1
|
Mao EY, Page SW, Sleebs BE, Gancheva MR, Wilson DW. A review of natural products as a source of next-generation drugs against apicomplexan parasites. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:51. [PMID: 40481189 PMCID: PMC12144137 DOI: 10.1038/s44259-025-00119-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 05/12/2025] [Indexed: 06/11/2025]
Abstract
Despite the substantial global health and economic burden of apicomplexan parasites in humans and livestock, treatment options remain limited. Natural products have long played an important role in combating these diseases, offering diverse chemical structures and bioactive compounds. This review summarises past and present natural-product-based therapies for six economically significant apicomplexans and explores the potential of revisiting natural products as a source of next-generation treatments.
Collapse
Affiliation(s)
- Emma Y Mao
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia.
- ARC Training Centre for Environmental and Agricultural Solutions to Antimicrobial Resistance (CEAStAR), St Lucia, Queensland, Australia.
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, 5005, SA, Australia.
| | | | - Brad E Sleebs
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Maria R Gancheva
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia
- ARC Training Centre for Environmental and Agricultural Solutions to Antimicrobial Resistance (CEAStAR), St Lucia, Queensland, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, 5005, SA, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia.
- ARC Training Centre for Environmental and Agricultural Solutions to Antimicrobial Resistance (CEAStAR), St Lucia, Queensland, Australia.
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, 5005, SA, Australia.
- Burnet Institute, Melbourne, 3004, Victoria, Australia.
| |
Collapse
|
2
|
Amanzougaghene N, Tajeri S, Franetich JF, Ashraf K, Soulard V, Bigeard P, Guindo CO, Bouillier C, Lemaitre J, Relouzat F, Legrand R, Kocken CHM, Zeeman AM, Roobsoong W, Sattabongkot J, Yang Z, Snounou G, Mazier D. Azithromycin disrupts apicoplast biogenesis in replicating and dormant liver stages of the relapsing malaria parasites Plasmodium vivax and Plasmodium cynomolgi. Int J Antimicrob Agents 2024; 63:107112. [PMID: 38367843 DOI: 10.1016/j.ijantimicag.2024.107112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024]
Abstract
The control and elimination of malaria caused by Plasmodium vivax is hampered by the threat of relapsed infection resulting from the activation of dormant hepatic hypnozoites. Currently, only the 8-aminoquinolines, primaquine and tafenoquine, have been approved for the elimination of hypnozoites, although their use is hampered by potential toxicity. Therefore, an alternative radical curative drug that safely eliminates hypnozoites is a pressing need. This study assessed the potential hypnozoiticidal activity of the antibiotic azithromycin, which is thought to exert antimalarial activity by inhibiting prokaryote-like ribosomal translation within the apicoplast, an indispensable organelle. The results show that azithromycin inhibited apicoplast development during liver-stage schizogony in P. vivax and Plasmodium cynomolgi, leading to impaired parasite maturation. More importantly, this study found that azithromycin is likely to impair the hypnozoite's apicoplast, resulting in the loss of this organelle. Subsequently, using a recently developed long-term hepatocyte culture system, this study found that this loss likely induces a delay in the hypnozoite activation rate, and that those parasites that do proceed to schizogony display liver-stage arrest prior to differentiating into hepatic merozoites, thus potentially preventing relapse. Overall, this work provides evidence for the potential use of azithromycin for the radical cure of relapsing malaria, and identifies apicoplast functions as potential drug targets in quiescent hypnozoites.
Collapse
Affiliation(s)
- Nadia Amanzougaghene
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France; Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Fontenay-aux-Roses and Kremlin-Bicêtre, France
| | - Shahin Tajeri
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Jean-François Franetich
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Kutub Ashraf
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Valérie Soulard
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Pierre Bigeard
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Cheick Oumar Guindo
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Camille Bouillier
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Fontenay-aux-Roses and Kremlin-Bicêtre, France
| | - Julien Lemaitre
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Fontenay-aux-Roses and Kremlin-Bicêtre, France
| | - Francis Relouzat
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Fontenay-aux-Roses and Kremlin-Bicêtre, France
| | - Roger Legrand
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Fontenay-aux-Roses and Kremlin-Bicêtre, France
| | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine Mahidol University, Bangkok, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine Mahidol University, Bangkok, Thailand
| | - Zhaoqing Yang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Chenggong New Town, Kunming, Yunnan Province,China
| | - Georges Snounou
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Fontenay-aux-Roses and Kremlin-Bicêtre, France.
| | - Dominique Mazier
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France.
| |
Collapse
|
3
|
Chan PA, Le Brazidec DL, Becasen JS, Martin H, Kapadia J, Reno H, Bachmann L, Barbee LA. Safety of Longer-Term Doxycycline Use: A Systematic Review and Meta-Analysis With Implications for Bacterial Sexually Transmitted Infection Chemoprophylaxis. Sex Transm Dis 2023; 50:701-712. [PMID: 37732844 PMCID: PMC10592014 DOI: 10.1097/olq.0000000000001865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
BACKGROUND Sexually transmitted infections (STIs) such as syphilis, gonorrhea, and chlamydia have significantly increased over the past decade in the United States. Doxycycline as chemoprophylaxis (i.e., postexposure prophylaxis) offers promise for addressing bacterial STIs. The goal of the current study was to evaluate the safety of longer-term doxycycline use (defined as 8 or more weeks) in the context of potential use as STI chemoprophylaxis through a systematic literature review and meta-analysis. METHODS This review used the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines to search MEDLINE/PubMed for clinical studies published from August 2003 to January 2023 that reported on adverse events with doxycycline use with a focus on side effects and metabolic effects of long-term use. RESULTS A total of 67 studies were included in the systematic review. Overall, studies on longer-term doxycycline use reported 0% to greater than 50% adverse events ranging from mild to severe. Most common adverse events included gastrointestinal symptoms (i.e., nausea, vomiting, and abdominal pain), dermatologic (i.e., rash), and neurological (i.e., headache and dizziness) symptoms. Discontinuation of doxycycline due to adverse events was relatively uncommon in most studies. A meta-analysis of placebo controlled clinical trials (N = 18) revealed that gastrointestinal and dermatological adverse events were more likely to occur in the doxycycline group. CONCLUSIONS Longer-term (8+ weeks) doxycycline use is generally safe and may be associated with minor side effects. Further research is needed on the potential metabolic impact of longer-term doxycycline use.
Collapse
Affiliation(s)
- Philip A. Chan
- Department of Medicine, Brown University, Providence, Rhode Island, USA
- Division of STD Prevention, National Center for HIV, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Jeffrey S. Becasen
- Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Harrison Martin
- Department of Medicine, Brown University, Providence, Rhode Island, USA
| | - Jhanavi Kapadia
- Department of Medicine, Brown University, Providence, Rhode Island, USA
| | - Hilary Reno
- Division of STD Prevention, National Center for HIV, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Laura Bachmann
- Division of STD Prevention, National Center for HIV, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Lindley A. Barbee
- Division of STD Prevention, National Center for HIV, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Burns AL, Sleebs BE, Gancheva M, McLean KT, Siddiqui G, Venter H, Beeson JG, O’Handley R, Creek DJ, Ma S, Frölich S, Goodman CD, McFadden GI, Wilson DW. Targeting malaria parasites with novel derivatives of azithromycin. Front Cell Infect Microbiol 2022; 12:1063407. [PMID: 36530422 PMCID: PMC9748569 DOI: 10.3389/fcimb.2022.1063407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/09/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction The spread of artemisinin resistant Plasmodium falciparum parasites is of global concern and highlights the need to identify new antimalarials for future treatments. Azithromycin, a macrolide antibiotic used clinically against malaria, kills parasites via two mechanisms: 'delayed death' by inhibiting the bacterium-like ribosomes of the apicoplast, and 'quick-killing' that kills rapidly across the entire blood stage development. Methods Here, 22 azithromycin analogues were explored for delayed death and quick-killing activities against P. falciparum (the most virulent human malaria) and P. knowlesi (a monkey parasite that frequently infects humans). Results Seventeen analogues showed improved quick-killing against both Plasmodium species, with up to 38 to 20-fold higher potency over azithromycin after less than 48 or 28 hours of treatment for P. falciparum and P. knowlesi, respectively. Quick-killing analogues maintained activity throughout the blood stage lifecycle, including ring stages of P. falciparum parasites (<12 hrs treatment) and were >5-fold more selective against P. falciparum than human cells. Isopentenyl pyrophosphate supplemented parasites that lacked an apicoplast were equally sensitive to quick-killing analogues, confirming that the quick killing activity of these drugs was not directed at the apicoplast. Further, activity against the related apicoplast containing parasite Toxoplasma gondii and the gram-positive bacterium Streptococcus pneumoniae did not show improvement over azithromycin, highlighting the specific improvement in antimalarial quick-killing activity. Metabolomic profiling of parasites subjected to the most potent compound showed a build-up of non-haemoglobin derived peptides that was similar to chloroquine, while also exhibiting accumulation of haemoglobin-derived peptides that was absent for chloroquine treatment. Discussion The azithromycin analogues characterised in this study expand the structural diversity over previously reported quick-killing compounds and provide new starting points to develop azithromycin analogues with quick-killing antimalarial activity.
Collapse
Affiliation(s)
- Amy L. Burns
- Research Centre for Infectious Diseases, School of Biological Sciences, the University of Adelaide, Adelaide, SA, Australia,School of Science and Technology, the University of New England, Armidale, NSW, Australia
| | - Brad E. Sleebs
- ACRF Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Maria Gancheva
- Research Centre for Infectious Diseases, School of Biological Sciences, the University of Adelaide, Adelaide, SA, Australia
| | - Kimberley T. McLean
- Research Centre for Infectious Diseases, School of Biological Sciences, the University of Adelaide, Adelaide, SA, Australia
| | - Ghizal Siddiqui
- Drug Delivery Disposition and Dynamics, Monash University, Parkville, VIC, Australia
| | - Henrietta Venter
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - James G. Beeson
- Healthy Mothers, Healthy Babies Program, Burnet Institute, Melbourne, VIC, Australia,Department of Medicine, University of Melbourne, Parkville, VIC, Australia,Central Clinical School, Monash University, Melbourne, Vic, Australia,Department of Microbiology, Monash University, Melbourne, Vic, Australia
| | - Ryan O’Handley
- School of Animal and Veterinary Science, University of Adelaide, Adelaide, SA, Australia,Australian Centre for Antimicrobial Resistance Ecology, The University of Adelaide, Adelaide, SA, Australia
| | - Darren J. Creek
- Drug Delivery Disposition and Dynamics, Monash University, Parkville, VIC, Australia
| | - Shutao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Sonja Frölich
- Research Centre for Infectious Diseases, School of Biological Sciences, the University of Adelaide, Adelaide, SA, Australia
| | | | | | - Danny W. Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, the University of Adelaide, Adelaide, SA, Australia,Healthy Mothers, Healthy Babies Program, Burnet Institute, Melbourne, VIC, Australia,Australian Centre for Antimicrobial Resistance Ecology, The University of Adelaide, Adelaide, SA, Australia,*Correspondence: Danny W. Wilson,
| |
Collapse
|
5
|
Effect of biannual azithromycin distribution on antibody responses to malaria, bacterial, and protozoan pathogens in Niger. Nat Commun 2022; 13:976. [PMID: 35190534 PMCID: PMC8861117 DOI: 10.1038/s41467-022-28565-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 02/02/2022] [Indexed: 11/30/2022] Open
Abstract
The MORDOR trial in Niger, Malawi, and Tanzania found that biannual mass distribution of azithromycin to children younger than 5 years led to a 13.5% reduction in all-cause mortality (NCT02048007). To help elucidate the mechanism for mortality reduction, we report IgG responses to 11 malaria, bacterial, and protozoan pathogens using a multiplex bead assay in pre-specified substudy of 30 communities in the rural Niger placebo-controlled trial over a three-year period (n = 5642 blood specimens, n = 3814 children ages 1–59 months). Mass azithromycin reduces Campylobacter spp. force of infection by 29% (hazard ratio = 0.71, 95% CI: 0.56, 0.89; P = 0.004) but serological measures show no significant differences between groups for other pathogens against a backdrop of high transmission. Results align with a recent microbiome study in the communities. Given significant sequelae of Campylobacter infection among preschool aged children, our results support an important mechanism through which biannual mass distribution of azithromycin likely reduces mortality in Niger. In a randomized placebo-controlled trial in rural Niger, biannual azithromycin distribution to children 1-59 months reduced all-cause mortality. Based on serology, Arzika et al. here report a reduction of Campylobacter infection, supporting one mechanism for the intervention’s impact on mortality.
