V.H., G.D.), Pediatrics (M.E.F.), and Pathology (G.R.) and the Center for AIDS Prevention Studies (E.D.C.), University of California, San Francisco, San Francisco; and the Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta (A.M.)
| |
Collapse
|
26
|
Moore KA, Simpson JA, Paw MK, Pimanpanarak M, Wiladphaingern J, Rijken MJ, Jittamala P, White NJ, Fowkes FJI, Nosten F, McGready R. Safety of artemisinins in first trimester of prospectively followed pregnancies: an observational study. THE LANCET. INFECTIOUS DISEASES 2016; 16:576-583. [PMID: 26869377 PMCID: PMC4835584 DOI: 10.1016/s1473-3099(15)00547-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Artemisinins, the most effective antimalarials available, are not recommended for falciparum malaria during the first trimester of pregnancy because of safety concerns. Therefore, quinine is used despite its poor effectiveness. Assessing artemisinin safety requires weighing the risks of malaria and its treatment. We aimed to assess the effect of first-trimester malaria and artemisinin treatment on miscarriage and major congenital malformations. METHODS In this observational study, we assessed data from antenatal clinics on the Thai-Myanmar border between Jan 1, 1994, and Dec 31, 2013. We included women who presented to antenatal clinics during their first trimester with a viable fetus. Women were screened for malaria, and data on malaria, antimalarial treatment, and birth outcomes were collected. The relationship between artemisinin treatments (artesunate, dihydroartemisinin, or artemether) and miscarriage or malformation was assessed using Cox regression with left-truncation and time-varying exposures. FINDINGS Of 55 636 pregnancies registered between 1994 and 2013, 25 485 pregnancies were analysed for first-trimester malaria and miscarriage, in which 2558 (10%) had first-trimester malaria. The hazard of miscarriage increased 1·61-fold after an initial first-trimester falciparum episode (95% CI 1·32-1·97; p<0·0001), 3·24-fold following falciparum recurrence (2·24-4·68; p<0·0001), and 2·44-fold (1·01-5·88; p=0·0473) following recurrent symptomatic vivax malaria. No difference was noted in miscarriage in first-line falciparum treatments with artemisinin (n=183) versus quinine (n=842; HR 0·78 [95% CI 0·45-1·34]; p=0·3645) or in risk of major congenital malformations (two [2%] of 109 [95% CI 0·22-6·47] versus eight (1%) of 641 [0·54-2·44], respectively). INTERPRETATION First-trimester falciparum and vivax malaria both increase the risk of miscarriage. We noted no evidence of an increased risk of miscarriage or of major congenital malformations associated with first-line treatment with an artemisinin derivative compared with quinine. In view of the low efficacy of quinine and wide availability of highly effective artemisinin-based combination therapies, it is time to reconsider first-trimester antimalarial treatment recommendations. FUNDING The Wellcome Trust and The Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Kerryn A Moore
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia; Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, Australia.
| | - Julie A Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Moo Kho Paw
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - MuPawJay Pimanpanarak
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Jacher Wiladphaingern
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Marcus J Rijken
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Podjanee Jittamala
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - 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, UK
| | - Freya J I Fowkes
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia; Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, Australia; Department of Epidemiology and Preventive Medicine and Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
27
|
Abstract
Severe malaria in pregnancy is a large contributor to maternal morbidity and mortality. Intravenous quinine has traditionally been the treatment drug of choice for severe malaria in pregnancy. However, recent randomized clinical trials (RCTs) indicate that intravenous artesunate is more efficacious for treating severe malaria, resulting in changes to the World Health Organization (WHO) treatment guidelines. Artemisinins, including artesunate, are embryo-lethal in animal studies and there is limited experience with their use in the first trimester. This review summarizes the current literature supporting 2010 WHO treatment guidelines for severe malaria in pregnancy and the efficacy, pharmacokinetics, and adverse event data for currently used antimalarials available for severe malaria in pregnancy. We identified ten studies on the treatment of severe malaria in pregnancy that reported clinical outcomes. In two studies comparing intravenous quinine with intravenous artesunate, intravenous artesunate was more efficacious and safe for use in pregnant women. No studies detected an increased risk of miscarriage, stillbirth, or congenital anomalies associated with first trimester exposure to artesunate. Although the WHO recommends using either quinine or artesunate for the treatment of severe malaria in first trimester pregnancies, our findings suggest that artesunate should be the preferred treatment option for severe malaria in all trimesters.
