1
|
Banda CG, Tarning J, Barnes KI. Use of population pharmacokinetic-pharmacodynamic modelling to inform antimalarial dose optimization in infants. Br J Clin Pharmacol 2025; 91:968-980. [PMID: 38858224 PMCID: PMC11992656 DOI: 10.1111/bcp.16132] [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: 10/13/2023] [Revised: 03/21/2024] [Accepted: 05/18/2024] [Indexed: 06/12/2024] Open
Abstract
Infants bear a significant malaria burden but are usually excluded from participating in early dose optimization studies that inform dosing regimens of antimalarial therapy. Unlike older children, infants' exclusion from early-phase trials has resulted in limited evidence to guide accurate dosing of antimalarial treatment for uncomplicated malaria or malaria-preventive treatment in this vulnerable population. Subsequently, doses used in infants are often extrapolated from older children or adults, with the potential for under- or overdosing. Population pharmacokinetic-pharmacodynamic (PK-PD) modelling, a quantitative methodology that applies mathematical and statistical techniques, can aid the design of clinical studies in infants that collect sparse pharmacokinetic data as well as support the analysis of such data to derive optimized antimalarial dosing in this complex and at-risk yet understudied subpopulation. In this review, we reflect on what PK-PD modelling can do in programmatic settings of most malaria-endemic areas and how it can be used to inform antimalarial dose optimization for preventive and curative treatment of uncomplicated malaria in infants. We outline key developmental physiological changes that affect drug exposure in early life, the challenges of conducting dose optimization studies in infants, and examples of how PK-PD modelling has previously informed antimalarial dose optimization in this subgroup. Additionally, we discuss the limitations and gaps of PK-PD modelling when used for dose optimization in infants. To utilize modelling well, there is a need to generate useful, sparse, PK and PD data in this subpopulation to inform antimalarial optimal dosing in infancy.
Collapse
Affiliation(s)
- Clifford G. Banda
- Malawi‐Liverpool‐Wellcome ProgrammeBlantyreMalawi
- Division of Clinical Pharmacology, Department of MedicineUniversity of Cape TownCape TownSouth Africa
- Kamuzu University of Health Sciences (formerly College of Medicine and Kamuzu College of Nursing, University of Malawi)BlantyreMalawi
| | - Joel Tarning
- Centre for Tropical Medicine and Global Health, Nuffield Department of MedicineUniversity of OxfordOxfordUK
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical MedicineMahidol UniversityBangkokThailand
| | - Karen I. Barnes
- Division of Clinical Pharmacology, Department of MedicineUniversity of Cape TownCape TownSouth Africa
- WorldWide Antimalarial Resistance Network (WWARN), Pharmacology Scientific GroupUniversity of Cape TownCape TownSouth Africa
| |
Collapse
|
2
|
Mwebaza N, Roh ME, Geng YZ, Opio L, Opira B, Marzan F, Mwima MW, Ssemukuye T, Guo K, Gingrich D, Fotaki N, Kakuru A, Kizza J, Aguti M, Adrama H, Kamya M, Dorsey G, Rosenthal PJ, Aweeka FT, Huang L. Drug-Drug Interaction Between Dihydroartemisinin-Piperaquine and Sulfadoxine-Pyrimethamine During Malaria Chemoprevention in Pregnant Women. Clin Pharmacol Ther 2025; 117:506-514. [PMID: 39402742 PMCID: PMC11747899 DOI: 10.1002/cpt.3471] [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: 06/06/2024] [Accepted: 09/21/2024] [Indexed: 11/20/2024]
Abstract
Co-administration of dihydroartemisinin-piperaquine (DP) and sulfadoxine-pyrimethamine (SP) for intermittent preventive treatment of malaria in pregnancy (IPTp) may be superior in preventing adverse birth outcomes compared with either therapy alone, but potential drug-drug interactions require investigation. We conducted intensive and sparse pharmacokinetic (PK) studies in a subset of Ugandan women participating in a randomized controlled trial of monthly IPTp with SP vs. DP vs. DP + SP. Intensive PK sampling was performed from day 0 to 23 after dosing at 28 weeks gestation in 87 participants across treatment arms. Sparse sampling was performed on day 28 (trough) after dosing at 20 and 28 gestational weeks in additional 196 participants receiving SP vs. DP + SP. Intensive PK analysis demonstrated that compared with SP alone, co-administration of DP + SP was associated with lower maximal concentrations, the area under the concentration-time curves (AUC), and day 23 concentrations of sulfadoxine (25%, 25%, and 27%) and pyrimethamine (26%, 34%, and 32%) (P < 0.05 for all comparisons). Sparse PK results demonstrated participants co-administered DP + SP had lower trough concentrations after dosing at 20 and 28 gestational weeks for sulfadoxine (6%, P = 0.68 and 31%, P = 0.023, respectively) and pyrimethamine (18%, P = 0.032 and 33%, P < 0.001, respectively) compared with SP alone. Co-administration of DP + SP was associated with a 19% reduction in piperaquine AUC (P = 0.046), but no significant difference in other PK parameters compared with DP alone. In summary, co-administration of DP + SP was associated with significantly reduced SP exposure, with a greater magnitude during the third vs. second trimester. The clinical consequences of this interaction are yet unknown.
Collapse
Affiliation(s)
- Norah Mwebaza
- Infectious Disease Research Collaboration, Kampala, Uganda
- Department of Pharmacology and Therapeutics, Makerere University, Kampala, Uganda
| | - Michelle E. Roh
- Institute for Global Health Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Yourong Z Geng
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - Leonard Opio
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Bishop Opira
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Florence Marzan
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - Moses W. Mwima
- Infectious Disease Research Collaboration, Kampala, Uganda
| | | | - Kevin Guo
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - David Gingrich
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - Nikoletta Fotaki
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - Abel Kakuru
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Jimmy Kizza
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Miriam Aguti
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Harriet Adrama
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Moses Kamya
- Infectious Disease Research Collaboration, Kampala, Uganda
- Department of Medicine, Makerere University, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, CA, USA
| | | | - Francesca T. Aweeka
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - Liusheng Huang
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| |
Collapse
|
3
|
Sok V, Marzan F, Roh M, Guo K, Legac J, Mwebaza N, Dorsey G, Rosenthal PJ, Aweeka FT, Huang L. Determination of sulfadoxine and pyrimethamine in microvolume human plasma using ultra high performance liquid chromatography-tandam mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1234:124030. [PMID: 38309043 PMCID: PMC11820842 DOI: 10.1016/j.jchromb.2024.124030] [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: 11/16/2023] [Revised: 12/31/2023] [Accepted: 01/22/2024] [Indexed: 02/05/2024]
Abstract
To support the pharmacokinetic study of sulfadoxine (SD) and pyrimethamine (PM) in pregnant women and children, sensitive methods with small sample volume are desirable. Here we report a method to determine SD and PM with microvolume plasma samples: 5 µL plasma samples were cleaned up by protein precipitation with acetonitrile. The deuterated analytes were used as the internal standards. The samples after cleanup were injected onto an ACE Excel SuperC18 column (50 × 2.1 mm, 1.7 μm, Hichrom Limited) connected to a Waters I class UPLC coupled with a Sciex Triple Quad 6500+ Mass Spectrometer and eluted with water and acetonitrile both containing 0.1% formic acid in a gradient mode at 0.8mL/min. Detection utilized ESI+ as the ion source and MRM as the quantification mode. The precursor-to-product ion transitions m/z 311→245 for SD and 249→233 for PM were selected for quantification. The ion transitions for the corresponding internal standards were 315→249 for SD-d4 and 254→235 for PM-d3. The simplest linear regression weighted by 1/x was used for the calibration curves. The calibration ranges were 1-200 µg/mL SD and 2 - 1000ng/mL PM. The mean (± standard deviation) recoveries were 94.3±3.2% (SD) and 97.0±1.5% (PM). The validated method was applied to analysis of 1719 clinical samples, demonstrating the method is suitable for the pharmacokinetic study with samples collected up to day 28 post-dose.
