1
|
Gebrie H, Yimer M, Ayehu A, Mohammed H, Hailgiorgis H, Wuletaw Y, Hailu M, Tolera G, Tasew G, Kassa M, Gidey B. Efficacy and safety of chloroquine plus primaquine for the treatment of Plasmodium vivax malaria in Hamusit site, Northwestern Ethiopia. Malar J 2024; 23:202. [PMID: 38971786 PMCID: PMC11227712 DOI: 10.1186/s12936-024-05031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/28/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Plasmodium vivax malaria is still an important public health problem in Ethiopia. Unlike Plasmodium falciparum, P. vivax has a dormant liver stage (hypnozoite) that can be a risk of recurrent vivax malaria unless treated by radical cure with primaquine. Drug resistance to chloroquine is threatening malaria control and elimination efforts. This study assessed the therapeutic efficacy and safety of chloroquine plus 14 days of primaquine on P. vivax infection based on parasitological, clinical, and haematological parameters. METHODS A single-arm in vivo prospective therapeutic efficacy study was conducted to assess the clinical and parasitological response to the first-line treatment of P. vivax in Ethiopia, chloroquine plus 14 days low dose of (0.25 mg/kg/day) primaquine between December 2022 and March 2023 at Hamusit Health Centre using the standard World Health Organization (WHO) protocol. A total of 100 study participants with P. vivax mono-infection who were over 6 months old were enrolled and monitored for adequate clinical and parasitological responses for 42 days. The WHO double-entry Excel sheet and SPSS v.25 software were used for Kaplan-Meier survival analysis, and a paired t-test was used for analysis of haemoglobin improvements between follow up days. RESULTS A total of 100 patients were enrolled among those, 96% cases were rural residents, 93% had previous malaria exposure, and predominant age group was 5-15 years (61%). 92.6% (95% CI 85.1-96.4%) of enrolled patients were adequate clinical and parasitological response, and 7.4% (95% CI 3.6-14.9%) recurrences were observed among treated patients. The fever and parasite clearance rate on day 3 were 98% and 94%, respectively. The baseline haemoglobin levels improved significantly compared to those days 14 and 42 (p < 0.001). No serious adverse event was observed during the study period. CONCLUSIONS In this study, co-administration of chloroquine with primaquine was efficacious and well-tolerated with fast resolution of fever and high parasites clearance rate. However, the 7.4% failure is reported is alarming that warrant further monitoring of the therapeutic efficacy study of P. vivax.
Collapse
Affiliation(s)
- Habtamu Gebrie
- Department of Medical Laboratory Science College of Medicine and Health Science, Dilla University, Dilla, Ethiopia.
| | - Mulat Yimer
- Department of Medical Laboratory Science College of Medicine and Health Science, Bahir Dar University, Bahir Dar, Ethiopia
| | - Animen Ayehu
- Department of Medical Laboratory Science College of Medicine and Health Science, Bahir Dar University, Bahir Dar, Ethiopia
| | | | | | - Yonas Wuletaw
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Mesay Hailu
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | | | - Geremew Tasew
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Mogess Kassa
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | | |
Collapse
|
2
|
Sridapan T, Rattanakoch P, Kijprasong K, Srisutham S. Drug resistance markers in Plasmodium vivax isolates from a Kanchanaburi province, Thailand between January to May 2023. PLoS One 2024; 19:e0304337. [PMID: 38968216 PMCID: PMC11226124 DOI: 10.1371/journal.pone.0304337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 05/10/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Plasmodium vivax has become the predominant species in the border regions of Thailand. The emergence and spread of antimalarial drug resistance in P. vivax is one of the significant challenges for malaria control. Continuous surveillance of drug resistance is therefore necessary for monitoring the development of drug resistance in the region. This study aims to investigate the prevalence of the mutation in the P. vivax multidrug resistant 1 (Pvmdr1), dihydrofolate reductase (Pvdhfr), and dihydropteroate synthetase (Pvdhps) genes conferred resistance to chloroquine (CQ), pyrimethamine (P) and sulfadoxine (S), respectively. METHOD 100 P. vivax isolates were obtained between January to May 2023 from a Kanchanaburi province, western Thailand. Nucleotide sequences of Pvmdr1, Pvdhfr, and Pvdhps genes were amplified and sequenced. The frequency of single nucleotide polymorphisms (SNPs)-haplotypes of drug-resistant alleles was assessed. The linkage disequilibrium (LD) tests were also analyzed. RESULTS In Pvmdr1, T958M, Y976F, and F1076L, mutations were detected in 100%, 21%, and 23% of the isolates, respectively. In Pvdhfr, the quadruple mutant allele (I57R58M61T117) prevailed in 84% of the samples, followed by (L57R58M61T117) in 11%. For Pvdhps, the double mutant allele (G383G553) was detected (48%), followed by the triple mutant allele (G383M512G553) (47%) of the isolates. The most prevalent combination of Pvdhfr (I57R58M61T117) and Pvdhps (G383G553) alleles was sextuple mutated haplotypes (48%). For LD analysis, the association in the SNPs pairs was found between the intragenic and intergenic regions of the Pvdhfr and Pvdhps genes. CONCLUSION The study has recently updated the high prevalence of three gene mutations associated with CQ and SP resistance. Genetic monitoring is therefore important to intensify in the regions to further assess the spread of drug resistant. Our data also provide evidence on the distribution of drug resistance for the early warning system, thereby threatening P. vivax malaria treatment policy decisions at the national level.
Collapse
Affiliation(s)
- Thanawat Sridapan
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Paweesuda Rattanakoch
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | | | - Suttipat Srisutham
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
3
|
Christensen P, Cinzah R, Suwanarusk R, Chua ACY, Kaneko O, Kyle DE, Aung HL, Matheson J, Bifani P, Rénia L, Cook GM, Snounou G, Russell B. Extended blood stage sensitivity profiles of Plasmodium cynomolgi to doxycycline and tafenoquine, as a model for Plasmodium vivax. Antimicrob Agents Chemother 2024; 68:e0028024. [PMID: 38587391 PMCID: PMC11064600 DOI: 10.1128/aac.00280-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 03/15/2024] [Indexed: 04/09/2024] Open
Abstract
Testing Plasmodium vivax antimicrobial sensitivity is limited to ex vivo schizont maturation assays, which preclude determining the IC50s of delayed action antimalarials such as doxycycline. Using Plasmodium cynomolgi as a model for P. vivax, we determined the physiologically significant delayed death effect induced by doxycycline [IC50(96 h), 1,401 ± 607 nM]. As expected, IC50(96 h) to chloroquine (20.4 nM), piperaquine (12.6 µM), and tafenoquine (1,424 nM) were not affected by extended exposure.
Collapse
Affiliation(s)
- Peter Christensen
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Rosy Cinzah
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Rossarin Suwanarusk
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Adeline Chiew Yen Chua
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), , Singapore
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Dennis E. Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Htin Lin Aung
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Jessica Matheson
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Pablo Bifani
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Laurent Rénia
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), , Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Gregory M. Cook
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Georges Snounou
- 11-Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Fontenay-aux-Roses & Kremlin- Bicêtre, Le Kremlin-Bicêtre, France
| | - Bruce Russell
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| |
Collapse
|
4
|
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
|
5
|
Mushtaq I, Sarwar MS, Chaudhry A, Shah SAH, Ahmad MM. Updates on traditional methods for combating malaria and emerging Wolbachia-based interventions. Front Cell Infect Microbiol 2024; 14:1330475. [PMID: 38716193 PMCID: PMC11074371 DOI: 10.3389/fcimb.2024.1330475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/14/2024] [Indexed: 05/24/2024] Open
Abstract
The escalating challenge of malaria control necessitates innovative approaches that extend beyond traditional control strategies. This review explores the incorporation of traditional vector control techniques with emerging Wolbachia-based interventions. Wolbachia, a naturally occurring bacteria, offers a novel approach for combatting vector-borne diseases, including malaria, by reducing the mosquitoes' ability to transmit these diseases. The study explores the rationale for this integration, presenting various case studies and pilot projects that have exhibited significant success. Employing a multi-dimensional approach that includes community mobilization, environmental modifications, and new biological methods, the paper posits that integrated efforts could mark a turning point in the struggle against malaria. Our findings indicate that incorporating Wolbachia-based strategies into existing vector management programs not only is feasible but also heightens the efficacy of malaria control initiatives in different countries especially in Pakistan. The paper concludes that continued research and international collaboration are imperative for translating these promising methods from the laboratory to the field, thereby offering a more sustainable and effective malaria control strategy.
Collapse
|
6
|
Obaldía N, Da Silva Filho JL, Núñez M, Glass KA, Oulton T, Achcar F, Wirjanata G, Duraisingh M, Felgner P, Tetteh KK, Bozdech Z, Otto TD, Marti M. Sterile protection against P. vivax malaria by repeated blood stage infection in the Aotus monkey model. Life Sci Alliance 2024; 7:e202302524. [PMID: 38158220 PMCID: PMC10756917 DOI: 10.26508/lsa.202302524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024] Open
Abstract
The malaria parasite Plasmodium vivax remains a major global public health challenge, and no vaccine is approved for use in humans. Here, we assessed whether P. vivax strain-transcendent immunity can be achieved by repeated infection in Aotus monkeys. Sterile immunity was achieved after two homologous infections, whereas subsequent heterologous challenge provided only partial protection. IgG levels based on P. vivax lysate ELISA and protein microarray increased with repeated infections and correlated with the level of homologous protection. Parasite transcriptional profiles provided no evidence of major antigenic switching upon homologous or heterologous challenge. However, we observed significant sequence diversity and transcriptional differences in the P. vivax core gene repertoire between the two strains used in the study, suggesting that partial protection upon heterologous challenge is due to molecular differences between strains rather than immune evasion by antigenic switching. Our study demonstrates that sterile immunity against P. vivax can be achieved by repeated homologous blood stage infection in Aotus monkeys, thus providing a benchmark to test the efficacy of candidate blood stage P. vivax malaria vaccines.
Collapse
Affiliation(s)
- Nicanor Obaldía
- Departamento de Investigaciones en Parasitologia, Instituto Conmemorativo Gorgas de Estudios de la Salud, Panamá City, Republic of Panamá
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Harvard University, Boston, MA, USA
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Joao Luiz Da Silva Filho
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Marlon Núñez
- Departamento de Investigaciones en Parasitologia, Instituto Conmemorativo Gorgas de Estudios de la Salud, Panamá City, Republic of Panamá
| | - Katherine A Glass
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Tate Oulton
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Fiona Achcar
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Grennady Wirjanata
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Manoj Duraisingh
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Philip Felgner
- Institute for Immunology, University of California, Irvine, CA, USA
| | - Kevin Ka Tetteh
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Thomas D Otto
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Matthias Marti
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Harvard University, Boston, MA, USA
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Pookmanee W, Thongthip S, Mungthin M, Sukasem C, Tankanitlert J, Chariyavilaskul P, Wittayalertpanya S. An increase in urinary primaquine and a reduction in urinary primaquine-5,6-orthoquinone in the Thai population with CYP2D6 reduced enzyme function. Heliyon 2024; 10:e24351. [PMID: 38293439 PMCID: PMC10827494 DOI: 10.1016/j.heliyon.2024.e24351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/27/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
Objectives Primaquine is metabolized by the cytochrome P450-2D6 enzyme (CYP2D6) to an active primaquine-5,6-orthoquinone (POQ). No relationships of CYP2D6 polymorphisms with the pharmacokinetics of primaquine and POQ were reported in the Thai population. Methods We evaluated the genetic distribution of CYP2D6 in 345 Thai army populations together with the pharmacokinetic profiles of primaquine and POQ in plasma and urine (n = 44, descriptive data are presented in median (range)). All dose-related pharmacokinetic parameters were normalized by primaquine dose per body weight before statistical analysis. Results CYP2D6*10 was the allele observed with the highest frequency (56.62%) corresponding to CYP2D6*10/*10 (32.94%) and CYP2D6*1/*10 (27.94%) genotypes. CYP2D6 intermediate metabolizers (CYP2D6 IM) were found in 44.41% of the cohort and had an increase in the cumulative amount of primaquine excreted (CAE) in urine compared to normal metabolizers of CYP2D6 (CYP2D6 NM); (CYP2D6 IM vs. CYP2D6 NM: 2444 (1697-3564) vs. 1757 (1092-2185) μg/mg/kg, p = 0.039), a reduction in urine CAE of POQ (CYP2D6 IM vs CYP2D6 NM: 115 (46-297) vs. 318 (92-498) μg/mg/kg, p = 0.047) and a reduction in the POQ/primaquine CAE ratio in urine (CYP2D6 IM vs. CYP2D6 NM: 0.06 (0.01-0.11) vs. 0.16 (0.06-0.26), p = 0.009). No significant differences were found in the pharmacokinetic profiles of plasma primaquine and POQ. Conclusions The CYP2D6 polymorphisms influenced the changes in primaquine and POQ that were noticeable in the urine, supporting the role of the CYP2D6 gene testing before drug administration.
Collapse
Affiliation(s)
- Waritda Pookmanee
- Interdisciplinary Program in Pharmacology, Graduate School, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Clinical Pharmacokinetics and Pharmacogenomics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Medical Depot Division, Royal Thai Army Medical Department, Bangkok, Thailand
| | - Siriwan Thongthip
- Maha Chakri Sirindhorn Clinical Research Center under the Royal Patronage, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Mathirut Mungthin
- Department of Pharmacology, Phramongkutklao College of Medicine, Bangkok, Thailand
| | - Chonlaphat Sukasem
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center, Ramathibodi Hospital, Bangkok, Thailand
- Pharmacogenomics and Precision Medicine, The Preventive Genomics & Family Check-up Services Center, Bumrungrad International Hospital, Bangkok, Thailand
| | | | - Pajaree Chariyavilaskul
- Center of Excellence in Clinical Pharmacokinetics and Pharmacogenomics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supeecha Wittayalertpanya
- Center of Excellence in Clinical Pharmacokinetics and Pharmacogenomics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
8
|
Salimo ZM, Barros AL, Adrião AAX, Rodrigues AM, Sartim MA, de Oliveira IS, Pucca MB, Baia-da-Silva DC, Monteiro WM, de Melo GC, Koolen HHF. Toxins from Animal Venoms as a Potential Source of Antimalarials: A Comprehensive Review. Toxins (Basel) 2023; 15:375. [PMID: 37368676 DOI: 10.3390/toxins15060375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Malaria is an infectious disease caused by Plasmodium spp. and it is mainly transmitted to humans by female mosquitoes of the genus Anopheles. Malaria is an important global public health problem due to its high rates of morbidity and mortality. At present, drug therapies and vector control with insecticides are respectively the most commonly used methods for the treatment and control of malaria. However, several studies have shown the resistance of Plasmodium to drugs that are recommended for the treatment of malaria. In view of this, it is necessary to carry out studies to discover new antimalarial molecules as lead compounds for the development of new medicines. In this sense, in the last few decades, animal venoms have attracted attention as a potential source for new antimalarial molecules. Therefore, the aim of this review was to summarize animal venom toxins with antimalarial activity found in the literature. From this research, 50 isolated substances, 4 venom fractions and 7 venom extracts from animals such as anurans, spiders, scorpions, snakes, and bees were identified. These toxins act as inhibitors at different key points in the biological cycle of Plasmodium and may be important in the context of the resistance of Plasmodium to currently available antimalarial drugs.