Collapse
|
28
|
Determination of the antimalarial drug piperaquine in small volume pediatric plasma samples by LC-MS/MS. Bioanalysis 2015; 6:3081-9. [PMID: 25529877 DOI: 10.4155/bio.14.254] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
AIM Determination of piperaquine (PQ) in pediatric plasma requires a method with a small sample volume. RESULTS We report a sensitive LC-MS/MS method for quantitation of PQ with only 25 µl human plasma. Using a deuterated internal standard (PQ-d6), an analytical PFP column, APCI(+) as the ion source and MRM (535/288 for PQ and 541/294 for the IS) for detection, the method has a linear calibration range of 1.5-250 ng/ml with a runtime of 3.0 min per sample. The method was applied to plasma samples from children. CONCLUSION The developed LC-MS/MS method is suitable for pediatric studies with small volume plasma samples collected via capillary tubes. One limitation was the performance of PFP columns varied among different brands.
Collapse
|
29
|
Poespoprodjo JR, Fobia W, Kenangalem E, Lampah DA, Sugiarto P, Tjitra E, Anstey NM, Price RN. Treatment policy change to dihydroartemisinin-piperaquine contributes to the reduction of adverse maternal and pregnancy outcomes. Malar J 2015; 14:272. [PMID: 26169783 PMCID: PMC4501192 DOI: 10.1186/s12936-015-0794-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 07/03/2015] [Indexed: 11/15/2022] Open
Abstract
Background In Papua, Indonesia, maternal malaria is prevalent, multidrug resistant and associated with adverse outcomes for mother and baby. In March 2006, anti-malarial policy was revised for the second and third trimester of pregnancy to dihydroartemisinin–piperaquine (DHP) for all species of malaria. This study presents the temporal analysis of adverse outcomes in pregnancy and early life following this policy change. Methods From April 2004 to May 2010, a standardized questionnaire was used to collect information from all pregnant women admitted to the maternity ward. A physical examination was performed on all live birth newborns. The relative risks (RR) and the associated population attributable risks (PAR) of adverse outcomes in women with a history of malaria treatment to the risk in those without a history of malaria during the current pregnancy were examined to evaluate the temporal trends before and after DHP deployment. Results Of 6,556 women enrolled with known pregnancy outcome, 1,018 (16%) reported prior anti-malarial treatment during their pregnancy. The proportion of women with malaria reporting treatment with DHP rose from 0% in 2004 to 64% (121/189) in 2010. In those with history of malaria during pregnancy, the increasing use of DHP was associated with a 54% fall in the proportion of maternal malaria at delivery and a 98% decrease in congenital malaria (from 7.1% prior to 0.1% after policy change). Overall policy change to more effective treatment was associated with an absolute 2% reduction of maternal severe anaemia and absolute 4.5% decrease in low birth weight babies. Conclusions Introduction of highly effective treatment in pregnancy was associated with a reduction of maternal malaria at delivery and improved neonatal outcomes. Ensuring universal access to arteminisin combination therapy (ACT) in pregnancy in an area of multidrug resistance has potential to impact significantly on maternal and infant health. Electronic supplementary material The online version of this article (doi:10.1186/s12936-015-0794-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jeanne Rini Poespoprodjo
- Mimika District Health Authority, District Government Building, Jl. Cendrawasih, Timika, 99910, Papua, Indonesia. .,Timika Malaria Research Programme, Papuan Health and Community Development Foundation, Jl. SP2-SP5, RSMM Area, Timika, 99910, Papua, Indonesia. .,Department of Child Health, Faculty of Medicine, University Gadjah Mada, Jl. Kesehatan no 1, Sekip, Yogyakarta, 55284, Indonesia.