Collapse
Affiliation(s)
- Vong Sok
- University of California, Department of Clinical Pharmacy, San Francisco, CA 94110, USA
| | - Florence Marzan
- University of California, Department of Clinical Pharmacy, San Francisco, CA 94110, USA
| | - Michelle Roh
- University of California, Institute for Global Health Sciences, San Francisco, CA 94110, USA
| | - Kevin Guo
- University of California, Department of Clinical Pharmacy, San Francisco, CA 94110, USA
| | - Jenny Legac
- Department of Medicine, University of California, San Francisco, CA 94110, USA
| | - Norah Mwebaza
- Infectious Disease Research Collaboration, Kampala, Uganda; Departments of Clinical Pharmacology and Therapeutics, Makerere University, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, CA 94110, USA
| | - Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, CA 94110, USA
| | - Francesca T Aweeka
- University of California, Department of Clinical Pharmacy, San Francisco, CA 94110, USA
| | - Liusheng Huang
- University of California, Department of Clinical Pharmacy, San Francisco, CA 94110, USA.
| |
Collapse
|
4
|
Blessborn D, Kaewkhao N, Tarning J. A high-throughput LC-MS/MS assay for piperaquine from dried blood spots: Improving malaria treatment in resource-limited settings. J Mass Spectrom Adv Clin Lab 2024; 31:19-26. [PMID: 38229676 PMCID: PMC10789632 DOI: 10.1016/j.jmsacl.2023.12.004] [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: 06/21/2023] [Revised: 12/04/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024] Open
Abstract
Background Malaria is a parasitic disease that affects many of the poorest economies, resulting in approximately 241 million clinical episodes and 627,000 deaths annually. Piperaquine, when administered with dihydroartemisinin, is an effective drug against the disease. Drug concentration measurements taken on day 7 after treatment initiation have been shown to be a good predictor of therapeutic success with piperaquine. A simple capillary blood collection technique, where blood is dried onto filter paper, is especially suitable for drug studies in remote areas or resource-limited settings or when taking samples from children, toddlers, and infants. Methods Three 3.2 mm discs were punched out from a dried blood spot (DBS) and then extracted in a 96-well plate using solid phase extraction on a fully automated liquid handling system. The analysis was performed using LC-MS/MS with a calibration range of 3 - 1000 ng/mL. Results The recovery rate was approximately 54-72 %, and the relative standard deviation was below 9 % for low, middle and high quality control levels. The LC-MS/MS quantification limit of 3 ng/mL is sensitive enough to detect piperaquine for up to 4-8 weeks after drug administration, which is crucial when evaluating recrudescence and drug resistance development. While different hematocrit levels can affect DBS drug measurements, the effect was minimal for piperaquine. Conclusion A sensitive LC-MS/MS method, in combination with fully automated extraction in a 96-well plate format, was developed and validated for the quantification of piperaquine in DBS. The assay was implemented in a bioanalytical laboratory for processing large-scale clinical trial samples.
Collapse
Affiliation(s)
- Daniel Blessborn
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Natpapat Kaewkhao
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Joel Tarning
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
5
|
Ali AM, Wallender E, Hughes E, Dorsey G, Savic RM. Interplay among malnutrition, chemoprevention, and the risk of malaria in young Ugandan children: Longitudinal pharmacodynamic and growth analysis. CPT Pharmacometrics Syst Pharmacol 2023; 12:656-667. [PMID: 36919202 DOI: 10.1002/psp4.12892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/30/2022] [Accepted: 10/06/2022] [Indexed: 03/16/2023] Open
Abstract
African children are at risk of malaria and malnutrition. We quantified relationships between malaria and malnutrition among young Ugandan children in a high malaria transmission region. Data were used from a randomized controlled trial where Ugandan HIV-unexposed (n = 393) and HIV-exposed (n = 186) children were randomized to receive no malaria chemoprevention, monthly sulfadoxine-pyrimethamine, daily trimethoprim-sulfamethoxazole, or monthly dihydroartemisinin-piperaquine (DP) from age 6-24 months, and then were followed off chemoprevention until age 36 months. Monthly height and weight, and time of incident malaria episodes were obtained; 89 children who received DP contributed piperaquine (PQ) concentrations. Malaria hazard was modeled using parametric survival analysis adjusted for repeated events, and height and weight were modeled using a Brody growth model. Among 579 children, stunting (height-for-age z-score [ZHA] < -2) was associated with a 17% increased malaria hazard (95% confidence interval [CI] 10-23%) compared with children with a ZHA of zero. DP was associated with a 35% lower malaria hazard (hazard ratio [HR] [95% CI], 0.65 [0.41-0.97]), compared to no chemoprevention. After accounting for PQ levels, stunted children who received DP had 2.1 times the hazard of malaria (HR [95% CI] 2.1 [1.6-3.0]) compared with children with a ZHA of zero who received DP. Each additional malaria episode was associated with a 0.4% reduced growth rate for height. Better dosing regimens are needed to optimize malaria prevention in malnourished populations, but, importantly, malaria chemoprevention may reduce the burden of malnutrition in early childhood.
Collapse
Affiliation(s)
- Ali Mohamed Ali
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA.,Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Erika Wallender
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, California, USA
| | - Emma Hughes
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Radojka M Savic
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
6
|
Mwankuna CJ, Kiros F, Mariki EE, Mabiki FP, Malebo HM, Mdegela RH, Styrishave B. Optimization of HPLC-MS/MS method for determination of antimalarial adulterants in herbal products. ANAL SCI 2023; 39:407-416. [PMID: 36633808 DOI: 10.1007/s44211-022-00255-8] [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: 09/30/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023]
Abstract
The use of herbal products is booming all over the world because of being believed as safer than conventional drugs and free of side effects. However, there are untrustworthy manufacturers who adulterate herbal products by adding conventional drugs which might eventually lead to microbial resistance and herb-to-drug interactions. There is a need to develop methods for detecting adulterants in herbal products. A high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) method for simultaneous identification and determination of conventional antimalarials (chloroquine, quinine, sulfadoxine, pyrimethamine, mefloquine, lumefantrine, amodiaquine, artemisinin, dihydroartemisinin, artesunate and artemether) in herbal products was developed. Stable isotopically labelled compounds (artemether-d3, quindine-d3, and sulfadoxine-d3) were used as internal standards (ISs) for quantitative analysis. Extraction of analytes was performed using methanol: water: formic acid (90:10:0.1, v/v) and chromatographic separation was done in a gradient mode using mobile phase A: Ultrapure water containing 0.1% formic acid and 1 mM ammonium formate and mobile phase B: Acetonitrile/methanol (50:50) containing 0.1% formic acid and 1 mM ammonium formate. The calibration curves were linear (r2 ≥ 0.991) over the range of 0.001-0.3 µg mL-1 for all compounds. The limit of detection (LOD) ranged from 0.002 to 0.02 μg mL-1 while the limit of quantification (LOQ) ranged from 0.006 to 0.08 μg mL-1. Accuracy, expressed as recovery of spiked herbal products ranged from 52 to 128%. The precision, expressed as percent relative standard deviation (%RSD) at two concentration levels, ranged from 1.0 to 13.8%. The matrix effect expressed as the matrix factor (MF) ranged from 0.77 to 0.97. The developed method was used to identify and quantify conventional antimalarials in herbal product samples from Tanzania. Ten out of 50 herbal products were found to contain amodiaquine, sulfadoxine, pyrimethamine, mefloquine, dihydroartemisinin, artemether and lumefantrine. The developed method is considered a valuable tool for getting a better understanding of the adulteration of conventional antimalarials in herbal products.
Collapse
Affiliation(s)
- Christopher J Mwankuna
- Department of Chemistry and Physics, College of Natural and Applied Sciences, Sokoine University of Agriculture, P.O. Box 3038, Morogoro, Tanzania.