Collapse
|
6
|
Phiri MD, Cairns M, Zongo I, Nikiema F, Diarra M, Yerbanga RS, Barry A, Tapily A, Coumare S, Thera I, Kuepfer I, Milligan P, Tinto H, Dicko A, Ouédraogo JB, Greenwood B, Chandramohan D, Sagara I. The Duration of Protection from Azithromycin Against Malaria, Acute Respiratory, Gastrointestinal, and Skin Infections When Given Alongside Seasonal Malaria Chemoprevention: Secondary Analyses of Data from a Clinical Trial in Houndé, Burkina Faso, and Bougouni, Mali. Clin Infect Dis 2021; 73:e2379-e2386. [PMID: 33417683 PMCID: PMC8492219 DOI: 10.1093/cid/ciaa1905] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Mass drug administration (MDA) with azithromycin (AZ) is being considered as a strategy to promote child survival in sub-Saharan Africa, but the mechanism by which AZ reduces mortality is unclear. To better understand the nature and extent of protection provided by AZ, we explored the profile of protection by time since administration, using data from a household-randomized, placebo-controlled trial in Burkina Faso and Mali. METHODS Between 2014 and 2016, 30 977 children aged 3-59 months received seasonal malaria chemoprevention (SMC) with sulfadoxine-pyrimethamine plus amodiaquine and either AZ or placebo monthly, on 4 occasions each year. Poisson regression with gamma-distributed random effects, accounting for the household randomization and within-individual clustering of illness episodes, was used to compare incidence of prespecified outcomes between SMC+AZ versus SMC+placebo groups in fixed time strata post-treatment. The likelihood ratio test was used to assess evidence for a time-treatment group interaction. RESULTS Relative to SMC+placebo, there was no evidence of protection from SMC+AZ against hospital admissions and deaths. Additional protection from SMC+AZ against malaria was confined to the first 2 weeks post-administration (protective efficacy (PE): 24.2% [95% CI: 17.8%, 30.1%]). Gastroenteritis and pneumonia were reduced by 29.9% [21.7; 37.3%], and 34.3% [14.9; 49.3%], respectively, in the first 2 weeks postadministration. Protection against nonmalaria fevers with a skin condition persisted up to 28 days: PE: 46.3% [35.1; 55.6%]. CONCLUSIONS The benefits of AZ-MDA are broad-ranging but short-lived. To maximize impact, timing of AZ-MDA must address the challenge of targeting asynchronous morbidity and mortality peaks from different causes.
Collapse
Affiliation(s)
- Mphatso Dennis Phiri
- Malaria Epidemiology Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Matthew Cairns
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Issaka Zongo
- Le Département Biomédical et de Santé Publique, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Frederic Nikiema
- Le Département Biomédical et de Santé Publique, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Modibo Diarra
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Rakiswendé Serge Yerbanga
- Le Département Biomédical et de Santé Publique, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Amadou Barry
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Amadou Tapily
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Samba Coumare
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Ismaila Thera
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Irene Kuepfer
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Paul Milligan
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Halidou Tinto
- Le Département Biomédical et de Santé Publique, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Alassane Dicko
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Jean Bosco Ouédraogo
- Le Département Biomédical et de Santé Publique, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Brian Greenwood
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Daniel Chandramohan
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Issaka Sagara
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| |
Collapse
|
7
|
Yang H, Wang J, Liu H, Zhao Y, Lakshmi S, Li X, Nie R, Li C, Wang H, Cao Y, Menezes L, Cui L. Efficacy and Safety of a Naphthoquine-Azithromycin Coformulation for Malaria Prophylaxis in Southeast Asia: A Phase 3, Double-blind, Randomized, Placebo-controlled Trial. Clin Infect Dis 2021; 73:e2470-e2476. [PMID: 32687174 DOI: 10.1093/cid/ciaa1018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND A prophylactic antimalarial drug that is both effective for protection and improves compliance is in high demand. METHODS We conducted a randomized, placebo-controlled, double-blinded phase 3 trial to evaluate the 1:1 fixed-dose combination of naphthoquine-azithromycin (NQAZ) for safety and protection against Plasmodium infections in villages along the China-Myanmar border. A total of 631 residents, 5-65 years of age, were randomized into the drug group (n = 319) and the placebo group (n = 312) to receive NZAQ and placebo, respectively, as a single-dose monthly treatment. Follow-ups were conducted weekly to monitor for adverse events and malaria infections. RESULTS Of the 531 subjects completing the trial, there were 46 and 3 blood smear-positive Plasmodium infections in the placebo and treatment groups, respectively. For the intent-to-treat analysis, the single-dose monthly NQAZ treatment had 93.62% protective efficacy (95% confidence interval [CI]: 91.72%-95.52%). For the per-protocol analysis, NQAZ treatment provided a 93.04% protective efficacy (95% CI: 90.98%-95.1%). Three smear-positive cases in the NQAZ group were all due to acute falciparum malaria. In comparison, NQAZ treatment provided 100% protection against the relapsing malaria Plasmodium vivax and Plasmodium ovale. The treatment group had 5.6% of participants experiencing transient elevation of liver aminotransferases compared with 2.2% in the placebo group (P > .05). CONCLUSIONS Monthly prophylaxis with NQAZ tablets was well tolerated and highly effective for preventing Plasmodium infections. It may prove useful for eliminating P. vivax in areas with a high prevalence of glucose-6-phosphate dehydrogenase deficiency in the population. CLINICAL TRIALS REGISTRATION ChiCTR1800020140.
Collapse
Affiliation(s)
- Henglin Yang
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Center of Malaria Research, Pu'er, Yunnan, China
| | - Jingyan Wang
- Institute of Microbiology and Epidemiology, Chinese Academy of Military Medical Sciences, Beijing, China
| | - Hui Liu
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Center of Malaria Research, Pu'er, Yunnan, China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Seetha Lakshmi
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Xingliang Li
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Center of Malaria Research, Pu'er, Yunnan, China
| | - Renhua Nie
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Center of Malaria Research, Pu'er, Yunnan, China
| | - Chunfu Li
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Center of Malaria Research, Pu'er, Yunnan, China
| | - Hengye Wang
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Center of Malaria Research, Pu'er, Yunnan, China
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Lynette Menezes
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
8
|
Shibeshi MA, Kifle ZD, Atnafie SA. Antimalarial Drug Resistance and Novel Targets for Antimalarial Drug Discovery. Infect Drug Resist 2020; 13:4047-4060. [PMID: 33204122 PMCID: PMC7666977 DOI: 10.2147/idr.s279433] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/29/2020] [Indexed: 12/16/2022] Open
Abstract
Malaria is among the most devastating and widespread tropical parasitic diseases in which most prevalent in developing countries. Antimalarial drug resistance is the ability of a parasite strain to survive and/or to multiply despite the administration and absorption of medicine given in doses equal to or higher than those usually recommended. Among the factors which facilitate the emergence of resistance to existing antimalarial drugs: the parasite mutation rate, the overall parasite load, the strength of drug selected, the treatment compliance, poor adherence to malaria treatment guideline, improper dosing, poor pharmacokinetic properties, fake drugs lead to inadequate drug exposure on parasites, and poor-quality antimalarial may aid and abet resistance. Malaria vaccines can be categorized into three categories: pre-erythrocytic, blood-stage, and transmission-blocking vaccines. Molecular markers of antimalarial drug resistance are used to screen for the emergence of resistance and assess its spread. It provides information about the parasite genetics associated with resistance, either single nucleotide polymorphisms or gene copy number variations which are associated with decreased susceptibility of parasites to antimalarial drugs. Glucose transporter PfHT1, kinases (Plasmodium kinome), food vacuole, apicoplast, cysteine proteases, and aminopeptidases are the novel targets for the development of new antimalarial drugs. Therefore, this review summarizes the antimalarial drug resistance and novel targets of antimalarial drugs.
Collapse
Affiliation(s)
- Melkamu Adigo Shibeshi
- Department of Pharmacology, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Zemene Demelash Kifle
- Department of Pharmacology, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Seyfe Asrade Atnafie
- Department of Pharmacology, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
9
|
An open label study of the safety and efficacy of a single dose of weekly chloroquine and azithromycin administered for malaria prophylaxis in healthy adults challenged with 7G8 chloroquine-resistant Plasmodium falciparum in a controlled human malaria infection model. Malar J 2020; 19:336. [PMID: 32938444 PMCID: PMC7493140 DOI: 10.1186/s12936-020-03409-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/04/2020] [Indexed: 11/15/2022] Open
Abstract
Background Malaria remains the top infectious disease threat facing the U.S. military in many forward operating environments. Compliance with malaria chemoprophylaxis remains a critical component in preventing malaria in the deployed Service Member. Studies of previous military operations show that compliance is consistently higher with weekly versus daily dosing regimens. Current FDA approved weekly chemoprophylaxis options have contraindications that can limit prescribing. The combination of chloroquine (CQ) with azithromycin (AZ) has previously been shown to be an efficacious treatment option for malaria, has pharmacokinetics compatible with weekly dosing, and has shown synergy when combined in vitro. Methods In this open label study, 18 healthy volunteers, aged 18–50 years (inclusive), were randomly assigned to receive either 300 mg CQ or 300 mg CQ and 2 gm azithromycin (CQAZ) of directly observed therapy, weekly for 3 weeks prior to undergoing mosquito bite challenge with chloroquine-resistant Plasmodium falciparum. Volunteers that remained asymptomatic and had no evidence of parasitaemia continued to receive weekly post-exposure chemoprophylaxis for 3 weeks following malaria challenge. The primary endpoint was the number of volunteers that remained asymptomatic and had no evidence of parasitaemia 28 days after the malaria challenge. Results All 6 (100%) volunteers randomized to the CQ control group became symptomatic with parasitaemia during the 28-day post-challenge period. Only 1/12 (8.3%) of volunteers in the CQAZ group developed symptoms and parasitaemia during the 28-day post-challenge period. However, after chemoprophylaxis was discontinued an additional 6 volunteers developed parasitaemia between days 28–41 after challenge, with 4 of 6 experiencing symptoms. 80% of subjects in the CQAZ group experienced treatment related gastrointestinal adverse events (including 13% that experienced severe nausea) compared to 38% in the CQ group. A comparison of the pharmacokinetics in the CQAZ group demonstrated higher azithromycin Cmax (p = 0.03) and AUC (p = 0.044) levels in those volunteers who never became parasitaemic compared to those who did. Conclusion Given the high rate of side effects and poor efficacy when administered for 3 weeks before and after challenge, the combination of weekly chloroquine and azithromycin is a suboptimal regimen combination for weekly malaria chemoprophylaxis. Trial registration ClinicalTrials.gov NCT03278808
Collapse
|
10
|
Alsowaida YS, Almulhim AS, Oh M, Erstad B, Abraham I. Sensorineural hearing loss with macrolide antibiotics exposure: a meta-analysis of the association. INTERNATIONAL JOURNAL OF PHARMACY PRACTICE 2020; 29:21-28. [PMID: 32871046 DOI: 10.1111/ijpp.12670] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Macrolide antibiotics are among the most commonly used antibiotics; the association of macrolide antibiotics exposure with sensorineural hearing loss (SNHL) has been hypothesized. A systematic search was conducted in PubMed, EMBASE and Cochrane Library from inception to 15 July 2019 to identify studies used macrolide antibiotics for any indication. The results were reported as odds ratio (OR) with 95% confidence interval (CI) using random-effects model to derive the association of macrolide antibiotics exposure with SNHL. The objective of this meta-analysis was to estimate the association of macrolide antibiotics exposure and SNHL from up-to-date evidence. KEY FINDINGS Nine studies met the inclusion criteria. There was no statistically significant association between macrolide antibiotics exposure and SNHL; the OR was 1.20 (95% CI: 0.96 to 1.49). No significant association was found with any of the subgroup meta-analyses. SUMMARY Whilst the frequency of SNHL was higher with macrolide antibiotics exposure compared with controls, overall, no association was found between macrolide antibiotics and SNHL.
Collapse
Affiliation(s)
- Yazed Saleh Alsowaida
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, University of Arizona, Tucson, AZ, USA.,Department of Pharmacy Services, Brigham and Women's Hospital, Boston, MA, USA.,Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Abdulaziz Saleh Almulhim
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, University of Arizona, Tucson, AZ, USA.,College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Mok Oh
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Brian Erstad
- Department of Pharmacy Practice & Science, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Ivo Abraham
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, University of Arizona, Tucson, AZ, USA.,University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA.,Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, USA.,Department of Family and Community Medicine, College of Medicine - Tucson, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
11
|
Repurposing Drugs to Fight Hepatic Malaria Parasites. Molecules 2020; 25:molecules25153409. [PMID: 32731386 PMCID: PMC7435416 DOI: 10.3390/molecules25153409] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022] Open
Abstract
Malaria remains one of the most prevalent infectious diseases worldwide, primarily affecting some of the most vulnerable populations around the globe. Despite achievements in the treatment of this devastating disease, there is still an urgent need for the discovery of new drugs that tackle infection by Plasmodium parasites. However, de novo drug development is a costly and time-consuming process. An alternative strategy is to evaluate the anti-plasmodial activity of compounds that are already approved for other purposes, an approach known as drug repurposing. Here, we will review efforts to assess the anti-plasmodial activity of existing drugs, with an emphasis on the obligatory and clinically silent liver stage of infection. We will also review the current knowledge on the classes of compounds that might be therapeutically relevant against Plasmodium in the context of other communicable diseases that are prevalent in regions where malaria is endemic. Repositioning existing compounds may constitute a faster solution to the current gap of prophylactic and therapeutic drugs that act on Plasmodium parasites, overall contributing to the global effort of malaria eradication.