| | - Wendelina Fobia
- Timika Malaria Research Programme, Papuan Health and Community Development Foundation, Jl. SP2-SP5, RSMM Area, Timika, 99910, Papua, Indonesia.
| | - Enny Kenangalem
- Mimika District Health Authority, District Government Building, Jl. Cendrawasih, Timika, 99910, Papua, Indonesia. .,Timika Malaria Research Programme, Papuan Health and Community Development Foundation, Jl. SP2-SP5, RSMM Area, Timika, 99910, Papua, Indonesia.
| | - Daniel A Lampah
- Timika Malaria Research Programme, Papuan Health and Community Development Foundation, Jl. SP2-SP5, RSMM Area, Timika, 99910, Papua, Indonesia.
| | - Paulus Sugiarto
- Mitra Masyarakat Hospital, Jl. SP2-SP5-Charitas, Timika, 99910, Indonesia.
| | - Emiliana Tjitra
- National Institute of Health Research and Development, Ministry of Health, Jl. Percetakan Negara, Jakarta, 10560, Indonesia.
| | - Nicholas M Anstey
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, PO Box 41096, Casuarina, NT, 0811, Australia. .,Division of Medicine, Royal Darwin Hospital, Darwin, NT, 0810, Australia.
| | - Richard N Price
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, PO Box 41096, Casuarina, NT, 0811, Australia. .,Nuffield Department of Clinical Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, OX37LJ, UK.
| |
Collapse
|
30
|
Population pharmacokinetics, tolerability, and safety of dihydroartemisinin-piperaquine and sulfadoxine-pyrimethamine-piperaquine in pregnant and nonpregnant Papua New Guinean women. Antimicrob Agents Chemother 2015; 59:4260-71. [PMID: 25963981 DOI: 10.1128/aac.00326-15] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 05/02/2015] [Indexed: 01/01/2023] Open
Abstract
The tolerability, safety, and disposition of dihydroartemisinin (DHA) and piperaquine (PQ) were assessed in 32 pregnant (second/third trimester) and 33 nonpregnant Papua New Guinean women randomized to adult treatment courses of DHA-PQ (three daily doses) or sulfadoxine-pyrimethamine (SP)-PQ (three daily PQ doses, single dose of SP). All dose adminstrations were observed, and subjects fasted for 2 h postdose. Plasma PQ was assayed by using high-performance liquid chromatography, and DHA was assessed by using liquid chromatography-mass spectrometry. Compartmental pharmacokinetic models were developed using a population-based approach. Both regimens were well tolerated. There was an expected increase in the rate-corrected electrocardiographic QT interval which was independent of pregnancy and treatment. Two pregnant and two nonpregnant women had Plasmodium falciparum parasitemia which cleared within 48 h, and no other subject became slide positive for malaria during 42 days of follow-up. Of 30 pregnant women followed to delivery, 27 (90%) delivered healthy babies and 3 (10%) had stillbirths; these obstetric outcomes are consistent with those in the general population. The area under the plasma PQ concentration-time curve (AUC0-∞) was lower in the pregnant patients (median [interquartile range], 23,721 μg · h/liter [21,481 to 27,951 μg · h/liter] versus 35,644 μg · h/liter [29,546 to 39,541 μg · h/liter]; P < 0.001) in association with a greater clearance relative to bioavailability (73.5 liters/h [69.4 to 78.4] versus 53.8 liters/h [49.7 to 58.2]; P < 0.001), but pregnancy did not influence the pharmacokinetics of DHA. The apparent pharmacokinetic differences between the present study and results from other studies of women with uncomplicated malaria that showed no effect of pregnancy on the AUC0-∞ of PQ and greater bioavailability may reflect differences in postdose fat intake, proportions of women with malaria, and/or racial differences in drug disposition.