Collapse
Affiliation(s)
- Zeca M Salimo
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus 69040-000, Brazil
- Grupo de Pesquisa em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
| | - André L Barros
- Setor de Medicina Veterinária, Universidade Nilton Lins, Manaus 69058-030, Brazil
| | - Asenate A X Adrião
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Grupo de Pesquisa em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia-Rede BIONORTE, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
| | - Aline M Rodrigues
- Grupo de Pesquisa em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
| | - Marco A Sartim
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia-Rede BIONORTE, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
- Pro-Reitoria de Pesquisa e Pós-Graduação, Universidade Nilton Lins, Manaus 69058-030, Brazil
| | - Isadora S de Oliveira
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-903, Brazil
| | - Manuela B Pucca
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Faculdade de Medicina, Universidade Federal de Roraima, Boa Vista 69317-810, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Roraima, Boa Vista 69317-810, Brazil
| | - Djane C Baia-da-Silva
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus 69040-000, Brazil
- Faculdade de Farmácia, Universidade Nilton Lins, Manaus 69058-030, Brazil
- Instituto Leônidas e Maria Deane, Fundação Oswaldo Cruz, Manaus 69057-070, Brazil
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Federal do Amazonas, Manaus 69080-900, Brazil
| | - Wuelton M Monteiro
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus 69040-000, Brazil
| | - Gisely C de Melo
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus 69040-000, Brazil
| | - Hector H F Koolen
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Grupo de Pesquisa em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia-Rede BIONORTE, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
| |
Collapse
|
9
|
Mekonnen DA, Abadura GS, Behaksra SW, Taffese HS, Bayissa GA, Bulto MG, Tessema TS, Tadesse FG, Gadisa E. Treatment of uncomplicated Plasmodium vivax with chloroquine plus radical cure with primaquine without G6PDd testing is safe in Arba Minch, Ethiopia: assessment of clinical and parasitological response. Malar J 2023; 22:135. [PMID: 37098510 PMCID: PMC10131480 DOI: 10.1186/s12936-023-04562-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/15/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND Ethiopia rolled out primaquine nationwide in 2018 for radical cure along with chloroquine for the treatment of uncomplicated Plasmodium vivax malaria in its bid for malaria elimination by 2030. The emergence of anti-malarial drug resistance would challenge the elimination goal. There is limited evidence on the emergence of chloroquine drug resistance. The clinical and parasitological outcomes of treatment of P. vivax with chloroquine plus radical cure using low dose 14 days primaquine were assessed in an endemic area of Ethiopia. METHODS A semi-directly observed 42-days follow up in-vivo therapeutic efficacy study was conducted from October 2019 to February 2020. Plasmodium vivax mono-species infected patients (n = 102) treated with a 14 days low dose (0.25 mg/kg body weight per day) primaquine plus chloroquine (a total dose of 25 mg base/kg for 3 days) were followed for 42 days to examine clinical and parasitological outcomes. Samples collected at recruitment and days of recurrence were examined by 18 S based nested polymerase chain reaction (nPCR) and Pvmsp3α nPCR-restriction fragment length polymorphism. Asexual parasitaemia and the presence of gametocytes were assessed on the scheduled days using microscopy. Clinical symptoms, haemoglobin levels, and Hillmen urine test were also assessed. RESULTS Of the 102 patients followed in this study, no early clinical and parasitological failure was observed. All patients had adequate clinical and parasitological responses within the 28 days of follow up. Late clinical (n = 3) and parasitological (n = 6) failures were observed only after day 28. The cumulative incidence of failure was 10.9% (95% confidence interval, 5.8-19.9%) on day 42. Among the paired recurrent samples, identical clones were detected only in two samples on day 0 and day of recurrences (day 30 and 42) using Pvmsp3α genotyping. No adverse effect was detected related to the low dose 14 days primaquine administrations. CONCLUSION Co-administration of CQ with PQ in the study area is well tolerated and there was no recurrence of P. vivax before 28 days of follow up. Interpretation of CQ plus PQ efficacy should be done with caution especially when the recurrent parasitaemia occurs after day 28. Therapeutic efficacy studies with appropriate design might be informative to rule out chloroquine or primaquine drug resistance and/or metabolism in the study area.
Collapse
Affiliation(s)
- Daniel Abebe Mekonnen
- Malaria and Neglected Tropical Diseases Directorate, Armauer Hansen Research Institute, 1005, Addis Ababa, Ethiopia.
- Institute of Biotechnology, Addis Ababa University, 1176, Addis Ababa, Ethiopia.
| | - Girma Shumie Abadura
- Malaria and Neglected Tropical Diseases Directorate, Armauer Hansen Research Institute, 1005, Addis Ababa, Ethiopia
| | - Sinknesh Wolde Behaksra
- Malaria and Neglected Tropical Diseases Directorate, Armauer Hansen Research Institute, 1005, Addis Ababa, Ethiopia
| | | | | | - Mikiyas Gebremichael Bulto
- Malaria and Neglected Tropical Diseases Directorate, Armauer Hansen Research Institute, 1005, Addis Ababa, Ethiopia
| | | | - Fitsum G Tadesse
- Malaria and Neglected Tropical Diseases Directorate, Armauer Hansen Research Institute, 1005, Addis Ababa, Ethiopia
| | - Endalamaw Gadisa
- Malaria and Neglected Tropical Diseases Directorate, Armauer Hansen Research Institute, 1005, Addis Ababa, Ethiopia
| |
Collapse
|
10
|
Bai J, Liu F, Yang F, Zhao Y, Jia X, Thongpoon S, Roobsoog W, Sattabongkot J, Zheng L, Cui Z, Zheng W, Cui L, Cao Y. Evaluation of transmission-blocking potential of Pv22 using clinical Plasmodium vivax infections and transgenic Plasmodium berghei. Vaccine 2023; 41:555-563. [PMID: 36503858 PMCID: PMC9812905 DOI: 10.1016/j.vaccine.2022.11.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 11/09/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022]
Abstract
Antigens expressed during the sexual development of malaria parasites are transmission-blocking vaccine (TBV) targets. Pb22, a protein expressed and localized to the plasma membrane of gametes and ookinetes in Plasmodium berghei, is an excellent TBV candidate. Here, we evaluated the TB potential of the Plasmodium vivax ortholog Pv22 using a transgenic P. berghei parasite line and P. vivax clinical isolates. The full-length recombinant Pv22 (rPv22) protein was produced and used to immunize mice and rabbits to obtain antibodies. We generated a transgenic P. berghei line (TrPv22Pb) by inserting the pv22 gene into the pb22 locus and showed that Pv22 expression completely rescued the defects in male gametogenesis of the pb22 deletion parasite. Since Pv22 in the transgenic parasite showed similar expression and localization patterns to Pb22, we used the TrPv22Pb parasite as a surrogate to evaluate the TB potential of Pv22. In mosquito feeding assays, mosquitoes feeding on rPv22-immunized mice infected with TrPv22Pb parasites showed a 49.3-53.3 % reduction in the oocyst density compared to the control group. In vitro assays showed that the rPv22 immune sera significantly inhibited exflagellation and ookinete formation of the TrPv22Pb parasites. In a direct membrane feeding assay using three clinical P. vivax isolates, the rabbit anti-rPv22 antibodies also significantly decreased the oocyst density by 53.7, 30.2, and 26.2 %, respectively. This study demonstrated the feasibility of using transgenic P. berghei parasites expressing P. vivax antigens as a potential tool to evaluate TBV candidates. However, the much weaker TB activity of Pv22 obtained from two complementary assays suggest that Pv22 may not be a promising TBV candidate for P. vivax.
Collapse
Affiliation(s)
- Jie Bai
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Fei Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Fan Yang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xitong Jia
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Sataporn Thongpoon
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
| | - Wanlapa Roobsoog
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
| | - Li Zheng
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Zeshi Cui
- College of Pharmacy, China Medical University, Shenyang, China
| | - Wenqi Zheng
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolian Medical University, Hohhot, China
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
11
|
Fabbri C, Quaresma Ramos G, Clarys Baia-da-Silva D, Oliveira Trindade A, Carlos Salazar-Alvarez L, Costa Ferreira Neves J, dos Santos Bastos I, Guimarães Costa A, Vinicius Guimarães Lacerda M, Marcelo Monteiro W, Trindade Maranhão Costa F, Costa Pinto Lopes S. The activity of methylene blue against asexual and sexual stages of Plasmodium vivax. Front Cell Infect Microbiol 2023; 13:1108366. [PMID: 37143740 PMCID: PMC10152470 DOI: 10.3389/fcimb.2023.1108366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 03/07/2023] [Indexed: 05/06/2023] Open
Abstract
Methylene blue (MB) is an alternative for combating drug-resistant malaria parasites. Its transmission-blocking potential has been demonstrated in vivo in murine models, in vitro, and in clinical trials. MB shows high efficacy against Plasmodium vivax asexual stages; however, its efficacy in sexual stages is unknown. In this study, we evaluated the potential of MB against asexual and sexual forms of P. vivax isolated from the blood of patients residing in the Brazilian Amazon. An ex vivo schizont maturation assay, zygote to ookinete transformation assay, direct membrane feed assay (DMFA), and standard membrane feed assay (SMFA) using P. vivax gametocytes with MB exposure were performed. A cytotoxicity assay was also performed on freshly collected peripheral blood mononuclear cells (PBMCs) and the hepatocyte carcinoma cell line HepG2. MB inhibited the P. vivax schizont maturation and demonstrated an IC50 lower than that of chloroquine (control drug). In the sexual forms, the MB demonstrated a high level of inhibition in the transformation of the zygotes into ookinetes. In the DMFA, MB did not considerably affect the infection rate and showed low inhibition, but it demonstrated a slight decrease in the infection intensity in all tested concentrations. In contrast, in the SMFA, MB was able to completely block the transmission at the highest concentration (20 µM). MB demonstrated low cytotoxicity to fresh PBMCs but demonstrated higher cytotoxicity to the hepatocyte carcinoma cell line HepG2. These results show that MB may be a potential drug for vivax malaria treatment.
Collapse
Affiliation(s)
- Camila Fabbri
- Instituto Leônidas & Maria Deane, Fiocruz Amazônia, Manaus, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- *Correspondence: Camila Fabbri, ; Stefanie Costa Pinto Lopes,
| | - Glenda Quaresma Ramos
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Escola Superior de Ciências da Saúde, Centro Multiusuário para Análise de Fenômenos Biomédicos da Universidade do Estado do Amazonas, Universidade do Estado do Amazonas, Manaus, Brazil
- Departamento de Morfologia, Universidade Federal do Amazonas, Manaus, Brazil
| | - Djane Clarys Baia-da-Silva
- Instituto Leônidas & Maria Deane, Fiocruz Amazônia, Manaus, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Departamento de Saúde Coletiva, Universidade Federal do Amazonas, Manaus, Brazil
- Faculdade de Farmácia, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
| | | | - Luis Carlos Salazar-Alvarez
- Departamento de Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas, Campinas, Brazil
| | - Juliana Costa Ferreira Neves
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
| | - Ivanildes dos Santos Bastos
- Instituto Leônidas & Maria Deane, Fiocruz Amazônia, Manaus, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Allyson Guimarães Costa
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas, Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Brazil
| | - Marcus Vinicius Guimarães Lacerda
- Instituto Leônidas & Maria Deane, Fiocruz Amazônia, Manaus, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
| | - Wuelton Marcelo Monteiro
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
| | | | - Stefanie Costa Pinto Lopes
- Instituto Leônidas & Maria Deane, Fiocruz Amazônia, Manaus, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- *Correspondence: Camila Fabbri, ; Stefanie Costa Pinto Lopes,
| |
Collapse
|
12
|
Mohammed H, Sime H, Hailgiorgis H, Chernet M, Alebachew M, Solomon H, Assefa G, Haile M, Girma S, Bekele W, Tasew G, Gidey B, Commons RJ, Assefa A. Efficacy and safety of pyronaridine-artesunate (Pyramax ®) for the treatment of uncomplicated Plasmodium vivax malaria in Northwest Ethiopia. Malar J 2022; 21:401. [PMID: 36587210 PMCID: PMC9805271 DOI: 10.1186/s12936-022-04422-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/19/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Declining efficacy of chloroquine for the treatment Plasmodium vivax malaria has been reported in different endemic settings in Ethiopia. This highlights the need to assess alternative options for P. vivax treatment with artemisinin-based combination therapy, such as pyronaridine-artesunate. This treatment regimen has shown high efficacy for uncomplicated malaria in both Africa and Asia. However, limited data are available from Ethiopia. This study was conducted to assess the efficacy and safety of pyronaridine-artesunate for the treatment of uncomplicated P. vivax malaria in Northwest Ethiopia. METHODS A single arm prospective efficacy study was conducted in the Hamusite area, Northwest Ethiopia. Fifty-one febrile adult patients with uncomplicated P. vivax malaria were enrolled between March and July 2021. Patients were treated with pyronaridine-artesunate once daily for three days. Clinical and parasitological parameters were monitored over a 42-day follow-up period using the standard World Health Organization protocol for therapeutic efficacy studies. RESULTS A total of 4372 febrile patients were screened with 51 patients enrolled and 49 completing the 42-day follow-up period. The PCR-uncorrected adequate clinical and parasitological response (ACPR) was 95.9% (47/49; 95% CI 84.9-99.0) on day 42. Two patients had recurrences [4.0% (2/49); 95% CI 0.7-12.1] on days 35 and 42. The parasite clearance rate was rapid with fast resolution of clinical symptoms; 100% of participants had cleared parasitaemia on day 1 and fever on day 2. All 16 (31.4%) patients with gametocyte carriage on day 0 had cleared by day 1. There were no serious adverse events. CONCLUSION In this small study, pyronaridine-artesunate was efficacious and well-tolerated for the treatment of uncomplicated P. vivax malaria. In adults in the study setting, it would be a suitable alternative option for case management.