Collapse
|
12
|
Biannual mass azithromycin distributions and malaria parasitemia in pre-school children in Niger: A cluster-randomized, placebo-controlled trial. PLoS Med 2019; 16:e1002835. [PMID: 31237871 PMCID: PMC6592520 DOI: 10.1371/journal.pmed.1002835] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 05/21/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Mass azithromycin distributions have been shown to reduce mortality in preschool children, although the factors mediating this mortality reduction are not clear. This study was performed to determine whether mass distribution of azithromycin, which has modest antimalarial activity, reduces the community burden of malaria. METHODS AND FINDINGS In a cluster-randomized trial conducted from 23 November 2014 until 31 July 2017, 30 rural communities in Niger were randomized to 2 years of biannual mass distributions of either azithromycin (20 mg/kg oral suspension) or placebo to children aged 1 to 59 months. Participants, field staff, and investigators were masked to treatment allocation. The primary malaria outcome was the community prevalence of parasitemia on thick blood smear, assessed in a random sample of children from each community at study visits 12 and 24 months after randomization. Analyses were performed in an intention-to-treat fashion. At the baseline visit, a total of 1,695 children were enumerated in the 15 azithromycin communities, and 3,029 children were enumerated in the 15 placebo communities. No communities were lost to follow-up. The mean prevalence of malaria parasitemia at baseline was 8.9% (95% CI 5.1%-15.7%; 52 of 552 children across all communities) in the azithromycin-treated group and 6.7% (95% CI 4.0%-12.6%; 36 of 542 children across all communities) in the placebo-treated group. In the prespecified primary analysis, parasitemia was lower in the azithromycin-treated group at month 12 (mean prevalence 8.8%, 95% CI 5.1%-14.3%; 51 of 551 children across all communities) and month 24 (mean 3.5%, 95% CI 1.9%-5.5%; 21 of 567 children across all communities) than it was in the placebo-treated group at month 12 (mean 15.3%, 95% CI 10.8%-20.6%; 81 of 548 children across all communities) and month 24 (mean 4.8%, 95% CI 3.3%-6.4%; 28 of 592 children across all communities) (P = 0.02). Communities treated with azithromycin had approximately half the odds of parasitemia compared to those treated with placebo (odds ratio [OR] 0.54, 95% CI 0.30 to 0.97). Parasite density was lower in the azithromycin group than the placebo group at 12 and 24 months (square root-transformed outcome; density estimates were 7,540 parasites/μl lower [95% CI -350 to -12,550 parasites/μl; P = 0.02] at a mean parasite density of 17,000, as was observed in the placebo arm). No significant difference in hemoglobin was observed between the 2 treatment groups at 12 and 24 months (mean 0.34 g/dL higher in the azithromycin arm, 95% CI -0.06 to 0.75 g/dL; P = 0.10). No serious adverse events were reported in either group, and among children aged 1 to 5 months, the most commonly reported nonserious adverse events (i.e., diarrhea, vomiting, and rash) were less common in the azithromycin-treated communities. Limitations of the trial include the timing of the treatments and monitoring visits, both of which took place before the peak malaria season, as well as the uncertain generalizability to areas with different malaria transmission dynamics. CONCLUSIONS Mass azithromycin distributions were associated with a reduced prevalence of malaria parasitemia in this trial, suggesting one possible mechanism for the mortality benefit observed with this intervention. TRIAL REGISTRATION The trial was registered on ClinicalTrials.gov (NCT02048007).
Collapse
|
13
|
Hansen MP, Scott AM, McCullough A, Thorning S, Aronson JK, Beller EM, Glasziou PP, Hoffmann TC, Clark J, Del Mar CB, Cochrane Acute Respiratory Infections Group. Adverse events in people taking macrolide antibiotics versus placebo for any indication. Cochrane Database Syst Rev 2019; 1:CD011825. [PMID: 30656650 PMCID: PMC6353052 DOI: 10.1002/14651858.cd011825.pub2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Macrolide antibiotics (macrolides) are among the most commonly prescribed antibiotics worldwide and are used for a wide range of infections. However, macrolides also expose people to the risk of adverse events. The current understanding of adverse events is mostly derived from observational studies, which are subject to bias because it is hard to distinguish events caused by antibiotics from events caused by the diseases being treated. Because adverse events are treatment-specific, rather than disease-specific, it is possible to increase the number of adverse events available for analysis by combining randomised controlled trials (RCTs) of the same treatment across different diseases. OBJECTIVES To quantify the incidences of reported adverse events in people taking macrolide antibiotics compared to placebo for any indication. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), which includes the Cochrane Acute Respiratory Infections Group Specialised Register (2018, Issue 4); MEDLINE (Ovid, from 1946 to 8 May 2018); Embase (from 2010 to 8 May 2018); CINAHL (from 1981 to 8 May 2018); LILACS (from 1982 to 8 May 2018); and Web of Science (from 1955 to 8 May 2018). We searched clinical trial registries for current and completed trials (9 May 2018) and checked the reference lists of included studies and of previous Cochrane Reviews on macrolides. SELECTION CRITERIA We included RCTs that compared a macrolide antibiotic to placebo for any indication. We included trials using any of the four most commonly used macrolide antibiotics: azithromycin, clarithromycin, erythromycin, or roxithromycin. Macrolides could be administered by any route. Concomitant medications were permitted provided they were equally available to both treatment and comparison groups. DATA COLLECTION AND ANALYSIS Two review authors independently extracted and collected data. We assessed the risk of bias of all included studies and the quality of evidence for each outcome of interest. We analysed specific adverse events, deaths, and subsequent carriage of macrolide-resistant bacteria separately. The study participant was the unit of analysis for each adverse event. Any specific adverse events that occurred in 5% or more of any group were reported. We undertook a meta-analysis when three or more included studies reported a specific adverse event. MAIN RESULTS We included 183 studies with a total of 252,886 participants (range 40 to 190,238). The indications for macrolide antibiotics varied greatly, with most studies using macrolides for the treatment or prevention of either acute respiratory tract infections, cardiovascular diseases, chronic respiratory diseases, gastrointestinal conditions, or urogynaecological problems. Most trials were conducted in secondary care settings. Azithromycin and erythromycin were more commonly studied than clarithromycin and roxithromycin.Most studies (89%) reported some adverse events or at least stated that no adverse events were observed.Gastrointestinal adverse events were the most commonly reported type of adverse event. Compared to placebo, macrolides caused more diarrhoea (odds ratio (OR) 1.70, 95% confidence interval (CI) 1.34 to 2.16; low-quality evidence); more abdominal pain (OR 1.66, 95% CI 1.22 to 2.26; low-quality evidence); and more nausea (OR 1.61, 95% CI 1.37 to 1.90; moderate-quality evidence). Vomiting (OR 1.27, 95% CI 1.04 to 1.56; moderate-quality evidence) and gastrointestinal disorders not otherwise specified (NOS) (OR 2.16, 95% CI 1.56 to 3.00; moderate-quality evidence) were also reported more often in participants taking macrolides compared to placebo.The number of additional people (absolute difference in risk) who experienced adverse events from macrolides was: gastrointestinal disorders NOS 85/1000; diarrhoea 72/1000; abdominal pain 62/1000; nausea 47/1000; and vomiting 23/1000.The number needed to treat for an additional harmful outcome (NNTH) ranged from 12 (95% CI 8 to 23) for gastrointestinal disorders NOS to 17 (9 to 47) for abdominal pain; 19 (12 to 33) for diarrhoea; 19 (13 to 30) for nausea; and 45 (22 to 295) for vomiting.There was no clear consistent difference in gastrointestinal adverse events between different types of macrolides or route of administration.Taste disturbances were reported more often by participants taking macrolide antibiotics, although there were wide confidence intervals and moderate heterogeneity (OR 4.95, 95% CI 1.64 to 14.93; I² = 46%; low-quality evidence).Compared with participants taking placebo, those taking macrolides experienced hearing loss more often, however only four studies reported this outcome (OR 1.30, 95% CI 1.00 to 1.70; I² = 0%; low-quality evidence).We did not find any evidence that macrolides caused more cardiac disorders (OR 0.87, 95% CI 0.54 to 1.40; very low-quality evidence); hepatobiliary disorders (OR 1.04, 95% CI 0.27 to 4.09; very low-quality evidence); or changes in liver enzymes (OR 1.56, 95% CI 0.73 to 3.37; very low-quality evidence) compared to placebo.We did not find any evidence that appetite loss, dizziness, headache, respiratory symptoms, blood infections, skin and soft tissue infections, itching, or rashes were reported more often by participants treated with macrolides compared to placebo.Macrolides caused less cough (OR 0.57, 95% CI 0.40 to 0.80; moderate-quality evidence) and fewer respiratory tract infections (OR 0.70, 95% CI 0.62 to 0.80; moderate-quality evidence) compared to placebo, probably because these are not adverse events, but rather characteristics of the indications for the antibiotics. Less fever (OR 0.73, 95% 0.54 to 1.00; moderate-quality evidence) was also reported by participants taking macrolides compared to placebo, although these findings were non-significant.There was no increase in mortality in participants taking macrolides compared with placebo (OR 0.96, 95% 0.87 to 1.06; I² = 11%; low-quality evidence).Only 24 studies (13%) provided useful data on macrolide-resistant bacteria. Macrolide-resistant bacteria were more commonly identified among participants immediately after exposure to the antibiotic. However, differences in resistance thereafter were inconsistent.Pharmaceutical companies supplied the trial medication or funding, or both, for 91 trials. AUTHORS' CONCLUSIONS The macrolides as a group clearly increased rates of gastrointestinal adverse events. Most trials made at least some statement about adverse events, such as "none were observed". However, few trials clearly listed adverse events as outcomes, reported on the methods used for eliciting adverse events, or even detailed the numbers of people who experienced adverse events in both the intervention and placebo group. This was especially true for the adverse event of bacterial resistance.
Collapse
Affiliation(s)
| | - Anna M Scott
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Amanda McCullough
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Sarah Thorning
- Gold Coast Hospital and Health ServiceGCUH LibraryLevel 1, Block E, GCUHSouthportQueenslandAustralia4215
| | - Jeffrey K Aronson
- Oxford UniversityNuffield Department of Primary Care Health SciencesOxfordOxonUKOX26GG
| | - Elaine M Beller
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Paul P Glasziou
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Tammy C Hoffmann
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Justin Clark
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Chris B Del Mar
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | | |
Collapse
|
14
|
Randomized, Double-Blind, Placebo-Controlled Studies to Assess Safety and Prophylactic Efficacy of Naphthoquine-Azithromycin Combination for Malaria Prophylaxis in Southeast Asia. Antimicrob Agents Chemother 2018; 62:AAC.00793-18. [PMID: 29987144 DOI: 10.1128/aac.00793-18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/05/2018] [Indexed: 01/01/2023] Open
Abstract
New prophylactic drugs against malaria infections are urgently needed. We conducted randomized, double-blind, placebo-controlled, phase 2 trials of a new antimalarial drug combination, naphthoquine-azithromycin (NQAZ), to determine its safety and protective efficacy in a low-endemicity area of Southeast Asia. In the first trial, 127 healthy volunteers were randomized to receive two single doses of either 400 mg of NQAZ (200 mg of each drug), 800 mg of NQAZ (400 mg of each drug), or placebo on day 0 and day 30. Weekly follow-ups were performed for 2 months, and physical and clinical laboratory exams were done during the second and eighth week. Both drug regimens were well tolerated, without any serious adverse events. Four adverse events (transient and slight elevations of serum transaminase concentrations) were found only in the two drug-treated groups and thus might be drug-related. In the second trial, 353 volunteer villagers were randomized into the same three groups as in the first trial, and malaria infections were followed for a month. For the intention-to-treat analysis, both regimens offered greater than 90% prophylactic efficacies against all malaria infections. When the analysis was done according to parasite species, 400 mg and 800 mg NQAZ provided 81.63 and 90.59% prophylactic efficacies, respectively, against Plasmodium falciparum infections, whereas both offered 100% prophylactic efficacy against Plasmodium vivax and Plasmodium ovale These trials showed that NQAZ had a good safety profile, and monthly single doses of 400 mg or 800 mg for adults offered excellent prophylaxis against malaria infections, especially the two relapsing species.