| | - Feven Kiros
- Toxicology Laboratory, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark Universitetsparken 2, DK-2100, Copenhagen, Denmark
| | - Eliapenda E Mariki
- Department of Chemistry and Physics, College of Natural and Applied Sciences, Sokoine University of Agriculture, P.O. Box 3038, Morogoro, Tanzania
| | - Faith P Mabiki
- Department of Chemistry and Physics, College of Natural and Applied Sciences, Sokoine University of Agriculture, P.O. Box 3038, Morogoro, Tanzania
| | - Hamisi M Malebo
- UNESCO National Commission of the United Republic of Tanzania, 7 Magogoni Street, P.O. Box 20384, Dar Es Salaam, Tanzania
| | - Robinson H Mdegela
- Department of Veterinary Medicine and Public Health, College of Veterinary Medicine and Biomedical Sciences, Sokoine University of Agriculture, P.O. Box 3015, Morogoro, Tanzania
| | - Bjarne Styrishave
- Toxicology Laboratory, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark Universitetsparken 2, DK-2100, Copenhagen, Denmark
| |
Collapse
|
7
|
Determination of unbound piperaquine in human plasma by ultra-high performance liquid chromatography tandem mass spectrometry. JOURNAL OF CHROMATOGRAPHY OPEN 2022; 2. [PMID: 35531322 PMCID: PMC9068709 DOI: 10.1016/j.jcoa.2022.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Piperaquine (PQ) is an antimalarial drug that is highly protein-bound. Variation in plasma protein contents may affect the pharmacokinetic (PK) exposure of unbound drug, leading to alteration of clinical outcomes. All published methods for determination of PQ in human plasma measure the total PQ including both bound and unbound PQ to plasma proteins. There is no published method for unbound PQ determination. Here we report an ultra-high performance liquid chromatography tandem mass spectrometry (UHPLC-MS/MS) method for determination of PQ in human plasma filtrate prepared by filtering human plasma through Millipore Microcon® centrifugal filters (10k NMWL). The filter cup had to be treated with 5% benzalkonium chloride to reduce non-specific binding to the filter devices before filtration of plasma samples. Multiple reactions monitoring (MRM) of the ion pairs m/z 535/288 for PQ and m/z 541/294 for the internal standard (IS) was selected for quantification. When electrospray ionization (ESI+) was used, paradoxical matrix effect was observed despite the structure similarity of the deuterated IS: Ion suppression for PQ versus ion enhancement for the PQ-d6, even though they were closely eluted: 0.62 min versus 0.61 min. Separation was achieved on Evo C18 column (50 × 2.1 mm, 1.7 μm, Phenomenex Inc.) eluted with 10 mM NH4OH and MeCN. When atmospheric pressure chemical ionization in positive mode (APCI+) was used for ion source, matrix effect diminished. Separation was achieved on a PFP column (30 × 2.1 mm, 1.7 μm, Waters, Corp.) eluted with aqueous 20 mM ammonium formate 0.14% trifluoroacetic acid (A) and methanol-acetonitrile (4:1, v/v) containing 0.1% trifluoroacetic acid (B) at 0.8 mL/min flow rate in a gradient mode: 30–30–80–80–30–30%B (0–0.1–1.0–1.40–1.41–1.50 min). The retention time was 0.67 min for both PQ and the IS. The method was validated with a linear calibration range from 20 to 5,000 pg/mL and applied to clinical samples.
Collapse
|
8
|
Phipps WS, Greene DN, Pflaum H, Laha TJ, Dickerson JA, Irvine J, Merrill AE, Ranjitkar P, Henderson CM, Hoofnagle AN. Small volume retinol binding protein measurement by liquid chromatography-tandem mass spectrometry. Clin Biochem 2022; 99:111-117. [PMID: 34678307 PMCID: PMC8671195 DOI: 10.1016/j.clinbiochem.2021.10.005] [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: 07/23/2021] [Revised: 10/13/2021] [Accepted: 10/17/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The measurement of plasma concentrations of retinol binding protein is a component of nutritional assessment in neonatal intensive care. However, serial testing in newborns is hampered by the limited amount of blood that can be sampled. Limitations are most severe with preterm infants, for whom close monitoring may be most important. METHODS We developed an assay to quantify retinol binding protein using trypsin digestion and liquid chromatography-tandem mass spectrometry, which requires a serum or plasma volume of 5 µl. Additionally, we validated the method according to current recommendations and performed comparison with a standard nephelometry platform in clinical use. RESULTS The assay demonstrated linearity from below 1 mg/dL (0.48 µM) to more than 20 mg/dL (9.7 µM), and an imprecision of 11.8% at 0.43 mg/dL (0.21 µM). The distribution of results observed with the new method was different when compared with nephelometry. CONCLUSION Liquid chromatography-tandem mass spectrometry facilitated testing a smaller sample volume, thereby increasing the ability to monitor key nutritional markers in premature infants. The differences in results compared with a commercially-available nephelometric assay revealed questionable results for lower concentrations by immunoassay.
Collapse
Affiliation(s)
- William S. Phipps
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Dina N. Greene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Hannah Pflaum
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Thomas J. Laha
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Jane A. Dickerson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA,Seattle Children’s Hospital, Seattle, WA
| | - Jill Irvine
- University of Washington Medical Center, Seattle, WA
| | - Anna E. Merrill
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Pratistha Ranjitkar
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Clark M. Henderson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Andrew N. Hoofnagle
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA,Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
9
|
Development and validation of an LC-MS/MS method for determination of hydroxychloroquine, its two metabolites, and azithromycin in EDTA-treated human plasma. PLoS One 2021; 16:e0247356. [PMID: 33667247 PMCID: PMC7935301 DOI: 10.1371/journal.pone.0247356] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/02/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Hydroxychloroquine (HCQ) and azithromycin (AZM) are antimalarial drugs recently reported to be active against severe acute respiratory syndrome coronavirus- 2 (SARS-CoV-2), which is causing the global COVID-19 pandemic. In an emergency response to the pandemic, we aimed to develop a quantitation method for HCQ, its metabolites desethylhydroxychloroquine (DHCQ) and bisdesethylchloroquine (BDCQ), and AZM in human plasma. METHODS Liquid chromatography tandem mass spectrometry was used to develop the method. Samples (20 μL) are extracted by solid-phase extraction and injected onto the LC-MS/MS system equipped with a PFP column (2.0 × 50 mm, 3 μm). ESI+ and MRM are used for detection. Ion pairs m/z 336.1→247.1 for HCQ, 308.1→179.1 for DHCQ, 264.1→179.1 for BDCQ, and 749.6→591.6 for AZM are selected for quantification. The ion pairs m/z 342.1→253.1, 314.1→181.1, 270.1→181.1, and 754.6→596.6 are selected for the corresponding deuterated internal standards (IS) HCQ-d4, DHCQ-d4, BDCQ-d4, and AZM-d5. The less abundant IS ions from 37Cl were used to overcome the interference from the analytes. RESULTS Under optimized conditions, retention times are 0.78 min for BDCQ, 0.79 min for DHCQ, 0.92 min for HCQ and 1.87 min for AZM. Total run time is 3.5 min per sample. The calibration ranges are 2-1000 ng/mL for HCQ and AZM, 1-500 ng/mL for DHCQ and 0.5-250 ng/mL for BDCQ; samples above the range are validated for up to 10-fold dilution. Recoveries of the method ranged from 88.9-94.4% for HCQ, 88.6-92.9% for DHCQ, 88.7-90.9% for BDCQ, and 98.6%-102% for AZM. The IS normalized matrix effect were within (100±10) % for all 4 analytes. Blood samples are stable for at least 6 hr at room temperature. Plasma samples are stable for at least 66 hr at room temperature, 38 days at -70°C, and 4 freeze-thaw cycles. CONCLUSIONS An LC-MS/MS method for simultaneous quantitation of HCQ, DHCQ, BDCQ, and AZM in human plasma was developed and validated for clinical studies requiring fast turnaround time and small samples volume.
Collapse
|
10
|
Chutvirasakul B, Joseph JF, Parr MK, Suntornsuk L. Development and applications of liquid chromatography-mass spectrometry for simultaneous analysis of anti-malarial drugs in pharmaceutical formulations. J Pharm Biomed Anal 2020; 195:113855. [PMID: 33406473 DOI: 10.1016/j.jpba.2020.113855] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 01/17/2023]
Abstract
The objective of this work was to develop a high-performance liquid chromatographic method coupled with a mass spectrometer (LC-MS) for the simultaneous analysis of artemisinin-based drugs (e.g. artemisinin, dihydroartemisinin, artesunate, artemether) and piperaquine in formulations. Simultaneous separation of the investigated drugs was achieved in 14 min on a C18 column (2.1 mm x 100 mm, particle size 1.8 μm) using a gradient elution of 0.05 % v/v formic acid in water and acetonitrile. MS detection was done in a positive ionization mode using electrospray ionization with acquisition in a single ion monitoring mode. Proper diluent and storage time in an autosampler played significant roles on the quantitation accuracy since the target analytes possessed varied solubility and stability in aqueous and organic solvents. The method was fully validated according to ICH guideline and data showed good linearity (R > 0.999, precision (RSD < 3.89 %) and accuracy (%recovery between 98.5 and 103.7) with low limits of detection (LOD < 24.7 ng/mL) and quantitation (LOQ < 82.40 ng/mL). Validation data indicated that the developed LC-MS method is fit for the intended purpose and was successfully applied to evaluate the drug contents in formulations. Among the tested samples, the percent labeled amounts found were between 93.1 and 105.0 % and one supplement capsule contained 0.039 %w/w of artemisinin. The newly developed method could benefit both the quality control departments in pharmaceutical industries and the authorities working on falsified drug problems since official methods for the analysis of these drugs are not available in pharmacopoeias. The method is fast and environmentally friendly due to the requirement of less chemicals and production of less wastes.