Collapse
Affiliation(s)
- Hussein Mohammed
- Bacterial, Parasitic and Zoonotic Diseases Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia.
| | - Heven Sime
- Bacterial, Parasitic and Zoonotic Diseases Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Henok Hailgiorgis
- Bacterial, Parasitic and Zoonotic Diseases Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Melkie Chernet
- Bacterial, Parasitic and Zoonotic Diseases Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Mihreteab Alebachew
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Hiwot Solomon
- National Malaria Elimination Program, Ministry of Health, Addis Ababa, Ethiopia
| | - Gudissa Assefa
- National Malaria Elimination Program, Ministry of Health, Addis Ababa, Ethiopia
| | - Mebrahtom Haile
- National Malaria Elimination Program, Ministry of Health, Addis Ababa, Ethiopia
| | | | - Worku Bekele
- World Health Organization, Addis Ababa, Ethiopia
| | - Geremew Tasew
- Bacterial, Parasitic and Zoonotic Diseases Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Bokretsion Gidey
- Bacterial, Parasitic and Zoonotic Diseases Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Robert J Commons
- Global Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
- General and Subspecialty Medicine, Grampians Health, Ballarat, Australia
| | - Ashenafi Assefa
- Bacterial, Parasitic and Zoonotic Diseases Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, USA
| |
Collapse
|
13
|
Kaur D, Sinha S, Sehgal R. Global scenario of Plasmodium vivax occurrence and resistance pattern. J Basic Microbiol 2022; 62:1417-1428. [PMID: 36125207 DOI: 10.1002/jobm.202200316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/20/2022] [Accepted: 09/04/2022] [Indexed: 11/06/2022]
Abstract
Malaria caused by Plasmodium vivax is comparatively less virulent than Plasmodium falciparum, which can also lead to severe disease and death. It shows a wide geographical distribution. Chloroquine serves as a drug of choice, with primaquine as a radical cure. However, with the appearance of resistance to chloroquine and treatment has been shifted to artemisinin combination therapy followed by primaquine as a radical cure. Sulphadoxine-pyrimethamine, mefloquine, and atovaquone-proguanil are other drugs of choice in chloroquine-resistant areas, and later resistance was soon reported for these drugs also. The emergence of drug resistance serves as a major hurdle to controlling and eliminating malaria. The discovery of robust molecular markers and regular surveillance for the presence of mutations in malaria-endemic areas would serve as a helpful tool to combat drug resistance. Here, in this review, we will discuss the endemicity of P. vivax, a historical overview of antimalarial drugs, the appearance of drug resistance and molecular markers with their global distribution along with different measures taken to reduce malaria burden due to P. vivax infection and their resistance.
Collapse
Affiliation(s)
- Davinder Kaur
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shweta Sinha
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rakesh Sehgal
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
14
|
Brenda CT, Norma RF, Marcela RL, Nelly LV, Teresa I F. Action mechanisms of metallic compounds on Plasmodium spp. J Trace Elem Med Biol 2022; 73:127028. [PMID: 35797926 DOI: 10.1016/j.jtemb.2022.127028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/10/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Malaria is a parasitic disease with the highest morbidity and mortality worldwide. Unfortunately, during the last decades, the causal agent, Plasmodium spp., has developed resistance to chloroquine and artemisinin. For this reason, metallic compounds have been proposed as an optional treatment since they have shown a potential antimalarial effect with diverse action mechanisms in the parasite and the host. OBJECTIVE To show the possible targets of metallic compounds in Plasmodium spp. CONCLUSION The metallic compounds are an option attractive to treatment for the malaria, for its low cost and its great activity to reduce parasitemia; however is necessary more studies principally in vivo in order to know the interactions that it can have in an experimental model.
Collapse
Affiliation(s)
- Casarrubias-Tabarez Brenda
- Departamento de Biología Celular y TIsular, Facultad de Medicina, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, C.P. 04510 Mexico City, Mexico.
| | - Rivera-Fernández Norma
- Departamento de Microbiología y Parasitología, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, C.P. 04510 Mexico City, Mexico.
| | - Rojas-Lemus Marcela
- Departamento de Biología Celular y TIsular, Facultad de Medicina, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, C.P. 04510 Mexico City, Mexico.
| | - López-Valdez Nelly
- Departamento de Biología Celular y TIsular, Facultad de Medicina, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, C.P. 04510 Mexico City, Mexico.
| | - Fortoul Teresa I
- Departamento de Biología Celular y TIsular, Facultad de Medicina, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, C.P. 04510 Mexico City, Mexico.
| |
Collapse
|
15
|
Valenciano AL, Gomez-Lorenzo MG, Vega-Rodríguez J, Adams JH, Roth A. In vitro models for human malaria: targeting the liver stage. Trends Parasitol 2022; 38:758-774. [PMID: 35780012 PMCID: PMC9378454 DOI: 10.1016/j.pt.2022.05.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
The Plasmodium liver stage represents a vulnerable therapeutic target to prevent disease progression as the parasite resides in the liver before clinical representation caused by intraerythrocytic development. However, most antimalarial drugs target the blood stage of the parasite's life cycle, and the few drugs that target the liver stage are lethal to patients with a glucose-6-phosphate dehydrogenase deficiency. Furthermore, implementation of in vitro liver models to study and develop novel therapeutics against the liver stage of human Plasmodium species remains challenging. In this review, we focus on the progression of in vitro liver models developed for human Plasmodium spp. parasites, provide a brief review on important assay requirements, and lastly present recommendations to improve models to enhance the discovery process of novel preclinical therapeutics.
Collapse
Affiliation(s)
- Ana Lisa Valenciano
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA; Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Maria G Gomez-Lorenzo
- Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
16
|
Zhang X, Wei H, Zhang Y, Zhao Y, Wang L, Hu Y, Nguitragool W, Sattabongkot J, Adams J, Cui L, Cao Y, Wang Q. Genetic diversity of Plasmodium vivax reticulocyte binding protein 2b in global parasite populations. Parasit Vectors 2022; 15:205. [PMID: 35698238 PMCID: PMC9191549 DOI: 10.1186/s13071-022-05296-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 04/23/2022] [Indexed: 11/19/2022] Open
Abstract
Background Plasmodium vivax reticulocyte binding protein 2b (PvRBP2b) plays a critical role in parasite invasion of reticulocytes by binding the transferrin receptor 1. PvRBP2b is a vaccine candidate based on the negative correlation between antibody titers against PvRBP2b recombinant proteins and parasitemia and risk of vivax malaria. The aim of this study was to analyze the genetic diversity of the PvRBP2b gene in the global P. vivax populations. Methods Near full-length PvRBP2b nucleotide sequences (190–8349 bp) were obtained from 88 P. vivax isolates collected from the China–Myanmar border (n = 44) and Thailand (n = 44). An additional 224 PvRBP2b sequences were retrieved from genome sequences from parasite populations worldwide. The genetic diversity, neutral selection, haplotype distribution and genetic differentiation of PvRBP2b were examined. Results The genetic diversity of PvRBP2b was distributed unevenly, with peak diversity found in the reticulocyte binding region in the N-terminus. Neutrality analysis suggested that this region is subjected to balancing selection or population bottlenecks. Several amino acid variants were found in all or nearly all P. vivax endemic regions. However, the critical residues responsible for reticulocyte binding were highly conserved. There was substantial population differentiation according to the geographical separation. The distribution of haplotypes in the reticulocyte binding region varied among regions; even the two major haplotypes Hap_6 and Hap_8 were found in only five populations. Conclusions Our data show considerable genetic variations of PvRBPb in global parasite populations. The geographic divergence may pose a challenge to PvRBP2b-based vaccine development. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05296-6.
Collapse
Affiliation(s)
- Xuexing Zhang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Haichao Wei
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.,Department of Blood Transfusion Medicine, General Hospital of Northern Theater Command, Shenyang, 110015, Liaoning, China
| | - Yangminghui Zhang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Lin Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.,Department of Blood Transfusion, Yantaishan Hospital, Yantai, 264000, Shandong, China
| | - Yubing Hu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.,Central Laboratory, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Wang Nguitragool
- 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
| | - John Adams
- College of Public Health, Global Health Infectious Disease Research (GHIDR) Program, Tampa, FL, USA
| | - Liwang Cui
- College of Public Health, Global Health Infectious Disease Research (GHIDR) Program, Tampa, FL, USA.,Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL, 33612, USA
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Qinghui Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
17
|
Highly efficient protein expression of Plasmodium vivax surface antigen, Pvs25, by silkworm and its biochemical analysis. Protein Expr Purif 2022; 195-196:106096. [PMID: 35460871 DOI: 10.1016/j.pep.2022.106096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 11/20/2022]
Abstract
Plasmodium vivax ookinete surface protein, Pvs25, is a candidate for a transmission-blocking vaccine (TBV) for malaria. Pvs25 has four EGF-like domains containing 22 cysteine residues forming 11 intramolecular disulfide bonds, a structural feature that makes its recombinant protein expression difficult. In this study, we report the high expression of recombinant Pvs25 as a soluble form in silkworm, Bombyx mori. The Pvs25 protein was purified from hemolymphs of larvae and pupae by affinity chromatography. In the Pvs25 expressed by silkworm, no isoforms with inappropriate disulfide bonds were found, requiring no further purification step, which is necessary in the case of Pichia pastoris-based expression systems. The Pvs25 from silkworm was confirmed to be molecularly uniform by sodium dodecyl sulfate gel electrophoresis and size-exclusion chromatography. To examine the immunogenicity, the Pvs25 from B. mori was administered to BALB/c mice subcutaneously with oil adjuvant. The Pvs25 produced by silkworm induced potent and robust immune responses, and the induced antisera correctly recognized P. vivax ookinetes in vitro, demonstrating the potency of Pvs25 from silkworm as a candidate for a malaria TBV. To the best of our knowledge, this is the first study to construct a system for mass-producing malaria TBV antigens using silkworm.
Collapse
|
18
|
Soe MT, Aung PL, Nyunt MH, Sein MM, Cho C, Yang Z, Menezes L, Parker DM, Kyaw MP, Cui L. Therapeutic efficacy of chloroquine for uncomplicated Plasmodium vivax malaria in southeastern and western border areas of Myanmar. Infection 2022; 50:681-688. [PMID: 35034327 DOI: 10.1007/s15010-021-01739-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/01/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND In the Greater Mekong Subregion of Southeast Asia, Plasmodium vivax malaria is endemic and causes significant morbidity. In this study, the efficacy of chloroquine for treating uncomplicated P. vivax malaria at the eastern and western borders of Myanmar was investigated. METHODS A total of 197 participants with microscopically confirmed P. vivax infection were enrolled from three townships of the southeastern (Thanbyuzayat and Kawthoung) and western (Kyauktaw) borders of Myanmar. Patients were treated with chloroquine according to the national malaria treatment guidelines and followed for 28 days. RESULTS Among the 197 enrollments, 172 completed the 28-day follow-up. Twelve recurrent P. vivax infections, all occurring in the third and fourth week, were detected, resulting in an overall cumulative rate of recurrence of 4.7% [95% confidence interval (CI) 1.5-7.8]. The incidence rate of recurrence varied among the three sites. In Thanbyuzayat township, no patients had recurrent parasitemia between days 7 and 28. In contrast, Kyauktaw township had a day 28 cumulative incidence rate of recurrence of 7.2% (95% CI 0.6-13.9%) compared to 6.9% (95% CI 0.6-13.2) in Kawthoung township. CONCLUSION While this study confirmed the relatively high clinical efficacy of chloroquine for treating P. vivax in Myanmar with modest rates of recurrent infections within 28 days of the treatment, it also revealed considerable geographical heterogeneity of chloroquine efficacy, which warrants continuous surveillance efforts.
Collapse
Affiliation(s)
- Myat Thu Soe
- Myanmar Health Network Organization, Yangon, Myanmar
| | | | - Myat Htut Nyunt
- Department of Medical Research, Ministry of Health and Sports, Yangon, Myanmar
| | - Myint Myint Sein
- Department of Microbiology, University of Medicine, Magway, Myanmar
| | - Cho Cho
- Myanmar Health Network Organization, Yangon, Myanmar
| | - Zhaoqing Yang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, 650500, Yunnan, People's Republic of China
| | - Lynette Menezes
- Division of Infectious Diseases and International Medicine, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL, 33612, USA
| | - Daniel M Parker
- Department of Population Health and Disease Prevention, Department of Epidemiology, University of California, Irvine, USA
| | | | - Liwang Cui
- Division of Infectious Diseases and International Medicine, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL, 33612, USA.
| |
Collapse
|
19
|
Yasri S, Wiwanitkit V. Artemisinin resistance: an important emerging clinical problem in tropical medicine. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2021; 13:152-157. [PMID: 35103097 PMCID: PMC8784654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/06/2021] [Indexed: 06/14/2023]
Abstract
Artemisinin is an important antimalarial drug which is originated and developed from Chinese traditional herbal regimen. At present, artemisinin is used as an antimalarial drug for treatment of drug resistant malarial infection. The effectiveness of artemisinin is clinically accepted. Hence, artemisinin is currently used as main drug for malaria treatment in many tropical countries. Artemisinin resistance is a new emerging clinical problem in tropical medicine. New mutation can result in artemisinin resistance and the resistance becomes important new emerging problem in clinical malariology. It is necessary to control of artemisinin use and searching for new effective drug against artemisinin resistant malaria. In this article, the authors summarizes on important updated information regarding artemisinin resistance.
Collapse
Affiliation(s)
- Sora Yasri
- KMT Primary Care CenterBangkok, Thailand
| | | |
Collapse
|
20
|
Bareng PN, Grignard L, Reyes R, Fornace K, Spencer F, Macalinao ML, Luchavez J, Espino FE, Drakeley C, Hafalla JCR. Prevalence and temporal changes of mutations linked to anti-malarial drug resistance in Plasmodium falciparum and Plasmodium vivax in Palawan, Philippines. Int J Infect Dis 2021; 116:174-181. [PMID: 34883232 PMCID: PMC8866131 DOI: 10.1016/j.ijid.2021.12.318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/24/2021] [Accepted: 12/02/2021] [Indexed: 11/30/2022] Open
Abstract
Plasmodium falciparum and Plasmodium vivax isolates from the Philippines were analysed. Varying mutations were found in markers linked to resistance to antimalarial drugs. None of the mutations were particularly of high prevalence. Clear temporal patterns in these mutations were observed within the past 15 years. Decrease in pfcrt and pfmdr mutations are in line with antimalarial policy change.
Objective This study provides 2016 data on the prevalence of key single nucleotide polymorphisms (SNPs) associated with antimalarial drug resistance in Palawan, Philippines. Findings were combined with historical data to model temporal changes in the prevalence of these SNPs in Plasmodium isolates. Methods Plasmodium isolates were genotyped using drug resistance markers pfmdr1, pfcrt, pfdhfr, pfdhps, kelch-13, pvmdr1, pvdhfr, and pvdhps. Temporal trends in the probability of mutations were estimated as a function of time using a binomial generalised linear model. Results All samples sequenced for Plasmodium falciparum chloroquine markers pfmdr1 and pfcrt had wild-type alleles. Varying mutation patterns were observed for the sulphadoxine/pyrimethamine markers pfdhps and pfdhfr; complete quintuplet mutations were not found. No SNPs were observed for the artemisinin marker kelch-13. For Plasmodium vivax, differing patterns were detected for pvmdr1, pvdhfr, and pvdhps. Conclusions The study findings suggest that the current drugs remain effective and that there is limited importation and establishment of resistant parasites in the area. Clear temporal trends were recognised, with prominent decreases in the proportions of pfcrt and pfmdr mutations detected within the past 15 years, consistent with a change in antimalarial drug policy. Continuous surveillance of antimalarial drug resistance is important to support malaria elimination efforts.