Collapse
|
15
|
Annual Versus Biannual Mass Azithromycin Distribution and Malaria Parasitemia During the Peak Transmission Season Among Children in Niger. Pediatr Infect Dis J 2018; 37:506-510. [PMID: 29088030 PMCID: PMC5924654 DOI: 10.1097/inf.0000000000001813] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Azithromycin has modest efficacy against malaria, and previous cluster randomized trials have suggested that mass azithromycin distribution for trachoma control may play a role in malaria control. We evaluated the effect of annual versus biannual mass azithromycin distribution over a 3-year period on malaria prevalence during the peak transmission season in a region with seasonal malaria transmission in Niger. METHODS Twenty-four communities in Matameye, Niger, were randomized to annual mass azithromycin distribution (3 distributions to the entire community during the peak transmission season) or biannual-targeted azithromycin distribution (6 distributions to children <12 years of age, including 3 in the peak transmission season and 3 in the low transmission season). Malaria indices were evaluated at 36 months during the high transmission season. RESULTS Parasitemia prevalence was 42.6% (95% confidence interval: 31.7%-53.6%) in the biannual distribution arm compared with 50.6% (95% confidence interval: 40.3%-60.8%) in the annual distribution arm (P = 0.29). There was no difference in parasite density or hemoglobin concentration in the 2 treatment arms. CONCLUSIONS Additional rounds of mass azithromycin distribution during low transmission may not have a significant impact on malaria parasitemia measured during the peak transmission season.
Collapse
|
16
|
Wangdi K, Furuya-Kanamori L, Clark J, Barendregt JJ, Gatton ML, Banwell C, Kelly GC, Doi SAR, Clements ACA. Comparative effectiveness of malaria prevention measures: a systematic review and network meta-analysis. Parasit Vectors 2018; 11:210. [PMID: 29587882 PMCID: PMC5869791 DOI: 10.1186/s13071-018-2783-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/06/2018] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Malaria causes significant morbidity and mortality worldwide. There are several preventive measures that are currently employed, including insecticide-treated nets (ITNs, including long-lasting insecticidal nets and insecticidal-treated bed nets), indoor residual spraying (IRS), prophylactic drugs (PD), and untreated nets (UN). However, it is unclear which measure is the most effective for malaria prevention. We therefore undertook a network meta-analysis to compare the efficacy of different preventive measures on incidence of malaria infection. METHODS A systematic literature review was undertaken across four medical and life sciences databases (PubMed, Cochrane Central, Embase, and Web of Science) from their inception to July 2016 to compare the effectiveness of different preventive measures on malaria incidence. Data from the included studies were analysed for the effectiveness of several measures against no intervention (NI). This was carried out using an automated generalized pairwise modeling (GPM) framework for network meta-analysis to generate mixed treatment effects against a common comparator of no intervention (NI). RESULTS There were 30 studies that met the inclusion criteria from 1998-2016. The GPM framework led to a final ranking of effectiveness of measures in the following order from best to worst: PD, ITN, IRS and UN, in comparison with NI. However, only ITN (RR: 0.49, 95% CI: 0.32-0.74) showed precision while other methods [PD (RR: 0.24, 95% CI: 0.004-15.43), IRS (RR: 0.55, 95% CI: 0.20-1.56) and UN (RR: 0.73, 95% CI: 0.28-1.90)] demonstrating considerable uncertainty associated with their point estimates. CONCLUSION Current evidence is strong for the protective effect of ITN interventions in malaria prevention. Even though ITNs were found to be the only preventive measure with statistical support for their effectiveness, the role of other malaria control measures may be important adjuncts in the global drive to eliminate malaria.
Collapse
Affiliation(s)
- Kinley Wangdi
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| | - Luis Furuya-Kanamori
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
- Department of Population Medicine, College of Medicine, Qatar University, Doha, Qatar
| | - Justin Clark
- Centre for Research in Evidence-Based Practice (CREBP), Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Queensland Australia
| | - Jan J. Barendregt
- School of Public Health, The University of Queensland, Brisbane, Queensland Australia
- Epigear International Pty Ltd, Sunrise Beach, Queensland Australia
| | - Michelle L. Gatton
- School of Public Health & Social Work, Queensland University of Technology, Brisbane, Queensland Australia
| | - Cathy Banwell
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| | - Gerard C. Kelly
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| | - Suhail A. R. Doi
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
- Department of Population Medicine, College of Medicine, Qatar University, Doha, Qatar
| | - Archie C. A. Clements
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| |
Collapse
|
17
|
O'Brien KS, Cotter SY, Amza A, Kadri B, Nassirou B, Stoller NE, Zhou Z, Cotter C, West SK, Bailey RL, Rosenthal PJ, Gaynor BD, Porco TC, Lietman TM. Mass Azithromycin and Malaria Parasitemia in Niger: Results from a Community-Randomized Trial. Am J Trop Med Hyg 2017; 97:696-701. [PMID: 28722569 PMCID: PMC5590561 DOI: 10.4269/ajtmh.16-0487] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Studies designed to determine the effects of mass administration of azithromycin on trachoma have suggested that mass azithromycin distributions may also reduce the prevalence of malaria. These studies have typically examined the impact of a small number of treatments over short durations. In this prespecified substudy of a cluster-randomized trial for trachoma, we compared malaria parasitemia prevalence in 24 communities in Niger randomized to receive either annual or biannual mass azithromycin distributions over 3 years. The 12 communities randomized to annual azithromycin received three treatments during the high-transmission season, and the 12 communities randomized to biannual azithromycin received a total of six treatments: three during the high-transmission season and three during the low-transmission season. Blood samples were taken to assess malariometric indices among children in all study communities at a single time point during the high-transmission season after 3 years of the intervention. No significant differences were identified in malaria parasitemia, parasite density, or hemoglobin concentration between the annual and biannual treatment arms. When compared with annual mass azithromycin alone, additional mass azithromycin distributions given during the low-transmission season did not significantly reduce the subsequent prevalence of malaria parasitemia or parasite density after 3 years, as measured during the high-transmission season.
Collapse
Affiliation(s)
- Kieran S O'Brien
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California
| | - Sun Y Cotter
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California
| | - Abdou Amza
- Programme FSS/Université Abdou Moumouni de Niamey, Programme National de Santé Oculaire, Niamey, Niger
| | - Boubacar Kadri
- Programme FSS/Université Abdou Moumouni de Niamey, Programme National de Santé Oculaire, Niamey, Niger
| | - Baido Nassirou
- Programme FSS/Université Abdou Moumouni de Niamey, Programme National de Santé Oculaire, Niamey, Niger
| | - Nicole E Stoller
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California
| | - Zhaoxia Zhou
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California
| | - Chris Cotter
- Global Health Group, University of California San Francisco, San Francisco, California
| | - Sheila K West
- Dana Center for Preventive Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland
| | - Robin L Bailey
- Clinical Research Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Philip J Rosenthal
- Department of Medicine, University of California San Francisco, California
| | - Bruce D Gaynor
- Department of Ophthalmology, University of California San Francisco, San Francisco, California.,Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California
| | - Travis C Porco
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California.,Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California
| | - Thomas M Lietman
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California.,Department of Ophthalmology, University of California San Francisco, San Francisco, California.,Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California
| |
Collapse
|
18
|
Salman S, Baiwog F, Page-Sharp M, Griffin S, Karunajeewa HA, Mueller I, Rogerson SJ, Siba PM, Ilett KF, Davis TME. Optimal Antimalarial Dose Regimens for Sulfadoxine-Pyrimethamine with or without Azithromycin in Pregnancy Based on Population Pharmacokinetic Modeling. Antimicrob Agents Chemother 2017; 61:e02291-16. [PMID: 28242669 PMCID: PMC5404578 DOI: 10.1128/aac.02291-16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/20/2017] [Indexed: 01/19/2023] Open
Abstract
Optimal dosing of sulfadoxine-pyrimethamine (SP) as intermittent preventive treatment in pregnancy remains to be established, particularly when coadministered with azithromycin (AZI). To further characterize SP pharmacokinetics in pregnancy, plasma concentration-time data from 45 nonpregnant and 45 pregnant women treated with SP-AZI (n = 15 in each group) and SP-chloroquine (n = 30 in each group) were analyzed. Population nonlinear mixed-effect pharmacokinetic models were developed for pyrimethamine (PYR), sulfadoxine (SDOX), and N-acetylsulfadoxine (the SDOX metabolite NASDOX), and potential covariates were included. Pregnancy increased the relative clearance (CL/F) of PYR, SDOX, and NASDOX by 48, 29, and 70%, respectively, as well as the relative volumes of distribution (V/F) of PYR (46 and 99%) and NASDOX (46%). Coadministration of AZI resulted in a greater increase in PYR CL/F (80%) and also increased NASDOX V/F by 76%. Apparent differences between these results and those of published studies of SP disposition may reflect key differences in study design, including the use of an early postpartum follow-up study rather than a nonpregnant comparator group. Simulations based on the final population model demonstrated that, compared to conventional single-dose SP in nonpregnant women, two such doses given 24 h apart should ensure that pregnant women have similar drug exposure, while three daily SP doses may be required if SP is given with AZI. The results of past and ongoing trials using recommended adult SP doses with or without AZI in pregnant women may need to be interpreted in light of these findings and consideration given to using increased doses in future trials.
Collapse
Affiliation(s)
- Sam Salman
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| | - Francisca Baiwog
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Madhu Page-Sharp
- School of Pharmacy, Curtin University of Technology, Bentley, Western Australia, Australia
| | - Susan Griffin
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Harin A Karunajeewa
- Population Health and Immunity, Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
| | - Ivo Mueller
- Population Health 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
| | - Peter M Siba
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Kenneth F Ilett
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| | - Timothy M E Davis
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
19
|
Berkley JA, Ngari M, Thitiri J, Mwalekwa L, Timbwa M, Hamid F, Ali R, Shangala J, Mturi N, Jones KDJ, Alphan H, Mutai B, Bandika V, Hemed T, Awuondo K, Morpeth S, Kariuki S, Fegan G. Daily co-trimoxazole prophylaxis to prevent mortality in children with complicated severe acute malnutrition: a multicentre, double-blind, randomised placebo-controlled trial. LANCET GLOBAL HEALTH 2016; 4:e464-73. [PMID: 27265353 PMCID: PMC6132285 DOI: 10.1016/s2214-109x(16)30096-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 04/08/2016] [Accepted: 05/11/2016] [Indexed: 01/13/2023]
Abstract
Background Children with complicated severe acute malnutrition (SAM) have a greatly increased risk of mortality from infections while in hospital and after discharge. In HIV-infected children, mortality and admission to hospital are prevented by daily co-trimoxazole prophylaxis, despite locally reported bacterial resistance to co-trimoxazole. We aimed to assess the efficacy of daily co-trimoxazole prophylaxis on survival in children without HIV being treated for complicated SAM. Methods We did a multicentre, double-blind, randomised, placebo-controlled study in four hospitals in Kenya (two rural hospitals in Kilifi and Malindi, and two urban hospitals in Mombasa and Nairobi) with children aged 60 days to 59 months without HIV admitted to hospital and diagnosed with SAM. We randomly assigned eligible participants (1:1) to 6 months of either daily oral co-trimoxazole prophylaxis (given as water-dispersible tablets; 120 mg per day for age <6 months, 240 mg per day for age 6 months to 5 years) or matching placebo. Assignment was done with computer-generated randomisation in permuted blocks of 20, stratified by centre and age younger or older than 6 months. Treatment allocation was concealed in opaque, sealed envelopes and patients, their families, and all trial staff were masked to treatment assignment. Children were given recommended medical care and feeding, and followed up for 12 months. The primary endpoint was mortality, assessed each month for the first 6 months, then every 2 months for the second 6 months. Secondary endpoints were nutritional recovery, readmission to hospital, and illness episodes treated as an outpatient. Analysis was by intention to treat. This trial was registered at ClinicalTrials.gov, number NCT00934492. Findings Between Nov 20, 2009, and March 14, 2013, we recruited and assigned 1778 eligible children to treatment (887 to co-trimoxazole prophylaxis and 891 to placebo). Median age was 11 months (IQR 7–16 months), 306 (17%) were younger than 6 months, 300 (17%) had oedematous malnutrition (kwashiorkor), and 1221 (69%) were stunted (length-for-age Z score <–2). During 1527 child-years of observation, 122 (14%) of 887 children in the co-trimoxazole group died, compared with 135 (15%) of 891 in the placebo group (unadjusted hazard ratio [HR] 0·90, 95% CI 0·71–1·16, p=0·429; 16·0 vs 17·7 events per 100 child-years observed (CYO); difference −1·7 events per 100 CYO, 95% CI −5·8 to 2·4]). In the first 6 months of the study (while participants received study medication), 63 suspected grade 3 or 4 associated adverse events were recorded among 57 (3%) children; 31 (2%) in the co-trimoxazole group and 32 (2%) in the placebo group (incidence rate ratio 0·98, 95% CI 0·58–1·65). The most common adverse events of these grades were urticarial rash (grade 3, equally common in both groups), neutropenia (grade 4, more common in the co-trimoxazole group), and anaemia (both grades equally common in both groups). One child in the placebo group had fatal toxic epidermal necrolysis with concurrent Pseudomonas aeruginosa bacteraemia. Interpretation Daily co-trimoxazole prophylaxis did not reduce mortality in children with complicated SAM without HIV. Other strategies need to be tested in clinical trials to reduce deaths in this population. Funding Wellcome Trust, UK
Collapse
Affiliation(s)
- James A Berkley
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya; Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.