Collapse
|
31
|
Pharmacokinetics of piperaquine transfer into the breast milk of Melanesian mothers. Antimicrob Agents Chemother 2015; 59:4272-8. [PMID: 25963980 DOI: 10.1128/aac.00327-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 05/02/2015] [Indexed: 12/29/2022] Open
Abstract
Transfer of piperaquine (PQ) into breast milk was examined in 27 Papua New Guinean women given a 3-day course of dihydroartemisinin-PQ or sulfadoxine-pyrimethamine-PQ during the second/third trimester. Breast milk was sampled on days 1, 2, 3 to 5, 7 to 11, and 14 to 17 postdelivery, a median of 70 days postdose (range, 6 to 145 days). A blood sample was taken at delivery, and additional serial samples were available from 9 women who delivered within 42 days of dosing. Milk and plasma PQ were assayed by high-performance liquid chromatography. A population-based approach was used to model the loge(plasma) and milk concentration-time data. A sigmoid Emax model best described PQ breast milk transfer. The population average milk:plasma PQ ratio was 0.58, with a peak of 2.5 at delivery. The model-derived maximum milk intake (148 ml/kg of body weight/day) was similar to the accepted value of 150 ml/kg/day. The median estimated absolute and relative cumulative infant PQ doses were 22 μg and 0.07%, respectively, corresponding to absolute and relative daily doses of 0.41 μg/kg and 0.004%. Model-based simulations for PQ treatment regimens given at birth, 1 week postdelivery, and 6 weeks postdelivery showed that the highest median estimated relative total infant dose (0.36%; median absolute total dose of 101 μg/kg) was seen after maternal PQ treatment 6 weeks postpartum. The maximum simulated relative total and daily doses from any scenario were 4.3% and 2.5%, respectively, which were lower than the recommended 10% upper limit. Piperaquine is transferred into breast milk after maternal treatment doses, but PQ exposure for suckling infants appears safe.
Collapse
|
32
|
Safe and efficacious artemisinin-based combination treatments for African pregnant women with malaria: a multicentre randomized control trial. Reprod Health 2015; 12:5. [PMID: 25592254 PMCID: PMC4326323 DOI: 10.1186/1742-4755-12-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/12/2015] [Indexed: 02/07/2023] Open
Abstract
Background Asymptomatic and symptomatic malaria during pregnancy has consequences for both mother and her offspring. Unfortunately, there is insufficient information on the safety and efficacy of most antimalarials in pregnancy. Indeed, clinical trials assessing antimalarial treatments systematically exclude pregnancy for fear of teratogenicity and embryotoxicity. The little available information originates from South East Asia while in sub-Saharan Africa such information is still limited and needs to be provided. Design A Phase 3, non-inferiority, multicentre, randomized, open-label clinical trial on safety and efficacy of 4 ACT when administered during pregnancy was carried out in 4 African countries: Burkina Faso, Ghana, Malawi and Zambia. This is a four arm trial using a balanced incomplete block design. Pregnant women diagnosed with malaria are randomised to receive either amodiaquine-artesunate (AQ-AS), dihydroartemisinin-piperaquine (DHA-PQ), artemether-lumefantrine (AL), or mefloquine-artesunate (MQAS). They are actively followed up until day 63 post-treatment and then monthly until 4–6 weeks post-delivery. The offspring is visited at the time of the first birthday. The primary endpoint is treatment failure (PCR adjusted) at day 63 and safety profiles. Secondary endpoints included PCR unadjusted treatment failure up to day 63, gametocyte carriage, Hb changes, placenta malaria, mean birth weight and low birth weight. The primary statistical analysis will use the combined data from all 4 centres, with adjustment for any centre effects, using an additive model for the response rates. This will allow