Collapse
Affiliation(s)
- Paolo N Bareng
- Department of Parasitology and National Reference Centre for Malaria and Other Parasites, Research Institute for Tropical Medicine, Department of Health, Muntinlupa City, Philippines.
| | - Lynn Grignard
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ralph Reyes
- Department of Parasitology and National Reference Centre for Malaria and Other Parasites, Research Institute for Tropical Medicine, Department of Health, Muntinlupa City, Philippines
| | - Kim Fornace
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Freya Spencer
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ma Lourdes Macalinao
- Department of Parasitology and National Reference Centre for Malaria and Other Parasites, Research Institute for Tropical Medicine, Department of Health, Muntinlupa City, Philippines
| | - Jennifer Luchavez
- Department of Parasitology and National Reference Centre for Malaria and Other Parasites, Research Institute for Tropical Medicine, Department of Health, Muntinlupa City, Philippines
| | - Fe Esperanza Espino
- Department of Parasitology and National Reference Centre for Malaria and Other Parasites, Research Institute for Tropical Medicine, Department of Health, Muntinlupa City, Philippines
| | - Chris Drakeley
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Julius Clemence R Hafalla
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| |
Collapse
|
21
|
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
|
22
|
de Pina-Costa A, Silvino ACR, Dos Santos EM, Pedro RS, Moreira J, Umana GL, da Silva ADT, da Rosa Santos OHL, de Deus Henriques KM, Daniel-Ribeiro CT, Brasil P, Sousa TN, Siqueira AM. Increased primaquine total dose prevents Plasmodium vivax relapses in patients with impaired CYP2D6 activity: report of three cases. Malar J 2021; 20:341. [PMID: 34391426 PMCID: PMC8364036 DOI: 10.1186/s12936-021-03869-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/28/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The relapsing nature of Plasmodium vivax infection is a major barrier to its control and elimination. Factors such as adequate dosing, adherence, drug quality, and pharmacogenetics can impact the effectiveness of radical cure of P. vivax and need to be adequately evaluated. CYP2D6 pathway mediates the activation of primaquine (primaquine) into an active metabolite(s) in hepatocytes, and impaired activity has been linked to a higher risk of relapse. CASES PRESENTATION Three patients diagnosed with P. vivax malaria presented repeated relapses after being initially treated with chloroquine (25 mg/kg) and primaquine (3.5 mg/kg in 14 days) at a non-endemic travel clinic. Recurring episodes were subsequently treated with a higher dose of primaquine (7 mg/kg in 14 days), which prevented further relapses in two patients. However, one patient still presented two episodes after a higher primaquine dose and was prescribed 300 mg of chloroquine weekly to prevent further episodes. Impaired CYP2D6 function was observed in all of them. CONCLUSION Lack of response to primaquine was associated with impaired CYP2D6 activity in three patients presenting multiple relapses followed in a non-endemic setting. Higher primaquine dosage was safe and effectively prevented relapses in two patients and should be further investigated as an option in Latin America. It is crucial to investigate the factors associated with unsuccessful radical cures and alternative therapeutic options.
Collapse
Affiliation(s)
- Anielle de Pina-Costa
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas-Instituto Nacional de Infectologia (INI) Evandro Chagas, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Centro de Pesquisa Diagnóstico e Treinamento em Malária-Fiocruz, Rio de Janeiro, Brazil
- Centro Universitário Serra Dos Órgãos (UNIFESO), Teresópolis, RJ, Brazil
| | | | - Edwiges Motta Dos Santos
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas-Instituto Nacional de Infectologia (INI) Evandro Chagas, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Renata Saraiva Pedro
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas-Instituto Nacional de Infectologia (INI) Evandro Chagas, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Assessoria Clínica-Instituto em Tecnologia em Imunobiológicos-Fiocruz, Rio de Janeiro, Brazil
| | - José Moreira
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas-Instituto Nacional de Infectologia (INI) Evandro Chagas, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Programa de Pós-Graduação Em Pesquisa Clínica, INI Evandro Chagas, Fiocruz, Rio de Janeiro, Brazil
| | - Gabriela Liseth Umana
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas-Instituto Nacional de Infectologia (INI) Evandro Chagas, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Ana Danielle Tavares da Silva
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas-Instituto Nacional de Infectologia (INI) Evandro Chagas, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Otília Helena Lupi da Rosa Santos
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas-Instituto Nacional de Infectologia (INI) Evandro Chagas, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Centro de Pesquisa Diagnóstico e Treinamento em Malária-Fiocruz, Rio de Janeiro, Brazil
| | - Karina Medeiros de Deus Henriques
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas-Instituto Nacional de Infectologia (INI) Evandro Chagas, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Programa de Pós-Graduação Em Pesquisa Clínica, INI Evandro Chagas, Fiocruz, Rio de Janeiro, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Centro de Pesquisa Diagnóstico e Treinamento em Malária-Fiocruz, Rio de Janeiro, Brazil
- Laboratório de Pesquisa Em Malária, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Patrícia Brasil
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas-Instituto Nacional de Infectologia (INI) Evandro Chagas, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Centro de Pesquisa Diagnóstico e Treinamento em Malária-Fiocruz, Rio de Janeiro, Brazil
- Programa de Pós-Graduação Em Pesquisa Clínica, INI Evandro Chagas, Fiocruz, Rio de Janeiro, Brazil
| | | | - André M Siqueira
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas-Instituto Nacional de Infectologia (INI) Evandro Chagas, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil.
- Centro de Pesquisa Diagnóstico e Treinamento em Malária-Fiocruz, Rio de Janeiro, Brazil.
- Programa de Pós-Graduação Em Pesquisa Clínica, INI Evandro Chagas, Fiocruz, Rio de Janeiro, Brazil.
| |
Collapse
|
23
|
Salles PF, Perce-da-Silva DS, Rossi AD, Raposo LR, Ramirez Ramirez AD, Pereira Bastos OM, Pratt-Riccio LR, Cassiano GC, Baptista ARS, Cardoso CC, Banic DM, Machado RLD. CYP2D6 Allele Frequency in Five Malaria Vivax Endemic Areas From Brazilian Amazon Region. Front Pharmacol 2021; 12:542342. [PMID: 34366834 PMCID: PMC8343396 DOI: 10.3389/fphar.2021.542342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/07/2021] [Indexed: 12/29/2022] Open
Abstract
Genetic variability was linked with individual responses to treatment and susceptibility to malaria by Plasmodium vivax. Polymorphisms in the CYP2D6 gene may modulate enzyme level and activity, thereby affecting individual responses to pharmacological treatment. The aim of the study was to investigate whether or not CYP2D6 single nucleotide polymorphisms rs1065852, rs38920-97, rs16947 and rs28371725 are unequally distributed in malaria by Plasmodium vivax individuals from the Brazilian Amazon region. The blood samples were collected from 220 unrelated Plasmodium vivax patients from five different endemic areas. Genotyping was performed using SNaPshot® and real-time polymerase chain reaction methods. In all five areas, the rs1065852 (CYP2D6*10, C.100C > T), rs3892097 (CYP2D6*4, 1846C > T) and rs16947 (CYP2D6*2, C.2850G > A), as a homozygous genotype, showed the lowest frequencies. The rs28371725 (CYP2D6*41, 2988G > A) homozygous genotype was not detected, while the allele A was found in a single patient from Macapá region. No deviations from Hardy-Weinberg equilibrium were found, although a borderline p-value was observed (p = 0.048) for the SNP rs3892097 in Goianésia do Pará, Pará state. No significant associations were detected in these frequencies among the five studied areas. For the SNP rs3892097, a higher frequency was observed for the C/T heterozygous genotype in the Plácido de Castro and Macapá, Acre and Amapá states, respectively. The distribution of the CYP2D6 alleles investigated in the different areas of the Brazilian Amazon is not homogeneous. Further investigations are necessary in order to determine which alleles might be informative to assure optimal drug dosing recommendations based on experimental pharmacogenetics.
Collapse
Affiliation(s)
- Paula Ferreira Salles
- Centro de Investigação de Microrganismos, Universidade Federal Fluminense, Niterói, Brazil
| | | | - Atila Duque Rossi
- Laboratório de Virologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luisa Riehl Raposo
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | | | | | - Gustavo Capatti Cassiano
- Saúde Global e Medicina Tropical, Instituto de Higiene e Medicina Tropical, Universidade de Lisboa, Lisbon, Portugal
| | | | - Cynthia Chester Cardoso
- Laboratório de Virologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dalma Maria Banic
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | |
Collapse
|
24
|
Asghari A, Nourmohammadi H, Majidiani H, Shariatzadeh SA, Shams M, Montazeri F. In silico analysis and prediction of immunogenic epitopes for pre-erythrocytic proteins of the deadly Plasmodium falciparum. INFECTION GENETICS AND EVOLUTION 2021; 93:104985. [PMID: 34214673 DOI: 10.1016/j.meegid.2021.104985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/23/2021] [Accepted: 06/27/2021] [Indexed: 12/18/2022]
Abstract
Malaria is the deadliest parasitic disease in tropical and subtropical areas around the world, with considerable morbidity and mortality, particularly due to the life-threatening Plasmodium falciparum. The present in silico investigation was performed to reveal the biophysical characteristics and immunogenic epitopes of the six pre-erythrocytic proteins of the P. falciparum using comprehensive immunoinformatics approaches. For this aim, different web servers were employed to predict subcellular localization, antigenicity, allergenicity, solubility, physico-chemical properties, post-translational modification sites (PTMs), the presence of signal peptide and transmembrane domains. Moreover, the secondary and tertiary structures of the proteins were revealed followed by refinement and validations. Finally, NetCTL server was used to predict cytotoxic T-lymphocyte (CTL) epitopes, followed by subsequent screening in terms of antigenicity and immunogenicity. Also, IEDB server was utilized to predict helper T-lymphocyte (HTL) epitopes, followed by screening regarding interferon gamma induction and population coverage. These proteins showed appropriate antigenicity, abundant PTMs as well as many CTL and HTL epitopes, which could be directed for future vaccination studies in the context of multi-epitope vaccine design.
Collapse
Affiliation(s)
- Ali Asghari
- Department of Medical Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Nourmohammadi
- Department of Internal Medicine, Shahid Mostafa Khomeini Hospital, Ilam University of Medical Sciences, Ilam, Iran; Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Hamidreza Majidiani
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Seyyed Ali Shariatzadeh
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Toxoplasmosis Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Morteza Shams
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran; Student Research Committee, Ilam University of Medical Sciences, Ilam, Iran.
| | - Fattaneh Montazeri
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
25
|
Mehlotra RK, Gaedigk A, Howes RE, Rakotomanga TA, Ratsimbasoa AC, Zimmerman PA. CYP2D6 Genetic Variation and Its Implication for Vivax Malaria Treatment in Madagascar. Front Pharmacol 2021; 12:654054. [PMID: 33959023 PMCID: PMC8093859 DOI: 10.3389/fphar.2021.654054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/01/2021] [Indexed: 12/19/2022] Open
Abstract
Plasmodium vivax is one of the five human malaria parasite species, which has a wide geographical distribution and can cause severe disease and fatal outcomes. It has the ability to relapse from dormant liver stages (hypnozoites), weeks to months after clearance of the acute blood-stage infection. An 8-aminoquinoline drug primaquine (PQ) can clear the hypnozoites, and thus can be used as an anti-relapse therapeutic agent. Recently, a number of studies have found that its efficacy is compromised by polymorphisms in the cytochrome P450 2D6 (CYP2D6) gene; decreased or absence of CYP2D6 activity contributes to PQ therapeutic failure. The present study sought to characterize CYP2D6 genetic variation in Madagascar, where populations originated from admixture between Asian and African populations, vivax malaria is endemic, and PQ can be deployed soon to achieve national malaria elimination. In a total of 211 samples collected from two health districts, CYP2D6 decreased function alleles CYP2D6*10, *17, *29, *36+*10, and *41 were observed at frequencies of 3.55-17.06%. In addition, nonfunctional alleles were observed, the most common of which were CYP2D6*4 (2.13%), *5 (1.66%), and the *4x2 gene duplication (1.42%). Given these frequencies, 34.6% of the individuals were predicted to be intermediate metabolizers (IM) with an enzyme activity score (AS) ≤ 1.0; both the IM phenotype and AS ≤ 1.0 have been found to be associated with PQ therapeutic failure. Furthermore, the allele and genotype frequency distributions add to the archaeological and genomic evidence of Malagasy populations constituting a unique, Asian-African admixed origin. The results from this exploratory study provide fresh insights about genomic characteristics that could affect the metabolism of PQ into its active state, and may enable optimization of PQ treatment across human genetic diversity, which is critical for achieving P. vivax elimination.
Collapse
Affiliation(s)
- Rajeev K Mehlotra
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy Kansas City, Kanas City, MO, United States
| | - Rosalind E Howes
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom.,Foundation for Innovative New Diagnostics, Geneva, Switzerland
| | - Tovonahary A Rakotomanga
- The National Malaria Control Program, Ministry of Health, Antananarivo, Madagascar.,University of Fianarantsoa, Fianarantsoa, Madagascar
| | - Arsene C Ratsimbasoa
- The National Malaria Control Program, Ministry of Health, Antananarivo, Madagascar.,University of Fianarantsoa, Fianarantsoa, Madagascar
| | - Peter A Zimmerman
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
26
|
Jiero S, Pasaribu AP. Haematological profile of children with malaria in Sorong, West Papua, Indonesia. Malar J 2021; 20:126. [PMID: 33663524 PMCID: PMC7931533 DOI: 10.1186/s12936-021-03638-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/09/2021] [Indexed: 11/13/2022] Open
Abstract
Background Malaria remains a major public health problem in Indonesian Papua, with children under five years of age being the most affected group. Haematological changes, such as cytopenia that occur during malaria infection have been suggested as potential predictors and can aid in the diagnosis of malaria. This study aimed to assess the haematological alterations associated with malaria infection in children presenting with signs and symptoms of malaria. Methods A retrospective study was performed by collecting data from the medical records of malaria patients at Sorong Regional General Hospital, Sorong, West Papua, Indonesia, both from outpatient and inpatient clinics, from January 2014 until December 2017. The laboratory profile of children suffering from malaria was evaluated. Results One hundred and eighty-two children aged 1 month to 18 years old were enrolled. The subjects were mostly male (112, 61.5%) with a mean age of 6.45 years (SD = 4.3 years). Children below 5 years of age suffered the most from malaria in this study (77, 42.3%). One hundred two subjects (56%) were infected with Plasmodium falciparum. Half of the enrolled subjects (50%) had haemoglobin level (Hb) between 5.1 and 10 gr/dL. A total of 41 children (53.2%) less than 5 years old suffered from P. falciparum infection. In the age group of 5–10 years, there were 34 children (57.6%) who suffered from P. falciparum, and in the age group > 10 years, 27 children (58.7%) suffered from P. falciparum infection. Only 4 subjects (5.2%) in the less than 5 years old age group had mixed malaria infection. Among eight predictors of the haematological profile, there were five predictors that were significantly associated with the diagnostic criteria, namely haemoglobin, haematocrit, leukocytes, platelets and monocytes (p < 0.05). Generally, clinical symptoms are not significantly associated with a malaria diagnosis, and only one variable showed a significant relationship, pale, with a P value of 0.001. Conclusions Children with malaria had changes in some haematological markers, with anaemia, low platelet count, white blood count, and lymphocyte count being the most important predictors of malaria infection in the study area. These markers could be used to raise suspicion of malaria in children living in high endemic areas, such as West Papua.