Collapse
Affiliation(s)
- Boonta Chutvirasakul
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayudhaya Rd., Rajathevee, Bangkok, 10400, Thailand; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Srinakharinwirot University, 63 Moo 7 Rangsit-Nakornnayok Road, Ongkharak, Nakornnayok, 26120, Thailand
| | - Jan Felix Joseph
- Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry (Pharmaceutical Analysis), Freie Universität Berlin, Berlin, Germany; Core Facility BiosupraMol, Department of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Maria Kristina Parr
- Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry (Pharmaceutical Analysis), Freie Universität Berlin, Berlin, Germany
| | - Leena Suntornsuk
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayudhaya Rd., Rajathevee, Bangkok, 10400, Thailand.
| |
Collapse
|
11
|
Piperaquine Exposure Is Altered by Pregnancy, HIV, and Nutritional Status in Ugandan Women. Antimicrob Agents Chemother 2020; 64:AAC.01013-20. [PMID: 33020153 DOI: 10.1128/aac.01013-20] [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: 05/18/2020] [Accepted: 09/29/2020] [Indexed: 11/20/2022] Open
Abstract
Dihydroartemisinin-piperaquine (DHA-PQ) provides highly effective therapy and chemoprevention for malaria in pregnant African women. PQ concentrations of >10.3 ng/ml have been associated with reduced maternal parasitemia, placental malaria, and improved birth outcomes. We characterized the population pharmacokinetics (PK) of PQ in a post hoc analysis of human immunodeficiency virus (HIV)-infected and -uninfected pregnant women receiving DHA-PQ as chemoprevention every 4 or 8 weeks. The effects of covariates such as pregnancy, nutritional status (body mass index [BMI]), and efavirenz (EFV)-based antiretroviral therapy were investigated. PQ concentrations from two chemoprevention trials were pooled to create a population PK database from 274 women and 2,218 PK observations. A three-compartment model with an absorption lag best fit the data. Consistent with our prior intensive PK evaluation, pregnancy and EFV use resulted in a 72% and 61% increased PQ clearance, compared to postpartum and HIV-uninfected pregnant women, respectively. Low BMI at 28 weeks of gestation was associated with increased clearance (2% increase per unit decrease in BMI). Low-BMI women given DHA-PQ every 8 weeks had a higher prevalence of parasitemia, malaria infection, and placental malaria compared to women with higher BMIs. The reduced piperaquine exposure in women with low BMI as well as during EFV coadministration, compared to pregnant women with higher BMIs and not taking EFV, suggests that these populations could benefit from weekly instead of monthly dosing for prevention of malaria parasitemia. Simulations indicated that because of the BMI-clearance relationship, weight-based regimens would not improve protection compared to a 2,880 mg fixed-dose regimen when provided monthly. (The clinical trials described in this paper have been registered at ClinicalTrials.gov under identifiers NCT02163447 and NCT02282293.).
Collapse
|
12
|
Mwebaza N, Cheah V, Forsman C, Kajubi R, Marzan F, Wallender E, Dorsey G, Rosenthal PJ, Aweeka F, Huang L. Determination of piperaquine concentration in human plasma and the correlation of capillary versus venous plasma concentrations. PLoS One 2020; 15:e0233893. [PMID: 32470030 PMCID: PMC7259774 DOI: 10.1371/journal.pone.0233893] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/14/2020] [Indexed: 11/20/2022] Open
Abstract
Background A considerable challenge in quantification of the antimalarial piperaquine in plasma is carryover of analyte signal between assays. Current intensive pharmacokinetic studies often rely on the merging of venous and capillary sampling. Drug levels in capillary plasma may be different from those in venous plasma, Thus, correlation between capillary and venous drug levels needs to be established. Methods Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) was used to develop the method. Piperaquine was measured in 205 pairs of capillary and venous plasma samples collected simultaneously at ≥24hr post dose in children, pregnant women and non-pregnant women receiving dihydroartemisinin-piperaquine as malaria chemoprevention. Standard three-dose regimen over three days applied to all participants with three 40mg dihydroartemisinin/320mg PQ tablets per dose for adults and weight-based dose for children. Correlation analysis was performed using the program Stata® SE12.1. Linear regression models were built using concentrations or logarithm transformed concentrations and the final models were selected based on maximal coefficient of determination (R2) and visual check. Results An LC-MS/MS method was developed and validated, utilizing methanol as a protein precipitation agent, a Gemini C18 column (50x2.0mm, 5μm) eluted with basic mobile phase solvents (ammonium hydroxide as the additive), and ESI+ as the ion source. This method had a calibration range of 10–1000 ng/mL and carryover was negligible. Correlation analysis revealed a linear relationship: Ccap = 1.04×Cven+4.20 (R2 = 0.832) without transformation of data, and lnCcap = 1.01×lnCven+0.0125, (R2 = 0.945) with natural logarithm transformation. The mean ratio (±SD) of Ccap/Cven was 1.13±0.42, and median (IQR) was 1.08 (0.917, 1.33). Conclusions Capillary and venous plasma PQ measures are nearly identical overall, but not readily exchangeable due to large variation. Further correlation study accounting for disposition phases may be necessary.
Collapse
Affiliation(s)
- Norah Mwebaza
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Vincent Cheah
- Drug Research Unit, Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, United States of America
| | - Camilla Forsman
- Drug Research Unit, Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, United States of America
| | - Richard Kajubi
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Florence Marzan
- Drug Research Unit, Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, United States of America
| | - Erika Wallender
- Drug Research Unit, Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States of America
| | - Grant Dorsey
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States of America
| | - Philip J. Rosenthal
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States of America
| | - Francesca Aweeka
- Drug Research Unit, Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, United States of America
| | - Liusheng Huang
- Drug Research Unit, Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, United States of America
- * E-mail:
| |
Collapse
|
13
|
Savic RM, Jagannathan P, Kajubi R, Huang L, Zhang N, Were M, Kakuru A, Muhindo MK, Mwebaza N, Wallender E, Clark TD, Opira B, Kamya M, Havlir DV, Rosenthal PJ, Dorsey G, Aweeka FT. Intermittent Preventive Treatment for Malaria in Pregnancy: Optimization of Target Concentrations of Dihydroartemisinin-Piperaquine. Clin Infect Dis 2019; 67:1079-1088. [PMID: 29547881 DOI: 10.1093/cid/ciy218] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 03/09/2018] [Indexed: 11/13/2022] Open
Abstract
Background Dihydroartemisinin-piperaquine (DHA-PQ) is highly efficacious as intermittent preventive therapy for malaria during pregnancy (IPTp). Determining associations between piperaquine (PQ) exposure, malaria risk, and adverse birth outcomes informs optimal dosing strategies. Methods Human immunodeficiency virus-uninfected pregnant women (n = 300) were enrolled in a placebo-controlled trial of IPTp at 12-20 weeks' gestation and randomized to sulfadoxine-pyrimethamine every 8 weeks, DHA-PQ every 8 weeks, or DHA-PQ every 4 weeks during pregnancy. Pharmacokinetic sampling for PQ was performed every 4 weeks, and an intensive pharmacokinetic substudy was performed in 30 women at 28 weeks' gestation. Concentration-effect relationships were assessed between exposure to PQ; the prevalence of Plasmodium falciparum infection during pregnancy; outcomes at delivery including placental malaria, low birth weight, and preterm birth; and risks for toxicity. Simulations of new dosing scenarios were performed. Results Model-defined PQ target venous plasma concentrations of 13.9 ng/mL provided 99% protection from P. falciparum infection during pregnancy. Each 10-day increase in time above target PQ concentrations was associated with reduced odds of placental parasitemia, preterm birth, and low birth weight, though increases in PQ concentrations were associated with QT interval prolongation. Modeling suggests that daily or weekly administration of lower dosages of PQ, compared to standard dosing, will maintain PQ trough levels above target concentrations with reduced PQ peak levels, potentially limiting toxicity. Conclusions The protective efficacy of IPTp with DHA-PQ was strongly associated with higher drug exposure. Studies of the efficacy and safety of alternative DHA-PQ IPTp dosing strategies are warranted. Clinical Trials Registration NCT02163447.