| | - Moses Ngari
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Johnstone Thitiri
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Laura Mwalekwa
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Molline Timbwa
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Fauzat Hamid
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Rehema Ali
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Jimmy Shangala
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Neema Mturi
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kelsey D J Jones
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya; Imperial College, London, UK
| | - Hassan Alphan
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya
| | | | | | | | - Ken Awuondo
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Susan Morpeth
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya; University College, London, UK
| | | | - Gregory Fegan
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya; Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
20
|
Rosenthal PJ. Azithromycin for Malaria? Am J Trop Med Hyg 2016; 95:2-4. [PMID: 27215296 PMCID: PMC4944689 DOI: 10.4269/ajtmh.16-0332] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 05/02/2016] [Indexed: 11/07/2022] Open
Affiliation(s)
- Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, California
| |
Collapse
|
21
|
Spring MD, Pichyangkul S, Lon C, Gosi P, Yongvanichit K, Srichairatanakul U, Limsalakpeth A, Chaisatit C, Chann S, Sriwichai S, Auayapon M, Chaorattanakawee S, Dutta S, Prom S, Meng Chour C, Walsh DS, Angov E, Saunders DL. Antibody profiles to plasmodium merozoite surface protein-1 in Cambodian adults during an active surveillance cohort with nested treatment study. Malar J 2016; 15:17. [PMID: 26747132 PMCID: PMC4706704 DOI: 10.1186/s12936-015-1058-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 12/17/2015] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND In addition to evidence for a protective role of antibodies to the malaria blood stage antigen merozoite surface protein 1 (MSP1), MSP1 antibodies are also considered as a marker of past malaria exposure in sero-epidemiological studies. METHODS In order to better assess the potential use of MSP1 serology in malaria chemoprophylaxis trials in endemic areas, an analysis for the prevalence of antibodies to both Plasmodium falciparum and Plasmodium vivax MSP142 in healthy Cambodian adults was conducted at two sites as part of an active, observational cohort evaluating the efficacy of dihydroartemisinin-piperaquine (DP) for uncomplicated malaria (ClinicalTrials.gov identifier NCT01280162). RESULTS Rates of baseline sero-positivity were high (59 and 73% for PfMSP142 and PvMSP142, respectively), and titers higher in those who lived in a higher transmission area, although there was little correlation in titers between the two species. Those volunteers who subsequently went on to develop malaria had higher baseline MSP142 titers than those who did not for both species. Titers to both antigens remained largely stable over the course of the 4-6 month study, except in those infected with P. falciparum who had multiple recurrences. CONCLUSION These findings illuminate the difficulties in using MSP142 serology as either a screening criterion and/or biomarker of exposure in chemoprophylaxis studies. Further work remains to identify useful markers of malarial infection and/or immunity.
Collapse
Affiliation(s)
- Michele D Spring
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand.
| | - Sathit Pichyangkul
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand.
| | - Chanthap Lon
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand.
| | - Panita Gosi
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand.
| | - Kosol Yongvanichit
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand.
| | | | - Amporn Limsalakpeth
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand.
| | - Chaiyaporn Chaisatit
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand.
| | - Soklyda Chann
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand.
| | - Sabaithip Sriwichai
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand.
| | - Montida Auayapon
- Center of Excellence for Biomedical and Public Health Informatics (BIOPHICS), Bangkok, Thailand.
| | | | - Sheetij Dutta
- Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | | | - Char Meng Chour
- National Center for Parasitology, Entomology and Malaria Control (CNM), Phnom Penh, Cambodia.
| | | | - Evelina Angov
- Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | - David L Saunders
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand.
| |
Collapse
|
22
|
Gaillard T, Madamet M, Pradines B. Tetracyclines in malaria. Malar J 2015; 14:445. [PMID: 26555664 PMCID: PMC4641395 DOI: 10.1186/s12936-015-0980-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 11/02/2015] [Indexed: 11/12/2022] Open
Abstract
Malaria, a parasite vector-borne disease, is one of the greatest health threats in tropical regions, despite the availability of malaria chemoprophylaxis. The emergence and rapid extension of Plasmodium falciparum resistance to various anti-malarial drugs has gradually limited the number of potential malaria therapeutics available to clinicians. In this context, doxycycline, a synthetically derived tetracycline, constitutes an interesting alternative for malaria treatment and prophylaxis. Doxycycline is a slow-acting blood schizontocidal agent that is highly effective at preventing malaria. In areas with chloroquine and multidrug-resistant P. falciparum parasites, doxycycline has already been successfully used in combination with quinine to treat malaria, and it has been proven to be effective and well-tolerated. Although not recommended for pregnant women and children younger than 8 years of age, severe adverse effects are rarely reported. In addition, resistance to doxycycline is rarely described. Prophylactic and clinical failures of doxycycline have been associated with both inadequate doses and poor patient compliance. The effects of tetracyclines on parasites are not completely understood. A better comprehension of the mechanisms underlying drug resistance would facilitate the identification of molecular markers of resistance to predict and survey the emergence of resistance.
Collapse
Affiliation(s)
- Tiphaine Gaillard
- Unité de Parasitologie, Département d'Infectiologie de Terrain, Institut de Recherche Biomédicale des Armées, Marseille, France. .,Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM 63, CNRS 7278, IRD 198, Inserm 1095, Aix Marseille Université, Marseille, France. .,Fédération des Laboratoires, Hôpital d'Instruction des Armées Saint Anne, Toulon, France.
| | - Marylin Madamet
- Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM 63, CNRS 7278, IRD 198, Inserm 1095, Aix Marseille Université, Marseille, France. .,Equipe Résidente de Recherche en Infectiologie Tropicale, Institut de Recherche Biomédicale des Armées, Hôpital d'Instruction des Armées, Marseille, France. .,Centre National de Référence du Paludisme, Marseille, France.
| | - Bruno Pradines
- Unité de Parasitologie, Département d'Infectiologie de Terrain, Institut de Recherche Biomédicale des Armées, Marseille, France. .,Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM 63, CNRS 7278, IRD 198, Inserm 1095, Aix Marseille Université, Marseille, France. .,Centre National de Référence du Paludisme, Marseille, France. .,Unité de Parasitologie et d'Entomologie, Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Brétigny sur Orge, France.
| |
Collapse
|
23
|
Čikoš A, Ćaleta I, Žiher D, Vine MB, Elenkov IJ, Dukši M, Gembarovski D, Ilijaš M, Dragojević S, Malnar I, Alihodžić S. Structure and conformational analysis of spiroketals from 6-O-methyl-9(E)-hydroxyiminoerythronolide A. Beilstein J Org Chem 2015; 11:1447-57. [PMID: 26425201 PMCID: PMC4578343 DOI: 10.3762/bjoc.11.157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/23/2015] [Indexed: 11/23/2022] Open
Abstract
Three novel spiroketals were prepared by a one-pot transformation of 6-O-methyl-9(E)-hydroxyiminoerythronolide A. We present the formation of a [4.5]spiroketal moiety within the macrolide lactone ring, but also the unexpected formation of a 10-C=11-C double bond and spontaneous change of stereochemistry at position 8-C. As a result, a thermodynamically stable structure was obtained. The structures of two new diastereomeric, unsaturated spiroketals, their configurations and conformations, were determined by means of NMR spectroscopy and molecular modelling. The reaction kinetics and mechanistic aspects of this transformation are discussed. These rearrangements provide a facile synthesis of novel macrolide scaffolds.
Collapse
Affiliation(s)
- Ana Čikoš
- GlaxoSmithKline Research Centre Zagreb Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Irena Ćaleta
- GlaxoSmithKline Research Centre Zagreb Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Dinko Žiher
- GlaxoSmithKline Research Centre Zagreb Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Mark B Vine
- GlaxoSmithKline, New Frontiers Science Park, Harlow, CM19 5AW, United Kingdom
| | - Ivaylo J Elenkov
- GlaxoSmithKline Research Centre Zagreb Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Marko Dukši
- GlaxoSmithKline Research Centre Zagreb Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Dubravka Gembarovski
- GlaxoSmithKline Research Centre Zagreb Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Marina Ilijaš
- GlaxoSmithKline Research Centre Zagreb Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Snježana Dragojević
- GlaxoSmithKline Research Centre Zagreb Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Ivica Malnar
- GlaxoSmithKline Research Centre Zagreb Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Sulejman Alihodžić
- GlaxoSmithKline Research Centre Zagreb Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| |
Collapse
|
24
|
Tevyashova AN, Olsufyeva EN, Preobrazhenskaya MN. Design of dual action antibiotics as an approach to search for new promising drugs. RUSSIAN CHEMICAL REVIEWS 2015. [DOI: 10.1070/rcr4448] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
25
|
Schachterle SE, Mtove G, Levens JP, Clemens E, Shi L, Raj A, Dumler JS, Munoz B, West S, Sullivan DJ. Short-term malaria reduction by single-dose azithromycin during mass drug administration for trachoma, Tanzania. Emerg Infect Dis 2015; 20:941-9. [PMID: 24865642 PMCID: PMC4036785 DOI: 10.3201/eid2006.131302] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
This drug might be beneficial in areas to which malaria and trachoma are endemic. Single-dose mass drug administration of azithromycin (AZT) is underway to eliminate trachoma worldwide. Studies in Ethiopia showed a reduction in all-cause childhood deaths after administration. To examine the effect of single-dose AZ MDA on prevalent malaria infections in a large prospective cohort of children and parents in Dodoma Province, Tanzania, we quantified the temporal prevalence of malaria parasitemia by real-time PCR for 6 months after single-dose AZT. In the first month after treatment but not in subsequent months, Plasmodium falciparum infections were reduced by 73% (95% CI 43%–89%) in treatment versus control villages and differences remained significant (p = 0.00497) in multivariate models with village-level random effects. Genetic sequencing of P. falciparum ribosomal L4 protein showed no mutations associated with AZT resistance. AZT mass drug administration caused a transient, 1-month antimalarial effect without selecting for P. falciparum ribosomal L4 resistance mutations in a region with a 10-year history of treating trachoma with this drug.
Collapse
|
26
|
Shibahara D, Kinjo T, Nishiyama N, Kami W, Nabeya D, Haranaga S, Higa F, Tateyama M, Shinzato T, Toma H, Kishimoto H, Fujita J. Falciparum Malaria Incidentally Pretreated with Azithromycin. Intern Med 2015; 54:2513-6. [PMID: 26424314 DOI: 10.2169/internalmedicine.54.5399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A 65-year-old man, who recently returned from Liberia, visited a clinic complaining of fever, and azithromycin was prescribed. The patient presented to a general hospital 5 days after the onset of symptoms, however, a blood smear examination failed to detect malaria. Contrary to the blood smear result, a rapid antigen test in our hospital was strongly-positive for falciparum malaria, indicating a high level of malarial antigen in the blood. Moreover, laboratory examinations on admission showed a tendency for improvement. We assumed that the administration of azithromycin partially treated malaria, thus complicating the blood smear diagnosis. We should be careful in prescribing azithromycin, which is widely used in clinics, to travelers returning from malaria-endemic countries.
Collapse
Affiliation(s)
- Daisuke Shibahara
- Department of Infectious Diseases, Respiratory and Digestive Medicine, Faculty of Medicine, University of the Ryukyus, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Efficacy and safety of a combination of azithromycin and chloroquine for the treatment of uncomplicated Plasmodium falciparum malaria in two multi-country randomised clinical trials in African adults. Malar J 2014; 13:458. [PMID: 25425434 PMCID: PMC4364337 DOI: 10.1186/1475-2875-13-458] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 11/18/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Given increasing rates of resistance to existing therapy, new options for treatment and prophylaxis of malaria are needed. METHODS Two randomised, comparative, non-inferiority studies were conducted in Africa, one double-blinded and one open-label. Adults with fever, a positive peripheral blood smear, and a positive rapid diagnostic test for Plasmodium falciparum were randomised in both studies to either azithromycin (AZ) 1,000 mg plus chloroquine (CQ) 600-mg base (AZCQ 1,000 mg) once daily for three days or mefloquine hydrochloride (MQ) 1,250 mg (split dose). In the first study, an additional regimen of AZ 500 mg plus CQ 600-mg base (AZCQ 500 mg) once daily for three days was included. All study participants were hospitalised until three consecutive daily blood smears were negative for asexual P. falciparum parasitaemia. Study participants were evaluated weekly for 42 days, with Day 28 polymerase chain reaction (PCR)-corrected parasitological clearance rate as primary endpoint. RESULTS A total of 467 subjects were randomised in the two studies. At 28 days' follow-up, PCR-corrected parasitological clearance rates in the per protocol population in the first study were 101/103 (98%) with AZCQ 1,000 mg compared with 102/103 (99%) with MQ (95% confidence interval [CI]: -5.2, 3.3). The AZCQ 500-mg regimen was stopped during an interim study review (six [86%] clearance of seven evaluable; two lost to follow-up). In the second study, clearance rates were similar: AZCQ 1,000 mg 107/107 (100%) vs MQ 111/112 (99%; 95% CI: -1.8, 3.6). Among the participating countries, in vitro CQ resistance based on pfcrt mutation frequency in the baseline isolates across both studies ranged from 20.8% (Zambia) to 96.1% (Uganda). Serious adverse events (AEs; all causality) were observed more frequently with MQ compared with AZCQ (four vs one, respectively), though discontinuations for AEs were similar (four vs three, respectively). Common AEs in the AZ-containing arms included pruritus, vomiting, dizziness, and headache. CONCLUSIONS Among adults with symptomatic uncomplicated falciparum malaria in Africa, the combination of AZ 1,000 mg and CQ 600-mg base once daily for three days resulted in Day 28 PCR-corrected parasitological clearance rates of ≥98% and was non-inferior to treatment with MQ. AZCQ was well tolerated. TRIAL REGISTRATION ClinicalTrials.gov identifiers NCT00082576 and NCT00367653.