Collapse
Affiliation(s)
- Syilvia Jiero
- Department of Child Health, Sorong Regional General Hospital, Sorong, West Papua, Indonesia
| | - Ayodhia Pitaloka Pasaribu
- Department of Child Health, Medical Faculty, Universitas Sumatera Utara, Dr. Mansur Street No. 5, 20156, Medan, Indonesia.
| |
Collapse
|
27
|
Bourgard C, Lopes SCP, Lacerda MVG, Albrecht L, Costa FTM. A suitable RNA preparation methodology for whole transcriptome shotgun sequencing harvested from Plasmodium vivax-infected patients. Sci Rep 2021; 11:5089. [PMID: 33658571 PMCID: PMC7930272 DOI: 10.1038/s41598-021-84607-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/06/2021] [Indexed: 12/03/2022] Open
Abstract
Plasmodium vivax is a world-threatening human malaria parasite, whose biology remains elusive. The unavailability of in vitro culture, and the difficulties in getting a high number of pure parasites makes RNA isolation in quantity and quality a challenge. Here, a methodological outline for RNA-seq from P. vivax isolates with low parasitemia is presented, combining parasite maturation and enrichment with efficient RNA extraction, yielding ~ 100 pg.µL−1 of RNA, suitable for SMART-Seq Ultra-Low Input RNA library and Illumina sequencing. Unbiased coding transcriptome of ~ 4 M reads was achieved for four patient isolates with ~ 51% of transcripts mapped to the P. vivax P01 reference genome, presenting heterogeneous profiles of expression among individual isolates. Amongst the most transcribed genes in all isolates, a parasite-staged mixed repertoire of conserved parasite metabolic, membrane and exported proteins was observed. Still, a quarter of transcribed genes remain functionally uncharacterized. In parallel, a P. falciparum Brazilian isolate was also analyzed and 57% of its transcripts mapped against IT genome. Comparison of transcriptomes of the two species revealed a common trophozoite-staged expression profile, with several homologous genes being expressed. Collectively, these results will positively impact vivax research improving knowledge of P. vivax biology.
Collapse
Affiliation(s)
- Catarina Bourgard
- Laboratory of Tropical Diseases, Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Stefanie C P Lopes
- Instituto Leônidas & Maria Deane, Fundação Oswaldo Cruz-Fiocruz, Manaus, AM, Brazil.,Fundação de Medicina Tropical Dr. Heitor Vieira Dourado-FMT-HVD, Gerência de Malária, Manaus, AM, Brazil
| | - Marcus V G Lacerda
- Instituto Leônidas & Maria Deane, Fundação Oswaldo Cruz-Fiocruz, Manaus, AM, Brazil.,Fundação de Medicina Tropical Dr. Heitor Vieira Dourado-FMT-HVD, Gerência de Malária, Manaus, AM, Brazil
| | - Letusa Albrecht
- Laboratory of Tropical Diseases, Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil. .,Instituto Carlos Chagas, Fundação Oswaldo Cruz-Fiocruz, Curitiba, PR, Brazil.
| | - Fabio T M Costa
- Laboratory of Tropical Diseases, Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil.
| |
Collapse
|
28
|
Yeshanew S, Dagne A, Taye B. Efficacy Study of Chloroquine to Plasmodium vivax Malaria in Darimu and Bure Districts, Southwest Ethiopia. Infect Drug Resist 2021; 14:795-803. [PMID: 33688214 PMCID: PMC7936702 DOI: 10.2147/idr.s300692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/22/2021] [Indexed: 11/27/2022] Open
Abstract
Background Chloroquine (CQ) is the first line treatment for vivax malaria in Ethiopia. However, the therapeutic efficacy of the drug is now declining. Several reports from different areas of the country showed CQ-vivax treatment failure increasing. This study therefore aimed to provide additional data on the therapeutic efficacy of CQ against Plasmodium vivax malaria from two districts of Southwestern Ethiopia. Methods An observational prospective study among P. vivax malaria infected individuals was conducted in two districts of Southwest Ethiopia for a period of 28 follow-up days. Study participants were treated with 25 mg/kg of standard CQ for 3 consecutive days according to the procedure. Microscopic blood film examinations and other clinical assessments were measured within the follow-up period on a weekly basis. Results A total of 115 patients were enrolled in the study. Sixty-five were from Darimu and 50 were from Bure districts. The majority (67%) of study participants were male and 86.1% (99/115) were below 35 years old. The study revealed that CQ treatment was able to clear vivax malaria parasites and febrile within a week. During the follow-up study period, recurrence of vivax parasitemia was not recorded. However, there was a marked heterogeneity with respect to fever clearance time, parasitemia load, and carriage of parasite gametocyte within 72 hours of post-treatment between the two study areas. Conclusion The present study revealed that CQ has good clinical and parasitological response to vivax malaria in the study areas. Thus, it can be continued as the first line P. vivax malaria treatment. However, further monitoring and evaluation of the drug should be considered.
Collapse
Affiliation(s)
- Solomon Yeshanew
- Department of Biology, Debre Markos University, Debre Markos, Ethiopia
| | | | - Behailu Taye
- Department of Biology, Mettu University, Mettu, Ethiopia
| |
Collapse
|
29
|
Xu S, Zeng W, Ngassa Mbenda HG, Liu H, Chen X, Xiang Z, Li C, Zhang Y, Baird JK, Yang Z, Cui L. Efficacy of directly-observed chloroquine-primaquine treatment for uncomplicated acute Plasmodium vivax malaria in northeast Myanmar: A prospective open-label efficacy trial. Travel Med Infect Dis 2020; 36:101499. [PMID: 31604130 PMCID: PMC7816571 DOI: 10.1016/j.tmaid.2019.101499] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Chloroquine (CQ) and primaquine (PQ) remain the frontline drugs for radical cure of uncomplicated P. vivax malaria in the Greater Mekong Sub-region (GMS). Recent reports of decreased susceptibility of P. vivax to CQ in many parts of the GMS raise concerns. METHODS From April 2014 to September 2016, 281 patients with uncomplicated P. vivax infection attending clinics in border settlements for internally displaced people in northeast Myanmar were recruited into this study. Patients were treated with standard regimen of 3-day CQ and concurrent 14-day PQ (3.5 mg/kg total dose) as directly observed therapy, and followed for recurrent parasitemia within 28 days post-patency. RESULTS Within the 28-day follow-up period, seven patients developed recurrent parasitemia, resulting in a cumulative rate of parasite recurrence of 2.6%. Five of the seven parasitemias recurred within two weeks, and two of those failed to clear within seven days, indicating high-grade resistance. CONCLUSION Although failure of CQ/PQ treatment of P. vivax was relatively infrequent in northeast Myanmar, this study nonetheless confirms that CQ/PQ-resistant strains do circulate in this area, some of them of a highly resistant phenotype. It is thus recommended that patients who acquire vivax malaria in Myanmar be treated an artemisinin-combination therapy along with hypnozoitocidal primaquine therapy to achieve radical cure.
Collapse
Affiliation(s)
- Shiling Xu
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| | - Weilin Zeng
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| | - Huguette Gaelle Ngassa Mbenda
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Blvd, Suite 304, Tampa, FL, 33612, USA
| | - Huaie Liu
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| | - Xi Chen
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| | - Zheng Xiang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| | - Cuiying Li
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| | - Yanmei Zhang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| | - J Kevin Baird
- Eijkman-Oxford Clinical Research Unit, Jalan Diponegoro No. 69, Jakarta, 10430, Indonesia; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7FZ, United Kingdom
| | - Zhaoqing Yang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan, 650500, PR China.
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Blvd, Suite 304, Tampa, FL, 33612, USA.
| |
Collapse
|
30
|
Influence of CYP2C8, CYP3A4, and CYP3A5 Host Genotypes on Early Recurrence of Plasmodium vivax. Antimicrob Agents Chemother 2020; 64:AAC.02125-19. [PMID: 32366712 DOI: 10.1128/aac.02125-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/25/2020] [Indexed: 11/20/2022] Open
Abstract
Cytochrome P450 (CYP) enzymes are involved in the biotransformation of chloroquine (CQ), but the role of the different profiles of metabolism of this drug in relation to Plasmodium vivax recurrences has not been properly investigated. To investigate the influence of the CYP genotypes associated with CQ metabolism on the rates of P. vivax early recurrences, a case-control study was carried out. The cases included patients presenting with an early recurrence (CQ-recurrent individuals), defined as a recurrence during the first 28 days after initial infection and plasma concentrations of CQ plus desethylchloroquine (DCQ; the major CQ metabolite) higher than 100 ng/ml. A control group with no parasite recurrence over the follow-up (the CQ-responsive group) was also included. CQ and DCQ plasma levels were measured on day 28. CQ-metabolizing CYP (CYP2C8, CYP3A4, and CYP3A5) genotypes were determined by real-time PCR. An ex vivo study was conducted to verify the efficacy of CQ and DCQ against P. vivax isolates. The frequency of alleles associated with normal and slow metabolism was similar between the cases and the controls for the CYP2C8 (odds ratio [OR] = 1.45, 95% confidence interval [CI] = 0.51 to 4.14, P = 0.570), CYP3A4 (OR = 2.38, 95% CI = 0.92 to 6.19, P = 0.105), and CYP3A5 (OR = 4.17, 95% CI = 0.79 to 22.04, P = 1.038) genes. DCQ levels were higher than CQ levels, regardless of the genotype. Regarding the DCQ/CQ ratio, there was no difference between groups or between those patients who had a normal genotype and those patients who had a mutant genotype. DCQ and CQ showed similar efficacy ex vivo CYP genotypes had no influence on early recurrence rates. The similar efficacy of CQ and DCQ ex vivo could explain the absence of therapeutic failure, despite the presence of alleles associated with slow metabolism.
Collapse
|
31
|
Li J, Zhang J, Li Q, Hu Y, Ruan Y, Tao Z, Xia H, Qiao J, Meng L, Zeng W, Li C, He X, Zhao L, Siddiqui FA, Miao J, Yang Z, Fang Q, Cui L. Ex vivo susceptibilities of Plasmodium vivax isolates from the China-Myanmar border to antimalarial drugs and association with polymorphisms in Pvmdr1 and Pvcrt-o genes. PLoS Negl Trop Dis 2020; 14:e0008255. [PMID: 32530913 PMCID: PMC7314094 DOI: 10.1371/journal.pntd.0008255] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 06/24/2020] [Accepted: 03/26/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Vivax malaria is an important public health problem in the Greater Mekong Subregion (GMS), including the China-Myanmar border. Previous studies have found that Plasmodium vivax has decreased sensitivity to antimalarial drugs in some areas of the GMS, but the sensitivity of P. vivax to antimalarial drugs is unclear in the China-Myanmar border. Here, we investigate the drug sensitivity profile and genetic variations for two drug resistance related genes in P. vivax isolates to provide baseline information for future drug studies in the China-Myanmar border. METHODOLOGY/PRINCIPAL FINDINGS A total of 64 P. vivax clinical isolates collected from the China-Myanmar border area were assessed for ex vivo susceptibility to eight antimalarial drugs by the schizont maturation assay. The medians of IC50 (half-maximum inhibitory concentrations) for chloroquine, mefloquine, pyronaridine, piperaquine, quinine, artesunate, artemether, dihydroartemisinin were 84.2 nM, 34.9 nM, 4.0 nM, 22.3 nM, 41.4 nM, 2.8 nM, 2.1 nM and 2.0 nM, respectively. Twelve P. vivax clinical isolates were found over the cut-off IC50 value (220 nM) for chloroquine resistance. In addition, sequence polymorphisms in pvmdr1 (P. vivax multidrug resistance-1), pvcrt-o (P. vivax chloroquine resistance transporter-o), and difference in pvmdr1 copy number were studied. Sequencing of the pvmdr1 gene in 52 samples identified 12 amino acid substitutions, among which two (G698S and T958M) were fixed, M908L were present in 98.1% of the isolates, while Y976F and F1076L were present in 3.8% and 78.8% of the isolates, respectively. Amplification of the pvmdr1 gene was only detected in 4.8% of the samples. Sequencing of the pvcrt-o in 59 parasite isolates identified a single lysine insertion at position 10 in 32.2% of the isolates. The pvmdr1 M908L substitutions in pvmdr1 in our samples was associated with reduced sensitivity to chloroquine, mefloquine, pyronaridine, piperaquine, quinine, artesunate and dihydroartemisinin. CONCLUSIONS Our findings depict a drug sensitivity profile and genetic variations of the P. vivax isolates from the China-Myanmar border area, and suggest possible emergence of chloroquine resistant P. vivax isolates in the region, which demands further efforts for resistance monitoring and mechanism studies.
Collapse
Affiliation(s)
- Jiangyan Li
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, Anhui Province, China
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Jie Zhang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan Province, China
| | - Qian Li
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, Anhui Province, China
- Xiangtan Blood Center, Xiangtan, Hunan Province, China
| | - Yue Hu
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yonghua Ruan
- Department of Pathology, Kunming Medical University, Kunming, Yunnan Province, China
| | - Zhiyong Tao
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, Anhui Province, China
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Hui Xia
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, Anhui Province, China
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Jichen Qiao
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, Anhui Province, China
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Lingwen Meng
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, Anhui Province, China
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Weilin Zeng
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan Province, China
| | - Cuiying Li
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan Province, China
| | - Xi He
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan Province, China
| | - Luyi Zhao
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan Province, China
| | - Faiza A. Siddiqui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Jun Miao
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Zhaoqing Yang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan Province, China
- * E-mail: (ZY); (QF)
| | - Qiang Fang
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, Anhui Province, China
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui Province, China
- * E-mail: (ZY); (QF)
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| |
Collapse
|
32
|
Rougeron V, Elguero E, Arnathau C, Acuña Hidalgo B, Durand P, Houze S, Berry A, Zakeri S, Haque R, Shafiul Alam M, Nosten F, Severini C, Gebru Woldearegai T, Mordmüller B, Kremsner PG, González-Cerón L, Fontecha G, Gamboa D, Musset L, Legrand E, Noya O, Pumpaibool T, Harnyuttanakorn P, Lekweiry KM, Mohamad Albsheer M, Mahdi Abdel Hamid M, Boukary AOMS, Trape JF, Renaud F, Prugnolle F. Human Plasmodium vivax diversity, population structure and evolutionary origin. PLoS Negl Trop Dis 2020; 14:e0008072. [PMID: 32150544 PMCID: PMC7082039 DOI: 10.1371/journal.pntd.0008072] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 03/19/2020] [Accepted: 01/18/2020] [Indexed: 11/19/2022] Open
Abstract
More than 200 million malaria clinical cases are reported each year due to Plasmodium vivax, the most widespread Plasmodium species in the world. This species has been neglected and understudied for a long time, due to its lower mortality in comparison with Plasmodium falciparum. A renewed interest has emerged in the past decade with the discovery of antimalarial drug resistance and of severe and even fatal human cases. Nonetheless, today there are still significant gaps in our understanding of the population genetics and evolutionary history of P. vivax, particularly because of a lack of genetic data from Africa. To address these gaps, we genotyped 14 microsatellite loci in 834 samples obtained from 28 locations in 20 countries from around the world. We discuss the worldwide population genetic structure and diversity and the evolutionary origin of P. vivax in the world and its introduction into the Americas. This study demonstrates the importance of conducting genome-wide analyses of P. vivax in order to unravel its complex evolutionary history. Among the five Plasmodium species infecting humans, P. vivax is the most prevalent parasite outside Africa. To date, there has been less research on this species than for Plasmodium falciparum, a more lethal species, principally because of the lack of an in vitro culture system and also because P. vivax is considered relatively benign. Nevertheless, P. vivax is responsible for severe and incapacitating clinical symptoms with significant effects on human health. The emergence of new drug resistance and the discovery of severe and even fatal cases due to P. vivax question the benign status of P. vivax malaria. In recent years, there has been increased interest in characterizing the distribution of genetic variation in P. vivax. However, these studies either generated genetic information from a regional geographic scale or combine genetic datasets generated in different molecular platforms, which is known to generate biased results. In this study, we used a single genotyping platform to genotype 14 microsatellite markers in 834 samples of P. vivax obtained from 28 locations in 20 countries from around the world, including several populations from East and West Africa. We discuss the worldwide population genetic structure and the evolutionary origins of P. vivax, as well as its introduction into the Americas.