Collapse
Affiliation(s)
- Rada M Savic
- Department of Bioengineering and Therapeutic Sciences
| | - Prasanna Jagannathan
- Department of Medicine, University of California, San Francisco.,Department of Medicine, Stanford University, California
| | - Richard Kajubi
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Liusheng Huang
- Department of Clinical Pharmacy, University of California, San Francisco
| | - Nan Zhang
- Department of Bioengineering and Therapeutic Sciences
| | - Moses Were
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Mary K Muhindo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Norah Mwebaza
- Department of Pharmacology and Therapeutics, Kampala, Uganda
| | - Erika Wallender
- Department of Medicine, University of California, San Francisco
| | - Tamara D Clark
- Department of Medicine, University of California, San Francisco
| | - Bishop Opira
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Moses Kamya
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | | | | | - Grant Dorsey
- Department of Medicine, Stanford University, California
| | | |
Collapse
|
14
|
Wallender E, Vucicevic K, Jagannathan P, Huang L, Natureeba P, Kakuru A, Muhindo M, Nakalembe M, Havlir D, Kamya M, Aweeka F, Dorsey G, Rosenthal PJ, Savic RM. Predicting Optimal Dihydroartemisinin-Piperaquine Regimens to Prevent Malaria During Pregnancy for Human Immunodeficiency Virus-Infected Women Receiving Efavirenz. J Infect Dis 2019; 217:964-972. [PMID: 29272443 DOI: 10.1093/infdis/jix660] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background A monthly treatment course of dihydroartemisinin-piperaquine (DHA-PQ) effectively prevents malaria during pregnancy. However, a drug-drug interaction pharmacokinetic (PK) study found that pregnant human immunodeficiency virus (HIV)-infected women receiving efavirenz-based antiretroviral therapy (ART) had markedly reduced piperaquine (PQ) exposure. This suggests the need for alternative DHA-PQ chemoprevention regimens in this population. Methods Eighty-three HIV-infected pregnant women who received monthly DHA-PQ and efavirenz contributed longitudinal PK and corrected QT interval (QTc) (n = 25) data. Population PK and PK-QTc models for PQ were developed to consider the benefits (protective PQ coverage) and risks (QTc prolongation) of alternative DHA-PQ chemoprevention regimens. Protective PQ coverage was defined as maintaining a concentration >10 ng/mL for >95% of the chemoprevention period. Results PQ clearance was 4540 L/day. With monthly DHA-PQ (2880 mg PQ), <1% of women achieved defined protective PQ coverage. Weekly (960 mg PQ) or low-dose daily (320 or 160 mg PQ) regimens achieved protective PQ coverage for 34% and >96% of women, respectively. All regimens were safe, with ≤2% of women predicted to have ≥30 msec QTc increase. Conclusions For HIV-infected pregnant women receiving efavirenz, low daily DHA-PQ dosing was predicted to improve protection against parasitemia and reduce risk of toxicity compared to monthly dosing. Clinical Trials Registration NCT02282293.
Collapse
Affiliation(s)
- Erika Wallender
- Department of Medicine, University of California, San Francisco
| | - Katarina Vucicevic
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco.,Department of Pharmacokinetics and Clinical Pharmacy, Faculty of Pharmacy, University of Belgrade, Serbia
| | | | - Liusheng Huang
- Department of Clinical Pharmacy, University of California, San Francisco
| | - Paul Natureeba
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Mary Muhindo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Diane Havlir
- Department of Medicine, University of California, San Francisco
| | - Moses Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda.,Makerere University College of Health Sciences, Kampala, Uganda
| | - Francesca Aweeka
- Department of Clinical Pharmacy, University of California, San Francisco
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco
| | | | - Radojka M Savic
- Department of Medicine, University of California, San Francisco.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco
| |
Collapse
|
15
|
Whalen ME, Kajubi R, Chamankhah N, Huang L, Orukan F, Wallender E, Kamya MR, Dorsey G, Jagannathan P, Rosenthal PJ, Mwebaza N, Aweeka FT. Reduced Exposure to Piperaquine, Compared to Adults, in Young Children Receiving Dihydroartemisinin-Piperaquine as Malaria Chemoprevention. Clin Pharmacol Ther 2019; 106:1310-1318. [PMID: 31173649 DOI: 10.1002/cpt.1534] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/07/2019] [Indexed: 11/10/2022]
Abstract
Dihydroartemisinin (DHA)-piperaquine is being evaluated as intermittent preventive therapy for malaria, but dosing has not been optimized for children. We assessed exposure to DHA and piperaquine in Ugandan children at two ages during infancy. Intensive sampling was performed in 32 children at 32 weeks of age, 31 children at 104 weeks, and 30 female adult controls. Compared with adults, DHA area under the concentration-time curve (AUC0-8 hr ) was 52% higher at 32 weeks and comparable at 104 weeks. Compared with adults, piperaquine AUC0-21 d was 35% lower at 32 weeks and 53% lower at 104 weeks. Terminal piperaquine concentrations on days 7, 14, and 21 were lower in children compared with adults and lower at 104 compared with 32 weeks. Piperaquine exposure was lower in young children compared with adults, and lower at 104 compared with 32 weeks of age, suggesting a need for age-based DHA-piperaquine dose optimization for chemoprevention.
Collapse
Affiliation(s)
- Meghan E Whalen
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Richard Kajubi
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda.,Department of Pharmacology and Therapeutics, Makerere University College of Health Sciences, Kampala, Uganda
| | - Nona Chamankhah
- Department of Pharmacy, Rady Children's Hospital, San Diego, California, USA
| | - Liusheng Huang
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Francis Orukan
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Erika Wallender
- Department of Medicine, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Moses R Kamya
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | | | - Philip J Rosenthal
- Department of Medicine, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Norah Mwebaza
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda.,Department of Pharmacology and Therapeutics, Makerere University College of Health Sciences, Kampala, Uganda
| | - Francesca T Aweeka
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| |
Collapse
|
16
|
Modeling Prevention of Malaria and Selection of Drug Resistance with Different Dosing Schedules of Dihydroartemisinin-Piperaquine Preventive Therapy during Pregnancy in Uganda. Antimicrob Agents Chemother 2019; 63:AAC.01393-18. [PMID: 30530597 DOI: 10.1128/aac.01393-18] [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] [Received: 07/02/2018] [Accepted: 11/24/2018] [Indexed: 11/20/2022] Open
Abstract
Dihydroartemisinin-piperaquine (DHA-PQ) is under study for intermittent preventive treatment during pregnancy (IPTp), but it may accelerate selection for drug resistance. Understanding the relationships between piperaquine concentration, prevention of parasitemia, and selection for decreased drug sensitivity can inform control policies and optimization of DHA-PQ dosing. Piperaquine concentrations, measures of parasitemia, and Plasmodium falciparum genotypes associated with decreased aminoquinoline sensitivity in Africa (pfmdr1 86Y, pfcrt 76T) were obtained from pregnant Ugandan women randomized to IPTp with sulfadoxine-pyrimethamine (SP) or DHA-PQ. Joint pharmacokinetic/pharmacodynamic models described relationships between piperaquine concentration and the probability of genotypes of interest using nonlinear mixed effects modeling. An increase in the piperaquine plasma concentration was associated with a log-linear decrease in risk of parasitemia. Our models predicted that higher median piperaquine concentrations would be required to provide 99% protection against mutant infections than against wild-type infections (pfmdr1: N86, 9.6 ng/ml; 86Y, 19.6 ng/ml; pfcrt: K76, 6.5 ng/ml; 76T, 19.6 ng/ml). Comparing monthly, weekly, and daily dosing, daily low-dose DHA-PQ was predicted to result in the fewest infections and the fewest mutant infections per 1,000 pregnancies (predicted mutant infections for pfmdr1 86Y: SP monthly, 607; DHA-PQ monthly, 198; DHA-PQ daily, 1; for pfcrt 76T: SP monthly, 1,564; DHA-PQ monthly, 283; DHA-PQ daily, 1). Our models predict that higher piperaquine concentrations are needed to prevent infections with the pfmdr1/pfcrt mutant compared to those with wild-type parasites and that, despite selection for mutants by DHA-PQ, the overall burden of mutant infections is lower for IPTp with DHA-PQ than for IPTp with SP. (This study has been registered at ClinicalTrials.gov under identifier NCT02282293.).