Collapse
|
28
|
Gao D, Amza A, Nassirou B, Kadri B, Sippl-Swezey N, Liu F, Ackley SF, Lietman TM, Porco TC. Optimal seasonal timing of oral azithromycin for malaria. Am J Trop Med Hyg 2014; 91:936-942. [PMID: 25223942 PMCID: PMC4228890 DOI: 10.4269/ajtmh.13-0474] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mass administration of azithromycin for trachoma has been shown to reduce malarial parasitemia. However, the optimal seasonal timing of such distributions for antimalarial benefit has not been established. We performed numerical analyses on a seasonally forced epidemic model (of Ross-Macdonald type) with periodic impulsive annual mass treatment to address this question. We conclude that when azithromycin-based trachoma elimination programs occur in regions of seasonal malaria transmission, such as Niger, the optimal seasonal timing of mass drug administration (MDA) may not occur during the season of maximum transmission.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Travis C. Porco
- *Address correspondence to Travis C. Porco, F.I. Proctor Foundation, UCSF, San Francisco, CA 94143-0412. E-mail:
| |
Collapse
|
29
|
Gaynor BD, Amza A, Kadri B, Nassirou B, Lawan O, Maman L, Stoller NE, Yu SN, Chin SA, West SK, Bailey RL, Rosenthal PJ, Keenan JD, Porco TC, Lietman TM. Impact of mass azithromycin distribution on malaria parasitemia during the low-transmission season in Niger: a cluster-randomized trial. Am J Trop Med Hyg 2014; 90:846-51. [PMID: 24615132 PMCID: PMC4015576 DOI: 10.4269/ajtmh.13-0379] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 12/23/2013] [Indexed: 12/22/2022] Open
Abstract
We assessed the effect of mass azithromycin treatment on malaria parasitemia in a trachoma trial in Niger. Twenty-four study communities received treatment during the wet, high-transmission season. Twelve of the 24 communities were randomized to receive an additional treatment during the dry, low-transmission season. Outcome measurements were conducted at the community-level in children < 1-72 months of age in May-June 2011. Parasitemia was higher in the 12 once-treated communities (29.8%, 95% confidence interval [CI] = 21.5-40.0%) than in the 12 twice-treated communities (19.5%, 95% CI = 13.0-26.5%, P = 0.03). Parasite density was higher in once-treated communities (354 parasites/μL, 95% CI = 117-528 parasites/μL) than in twice-treated communities (74 parasites/μL, 95% CI = 41-202 parasites/μL, P = 0.03). Mass distribution of azithromycin reduced malaria parasitemia 4-5 months after the intervention. The results suggest that drugs with antimalaria activity can have long-lasting impacts on malaria during periods of low transmission.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Thomas M. Lietman
- F.I. Proctor Foundation, Department of Ophthalmology, Department of Epidemiology and Biostatistics, Institute for Global Health, Department of Medicine, University of California, San Francisco, California; Programme National de Lutte Contre la Cecité Niamey, Niger; Zinder Hospital, Zinder, Niger; Dana Center for Preventive Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland; Clinical Research Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
30
|
Hart JD, Edwards T, Burr SE, Harding-Esch EM, Takaoka K, Holland MJ, Sillah A, Mabey DCW, Bailey RL. Effect of azithromycin mass drug administration for trachoma on spleen rates in Gambian children. Trop Med Int Health 2014; 19:207-11. [PMID: 24433194 DOI: 10.1111/tmi.12234] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE To assess the effect of azithromycin mass drug administration regimens on spleen rates in children aged 0-5 years. METHODS Clinical assessment of spleen size was carried out during a cluster-randomised trial of azithromycin mass treatment for trachoma elimination in The Gambia. Twenty-four communities received three annual mass treatments with azithromycin, and 24 communities received treatment at baseline only. RESULTS At the 30-month follow-up, 3646 children aged 0-5 years had spleen examination and measurement. Palpable splenomegaly was significantly lower in annually treated vs. baseline-only treatment communities and in treated vs. untreated children at 24 months in the annual treatment arm. CONCLUSION The results suggest an effect of azithromycin on spleen rates at the individual level and are most plausibly due to the antimalarial effects of azithromycin.
Collapse
Affiliation(s)
- John D Hart
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Petersen E. Malaria chemoprophylaxis: when should we use it and what are the options? Expert Rev Anti Infect Ther 2014; 2:119-32. [PMID: 15482177 DOI: 10.1586/14787210.2.1.119] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Malaria chemoprophylaxis concerns prescribing healthy individuals medication for an infection they have an unknown chance of getting. Sensible use of malaria chemoprophylaxis is a balance between the risk of infection and death, and the risk of side effects. The risk of infection can be broken down into the risk of being bitten by a malaria-infected mosquito and the risk of the malaria parasites being resistant to the drug used for prophylaxis. Our knowledge of these parameters is patchy. The risk of infection is not uniform at a given location and the standard of living will greatly influence risk. It is suggested that chemoprophylaxis should not be recommended in areas with less than ten reported cases of P. falciparum malaria per 1000 inhabitants per year. The resistance pattern is known to a certain extent but, for instance, diverging opinion of how much resistance to chloroquine there is in West Africa illustrates the lack of data. There is much debate on rare adverse events, which usually escape Phase III studies prior to registration and are only picked up by passive, postmarketing surveillance. The lessons over the past 20 years with the introduction of amodiaquine, pyrimethamine/dapsone (Maloprim, GlaxoSmithKline) and pyrimethamine/sulfadoxine (Fansidar, Roche), which were all withdrawn for prophylaxis after a few years, show how sensitive drugs for chemoprophylaxis are to side effects. Three levels of chemoprophylaxis are used: chloroquine in areas with sensitive P. falciparum, chloroquine plus proguanil in areas with low level chloroquine resistance, and atovaquone/proguanil (Malarone, GlaxoSmithKline), doxycycline or mefloquine (Lariam, Roche) in areas with extensive resistance against chloroquine and proguanil. Primaquine and the primaquone analog tafenoquine may be future alternatives but otherwise there are few new drugs for chemoprophylaxis on the horizon.
Collapse
Affiliation(s)
- Eskild Petersen
- Department of Infectious Diseases, Aarhus University Hospital Skejby, DK-8200 Aarhus N, Denmark.
| |
Collapse
|
32
|
Randomized, open-label trial of primaquine against vivax malaria relapse in Indonesia. Antimicrob Agents Chemother 2012; 57:1128-35. [PMID: 23254437 DOI: 10.1128/aac.01879-12] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Radical cure of Plasmodium vivax infection applies blood schizontocidal therapy against the acute attack and hypnozoitocidal therapy against later relapse. Chloroquine and primaquine have been used for 60 years in this manner. Resistance to chloroquine by the parasite now requires partnering other blood schizontocides with primaquine. However, the safety and efficacy of primaquine against relapse when combined with other drugs have not been demonstrated. This randomized, open-label, and relapse-controlled trial estimated the efficacy of primaquine against relapse when administered with quinine or dihydroartemisinin-piperaquine for treatment of the acute infection. Among 650 soldiers who had returned to their malaria-free base in Java, Indonesia, after 12 months in malarious Papua, Indonesia, 143 with acute P. vivax malaria were eligible for study. One hundred sixteen enrolled subjects were randomized to these treatments: artesunate (200-mg dose followed by 100 mg/day for 6 days), quinine (1.8 g/day for 7 days) plus concurrent primaquine (30 mg/day for 14 days), or dihydroartemisinin (120 mg) plus piperaquine (960 mg) daily for 3 days followed 25 days later by primaquine (30 mg/day for 14 days). Follow-up was for 12 months. One hundred thirteen subjects were analyzable. Relapse occurred in 32 of 41 (78%) subjects administered artesunate alone (2.71 attacks/person-year), 7 of 36 (19%) administered quinine plus primaquine (0.23 attack/person-year), and 2 of 36 (6%) administered dihydroartemisinin-piperaquine plus primaquine (0.06 attack/person-year). The efficacy of primaquine against relapse was 92% (95% confidence interval [CI] = 81% to 96%) for quinine plus primaquine and 98% (95% CI = 91% to 99%) for dihydroartemisinin-piperaquine plus primaquine. Antirelapse therapy with primaquine begun a month after treatment of the acute attack with dihydroartemisinin-piperaquine proved safe and highly efficacious against relapse by P. vivax acquired in Papua, Indonesia.
Collapse
|
33
|
Kosol S, Schrank E, Krajačić MB, Wagner GE, Meyer NH, Göbl C, Rechberger GN, Zangger K, Novak P. Probing the Interactions of Macrolide Antibiotics with Membrane-Mimetics by NMR Spectroscopy. J Med Chem 2012; 55:5632-6. [DOI: 10.1021/jm300647f] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Simone Kosol
- Institute
of Chemistry/Organic
and Bioorganic Chemistry, University of Graz, Heinrichstrasse 28,
A-8010 Graz, Austria
| | - Evelyne Schrank
- Institute
of Chemistry/Organic
and Bioorganic Chemistry, University of Graz, Heinrichstrasse 28,
A-8010 Graz, Austria
| | | | - Gabriel E. Wagner
- Institute
of Chemistry/Organic
and Bioorganic Chemistry, University of Graz, Heinrichstrasse 28,
A-8010 Graz, Austria
| | - N. Helge Meyer
- Institute
of Chemistry/Organic
and Bioorganic Chemistry, University of Graz, Heinrichstrasse 28,
A-8010 Graz, Austria
| | - Christoph Göbl
- Institute
of Chemistry/Organic
and Bioorganic Chemistry, University of Graz, Heinrichstrasse 28,
A-8010 Graz, Austria
| | - Gerald N. Rechberger
- Institute of Molecular Biosciences,
University of Graz, Humboldtstrasse 50, A-8010 Graz, Austria
| | - Klaus Zangger
- Institute
of Chemistry/Organic
and Bioorganic Chemistry, University of Graz, Heinrichstrasse 28,
A-8010 Graz, Austria
| | - Predrag Novak
- Department of Chemistry, Faculty
of Natural Science, University of Zagreb, Laboratory for Analytical
Chemistry, Horvatovac 102a, HR-10000 Zagreb, Croatia
| |
Collapse
|
34
|
Pešić D, Starčević K, Toplak A, Herreros E, Vidal J, Almela MJ, Jelić D, Alihodžić S, Spaventi R, Perić M. Design, Synthesis, and in Vitro Activity of Novel 2′-O-Substituted 15-Membered Azalides. J Med Chem 2012; 55:3216-27. [DOI: 10.1021/jm201676t] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Dijana Pešić
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića
29, 10000 Zagreb, Croatia
| | - Kristina Starčević
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića
29, 10000 Zagreb, Croatia
| | - Ana Toplak
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića
29, 10000 Zagreb, Croatia
| | - Esperanza Herreros
- Tres Cantos Medicines Development
Campus, Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos (Madrid), Spain
| | - Jaume Vidal
- Tres Cantos Medicines Development
Campus, Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos (Madrid), Spain
| | - Maria Jesus Almela
- Tres Cantos Medicines Development
Campus, Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos (Madrid), Spain
| | - Dubravko Jelić
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića
29, 10000 Zagreb, Croatia
| | - Sulejman Alihodžić
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića
29, 10000 Zagreb, Croatia
| | - Radan Spaventi
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića
29, 10000 Zagreb, Croatia
| | - Mihaela Perić
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića
29, 10000 Zagreb, Croatia
| |
Collapse
|
35
|
|
36
|
Novel hybrid molecules based on 15-membered azalide as potential antimalarial agents. Eur J Med Chem 2012; 49:365-78. [DOI: 10.1016/j.ejmech.2012.01.039] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 01/16/2012] [Accepted: 01/17/2012] [Indexed: 11/19/2022]
|
37
|
Steinhardt LC, Magill AJ, Arguin PM. Review: Malaria chemoprophylaxis for travelers to Latin America. Am J Trop Med Hyg 2012; 85:1015-24. [PMID: 22144437 DOI: 10.4269/ajtmh.2011.11-0464] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Because of recent declining malaria transmission in Latin America, some authorities have recommended against chemoprophylaxis for most travelers to this region. However, the predominant parasite species in Latin America, Plasmodium vivax, can form hypnozoites sequestered in the liver, causing malaria relapses. Additionally, new evidence shows the potential severity of vivax infections, warranting continued consideration of prophylaxis for travel to Latin America. Individualized travel risk assessments are recommended and should consider travel locations, type, length, and season, as well as probability of itinerary changes. Travel recommendations might include no precautions, mosquito avoidance only, or mosquito avoidance and chemoprophylaxis. There are a range of good options for chemoprophylaxis in Latin America, including atovaquone-proguanil, doxycycline, mefloquine, and--in selected areas--chloroquine. Primaquine should be strongly considered for nonpregnant, G6PD-nondeficient patients traveling to vivax-endemic areas of Latin America, and it has the added benefit of being the only drug to protect against malaria relapses.