Collapse
Affiliation(s)
- Virginie Rougeron
- Laboratoire MIVEGEC (Université de Montpellier-CNRS-IRD), CREES, Montpellier, France
- * E-mail: ,
| | - Eric Elguero
- Laboratoire MIVEGEC (Université de Montpellier-CNRS-IRD), CREES, Montpellier, France
| | - Céline Arnathau
- Laboratoire MIVEGEC (Université de Montpellier-CNRS-IRD), CREES, Montpellier, France
| | - Beatriz Acuña Hidalgo
- Laboratoire MIVEGEC (Université de Montpellier-CNRS-IRD), CREES, Montpellier, France
| | - Patrick Durand
- Laboratoire MIVEGEC (Université de Montpellier-CNRS-IRD), CREES, Montpellier, France
| | - Sandrine Houze
- Service de Parasitologie-mycologie CNR du Paludisme, AP-HP Hôpital Bichat, Paris, France
| | - Antoine Berry
- Centre de Physiopathologie de Toulouse-Purpan (CPTP), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1043, CNRS UMR5282, Université de Toulouse Paul Sabatier, F-31300 Toulouse, France
- Service de Parasitologie-Mycologie, Institut Fédératif de Biologie, Centre Hospitalier Universitaire de Toulouse, F-31300 Toulouse, France
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Rashidul Haque
- Emerging Infections & Parasitology Laboratory, icddr,b, Mohakhali, Dhaka, Bangladesh
| | - Mohammad Shafiul Alam
- Emerging Infections & Parasitology Laboratory, icddr,b, Mohakhali, Dhaka, Bangladesh
| | - François Nosten
- Centre for Tropical Medicine and Global Health,Oxford, United Kingdom
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Carlo Severini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Tamirat Gebru Woldearegai
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
- Department of Medical Laboratory Sciences, College of Medical and Health Sciences, Haramaya University, Harar, Ethiopia
| | - Benjamin Mordmüller
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | | | - Lilia González-Cerón
- Regional Centre of Research in Public Health, National Institute of Public Health, Tapachula, Chiapas, Mexico
| | - Gustavo Fontecha
- Microbiology Research Institute, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Dionicia Gamboa
- Instituto de Medicina Tropical Alexander Von Humboldt, Universidad Peruana Cayetano Heredia, AP, Lima, Peru
| | - Lise Musset
- Unit, Institut Pasteur de Guyane, BP6010, French Guiana
| | - Eric Legrand
- Malaria Genetic and Resistance Group, Biology of Host-Parasite Interactions Unit, Institut Pasteur, Paris, France
| | - Oscar Noya
- Centro para Estudios Sobre Malaria, Instituto de Altos Estudios en Salud “Dr. Arnoldo Gabaldón”, Ministerio del Poder Popular para la Salud and Instituto de Medicina Tropical, Universidad Central de Venezuela, Maracay, Caracas, Venezuela
| | - Tepanata Pumpaibool
- Biomedical Science, Graduate School, Chulalongkorn University, Bangkok, Thailand
- Malaria Research Programme, College of Public Health Science, Chulalongkorn University, Bangkok, Thailand
| | - Pingchai Harnyuttanakorn
- Malaria Research Programme, College of Public Health Science, Chulalongkorn University, Bangkok, Thailand
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Khadijetou Mint Lekweiry
- UR-Génomes et milieux, Faculté des Sciences et Techniques, Université de Nouakchott Al-Aasriya, Mauritania
| | - Musab Mohamad Albsheer
- Department of Parasitology and Medical Entomology, Medical Campus, University of Khartoum, Sudan
| | | | - Ali Ould Mohamed Salem Boukary
- UR-Génomes et milieux, Faculté des Sciences et Techniques, Université de Nouakchott Al-Aasriya, Mauritania
- Aix Marseille Univ, IRD, AP-HM, SSA, VITROME, Marseille, France
| | - Jean-François Trape
- Laboratoire MIVEGEC (Université de Montpellier-CNRS-IRD), CREES, Montpellier, France
| | - François Renaud
- Laboratoire MIVEGEC (Université de Montpellier-CNRS-IRD), CREES, Montpellier, France
| | - Franck Prugnolle
- Laboratoire MIVEGEC (Université de Montpellier-CNRS-IRD), CREES, Montpellier, France
| |
Collapse
|
33
|
Heterogeneous Network Model to Identify Potential Associations Between Plasmodium vivax and Human Proteins. Int J Mol Sci 2020; 21:ijms21041310. [PMID: 32075230 PMCID: PMC7072978 DOI: 10.3390/ijms21041310] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 01/29/2020] [Accepted: 02/12/2020] [Indexed: 02/06/2023] Open
Abstract
Integration of multiple sources and data levels provides a great insight into the complex associations between human and malaria systems. In this study, a meta-analysis framework was developed based on a heterogeneous network model for integrating human-malaria protein similarities, a human protein interaction network, and a Plasmodium vivax protein interaction network. An iterative network propagation was performed on the heterogeneous network until we obtained stabilized weights. The association scores were calculated for qualifying a novel potential human-malaria protein association. This method provided a better performance compared to random experiments. After that, the stabilized network was clustered into association modules. The potential association candidates were then thoroughly analyzed by statistical enrichment analysis with protein complexes and known drug targets. The most promising target proteins were the succinate dehydrogenase protein complex in the human citrate (TCA) cycle pathway and the nicotinic acetylcholine receptor in the human central nervous system. Promising associations and potential drug targets were also provided for further studies and designs in therapeutic approaches for malaria at a systematic level. In conclusion, this method is efficient to identify new human-malaria protein associations and can be generalized to infer other types of association studies to further advance biomedical science.
Collapse
|
34
|
Ngassa Mbenda HG, Wang M, Guo J, Siddiqui FA, Hu Y, Yang Z, Kittichai V, Sattabongkot J, Cao Y, Jiang L, Cui L. Evolution of the Plasmodium vivax multidrug resistance 1 gene in the Greater Mekong Subregion during malaria elimination. Parasit Vectors 2020; 13:67. [PMID: 32051017 PMCID: PMC7017538 DOI: 10.1186/s13071-020-3934-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/03/2020] [Indexed: 11/10/2022] Open
Abstract
Background The malaria elimination plan of the Greater Mekong Subregion (GMS) is jeopardized by the increasing number of Plasmodium vivax infections and emergence of parasite strains with reduced susceptibility to the frontline drug treatment chloroquine/primaquine. This study aimed to determine the evolution of the P. vivax multidrug resistance 1 (Pvmdr1) gene in P. vivax parasites isolated from the China–Myanmar border area during the major phase of elimination. Methods Clinical isolates were collected from 275 P. vivax patients in 2008, 2012–2013 and 2015 in the China–Myanmar border area and from 55 patients in central China. Comparison was made with parasites from three border regions of Thailand. Results Overall, genetic diversity of the Pvmdr1 was relatively high in all border regions, and over the seven years in the China–Myanmar border, though slight temporal fluctuation was observed. Single nucleotide polymorphisms previously implicated in reduced chloroquine sensitivity were detected. In particular, M908L approached fixation in the China–Myanmar border area. The Y976F mutation sharply decreased from 18.5% in 2008 to 1.5% in 2012–2013 and disappeared in 2015, whereas F1076L steadily increased from 33.3% in 2008 to 77.8% in 2015. While neutrality tests suggested the action of purifying selection on the pvmdr1 gene, several likelihood-based algorithms detected positive as well as purifying selections operating on specific amino acids including M908L, T958M and F1076L. Fixation and selection of the nonsynonymous mutations are differently distributed across the three border regions and central China. Comparison with the global P. vivax populations clearly indicated clustering of haplotypes according to geographic locations. It is noteworthy that the temperate-zone parasites from central China were completely separated from the parasites from other parts of the GMS. Conclusions This study showed that P. vivax populations in the China–Myanmar border has experienced major changes in the Pvmdr1 residues proposed to be associated with chloroquine resistance, suggesting that drug selection may play an important role in the evolution of this gene in the parasite populations.![]()
Collapse
Affiliation(s)
- Huguette Gaelle Ngassa Mbenda
- Division of Infectious Diseases and International Medicine, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Meilian Wang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110001, China
| | - Jian Guo
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji School of Medicine, Shanghai, China
| | - Faiza Amber Siddiqui
- Division of Infectious Diseases and International Medicine, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Yue Hu
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan, China
| | - Zhaoqing Yang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan, China
| | - Veerayuth Kittichai
- 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
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110001, China
| | - Lubin Jiang
- Unit of Human Parasite Molecular and Cell Biology, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Liwang Cui
- Division of Infectious Diseases and International Medicine, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
35
|
Waldman BS, Schwarz D, Wadsworth MH, Saeij JP, Shalek AK, Lourido S. Identification of a Master Regulator of Differentiation in Toxoplasma. Cell 2020; 180:359-372.e16. [PMID: 31955846 PMCID: PMC6978799 DOI: 10.1016/j.cell.2019.12.013] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 10/28/2019] [Accepted: 12/10/2019] [Indexed: 01/06/2023]
Abstract
Toxoplasma gondii chronically infects a quarter of the world's population, and its recrudescence can cause life-threatening disease in immunocompromised individuals and recurrent ocular lesions in the immunocompetent. Acute-stage tachyzoites differentiate into chronic-stage bradyzoites, which form intracellular cysts resistant to immune clearance and existing therapies. The molecular basis of this differentiation is unknown, despite being efficiently triggered by stresses in culture. Through Cas9-mediated screening and single-cell profiling, we identify a Myb-like transcription factor (BFD1) necessary for differentiation in cell culture and in mice. BFD1 accumulates during stress and its synthetic expression is sufficient to drive differentiation. Consistent with its function as a transcription factor, BFD1 binds the promoters of many stage-specific genes and represents a counterpoint to the ApiAP2 factors that dominate our current view of parasite gene regulation. BFD1 provides a genetic switch to study and control Toxoplasma differentiation and will inform prevention and treatment of chronic infections.
Collapse
Affiliation(s)
- Benjamin S Waldman
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Dominic Schwarz
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Marc H Wadsworth
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02319, USA
| | - Jeroen P Saeij
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Alex K Shalek
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02319, USA
| | - Sebastian Lourido
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
36
|
Cowell AN, Winzeler EA. The genomic architecture of antimalarial drug resistance. Brief Funct Genomics 2019; 18:314-328. [PMID: 31119263 PMCID: PMC6859814 DOI: 10.1093/bfgp/elz008] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 03/19/2019] [Accepted: 04/09/2019] [Indexed: 11/15/2022] Open
Abstract
Plasmodium falciparum and Plasmodium vivax, the two protozoan parasite species that cause the majority of cases of human malaria, have developed resistance to nearly all known antimalarials. The ability of malaria parasites to develop resistance is primarily due to the high numbers of parasites in the infected person's bloodstream during the asexual blood stage of infection in conjunction with the mutability of their genomes. Identifying the genetic mutations that mediate antimalarial resistance has deepened our understanding of how the parasites evade our treatments and reveals molecular markers that can be used to track the emergence of resistance in clinical samples. In this review, we examine known genetic mutations that lead to resistance to the major classes of antimalarial medications: the 4-aminoquinolines (chloroquine, amodiaquine and piperaquine), antifolate drugs, aryl amino-alcohols (quinine, lumefantrine and mefloquine), artemisinin compounds, antibiotics (clindamycin and doxycycline) and a napthoquinone (atovaquone). We discuss how the evolution of antimalarial resistance informs strategies to design the next generation of antimalarial therapies.
Collapse
Affiliation(s)
- Annie N Cowell
- Division of Infectious Diseases and Global Health, Department of Medicine, University of California, San Diego, Gilman Dr., La Jolla, CA, USA
| | - Elizabeth A Winzeler
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California, San Diego, Gilman Dr., La Jolla, CA, USA
| |
Collapse
|
37
|
Sá JM, Kaslow SR, Moraes Barros RR, Brazeau NF, Parobek CM, Tao D, Salzman RE, Gibson TJ, Velmurugan S, Krause MA, Melendez-Muniz V, Kite WA, Han PK, Eastman RT, Kim A, Kessler EG, Abebe Y, James ER, Chakravarty S, Orr-Gonzalez S, Lambert LE, Engels T, Thomas ML, Fasinu PS, Serre D, Gwadz RW, Walker L, DeConti DK, Mu J, Bailey JA, Sim BKL, Hoffman SL, Fay MP, Dinglasan RR, Juliano JJ, Wellems TE. Plasmodium vivax chloroquine resistance links to pvcrt transcription in a genetic cross. Nat Commun 2019; 10:4300. [PMID: 31541097 PMCID: PMC6754410 DOI: 10.1038/s41467-019-12256-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/26/2019] [Indexed: 12/30/2022] Open
Abstract
Mainstay treatment for Plasmodium vivax malaria has long relied on chloroquine (CQ) against blood-stage parasites plus primaquine against dormant liver-stage forms (hypnozoites), however drug resistance confronts this regimen and threatens malaria control programs. Understanding the basis of P. vivax chloroquine resistance (CQR) will inform drug discovery and malaria control. Here we investigate the genetics of P. vivax CQR by a cross of parasites differing in drug response. Gametocytogenesis, mosquito infection, and progeny production are performed with mixed parasite populations in nonhuman primates, as methods for P. vivax cloning and in vitro cultivation remain unavailable. Linkage mapping of progeny surviving >15 mg/kg CQ identifies a 76 kb region in chromosome 1 including pvcrt, an ortholog of the Plasmodium falciparum CQR transporter gene. Transcriptional analysis supports upregulated pvcrt expression as a mechanism of CQR.