the assessment of all 6 possible pair-wise treatment comparisons using all available data. Discussion The strength of this trial is the involvement of several African countries, increasing the generalisability of the results. In addition, it assesses most ACTs currently available, determining their relative ‘-value-’ compared to others. The balanced incomplete block design was chosen because using all 4-arms in each site would have increased complexity in terms of implementation. Excluding HIV-positive pregnant women on antiretroviral drugs may be seen as a limitation because of the possible interactions between antiretroviral and antimalarial treatments. Nevertheless, the results of this trial will provide the evidence base for the formulation of malaria treatment policy for pregnant women in sub-Saharan Africa. Trial registration NCT00852423
Collapse
|
33
|
Padberg S. Anti-infective Agents. DRUGS DURING PREGNANCY AND LACTATION 2015. [PMCID: PMC7150338 DOI: 10.1016/b978-0-12-408078-2.00007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
|
34
|
Wang L, Świtalska M, Wang N, Du ZJ, Fukumoto Y, Diep NK, Kiguchi R, Nokami J, Wietrzyk J, Inokuchi T. Design, synthesis, and biological evaluation of artemisinin-indoloquinoline hybrids as potent antiproliferative agents. Molecules 2014; 19:19021-35. [PMID: 25412047 PMCID: PMC6271626 DOI: 10.3390/molecules191119021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/10/2014] [Accepted: 11/12/2014] [Indexed: 11/16/2022] Open
Abstract
A series of artemisinin-indoloquinoline hybrids were designed and synthesized in an attempt to develop potent and selective anti-tumor agents. Compounds 7a–7f, 8 and 9 were prepared and characterized. Their antiproliferative activities against MV4-11, HCT-116, A549, and BALB/3T3 cell lines in vitro were tested. Nearly all of the tested compounds (7–9, except for compounds 7d and 7e against HCT-116) showed an increased antitumor activity against HCT-116 and A549 cell lines when compared to the dihydroartemisinin control. Especially for the artemisinin-indoloquinoline hybrid 8, with an 11-aminopropylamino-10H-indolo[3,2-b]quinoline substituent, the antiproliferative activity against the A549 cell line had improved more than ten times. The IC50 value of hybrid 8 against A549 cell lines was decreased to 1.328 ± 0.586 μM, while dihydroartemisin showed IC50 value of >20 µM in the same cell line. Thus, these results have proven that the strategy of introducing a planar basic fused aromatic moiety, such as the indoloquinoline skeleton, could improve the antiproliferative activity and selectivity towards cancer cell lines.
Collapse
Affiliation(s)
- Li Wang
- Division of Chemistry and Biotechnology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| | - Marta Świtalska
- Institute of Immunology and Experimental Therapy, Polish Academy of Science, 12, R. Weigl Street, Wroclaw 53-114, Poland.
| | - Ning Wang
- Division of Chemistry and Biotechnology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| | - Zhen-Jun Du
- Department of Applied Chemistry, Faculty of Engineering, Okayama University of Science, Ridai-cho, Kita-ku, Okayama, Japan.
| | - Yuta Fukumoto
- Division of Chemistry and Biotechnology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| | - Nguyen Kim Diep
- Division of Chemistry and Biotechnology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| | - Ryo Kiguchi
- Division of Chemistry and Biotechnology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| | - Junzo Nokami
- Department of Applied Chemistry, Faculty of Engineering, Okayama University of Science, Ridai-cho, Kita-ku, Okayama, Japan.
| | - Joanna Wietrzyk
- Institute of Immunology and Experimental Therapy, Polish Academy of Science, 12, R. Weigl Street, Wroclaw 53-114, Poland.
| | - Tsutomu Inokuchi
- Division of Chemistry and Biotechnology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| |
Collapse
|