Collapse
Affiliation(s)
- Laura C Steinhardt
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Prevention and Control, Atlanta, Georgia 30333, USA.
| | | | | |
Collapse
|
38
|
Perić M, Fajdetić A, Rupčić R, Alihodžić S, Ziher D, Bukvić Krajačić M, Smith KS, Ivezić-Schönfeld Z, Padovan J, Landek G, Jelić D, Hutinec A, Mesić M, Ager A, Ellis WY, Milhous WK, Ohrt C, Spaventi R. Antimalarial activity of 9a-N substituted 15-membered azalides with improved in vitro and in vivo activity over azithromycin. J Med Chem 2012; 55:1389-401. [PMID: 22148880 DOI: 10.1021/jm201615t] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Novel classes of antimalarial drugs are needed due to emerging drug resistance. Azithromycin, the first macrolide investigated for malaria treatment and prophylaxis, failed as a single agent and thus novel analogues were envisaged as the next generation with improved activity. We synthesized 42 new 9a-N substituted 15-membered azalides with amide and amine functionalities via simple and inexpensive chemical procedures using easily available building blocks. These compounds exhibited marked advances over azithromycin in vitro in terms of potency against Plasmodium falciparum (over 100-fold) and high selectivity for the parasite and were characterized by moderate oral bioavailability in vivo. Two amines and one amide derivative showed improved in vivo potency in comparison to azithromycin when tested in a mouse efficacy model. Results obtained for compound 6u, including improved in vitro potency, good pharmacokinetic parameters, and in vivo efficacy higher than azithromycin and comparable to chloroquine, warrant its further development for malaria treatment and prophylaxis.
Collapse
Affiliation(s)
- Mihaela Perić
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Deye GA, Miller RS, Miller L, Salas CJ, Tosh D, Macareo L, Smith BL, Fracisco S, Clemens EG, Murphy J, Sousa JC, Dumler JS, Magill AJ. Prolonged protection provided by a single dose of atovaquone-proguanil for the chemoprophylaxis of Plasmodium falciparum malaria in a human challenge model. Clin Infect Dis 2012; 54:232-9. [PMID: 22052893 DOI: 10.1093/cid/cir770] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND We conducted a randomized, placebo-controlled, double-blind trial to establish the efficacy of atovaquone-proguanil to prevent malaria with the goal of simulating weekly dosing in a human Plasmodium falciparum challenge model. METHODS Thirty volunteers randomly received 1 of the following dose regimens: (1) 250 milligrams of atovaquone and 100 milligrams of proguanil (250/100 milligrams) 1 day prior to infectious mosquito challenge (day -1), (2) 250/100 milligrams on day 4 after challenge, (3) 250/100 milligrams on day -7, (4) 500 milligrams of atovaquone and 200 milligrams of proguanil (500/200 milligrams) on day -7 or, (5) 1000 milligrams of atovaquone and 400 milligrams of proguanil (1000/400 milligrams) on day -7. All regimens included matching placebo such that all volunteers received identical pill numbers. Six volunteers served as open-label infectivity controls. Volunteers underwent mosquito sporozoite challenge with P. falciparum 3D7 strain. Follow-up consisted of serial microscopy and close clinical monitoring for 90 days. RESULTS Six of 6 infectivity controls developed parasitemia as expected. Two of 5 evaluable volunteers receiving 250/100 milligrams 7 days prior to challenge and 1 of 6 volunteers receiving 1000/400 milligrams 7 days prior to challenge were microscopically diagnosed with malaria. All other volunteers were protected. Atovaquone exposure (area under the curve) during liver stage development was low in 2 of 3 volunteers with prophylactic failure (423 and 199 ng/mL × days compared with a mean for protected volunteers of 1903 ng/mL × days), as was peak concentration (165 and 81 ng/mL compared with a mean of 594 ng/mL in volunteers with prophylactic success). Elimination half-life was short in volunteers with prophylactic failure (2.4, 2.0, and 3.3 days compared with a mean of 4.1 days in volunteers with prophylactic success). CONCLUSIONS Single-dose atovaquone-proguanil provides effective malaria chemoprophylaxis against P. falciparum challenge at dosing intervals supportive of weekly dosing. Postexposure prophylaxis 4 days after challenge was 100% effective.
Collapse
Affiliation(s)
- Gregory A Deye
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring,Maryland 20910, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Dow GS, Magill AJ, Ohrt C. Clinical development of new prophylactic antimalarial drugs after the 5th Amendment to the Declaration of Helsinki. Ther Clin Risk Manag 2011; 4:803-19. [PMID: 19209263 PMCID: PMC2621393 DOI: 10.2147/tcrm.s1025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Malaria is of continuing concern in nonimmune traveling populations. Traditionally, antimalarial drugs have been developed as agents for dual indications (treatment and prophylaxis). However, since 2000, when the 5th Amendment to the Declaration of Helsinki (DH2000) was adopted, development of new malaria prophylaxis drugs in this manner has ceased. As a consequence, there may not be any new drugs licensed for this indication in the foreseeable future. Major pharmaceutical companies have interpreted DH2000 to mean that the traditional development paradigm may be considered unethical because of doubt over the likelihood of benefit to endemic populations participating in clinical studies, the use of placebo, and the sustainability of post-trial access to study medications. In this article, we explore the basis of these concerns and suggest that the traditional development paradigm remains ethical under certain circumstances. We also consider alternative approaches that may be more attractive to sponsors as they either do not use placebo, or utilize populations in endemic countries who may unambiguously benefit. These approaches represent the way forward in the future, but are at present unproven in clinical practice, and face numerous regulatory, logistical and technical challenges. Consequently, in the short term, we argue that the traditional clinical development paradigm remains the most feasible approach and is ethical and consistent with the spirit of DH2000 under the appropriate circumstances.
Collapse
Affiliation(s)
- Geoffrey S Dow
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | | | | |
Collapse
|
41
|
Chico RM, Chandramohan D. Azithromycin plus chloroquine: combination therapy for protection against malaria and sexually transmitted infections in pregnancy. Expert Opin Drug Metab Toxicol 2011; 7:1153-67. [PMID: 21736423 PMCID: PMC3170143 DOI: 10.1517/17425255.2011.598506] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Introduction: The first-line therapy for the intermittent preventive treatment of malaria in pregnancy (IPTp) is sulphadoxine-pyrimethamine (SP). There is an urgent need to identify safe, well-tolerated and efficacious alternatives to SP due to widespread Plasmodium falciparum resistance. Combination therapy using azithromycin and chloroquine is one possibility that has demonstrated adequate parasitological response > 95% in clinical trials of non-pregnant adults in sub-Saharan Africa and where IPTp is a government policy in 33 countries. Areas covered: Key safety, tolerability and efficacy data are presented for azithromycin and chloroquine, alone and/or in combination, when used to prevent and/or treat P. falciparum, P. vivax, and several curable sexually transmitted and reproductive tract infections (STI/RTI). Pharmacokinetic evidence from pregnant women is also summarized for both compounds. Expert opinion: The azithromycin-chloroquine regimen that has demonstrated consistent efficacy in non-pregnant adults has been a 3-day course containing daily doses of 1 g of azithromycin and 600 mg base of chloroquine. The pharmacokinetic evidence of these compounds individually suggests that dose adjustments may not be necessary when used in combination for treatment efficacy against P. falciparum, P. vivax, as well as several curable STI/ RTI among pregnant women, although clinical confirmation will be necessary. Mass trachoma-treatment campaigns have shown that azithromycin selects for macrolide resistance in the pneumococcus, which reverses following the completion of therapy. Most importantly, no evidence to date suggests that azithromycin induces pneumococcal resistance to penicillin.
Collapse
Affiliation(s)
- R Matthew Chico
- London School of Hygiene and Tropical Medicine, Faculty of Infectious and Tropical Diseases, Disease Control Department, UK.
| | | |
Collapse
|
42
|
Tan KR, Magill AJ, Parise ME, Arguin PM. Doxycycline for malaria chemoprophylaxis and treatment: report from the CDC expert meeting on malaria chemoprophylaxis. Am J Trop Med Hyg 2011; 84:517-31. [PMID: 21460003 PMCID: PMC3062442 DOI: 10.4269/ajtmh.2011.10-0285] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Doxycycline, a synthetically derived tetracycline, is a partially efficacious causal prophylactic (liver stage of Plasmodium) drug and a slow acting blood schizontocidal agent highly effective for the prevention of malaria. When used in conjunction with a fast acting schizontocidal agent, it is also highly effective for malaria treatment. Doxycycline is especially useful as a prophylaxis in areas with chloroquine and multidrug-resistant Plasmodium falciparum malaria. Although not recommended for pregnant women and children < 8 years of age, severe adverse events are rarely reported for doxycycline. This report examines the evidence behind current recommendations for the use of doxycycline for malaria and summarizes the available literature on its safety and tolerability.
Collapse
Affiliation(s)
- Kathrine R Tan
- Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, 4770 Buford Hwy., Atlanta, GA 30341, USA.
| | | | | | | | | |
Collapse
|
43
|
Fernando SD, Rodrigo C, Rajapakse S. Chemoprophylaxis in malaria: drugs, evidence of efficacy and costs. ASIAN PAC J TROP MED 2011; 4:330-6. [PMID: 21771482 DOI: 10.1016/s1995-7645(11)60098-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 01/27/2010] [Accepted: 02/15/2011] [Indexed: 11/18/2022] Open
Abstract
This review concentrates on different aspects of malaria chemoprophylaxis, namely drug combinations, resistance, impact of malaria prevention in pregnancy and cost effectiveness. A MEDLINE search was performed for all articles with the key word 'Malaria' in the title field and 'Prophylaxis' in any field. The search was restricted to articles published in English within the last decade (1999-2009). Data sources included review articles published in core clinical journals, cohort studies, interventional studies, case control studies and cross sectional analyses. The mechanism of action, trial evidence of efficacy, side effects and geographical distribution of resistance is discussed for each prophylactic drug regimen. Impact of prophylaxis in pregnancy and the cost considerations are discussed under two separate sub topics.
Collapse
|
44
|
Bukvić Krajačić M, Perić M, Smith KS, Schönfeld ZI, Žiher D, Fajdetić A, Kujundžić N, Schönfeld W, Landek G, Padovan J, Jelić D, Ager A, Milhous WK, Ellis W, Spaventi R, Ohrt C. Synthesis, structure-activity relationship, and antimalarial activity of ureas and thioureas of 15-membered azalides. J Med Chem 2011; 54:3595-605. [PMID: 21476508 DOI: 10.1021/jm2001585] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Azithromycin, a first member of the azalide family of macrolides, while having substantial antimalarial activity, failed as a single agent for malaria prophylaxis. In this paper we present the first analogue campaign to identify more potent compounds from this class. Ureas and thioureas of 15-membered azalides, N''-substituted 9a-(N'-carbamoyl-β-aminoethyl), 9a-(N'-thiocarbamoyl-β-aminoethyl), 9a-[N'-(β-cyanoethyl)-N'-(carbamoyl-β-aminoethyl)], 9a-[N'-(β-cyanoethyl)-N'-(thiocarbamoyl-β-aminoethyl)], 9a-{N'-[β-(ethoxycarbonyl)ethyl]-N'(carbamoyl-β-aminoethyl)}, and 9a-[N'-(β-amidoethyl)-N'-(carbamoyl-β-aminoethyl)] of 9-deoxo-9-dihydro-9a-aza-9a-homoerythromycin A, were synthesized and their biological properties evaluated. The results obtained indicate a substantial improvement of the in vitro activity against P. falciparum (up to 88 times over azithromycin), particularly for compounds containing both sugars on the macrocyclic ring and aromatic moiety on 9a-position. The improved in vitro activity was not confirmed in the mouse model, likely due to an increase in lipophilicity of these analogues leading to a higher volume of distribution. Overall, with increased in vitro activity, promising PK properties, and modest in vivo efficacy, this series of molecules represents a good starting platform for the design of novel antimalarial azalides.