Collapse
|
17
|
Jagannathan P, Kakuru A, Okiring J, Muhindo MK, Natureeba P, Nakalembe M, Opira B, Olwoch P, Nankya F, Ssewanyana I, Tetteh K, Drakeley C, Beeson J, Reiling L, Clark TD, Rodriguez-Barraquer I, Greenhouse B, Wallender E, Aweeka F, Prahl M, Charlebois ED, Feeney ME, Havlir DV, Kamya MR, Dorsey G. Dihydroartemisinin-piperaquine for intermittent preventive treatment of malaria during pregnancy and risk of malaria in early childhood: A randomized controlled trial. PLoS Med 2018; 15:e1002606. [PMID: 30016328 PMCID: PMC6049882 DOI: 10.1371/journal.pmed.1002606] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 06/08/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Intermittent preventive treatment of malaria in pregnancy (IPTp) with dihydroartemisinin-piperaquine (IPTp-DP) has been shown to reduce the burden of malaria during pregnancy compared to sulfadoxine-pyrimethamine (IPTp-SP). However, limited data exist on how IPTp regimens impact malaria risk during infancy. We conducted a double-blinded randomized controlled trial (RCT) to test the hypothesis that children born to mothers given IPTp-DP would have a lower incidence of malaria during infancy compared to children born to mothers who received IPTp-SP. METHODS AND FINDINGS We compared malaria metrics among children in Tororo, Uganda, born to women randomized to IPTp-SP given every 8 weeks (SP8w, n = 100), IPTp-DP every 8 weeks (DP8w, n = 44), or IPTp-DP every 4 weeks (DP4w, n = 47). After birth, children were given chemoprevention with DP every 12 weeks from 8 weeks to 2 years of age. The primary outcome was incidence of malaria during the first 2 years of life. Secondary outcomes included time to malaria from birth and time to parasitemia following each dose of DP given during infancy. Results are reported after adjustment for clustering (twin gestation) and potential confounders (maternal age, gravidity, and maternal parasitemia status at enrolment).The study took place between June 2014 and May 2017. Compared to children whose mothers were randomized to IPTp-SP8w (0.24 episodes per person year [PPY]), the incidence of malaria was higher in children born to mothers who received IPTp-DP4w (0.42 episodes PPY, adjusted incidence rate ratio [aIRR] 1.92; 95% CI 1.00-3.65, p = 0.049) and nonsignificantly higher in children born to mothers who received IPT-DP8w (0.30 episodes PPY, aIRR 1.44; 95% CI 0.68-3.05, p = 0.34). However, these associations were modified by infant sex. Female children whose mothers were randomized to IPTp-DP4w had an apparently 4-fold higher incidence of malaria compared to female children whose mothers were randomized to IPTp-SP8w (0.65 versus 0.20 episodes PPY, aIRR 4.39, 95% CI 1.87-10.3, p = 0.001), but no significant association was observed in male children (0.20 versus 0.28 episodes PPY, aIRR 0.66, 95% CI 0.25-1.75, p = 0.42). Nonsignificant increases in malaria incidence were observed among female, but not male, children born to mothers who received DP8w versus SP8w. In exploratory analyses, levels of malaria-specific antibodies in cord blood were similar between IPTp groups and sex. However, female children whose mothers were randomized to IPTp-DP4w had lower mean piperaquine (PQ) levels during infancy compared to female children whose mothers received IPTp-SP8w (coef 0.81, 95% CI 0.65-1.00, p = 0.048) and male children whose mothers received IPTp-DP4w (coef 0.72, 95% CI 0.57-0.91, p = 0.006). There were no significant sex-specific differences in PQ levels among children whose mothers were randomized to IPTp-SP8w or IPTp-DP8w. The main limitations were small sample size and childhood provision of DP every 12 weeks in infancy. CONCLUSIONS Contrary to our hypothesis, preventing malaria in pregnancy with IPTp-DP in the context of chemoprevention with DP during infancy does not lead to a reduced incidence of malaria in childhood; in this setting, it may be associated with an increased incidence of malaria in females. Future studies are needed to better understand the biological mechanisms of in utero drug exposure on drug metabolism and how this may affect the dosing of antimalarial drugs for treatment and prevention during infancy. TRIAL REGISTRATION ClinicalTrials.gov number NCT02163447.
Collapse
MESH Headings
- Adolescent
- Adult
- Antimalarials/administration & dosage
- Antimalarials/adverse effects
- Artemisinins/administration & dosage
- Artemisinins/adverse effects
- Child, Preschool
- Double-Blind Method
- Drug Administration Schedule
- Drug Combinations
- Female
- Humans
- Incidence
- Infant
- Infant, Newborn
- Infectious Disease Transmission, Vertical/prevention & control
- Malaria, Falciparum/epidemiology
- Malaria, Falciparum/parasitology
- Malaria, Falciparum/prevention & control
- Malaria, Falciparum/transmission
- Pregnancy
- Pregnancy Complications, Parasitic/epidemiology
- Pregnancy Complications, Parasitic/parasitology
- Pregnancy Complications, Parasitic/prevention & control
- Pyrimethamine/administration & dosage
- Pyrimethamine/adverse effects
- Quinolines/administration & dosage
- Quinolines/adverse effects
- Sulfadoxine/administration & dosage
- Sulfadoxine/adverse effects
- Time Factors
- Treatment Outcome
- Uganda/epidemiology
- Young Adult
Collapse
Affiliation(s)
- Prasanna Jagannathan
- Department of Medicine, Stanford University, Stanford, California, United States of America
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Jaffer Okiring
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Paul Natureeba
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Miriam Nakalembe
- Department of Obstetrics and Gynecology, Makerere University College of Health Sciences, Kampala, Uganda
| | - Bishop Opira
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Peter Olwoch
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | - Kevin Tetteh
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Chris Drakeley
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | | | - Tamara D. Clark
- Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
| | - Isabel Rodriguez-Barraquer
- Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
| | - Bryan Greenhouse
- Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
| | - Erika Wallender
- Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
| | - Francesca Aweeka
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, California, United States of America
| | - Mary Prahl
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
| | - Edwin D. Charlebois
- Center for AIDS Prevention Studies, University of California, San Francisco, San Francisco, California, United States of America
| | - Margaret E. Feeney
- Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
| | - Diane V. Havlir
- Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
| | - Moses R. Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
| |
Collapse
|
18
|
Sun Y, Wang S, Ji J, Zhai G, Xing J. Metabolite identification of the antimalarial naphthoquine using liquid chromatography-tandem high-resolution mass spectrometry in combination with multiple data-mining tools. Biomed Chromatogr 2018; 32:e4207. [DOI: 10.1002/bmc.4207] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Yanhong Sun
- School of Pharmaceutical Sciences; Shandong University; Jinan China
| | - Shuqi Wang
- School of Pharmaceutical Sciences; Shandong University; Jinan China
| | - Jianbo Ji
- School of Pharmaceutical Sciences; Shandong University; Jinan China
| | - Guangxi Zhai
- School of Pharmaceutical Sciences; Shandong University; Jinan China
| | - Jie Xing
- School of Pharmaceutical Sciences; Shandong University; Jinan China
| |
Collapse
|
19
|
Conrad MD, Mota D, Foster M, Tukwasibwe S, Legac J, Tumwebaze P, Whalen M, Kakuru A, Nayebare P, Wallender E, Havlir DV, Jagannathan P, Huang L, Aweeka F, Kamya MR, Dorsey G, Rosenthal PJ. Impact of Intermittent Preventive Treatment During Pregnancy on Plasmodium falciparum Drug Resistance-Mediating Polymorphisms in Uganda. J Infect Dis 2017; 216:1008-1017. [PMID: 28968782 PMCID: PMC5853776 DOI: 10.1093/infdis/jix421] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/17/2017] [Indexed: 11/13/2022] Open
Abstract
Background In a recent trial of intermittent preventive treatment in pregnancy (IPTp) in Uganda, dihydroartemisinin-piperaquine (DP) was superior to sulfadoxine-pyrimethamine (SP) in preventing maternal and placental malaria. Methods We compared genotypes using sequencing, fluorescent microsphere, and quantitative polymerase chain reaction assays at loci associated with drug resistance in Plasmodium falciparum isolated from subjects receiving DP or SP. Results Considering aminoquinoline resistance, DP was associated with increased prevalences of mutations at pfmdr1 N86Y, pfmdr1 Y184F, and pfcrt K76T compared to SP (64.6% vs 27.4%, P < .001; 93.9% vs 59.2%, P < .001; and 87.7% vs 75.4%, P = .03, respectively). Increasing plasma piperaquine concentration at the time of parasitemia was associated with increasing pfmdr1 86Y prevalence; no infections with the N86 genotype occurred with piperaquine >2.75 ng/mL. pfkelch13 propeller domain polymorphisms previously associated with artemisinin resistance were not identified. Recently identified markers of piperaquine resistance were uncommon and not associated with DP. Considering antifolate resistance, SP was associated with increased prevalence of a 5-mutation haplotype (pfdhfr 51I, 59R, and 108N; pfdhps 437G and 581G) compared to DP (90.8% vs 60.0%, P = .001). Conclusions IPTp selected for genotypes associated with decreased sensitivity to treatment regimens, but genotypes associated with clinically relevant DP resistance in Asia have not emerged in Uganda.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | | | - Prasanna Jagannathan
- University of California, San Francisco,Stanford University, Palo Alto, California
| | | | | | - Moses R Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda,Makerere University College of Health Sciences, Kampala, Uganda
| | | | - Philip J Rosenthal
- University of California, San Francisco,Correspondence: P. J. Rosenthal, University of California, San Francisco, Box 0811, San Francisco, CA 94143 ()
| |
Collapse
|
20
|
LC-MS/MS quantitation of antimalarial drug piperaquine and metabolites in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1063:253-258. [PMID: 28863865 DOI: 10.1016/j.jchromb.2017.06.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/31/2017] [Accepted: 06/18/2017] [Indexed: 11/22/2022]
Abstract
PURPOSE This study aimed to develop a sensitive, quantitative assay for the antimalarial piperaquine (PQ) and its metabolites M1 and M2 in human plasma. RESULTS Analytes were gradiently separated on a C18 column and detected with a Sciex API 4000 MS/MS with an ESI source operated in the positive ion mode with deuterated PQ as internal standard. The response was linear in the range 3.9-2508nM with a runtime of 7.0min per sample. The method was applied to clinical samples from healthy volunteers. CONCLUSION This LC-MS/MS method for the simultaneous quantitation of PQ and two of its metabolites in plasma may prove helpful for assessment of metabolite safety issues in vivo.