Collapse
Affiliation(s)
- Juliana M Sá
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sarah R Kaslow
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Roberto R Moraes Barros
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nicholas F Brazeau
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Christian M Parobek
- Curriculum in Genetics and Molecular Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Dingyin Tao
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Rebecca E Salzman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tyler J Gibson
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Michael A Krause
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Viviana Melendez-Muniz
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Whitney A Kite
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Paul K Han
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Richard T Eastman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Adam Kim
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Evan G Kessler
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | | | - Sachy Orr-Gonzalez
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lynn E Lambert
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Theresa Engels
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marvin L Thomas
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Pius S Fasinu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC, 27506, USA
| | - David Serre
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Robert W Gwadz
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Larry Walker
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC, 27506, USA
| | - Derrick K DeConti
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Jianbing Mu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jeffrey A Bailey
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC, 27506, USA
- Division of Transfusion Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | | | | | - Michael P Fay
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Rhoel R Dinglasan
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Jonathan J Juliano
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Genetics and Molecular Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Thomas E Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
38
|
Elgoraish AG, Elzaki SEG, Ahmed RT, Ahmed AI, Fadlalmula HA, Abdalgader Mohamed S, Abdallah NI, Abdelgadir O, Ageep TB, El-Sayed BB. Epidemiology and distribution of Plasmodium vivax malaria in Sudan. Trans R Soc Trop Med Hyg 2019; 113:517-524. [PMID: 31162590 DOI: 10.1093/trstmh/trz044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/18/2019] [Accepted: 05/06/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Plasmodium vivax malaria has been recognised as an important cause of morbidity in several African countries. The prevalence was previously estimated as 2-5% in eastern Sudan. These estimates are observed to be rising and spreading continuously. The present study was undertaken to investigate the situation of distribution and epidemiology of P. vivax malaria in Sudan. METHODS Cross-sectional malaria surveys carried out in hospitals and health centres covered 21 sites in 10 states. Data and blood samples were collected from 1226 clinically investigated suspected malaria cases of both genders and all ages. Microscopically detected malaria parasites were confirmed by PCR. RESULTS The overall prevalence of P. vivax among the malaria cases was 26.6%. The prevalence showed significant variations between the states (p<0.001), which could be explained by differences in population movement, the presence of refugees and proximity to endemic neighbouring countries. It also varied significantly with residence status (p<0.001), reflecting the stability of transmission. CONCLUSION Although malaria in Sudan is still largely attributed to Plasmodium falciparum, P. vivax has been rising with worrying proportions and spreading to new areas. The emergence and marked increase of P. vivax poses new challenges to malaria treatment and control in Sudan.
Collapse
Affiliation(s)
- Amanda G Elgoraish
- Department of Epidemiology, Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
| | - Salah Eldin G Elzaki
- Department of Epidemiology, Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
| | - Rania TagElsir Ahmed
- Department of Epidemiology, Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
| | - Arwa Ibrahim Ahmed
- Department of Epidemiology, Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
| | - Huda A Fadlalmula
- Department of Epidemiology, Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
| | - Shymaa Abdalgader Mohamed
- Department of Epidemiology, Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
- Department of Zoology, Faculty of Sciences, University of Khartoum, Khartoum, Sudan
| | - Nusiba I Abdallah
- Department of Epidemiology, Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
- National University Research Institute, National University, Khartoum, Sudan
| | - Osman Abdelgadir
- Department of Epidemiology, Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
- Department of Parasitology and Medical Entomology, Faculty of Medicine and Health Science, University of Kordofan, Elobeid, Sudan
| | - Tellal B Ageep
- Department of Epidemiology, Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
| | - Badria B El-Sayed
- Department of Epidemiology, Tropical Medicine Research Institute, National Centre for Research, Khartoum, Sudan
| |
Collapse
|
39
|
Development of a film-based immunochromatographic microfluidic device for malaria diagnosis. Biomed Microdevices 2019; 21:86. [PMID: 31451957 DOI: 10.1007/s10544-019-0431-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In this study, a novel film-based immunochromatographic microfluidic device (IMD) has been developed for malaria diagnosis. A microfluidic channel was patterned on a polyethylene terephthalate (PET) double-sided adhesive film using a plotting cutter and was assembled with a polycarbonate (PC) film. The PC film used for the probe immobilization layer was activated using oxygen plasma treatment to modify the film surface with avidin-biotin linker to immobilize a capture antibody. A fluorescent labeled Pan type mAb conjugate was prepared for signal indicator after undergoing a sandwich enzyme-linked immunosorbent assay (ELISA). Target antigens include Plasmodium falciparum (P. falciparum) lactate dehydrogenase (LDH) and Plasmodium vivax (P. vivax) LDH which were injected into the sample inlet. Target antigens combined with the conjugate and then flowed to the detection chamber where two test dots and a control dot (Ctrl) exist. In the presence of P. falciparum LDH, three detection dots including test dot 1 (T1), test dot 2 (T2) and Ctrl revealed fluorescence signals where P. falciparum mAb, Pan type pLDH mAb and goat anti-mouse IgG were immobilized, respectively. When P. vivax LDH was present, T2 and Ctrl dots showed fluorescence signals while no signal was detected with the negative control. P. falciparum LDH and P. vivax LDH were successfully detected on the IMD with a detection limit of 50 ng/mL and 100 ng/mL, respectively. The IMD provides a point-of-care diagnosis platform which is able to analyze pathogenic bacteria and viruses that can be applied in the field of clinical diagnosis and food safety testing.
Collapse
|
40
|
Plasmodium Genomics and Genetics: New Insights into Malaria Pathogenesis, Drug Resistance, Epidemiology, and Evolution. Clin Microbiol Rev 2019; 32:32/4/e00019-19. [PMID: 31366610 DOI: 10.1128/cmr.00019-19] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Protozoan Plasmodium parasites are the causative agents of malaria, a deadly disease that continues to afflict hundreds of millions of people every year. Infections with malaria parasites can be asymptomatic, with mild or severe symptoms, or fatal, depending on many factors such as parasite virulence and host immune status. Malaria can be treated with various drugs, with artemisinin-based combination therapies (ACTs) being the first-line choice. Recent advances in genetics and genomics of malaria parasites have contributed greatly to our understanding of parasite population dynamics, transmission, drug responses, and pathogenesis. However, knowledge gaps in parasite biology and host-parasite interactions still remain. Parasites resistant to multiple antimalarial drugs have emerged, while advanced clinical trials have shown partial efficacy for one available vaccine. Here we discuss genetic and genomic studies of Plasmodium biology, host-parasite interactions, population structures, mosquito infectivity, antigenic variation, and targets for treatment and immunization. Knowledge from these studies will advance our understanding of malaria pathogenesis, epidemiology, and evolution and will support work to discover and develop new medicines and vaccines.
Collapse
|
41
|
Verzier LH, Coyle R, Singh S, Sanderson T, Rayner JC. Plasmodium knowlesi as a model system for characterising Plasmodium vivax drug resistance candidate genes. PLoS Negl Trop Dis 2019; 13:e0007470. [PMID: 31158222 PMCID: PMC6564043 DOI: 10.1371/journal.pntd.0007470] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 06/13/2019] [Accepted: 05/15/2019] [Indexed: 12/29/2022] Open
Abstract
Plasmodium vivax causes the majority of malaria outside Africa, but is poorly understood at a cellular level partly due to technical difficulties in maintaining it in in vitro culture conditions. In the past decades, drug resistant P. vivax parasites have emerged, mainly in Southeast Asia, but while some molecular markers of resistance have been identified, none have so far been confirmed experimentally, which limits interpretation of the markers, and hence our ability to monitor and control the spread of resistance. Some of these potential markers have been identified through P. vivax genome-wide population genetic analyses, which highlighted genes under recent evolutionary selection in Southeast Asia, where chloroquine resistance is most prevalent. These genes could be involved in drug resistance, but no experimental proof currently exists to support this hypothesis. In this study, we used Plasmodium knowlesi, the most closely related species to P. vivax that can be cultured in human erythrocytes, as a model system to express P. vivax genes and test for their role in drug resistance. We adopted a strategy of episomal expression, and were able to express fourteen P. vivax genes, including two allelic variants of several hypothetical resistance genes. Their expression level and localisation were assessed, confirming cellular locations conjectured from orthologous species, and suggesting locations for several previously unlocalised proteins, including an apical location for PVX_101445. These findings establish P. knowlesi as a suitable model for P. vivax protein expression. We performed chloroquine and mefloquine drug assays, finding no significant differences in drug sensitivity: these results could be due to technical issues, or could indicate that these genes are not actually involved in drug resistance, despite being under positive selection pressure in Southeast Asia. These data confirm that in vitro P. knowlesi is a useful tool for studying P. vivax biology. Its close evolutionary relationship to P. vivax, high transfection efficiency, and the availability of markers for colocalisation, all make it a powerful model system. Our study is the first of its kind using P. knowlesi to study unknown P. vivax proteins and investigate drug resistance mechanisms.
Collapse
Affiliation(s)
- Lisa H. Verzier
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, United Kingdom
| | - Rachael Coyle
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, United Kingdom
| | - Shivani Singh
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, United Kingdom
| | - Theo Sanderson
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, United Kingdom
| | - Julian C. Rayner
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, United Kingdom
| |
Collapse
|
42
|
Aracil A, Green J. Plants with antimalarial properties: A systematic review of the current clinical evidence. Eur J Integr Med 2019. [DOI: 10.1016/j.eujim.2019.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
43
|
Malaria morbidity and mortality following introduction of a universal policy of artemisinin-based treatment for malaria in Papua, Indonesia: A longitudinal surveillance study. PLoS Med 2019; 16:e1002815. [PMID: 31167228 PMCID: PMC6541239 DOI: 10.1371/journal.pmed.1002815] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 04/30/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Malaria control activities can have a disproportionately greater impact on Plasmodium falciparum than on P. vivax in areas where both species are coendemic. We investigated temporal trends in malaria-related morbidity and mortality in Papua, Indonesia, before and after introduction of a universal, artemisinin-based antimalarial treatment strategy for all Plasmodium species. METHODS AND FINDINGS A prospective, district-wide malariometric surveillance system was established in April 2004 to record all cases of malaria at community clinics and the regional hospital and maintained until December 2013. In March 2006, antimalarial treatment policy was changed to artemisinin combination therapy for uncomplicated malaria and intravenous artesunate for severe malaria due to any Plasmodium species. Over the study period, a total of 418,238 patients presented to the surveillance facilities with malaria. The proportion of patients with malaria requiring admission to hospital fell from 26.9% (7,745/28,789) in the pre-policy change period (April 2004 to March 2006) to 14.0% (4,786/34,117) in the late transition period (April 2008 to December 2009), a difference of -12.9% (95% confidence interval [CI] -13.5% to -12.2%). There was a significant fall in the mortality of patients presenting to the hospital with P. falciparum malaria (0.53% [100/18,965] versus 0.32% [57/17,691]; difference = -0.21% [95% CI -0.34 to -0.07]) but not in patients with P. vivax malaria (0.28% [21/7,545] versus 0.23% [28/12,397]; difference = -0.05% [95% CI -0.20 to 0.09]). Between the same periods, the overall proportion of malaria due to P. vivax rose from 44.1% (30,444/69,098) to 53.3% (29,934/56,125) in the community clinics and from 32.4% (9,325/28,789) to 44.1% (15,035/34,117) at the hospital. After controlling for population growth and changes in treatment-seeking behaviour, the incidence of P. falciparum malaria fell from 511 to 249 per 1,000 person-years (py) (incidence rate ratio [IRR] = 0.49 [95% CI 0.48-0.49]), whereas the incidence of P. vivax malaria fell from 331 to 239 per 1,000 py (IRR = 0.72 [95% CI 0.71-0.73]). The main limitations of our study were possible confounding from changes in healthcare provision, a growing population, and significant shifts in treatment-seeking behaviour following implementation of a new antimalarial policy. CONCLUSIONS In this area with high levels of antimalarial drug resistance, adoption of a universal policy of efficacious artemisinin-based therapy for malaria infections due to any Plasmodium species was associated with a significant reduction in total malaria-attributable morbidity and mortality. The burden of P. falciparum malaria was reduced to a greater extent than that of P. vivax malaria. In coendemic regions, the timely elimination of malaria will require that safe and effective radical cure of both the blood and liver stages of the parasite is widely available for all patients at risk of malaria.
Collapse
|
44
|
Lawpoolsri S, Sattabongkot J, Sirichaisinthop J, Cui L, Kiattibutr K, Rachaphaew N, Suk-Uam K, Khamsiriwatchara A, Kaewkungwal J. Epidemiological profiles of recurrent malaria episodes in an endemic area along the Thailand-Myanmar border: a prospective cohort study. Malar J 2019; 18:124. [PMID: 30961583 PMCID: PMC6454765 DOI: 10.1186/s12936-019-2763-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/02/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND In low malaria transmission areas, many people acquire multiple malaria infections within a single season. This study aimed to describe the pattern and epidemiological profile of malaria recurrence in a hypoendemic area of western Thailand and identify factors associated with having multiple malaria episodes. METHODS An open cohort of 7000 residents in seven clusters along the Thai-Myanmar border was followed during a 6.5-year period (2011-mid 2017). Symptomatic and asymptomatic malaria infections were detected by passive case detection (PCD), weekly household visit, and mass blood surveys every 4-6 months. Malaria recurrence was defined as subsequent parasitaemic episodes occurred later than 7 days after receiving anti-malarial treatment. This study focused on analysis of recurrent episodes that occurred within 1 year after treatment. Numbers of malaria cases with single and multiple episodes were compared between clusters. Kaplan-Meier curve was performed to determine the intervals of recurrent episodes by Plasmodium species and age groups. The ordinal logistic model was used to determine factors associated with multiple malaria episodes, and to compare with single episodes, and those with no malaria infection. RESULTS The cumulative incidence of malaria in the study area was 5.2% over the 6.5 years. Overall, 410 malaria patients were detected. Of these patients, 20% and 16% had multiple malaria episodes during the entire period and within 1 year after initial treatment, respectively. About 80% of repeated malaria episodes were caused by the same Plasmodium species as the primary infections. The median interval and interquartile range (IQR) between the first and second episode was 88 (43-175) days for all parasites, 56 (35-133) days for two Plasmodium falciparum episodes, and 90 (59-204) days for two Plasmodium vivax episodes. The interval between the episodes was increased with age. Factors significantly associated with multiple episodes of malaria infection included male sex, young age, Karen ethnicity, forest-related occupation, and having other malaria infected persons in the same house in the same period. CONCLUSIONS People who have multiple malaria episodes may play an important role in maintaining malaria transmission in the area. Understanding epidemiological profiles of this group is important for planning strategies to achieve the elimination goal.