Collapse
Affiliation(s)
- Mirjana Bukvić Krajačić
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz Baruna Filipovića 29, 10000 Zagreb, Croatia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
BACKGROUND To prevent the development of drug resistance, the World Health Organization (WHO) recommends treating malaria with combination therapy. Azithromycin, an antibiotic with antimalarial properties, may be a useful additional option for antimalarial therapy. OBJECTIVES To compare the use of azithromycin alone or in combination with other antimalarial drugs with the use of alternative antimalarial drugs for treating uncomplicated malaria caused by Plasmodium falciparum or Plasmodium vivax. SEARCH STRATEGY We searched the Cochrane Infectious Diseases Group Specialized Register (August 2010); CENTRAL (The Cochrane Library Issue 3, 2010); MEDLINE (1966 to August 2010); EMBASE (1974 to August 2010); LILACS (August 2010); the metaRegister of Controlled Trials (mRCT, August 2010); conference proceedings; and reference lists. We also contacted researchers and a pharmaceutical company. SELECTION CRITERIA Randomized controlled trials comparing azithromycin, either alone or combined with another antimalarial drug, with another antimalarial drug used alone or combined with another antimalarial drug, or with azithromycin combined with another antimalarial drug if different combinations or doses of azithromycin were used. The primary outcome was treatment failure by day 28, defined as parasitological or clinical evidence of treatment failure between the start of treatment and day 28. Secondary outcomes included treatment failure by day 28 corrected for new infections confirmed by polymerase chain reaction (PCR), fever and parasite clearance time, and adverse events. DATA COLLECTION AND ANALYSIS Two people independently applied the inclusion criteria, extracted data and assessed methodological quality. We used risk ratio (RR) and 95% confidence intervals (CI). MAIN RESULTS Fifteen trials met the inclusion criteria (2284 participants, 69% males, 16% children). They were conducted in disparate malaria endemic areas, with the earlier studies conducted in Thailand (five) and India (two), and the more recent studies (eight) spread across three continents (South America, Africa, Asia). The 15 studies involved 41 treatment arms, 12 different drugs, and 28 different treatment regimens. Two studies examined P. vivax.Three-day azithromycin (AZ) monotherapy did not perform well for P. vivax or P. falciparum (Thailand: P. vivax failure rate 0.5 g daily, 56%, 95% CI 31 to 78. India: P. vivax failure rate 1 g daily,12%, 95% CI 7 to 21; P. falciparum failure rate 1 g daily, 64%, 95% CI 36 to 86.) A 1 g azithromycin and 0.6 g chloroquine combination daily for three days for uncomplicated P. falciparum infections was associated with increased treatment failure in India and Indonesia compared with the combination of sulphadoxine-pyrimethamine and chloroquine (pooled RR 2.66, 95% CI 1.25 to 5.67), and compared with the combination atovaquone-proguanil in a multicentre trial in Columbia and Surinam (RR 24.72, 95% CI 6.16 to 99.20). No increased risk of treatment failure was seen in two studies in Africa with mefloquine as the comparator drug (pooled RR 2.02, 95% CI 0.51 to 7.96, P = 0.3); the pooled RR for PCR-corrected data for the combination versus mefloquine was 1.01, 95% CI 0.18 to 5.84 (P = 1.0). An increased treatment failure risk was seen when comparing azithromycin in a dose of 1.2 to 1.5 mg in combination with artesunate (200 mg per day for three days) with artemether-lumefantrine (pooled RR 3.08, 95% CI 2.09 to 4.55; PCR-corrected pooled RR 3.63, 95% CI 2.02 to 6.52).Serious adverse events and treatment discontinuation were similar across treatment arms. More adverse events were reported when comparing the 1 g azithromycin/ 0.6 g chloroquine combination with mefloquine (pooled RR 1.20, 95% CI 1.06 to 1.36) or atovaquone-proguanil (RR 1.41, 95% CI 1.09 to1.83). AUTHORS' CONCLUSIONS Currently, there is no evidence for the superiority or equivalence of azithromycin monotherapy or combination therapy for the treatment of P. falciparum or P. vivax compared with other antimalarials or with the current first-line antimalarial combinations. The available evidence suggests that azithromycin is a weak antimalarial with some appealing safety characteristics. Unless the ongoing dose, formulation and product optimisation process results in a universally efficacious product, or a specific niche application is identified that is complementary to the current scala of more efficacious antimalarial combinations, azithromycin's future for the treatment of malaria does not look promising.
Collapse
Affiliation(s)
- Anna M van Eijk
- Liverpool School of Tropical MedicineChild & Reproductive Health GroupPembroke PlaceLiverpoolUKL3 5QA
| | - Dianne J Terlouw
- Liverpool School of Tropical MedicineChild & Reproductive Health GroupPembroke PlaceLiverpoolUKL3 5QA
| |
Collapse
|
46
|
Hutinec A, Rupčić R, Ziher D, Smith KS, Milhous W, Ellis W, Ohrt C, Schönfeld ZI. An automated, polymer-assisted strategy for the preparation of urea and thiourea derivatives of 15-membered azalides as potential antimalarial chemotherapeutics. Bioorg Med Chem 2011; 19:1692-701. [PMID: 21316974 DOI: 10.1016/j.bmc.2011.01.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2010] [Revised: 01/13/2011] [Accepted: 01/15/2011] [Indexed: 10/18/2022]
Abstract
A series of 15-membered azalide urea and thiourea derivatives has been synthesized and evaluated for their in vitro antimalarial activity against chloroquine-sensitive (D6), chloroquine/pyremethamine resistant (W2) and multidrug resistant (TM91C235) strains of Plasmodium falciparum. We have developed an effective automated synthetic strategy for the rapid synthesis of urea/thiourea libraries of a macrolide scaffold. Compounds have been synthesized using a solution phase strategy with overall yields of 50-80%. Most of the synthesized compounds had inhibitory effects. The top 10 compounds were 30-65 times more potent than azithromycin, an azalide with antimalarial activity, against all three strains.
Collapse
Affiliation(s)
- Antun Hutinec
- GlaxoSmithKline Research Centre Zagreb Ltd, Zagreb, Croatia.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Luntamo M, Kulmala T, Mbewe B, Cheung YB, Maleta K, Ashorn P. Effect of repeated treatment of pregnant women with sulfadoxine-pyrimethamine and azithromycin on preterm delivery in Malawi: a randomized controlled trial. Am J Trop Med Hyg 2011; 83:1212-20. [PMID: 21118924 DOI: 10.4269/ajtmh.2010.10-0264] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Preterm delivery, which is associated with infections during pregnancy, is common in sub-Saharan Africa. We enrolled 1,320 pregnant women into a randomized, controlled trial in Malawi to study whether preterm delivery and low birth weight (LBW) incidence can be reduced by intermittent preventive treatment of maternal malaria and reproductive tract infections. The participants received either sulfadoxine-pyrimethamine (SP) twice (controls), monthly SP, or monthly SP and two doses of azithromycin (AZI-SP). The incidence of preterm delivery was 17.9% in controls, 15.4% in the monthly SP group (P = 0.32), and 11.8% in AZI-SP group (risk ratio = 0.66, P = 0.01). Compared with controls, those in AZI-SP group had a risk ratio of 0.61 (P = 0.02) for LBW. Incidence of serious adverse events was low in all groups. In conclusion, the incidence of preterm delivery and LBW can in some conditions be reduced by treating pregnant women with monthly SP and two azithromycin doses.
Collapse
Affiliation(s)
- Mari Luntamo
- Department of International Health, University of Tampere Medical School, Tampere, Finland.
| | | | | | | | | | | |
Collapse
|
48
|
Stepanić V, Koštrun S, Malnar I, Hlevnjak M, Butković K, Ćaleta I, Dukši M, Kragol G, Makaruha-Stegić O, Mikac L, Ralić J, Tatić I, Tavčar B, Valko K, Zulfikari S, Munić V. Modeling Cellular Pharmacokinetics of 14- and 15-Membered Macrolides with Physicochemical Properties. J Med Chem 2011; 54:719-33. [DOI: 10.1021/jm101317f] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Višnja Stepanić
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Sanja Koštrun
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Ivica Malnar
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Mario Hlevnjak
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Kristina Butković
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Irena Ćaleta
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Marko Dukši
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Goran Kragol
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | | | - Lara Mikac
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Jovica Ralić
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Iva Tatić
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Branka Tavčar
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Klara Valko
- Analytical Chemistry, Molecular Discovery Research, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Herts SG1 2NY, U.K
| | | | - Vesna Munić
- GlaxoSmithKline Research Centre Zagreb Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
| |
Collapse
|
49
|
Salman S, Rogerson SJ, Kose K, Griffin S, Gomorai S, Baiwog F, Winmai J, Kandai J, Karunajeewa HA, O'Halloran SJ, Siba P, Ilett KF, Mueller I, Davis TME. Pharmacokinetic properties of azithromycin in pregnancy. Antimicrob Agents Chemother 2010; 54:360-6. [PMID: 19858250 PMCID: PMC2798488 DOI: 10.1128/aac.00771-09] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 09/17/2009] [Accepted: 10/19/2009] [Indexed: 11/20/2022] Open
Abstract
Azithromycin (AZI) is an azalide antibiotic with antimalarial activity that is considered safe in pregnancy. To assess its pharmacokinetic properties when administered as intermittent preventive treatment in pregnancy (IPTp), two 2-g doses were given 24 h apart to 31 pregnant and 29 age-matched nonpregnant Papua New Guinean women. All subjects also received single-dose sulfadoxine-pyrimethamine (SP) (1,500 mg or 75 mg) or chloroquine (450-mg base daily for 3 days). Blood samples were taken at 0, 1, 2, 3, 6, 12, 24, 32, 40, 48, and 72 h and on days 4, 5, 7, 10, and 14 for AZI assay by ultra-high-performance liquid chromatography-tandem mass spectrometry. The treatments were well tolerated. Using population pharmacokinetic modeling, a three-compartment model with zero-order followed by first-order absorption and no lag time provided the best fit. The areas under the plasma concentration-time curve (AUC(0-infinity)) (28.7 and 31.8 mg.h liter(-1) for pregnant and nonpregnant subjects, respectively) were consistent with the results of previous studies, but the estimated terminal elimination half-lives (78 and 77 h, respectively) were generally longer. The only significant relationship for a range of potential covariates, including malarial parasitemia, was with pregnancy, which accounted for an 86% increase in the volume of distribution of the central compartment relative to bioavailability without a significant change in the AUC(0-infinity). These data suggest that AZI can be combined with compounds with longer half-lives, such as SP, in combination IPTp without the need for dose adjustment.
Collapse
Affiliation(s)
- Sam Salman
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Stephen J. Rogerson
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Kay Kose
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Susan Griffin
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Servina Gomorai
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Francesca Baiwog
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Josephine Winmai
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Josin Kandai
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Harin A. Karunajeewa
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Sean J. O'Halloran
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Peter Siba
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Kenneth F. Ilett
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Ivo Mueller
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| | - Timothy M. E. Davis
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia, Faculty of Medicine, University of Melbourne, Melbourne, Australia, Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea, Western Health, Melbourne, Australia, Clinical Pharmacology and Toxicology Laboratory, Path West Laboratory Medicine, Nedlands, Australia
| |
Collapse
|
50
|
Aminzadeh A, Demircay Z, Ocak K, Soyletir G. Prevention of chronic furunculosis with low‐dose azithromycin. J DERMATOL TREAT 2009; 18:105-8. [PMID: 17520467 DOI: 10.1080/09546630601165125] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Chronic furunculosis is a recurrent staphylococcal abscess of the hair follicle. Besides ensuring personal hygiene, the management consists of long-term treatment with topical and systemic antibiotics. OBJECTIVE An open-labeled, prospective study was conducted to assess the clinical and in vitro efficacy of azithromycin in the long-term suppressive treatment of chronic furunculosis. METHODS Patients with a history of three or more episodes of furuncles were assigned to receive 12 weeks of suppressive treatment with azithromycin at a weekly dosage of 500 mg. In vitro susceptibility of azithromycin was evaluated with E-test. The primary efficacy parameter was complete absence of furuncles during the 3 months of azithromycin treatment. The secondary efficacy parameter was further absence of furuncles during the 3-month follow-up period. RESULTS At the end of 3 months of therapy, azithromycin was found to be effective in 19 (79.2%) of 24 patients; 18 of these patients remained in remission during the 3 months of follow-up. All of the strains were methicillin-sensitive. The results of the E-test showed that 15 of 18 strains (83.3%) were susceptible to azithromycin. CONCLUSION The results of this study indicate that azithromycin is an effective and safe alternative in the treatment of chronic furunculosis caused by methicillin-sensitive Staphylococcus aureus.
Collapse
Affiliation(s)
- Arya Aminzadeh
- Departments of Dermatology, Marmara University School of Medicine, Istanbul, Turkey.
| | | | | | | |
Collapse
|