Collapse
|
21
|
Kajubi R, Huang L, Jagannathan P, Chamankhah N, Were M, Ruel T, Koss CA, Kakuru A, Mwebaza N, Kamya M, Havlir D, Dorsey G, Rosenthal PJ, Aweeka FT. Antiretroviral Therapy With Efavirenz Accentuates Pregnancy-Associated Reduction of Dihydroartemisinin-Piperaquine Exposure During Malaria Chemoprevention. Clin Pharmacol Ther 2017; 102:520-528. [PMID: 28187497 PMCID: PMC5546920 DOI: 10.1002/cpt.664] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 02/02/2017] [Accepted: 02/03/2017] [Indexed: 11/08/2022]
Abstract
Dihydroartemisinin (DHA)-piperaquine is promising for malaria chemoprevention in pregnancy. We assessed the impacts of pregnancy and efavirenz-based antiretroviral therapy on exposure to DHA and piperaquine in pregnant Ugandan women. Intensive sampling was performed at 28 weeks gestation in 31 HIV-uninfected pregnant women, in 27 HIV-infected pregnant women receiving efavirenz, and in 30 HIV-uninfected nonpregnant women. DHA peak concentration and area under the concentration time curve (AUC0-8hr ) were 50% and 47% lower, respectively, and piperaquine AUC0-21d was 40% lower in pregnant women compared to nonpregnant women. DHA AUC0-8hr and piperaquine AUC0-21d were 27% and 38% lower, respectively, in pregnant women receiving efavirenz compared to HIV-uninfected pregnant women. Exposure to DHA and piperaquine were lower among pregnant women and particularly in women on efavirenz, suggesting a need for dose modifications. The study of modified dosing strategies for these populations is urgently needed.
Collapse
Affiliation(s)
- Richard Kajubi
- Infectious Disease Research Collaboration, Makerere University
College of Health Sciences- all in Kampala, Uganda
- Department of Pharmacology and Therapeutics, Makerere University
College of Health Sciences- all in Kampala, Uganda
| | - Liusheng Huang
- Department of Clinical Pharmacy, University of California, San
Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - Prasanna Jagannathan
- Department of Medicine, University of California, San Francisco, San
Francisco General Hospital, San Francisco, CA, USA
| | - Nona Chamankhah
- Department of Clinical Pharmacy, University of California, San
Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - Moses Were
- Infectious Disease Research Collaboration, Makerere University
College of Health Sciences- all in Kampala, Uganda
| | - Theodore. Ruel
- Department of Pediatrics, University of California, San Francisco,
San Francisco General Hospital, San Francisco, CA, USA
| | - Catherine A. Koss
- Department of Medicine, University of California, San Francisco, San
Francisco General Hospital, San Francisco, CA, USA
| | - Abel Kakuru
- Infectious Disease Research Collaboration, Makerere University
College of Health Sciences- all in Kampala, Uganda
| | - Norah Mwebaza
- Infectious Disease Research Collaboration, Makerere University
College of Health Sciences- all in Kampala, Uganda
- Department of Pharmacology and Therapeutics, Makerere University
College of Health Sciences- all in Kampala, Uganda
| | - Moses Kamya
- Infectious Disease Research Collaboration, Makerere University
College of Health Sciences- all in Kampala, Uganda
| | - Diane Havlir
- Department of Medicine, University of California, San Francisco, San
Francisco General Hospital, San Francisco, CA, USA
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, San
Francisco General Hospital, San Francisco, CA, USA
| | - Philip J. Rosenthal
- Department of Medicine, University of California, San Francisco, San
Francisco General Hospital, San Francisco, CA, USA
| | - Francesca T. Aweeka
- Department of Clinical Pharmacy, University of California, San
Francisco, San Francisco General Hospital, San Francisco, CA, USA
| |
Collapse
|
22
|
Liu H, Zang M, Yang A, Ji J, Xing J. Simultaneous determination of piperaquine and its N-oxidated metabolite in rat plasma using LC-MS/MS. Biomed Chromatogr 2017; 31. [PMID: 28299804 DOI: 10.1002/bmc.3974] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/06/2017] [Accepted: 03/13/2017] [Indexed: 01/05/2023]
Abstract
A sensitive and efficient liquid chromatography tandem mass spectrometry method was developed and validated for the simultaneous determination of piperaquine (PQ) and its N-oxidated metabolite (PQ-M) in plasma. A simple protein precipitation procedure was used for sample preparation. Adequate chromatographic retention was achieved on a C18 column under gradient elution with acetonitrile and 2 mm aqueous ammonium acetate containing 0.15% formic acid and 0.05% trifluoroacetic acid. A triple-quadrupole mass spectrometer equipped with an electrospray source was set up in the positive ion mode and multiple reaction monitoring mode. The method was linear in the range of 2.0-400.0 ng/mL for PQ and 1.0-50.0 ng/mL for PQ-M with suitable accuracy, precision and extraction recovery. The lower limits of detection (LLOD) were established at 0.4 and 0.2 ng/mL for PQ and PQ-M, respectively, using 40 μL of plasma sample. The matrix effect was negligible under the current conditions. No effect was found for co-administrated artemisinin drugs or hemolysis on the quantification of PQ and PQ-M. Stability testing showed that two analytes remained stable under all relevant analytical conditions. The validated method was successfully applied to a pharmacokinetic study performed in rats after a single oral administration of PQ (60 mg/kg).