Collapse
Affiliation(s)
- Saranath Lawpoolsri
- Center of Excellence in Biomedical and Public Health Informatics (BIOPHICS), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand. .,Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jeeraphat Sirichaisinthop
- Bureau of Vector-Borne Diseases, Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | - Liwang Cui
- Division of Infectious Diseases and Internal Medicine, Department of Internal Medicine, University of South Florida, Tampa, USA
| | - Kirakorn Kiattibutr
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nattawan Rachaphaew
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kritsana Suk-Uam
- Vector-Borne Disease Control Center 2.3, Ministry of Public Health, Tak, Thailand
| | - Amnat Khamsiriwatchara
- Center of Excellence in Biomedical and Public Health Informatics (BIOPHICS), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jaranit Kaewkungwal
- Center of Excellence in Biomedical and Public Health Informatics (BIOPHICS), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
45
|
Phong NC, Chavchich M, Quang HH, San NN, Birrell GW, Chuang I, Martin NJ, Manh ND, Edstein MD. Susceptibility of Plasmodium falciparum to artemisinins and Plasmodium vivax to chloroquine in Phuoc Chien Commune, Ninh Thuan Province, south-central Vietnam. Malar J 2019; 18:10. [PMID: 30654808 PMCID: PMC6335800 DOI: 10.1186/s12936-019-2640-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/08/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Reduced artemisinin susceptibility and artemisinin-based combination therapy (ACT)-resistance against Plasmodium falciparum and chloroquine (CQ)-resistant P. vivax malaria has been reported in Vietnam. Two therapeutic efficacy studies were conducted in Thuan Bac District (Ninh Thuan Province, Vietnam) in 2015 and 2016 to determine the extent of reduced artemisinin susceptibility and ACT resistant falciparum malaria, and CQ-resistant vivax malaria were present. METHODS Twenty-seven patients with falciparum malaria were randomized to receive artesunate alone (AS ~ 4 mg/kg/day) for 4 days followed by dihydroartemisinin (DHA) (2.2 mg/kg)-piperaquine (PPQ) (18 mg/kg) daily for 3 days or artemether (AM) (1.7 mg/kg)-lumefantrine (LUM) (12 mg/kg) twice daily for 3 days. Sixteen subjects with vivax malaria received CQ (total 25 mg/kg over 3 days). The therapeutic efficacy study for treating falciparum malaria was complemented with molecular analysis for artemisinin and piperaquine resistance, and in vitro drug susceptibility testing. Patient's drug exposure following both falciparum and vivax treatment studies was determined. RESULTS Twenty-five of 27 patients treated with the artemisinin regimens completed the 42-day follow-up period. None had parasites present on day 3 after commencing treatment with no incidence of recrudescence (100% curative rate). One patient on AS + DHA-PPQ was lost to follow-up and one patient had Plasmodium falciparum and Plasmodium vivax infection on day 0 by PCR. Of the vivax patients, 15 of 16 completed CQ treatment and two had a recurrence of vivax malaria on day 28, a failure rate of 13.3% (2/15). No mutations in the Pfkelch-13 gene for artemisinin resistance or exo-E415G gene polymorphism and amplification in plasmepsins 2 and 3 for piperaquine resistance were observed. In vitro testing of patient's falciparum parasites indicated susceptibility (low IC50 nM values) to dihydroartemisinin, lumefantrine, piperaquine and pyronaridine. Patient's drug exposure to artesunate and lumefantrine was comparable to published data, however, blood CQ concentrations were lower. CONCLUSIONS Clinical findings, molecular analysis and in vitro testing revealed that the falciparum parasites at Phuoc Chien Commune were artemisinin susceptible. The clinical failure rate of the 15 vivax patients who completed CQ treatment was 13%. Further studies are required to determine whether CQ-resistant vivax malaria is present at the commune.
Collapse
Affiliation(s)
- Nguyen Chinh Phong
- Vietnam People's Army Military Institute of Preventive Medicine, Hanoi, Vietnam
| | - Marina Chavchich
- Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - Huynh Hong Quang
- Institute of Malariology, Parasitology and Entomology, Quy Nhon, Vietnam
| | - Nguyen Ngoc San
- Vietnam People's Army Military Institute of Preventive Medicine, Hanoi, Vietnam
| | - Geoffrey W Birrell
- Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - Ilin Chuang
- Naval Medical Research Center, Silver Spring, USA
| | | | - Nguyen Duc Manh
- Vietnam People's Army Military Institute of Preventive Medicine, Hanoi, Vietnam
| | - Michael D Edstein
- Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, Australia.
| |
Collapse
|
46
|
Oboh MA, Badiane AS, Ntadom G, Ndiaye YD, Diongue K, Diallo MA, Ndiaye D. Molecular identification of Plasmodium species responsible for malaria reveals Plasmodium vivax isolates in Duffy negative individuals from southwestern Nigeria. Malar J 2018; 17:439. [PMID: 30486887 PMCID: PMC6263541 DOI: 10.1186/s12936-018-2588-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/22/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Malaria in Nigeria is principally due to Plasmodium falciparum and, to a lesser extent to Plasmodium malariae and Plasmodium ovale. Plasmodium vivax is thought to be absent in Nigeria in particular and sub-Saharan Africa in general, due to the near fixation of the Duffy negative gene in this population. Nevertheless, there are frequent reports of P. vivax infection in Duffy negative individuals in the sub-region, including reports from two countries sharing border with Nigeria to the west (Republic of Benin) and east (Cameroon). Additionally, there were two cases of microscopic vivax-like malaria from Nigerian indigenous population. Hence molecular surveillance of the circulating Plasmodium species in two states (Lagos and Edo) of southwestern Nigeria was carried out. METHODS A cross-sectional survey between September 2016 and March 2017 was conducted. 436 febrile patients were included for the present work. Venous blood of these patients was subjected to RDT as well as microscopy. Further, parasite DNA was isolated from positive samples and PCR diagnostic was employed followed by direct sequencing of the 18S rRNA of Plasmodium species as well as sequencing of a portion of the promoter region of the Duffy antigen receptor for chemokines. Samples positive for P. vivax were re-amplified several times and finally using the High Fidelity Taq to rule out any bias introduced. RESULTS Of the 256 (58.7%) amplifiable malaria parasite DNA, P. falciparum was, as expected, the major cause of infection, either alone 85.5% (219/256; 97 from Edo and 122 from Lagos), or mixed with P. malariae 6.3% (16/256) or with P. vivax 1.6% (4/256). Only one of the five P. vivax isolates was found to be a single infection. DNA sequencing and subsequent alignment of the 18S rRNA of P. vivax with the reference strains displayed very high similarities (100%). Remarkably, the T-33C was identified in all P. vivax samples, thus confirming that all vivax-infected patients in the current study are Duffy negative. CONCLUSION The present study gave the first molecular evidence of P. vivax in Nigeria in Duffy negative individuals. Though restricted to two states; Edo in South-South and Lagos in South-west Nigeria, the real burden of this species in Nigeria and sub-Saharan Africa might have been underestimated, hence there is need to put in place a country-wide, as well as a sub-Saharan Africa-wide surveillance and appropriate control measures.
Collapse
MESH Headings
- Child, Preschool
- Cross-Sectional Studies
- DNA, Protozoan/chemistry
- DNA, Protozoan/genetics
- DNA, Ribosomal/chemistry
- DNA, Ribosomal/genetics
- Duffy Blood-Group System/genetics
- Female
- Humans
- Infant
- Infant, Newborn
- Malaria, Vivax/epidemiology
- Malaria, Vivax/parasitology
- Male
- Nigeria/epidemiology
- Plasmodium vivax/classification
- Plasmodium vivax/genetics
- Plasmodium vivax/isolation & purification
- RNA, Ribosomal, 18S/genetics
- Receptors, Cell Surface/genetics
- Sequence Analysis, DNA
Collapse
Affiliation(s)
- Mary Aigbiremo Oboh
- Parasitology and Mycology Laboratory, Université Cheikh Anta Diop, Dakar, Senegal.
| | - Aida Sadikh Badiane
- Parasitology and Mycology Laboratory, Université Cheikh Anta Diop, Dakar, Senegal
| | - Godwin Ntadom
- National Malaria Elimination Programme/Epidemiology Division, Department of Public Health, Federal Ministry of Health, Abuja, Nigeria
| | - Yaye Die Ndiaye
- Parasitology and Mycology Laboratory, Université Cheikh Anta Diop, Dakar, Senegal
| | - Khadim Diongue
- Parasitology and Mycology Laboratory, Université Cheikh Anta Diop, Dakar, Senegal
| | - Mamadou Alpha Diallo
- Parasitology and Mycology Laboratory, Université Cheikh Anta Diop, Dakar, Senegal
| | - Daouda Ndiaye
- Parasitology and Mycology Laboratory, Université Cheikh Anta Diop, Dakar, Senegal
| |
Collapse
|
47
|
Emran TB, Iyori M, Ono Y, Amelia F, Yusuf Y, Islam A, Alam A, Tamura M, Ogawa R, Matsuoka H, Yamamoto DS, Yoshida S. Baculovirus-Induced Fast-Acting Innate Immunity Kills Liver-Stage Plasmodium. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2441-2451. [PMID: 30209187 DOI: 10.4049/jimmunol.1800908] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/15/2018] [Indexed: 12/14/2022]
Abstract
Baculovirus (BV), an enveloped insect virus with a circular dsDNA genome, possesses unique characteristics that induce strong innate immune responses in mammalian cells. In this study, we show that BV administration in BALB/c mice not only provides complete protection against a subsequent Plasmodium berghei sporozoite infection for up to 7 d after the injection but also eliminates existing liver-stage parasites completely. The elimination of sporozoites by BV was superior to that by primaquine, and this effect occurred in a TLR9-independent manner. At 6 h after BV administration, IFN-α and IFN-γ were robustly produced in the serum, and RNA transcripts of IFN-stimulated genes were markedly upregulated in the liver compared with control mice. The in vivo passive transfer of serum after BV administration effectively eliminated liver-stage parasites, and IFN-α neutralization abolished this effect, indicating that the BV liver-stage parasite-killing mechanism is downstream of the type I IFN signaling pathway. These findings provide evidence that BV-induced, fast-acting innate immunity completely kills liver-stage parasites and, thus, may lead to new malaria drug and vaccine strategies.
Collapse
Affiliation(s)
- Talha Bin Emran
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yuki Ono
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Fitri Amelia
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Ashekul Islam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Asrar Alam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Megumi Tamura
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Ryohei Ogawa
- Department of Radiological Sciences, University of Toyama, Toyama 930-0194, Japan; and
| | - Hiroyuki Matsuoka
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0431, Japan
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0431, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan;
| |
Collapse
|
48
|
Rajendran V, Singh C, Ghosh PC. Improved efficacy of doxycycline in liposomes against Plasmodium falciparum in culture and Plasmodium berghei infection in mice. Can J Physiol Pharmacol 2018; 96:1145-1152. [PMID: 30075085 DOI: 10.1139/cjpp-2018-0067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The rate at which Plasmodium falciparum is developing resistance to clinically used antimalarial drugs is alarming. Therefore, there is a compelling need to develop an efficient drug delivery system to improve the efficacy of existing antimalarial agents and circumvent drug resistance. Here, we report the antibacterial drug doxycycline (DOXY) in liposomal formulations exhibits enhanced antiplasmodial activity against blood stage forms of P. falciparum (3D7) in culture and established Plasmodium berghei NK-65 infection in murine model. Parasite killing on blood stage forms in culture was determined by a radiolabeled [3H] hypoxanthine incorporation assay and infected erythrocytes stained with Giemsa were counted using microscopy in vivo. The 50% inhibitory concentration (IC50) of DOXY-stearylamine liposome (IC50 0.36 μM) and DOXY-SPC:Chol-liposome (IC50 0.85 μM) exhibited marked growth inhibition of parasites compared with free DOXY (IC50 14 μM), with minimal toxicity to normal erythrocytes. Administration of polyethylene glycol distearoyl phosphatidylethanolamine-methoxy-polyethylene glycol2000 (DSPE-mPEG-2000) coated liposomes loaded with DOXY at 2.5 mg/kg per day resulted in efficacious killing of blood parasites with improved survival in mice relative to the free drug in both chloroquine sensitive and resistant strains of P. berghei infection. This is the first report to demonstrate that DOXY in liposomal system has immense chemotherapeutic potential against plasmodial infections at lower dosages.
Collapse
Affiliation(s)
- Vinoth Rajendran
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India.,Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Chanchal Singh
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India.,Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Prahlad C Ghosh
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India.,Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| |
Collapse
|
49
|
Pandey RK, Ali M, Ojha R, Bhatt TK, Prajapati VK. Development of multi-epitope driven subunit vaccine in secretory and membrane protein of Plasmodium falciparum to convey protection against malaria infection. Vaccine 2018; 36:4555-4565. [PMID: 29921492 DOI: 10.1016/j.vaccine.2018.05.082] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/20/2018] [Accepted: 05/22/2018] [Indexed: 10/28/2022]
Abstract
Malaria infection is the severe health concern for a long time. As per the WHO reports, the malarial infection causes huge mortality all around the world and is incomparable with any other infectious diseases. The absence of effective treatment options and increasing drug resistance to the available therapeutics like artemisinin and other derivatives demand an efficient alternative to overcome this death burden. Here, we performed the literature survey and sorted the Plasmodium falciparum secretory and membrane proteins to design multi-epitope subunit vaccine using an adjuvant, B-cell- and T-cell epitopes. Every helper T-lymphocyte (HTL) epitope was IFN-γ positive and IL-4 non-inducer. The physicochemical properties, allergenicity, and antigenicity of designed vaccine were analyzed for the safety concern. Homology modeling and refinement were performed to obtain the functional tertiary structure of vaccine protein followed by its molecular docking with the toll-like receptor-4 (TLR-4) immune receptor. Molecular dynamics simulation was performed to check the interaction and stability of the receptor-ligand complex. Lastly, in silico cloning was performed to generate the restriction clone of designed vaccine for the futuristic expression in a microbial expression system. This way, we designed the multi-epitope subunit vaccine to serve the people living in the global endemic zone.
Collapse
Affiliation(s)
- Rajan Kumar Pandey
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer, Rajasthan 305817, India
| | - Mudassar Ali
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer, Rajasthan 305817, India
| | - Rupal Ojha
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer, Rajasthan 305817, India
| | - Tarun Kumar Bhatt
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer, Rajasthan 305817, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer, Rajasthan 305817, India.
| |
Collapse
|
50
|
Emran TB, Iyori M, Ono Y, Amelia F, Yusuf Y, Islam A, Alam A, Ogawa R, Matsuoka H, Yamamoto D, Yoshida S. Baculovirus-inducing fast-acting innate immunity kills Plasmodium liver stages.. [DOI: 10.1101/320036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
ABSTRACTBaculovirus (BV), an enveloped insect virus with a circular double-stranded DNA genome, possesses unique characteristics that induce strong innate immune responses in mammalian cells. Here, we show that BV administration not only sterilely protects BALB/c mice for at least 7 days from subsequent Plasmodium berghei sporozoite infection but also eliminates existing liver-stage parasites completely, effects superior to those of primaquine, and does so in a TLR9-independent manner. Six hours post-BV administration, IFN-α and IFN-γ were robustly produced in serum, and RNA transcripts of interferon-stimulated genes were drastically upregulated in the liver. The in vivo passive transfer of post-BV administration serum effectively eliminated liver-stage parasites, and IFN-α neutralization abolished this effect, indicating that the BV liver-stage parasite killing mechanism is downstream of the type I IFN signaling pathway. Our results demonstrate that BV is a potent IFN-inducing prophylactic and therapeutic agent with great potential for further development as a new malaria vaccine and/or anti-hypnozoite drug.
Collapse
|