Collapse
Affiliation(s)
- Huixiang Liu
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Meitong Zang
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Aijuan Yang
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Jianbo Ji
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Jie Xing
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
23
|
Jagannathan P, Bowen K, Nankya F, McIntyre TI, Auma A, Wamala S, Sikyomu E, Naluwu K, Nalubega M, Boyle MJ, Farrington LA, Bigira V, Kapisi J, Aweeka F, Greenhouse B, Kamya M, Dorsey G, Feeney ME. Effective Antimalarial Chemoprevention in Childhood Enhances the Quality of CD4+ T Cells and Limits Their Production of Immunoregulatory Interleukin 10. J Infect Dis 2016; 214:329-38. [PMID: 27067196 DOI: 10.1093/infdis/jiw147] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 04/04/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Experimental inoculation of viable Plasmodium falciparum sporozoites administered with chemoprevention targeting blood-stage parasites results in protective immunity. It is unclear whether chemoprevention similarly enhances immunity following natural exposure to malaria. METHODS We assessed P. falciparum-specific T-cell responses among Ugandan children who were randomly assigned to receive monthly dihydroartemisinin-piperaquine (DP; n = 87) or no chemoprevention (n = 90) from 6 to 24 months of age, with pharmacologic assessments for adherence, and then clinically followed for an additional year. RESULTS During the intervention, monthly DP reduced malaria episodes by 55% overall (P < .001) and by 97% among children who were highly adherent to DP (P < .001). In the year after the cessation of chemoprevention, children who were highly adherent to DP had a 55% reduction in malaria incidence as compared to children given no chemoprevention (P = .004). Children randomly assigned to receive DP had higher frequencies of blood-stage specific CD4(+) T cells coproducing interleukin-2 and tumor necrosis factor α (P = .003), which were associated with protection from subsequent clinical malaria and parasitemia, and fewer blood-stage specific CD4(+) T cells coproducing interleukin-10 and interferon γ (P = .001), which were associated with increased risk of malaria. CONCLUSIONS In this setting, effective antimalarial chemoprevention fostered the development of CD4(+) T cells that coproduced interleukin 2 and tumor necrosis factor α and were associated with prospective protection, while limiting CD4(+) T-cell production of the immunoregulatory cytokine IL-10.
Collapse
Affiliation(s)
| | | | | | | | - Ann Auma
- Infectious Diseases Research Collaboration
| | | | | | | | | | - Michelle J Boyle
- Department of Medicine, San Francisco General Hospital Center for Biomedical Research, The Burnet Institute, Melbourne, Australia
| | | | | | | | | | | | - Moses Kamya
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, San Francisco General Hospital
| | - Margaret E Feeney
- Department of Medicine, San Francisco General Hospital Department of Pediatrics, University of California-San Francisco
| |
Collapse
|
24
|
Yang A, Zang M, Liu H, Fan P, Xing J. Metabolite identification of the antimalarial piperaquinein vivousing liquid chromatography-high-resolution mass spectrometry in combination with multiple data-mining tools in tandem. Biomed Chromatogr 2016; 30:1324-30. [DOI: 10.1002/bmc.3689] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/22/2015] [Accepted: 01/24/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Aijuan Yang
- School of Pharmaceutical Sciences; Shandong University; Jinan China
| | - Meitong Zang
- School of Pharmaceutical Sciences; Shandong University; Jinan China
| | - Huixiang Liu
- School of Pharmaceutical Sciences; Shandong University; Jinan China
| | - Peihong Fan
- School of Pharmaceutical Sciences; Shandong University; Jinan China
| | - Jie Xing
- School of Pharmaceutical Sciences; Shandong University; Jinan China
| |
Collapse
|
25
|
Farrington LA, Jagannathan P, McIntyre TI, Vance HM, Bowen K, Boyle MJ, Nankya F, Wamala S, Auma A, Nalubega M, Sikyomu E, Naluwu K, Bigira V, Kapisi J, Dorsey G, Kamya MR, Feeney ME. Frequent Malaria Drives Progressive Vδ2 T-Cell Loss, Dysfunction, and CD16 Up-regulation During Early Childhood. J Infect Dis 2015; 213:1483-90. [PMID: 26667315 DOI: 10.1093/infdis/jiv600] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/04/2015] [Indexed: 12/13/2022] Open
Abstract
γδ T cells expressing Vδ2 may be instrumental in the control of malaria, because they inhibit the replication of blood-stage parasites in vitro and expand during acute malaria infection. However, Vδ2 T-cell frequencies and function are lower among children with heavy prior malaria exposure. It remains unclear whether malaria itself is driving this loss. Here we measure Vδ2 T-cell frequency, cytokine production, and degranulation longitudinally in Ugandan children enrolled in a malaria chemoprevention trial from 6 to 36 months of age. We observed a progressive attenuation of the Vδ2 response only among children incurring high rates of malaria. Unresponsive Vδ2 T cells were marked by expression of CD16, which was elevated in the setting of high malaria transmission. Moreover, chemoprevention during early childhood prevented the development of dysfunctional Vδ2 T cells. These observations provide insight into the role of Vδ2 T cells in the immune response to chronic malaria.
Collapse
Affiliation(s)
| | | | - Tara I McIntyre
- Departments of Medicine, University of California San Francisco
| | - Hilary M Vance
- Departments of Medicine, University of California San Francisco
| | - Katherine Bowen
- Departments of Medicine, University of California San Francisco
| | - Michelle J Boyle
- Departments of Medicine, University of California San Francisco Center for Biomedical Research, The Burnet Institute, Melbourne, Victoria, Australia
| | - Felistas Nankya
- Infectious Diseases Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Samuel Wamala
- Infectious Diseases Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Ann Auma
- Infectious Diseases Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Mayimuna Nalubega
- Infectious Diseases Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Esther Sikyomu
- Infectious Diseases Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Kate Naluwu
- Infectious Diseases Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Victor Bigira
- Infectious Diseases Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - James Kapisi
- Infectious Diseases Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Grant Dorsey
- Departments of Medicine, University of California San Francisco
| | - Moses R Kamya
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Margaret E Feeney
- Departments of Medicine, University of California San Francisco Pediatrics, University of California San Francisco
| |
Collapse
|
26
|
Sundell K, Jagannathan P, Huang L, Bigira V, Kapisi J, Kakuru MM, Savic R, Kamya MR, Dorsey G, Aweeka F. Variable piperaquine exposure significantly impacts protective efficacy of monthly dihydroartemisinin-piperaquine for the prevention of malaria in Ugandan children. Malar J 2015; 14:368. [PMID: 26403465 PMCID: PMC4582734 DOI: 10.1186/s12936-015-0908-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/15/2015] [Indexed: 11/10/2022] Open
Abstract
Background Anti-malarial chemoprevention with dihydroartemisinin-piperaquine (DHA/PQ) is a promising tool for malaria control, but its efficacy in children may be limited by inadequate drug exposure. Methods Children were enrolled in a non directly-observed trial of DHA/PQ chemoprevention in a high transmission setting in Uganda. Children were randomized at 6 months of age to no chemoprevention (n = 89) or monthly DHA/PQ (n = 87) and followed through 24 months of age, with pharmacokinetic sampling performed at variable times following monthly dosing of DHA/PQ. A previously published pharmacokinetic model was used to estimate piperaquine (PQ) exposure in each child, and associations between PQ exposure and the protective efficacy (PE) of DHA/PQ were explored. Results The incidence of malaria was 6.83 and 3.09 episodes per person year at risk in the no chemoprevention and DHA/PQ arms, respectively (PE 54 %, 95 % CI 39–66 %, P < 0.001). Among children randomized to DHA/PQ, 493 pharmacokinetic samples were collected. Despite nearly 100 % reported adherence to study drug administration at home, there was wide variability in PQ exposure, and children were stratified into three groups based on average PQ exposure during the intervention that was determined by model generated percentiles (low, n = 40; medium, n = 37, and high, n = 10). Gender and socioeconomic factors were not significantly associated with PQ exposure. In multivariate models, the PE of DHA/PQ was 31 % in the low PQ exposure group (95 % CI 6–49 %, P = 0.02), 67 % in the medium PQ exposure group (95 % CI 54–76 %, P < 0.001), and 97 % in the high PQ exposure group (95 % CI 89–99 %, P < 0.001). Conclusions The protective efficacy of DHA/PQ chemoprevention in young children was strongly associated with higher drug exposure; in children with the highest PQ exposure, monthly DHA/PQ chemoprevention was nearly 100 % protective against malaria. Strategies to ensure good adherence to monthly dosing and optimize drug exposure are critical to maximize the efficacy of this promising malaria control strategy. Trial Registration: Current Controlled Trials Identifier NCT00948896 Electronic supplementary material The online version of this article (doi:10.1186/s12936-015-0908-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kerstin Sundell
- Department of Pharmaceutical Biosciences, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden.
| | - Prasanna Jagannathan
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, USA.
| | - Liusheng Huang
- Department of Clinical Pharmacy, University of California, San Francisco, USA.
| | - Victor Bigira
- Infectious Diseases Research Collaboration, Kampala, Uganda.
| | - James Kapisi
- Infectious Diseases Research Collaboration, Kampala, Uganda.
| | - Mary M Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda.
| | - Rada Savic
- Department of Bioengineering and Therapeutics, University of California, San Francisco, USA.
| | - Moses R Kamya
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda.
| | - Grant Dorsey
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, USA.
| | - Francesca Aweeka
- Department of Clinical Pharmacy, University of California, San Francisco, USA.
| |
Collapse
|
27
|
|