Haemolysis in G6PD Heterozygous Females Treated with Primaquine for Plasmodium vivax Malaria: A Nested Cohort in a Trial of Radical Curative Regimens

Cindy S Chu, Germana Bancone, Kerryn A Moore, Htun Htun Win, Niramon Thitipanawan, Christina Po, Nongnud Chowwiwat, Rattanaporn Raksapraidee, Pornpimon Wilairisak, Aung Pyae Phyo, Lily Keereecharoen, Stéphane Proux, Prakaykaew Charunwatthana, François Nosten, Nicholas J White, Cindy S Chu, Germana Bancone, Kerryn A Moore, Htun Htun Win, Niramon Thitipanawan, Christina Po, Nongnud Chowwiwat, Rattanaporn Raksapraidee, Pornpimon Wilairisak, Aung Pyae Phyo, Lily Keereecharoen, Stéphane Proux, Prakaykaew Charunwatthana, François Nosten, Nicholas J White

Abstract

Background: Radical cure of Plasmodium vivax malaria with 8-aminoquinolines (primaquine or tafenoquine) is complicated by haemolysis in individuals with glucose-6-phosphate dehydrogenase (G6PD) deficiency. G6PD heterozygous females, because of individual variation in the pattern of X-chromosome inactivation (Lyonisation) in erythroid cells, may have low G6PD activity in the majority of their erythrocytes, yet are usually reported as G6PD "normal" by current phenotypic screening tests. Their haemolytic risk when treated with 8-aminoquinolines has not been well characterized.

Methods and findings: In a cohort study nested within a randomised clinical trial that compared different treatment regimens for P. vivax malaria, patients with a normal standard NADPH fluorescent spot test result (≳30%-40% of normal G6PD activity) were randomised to receive 3 d of chloroquine or dihydroartemisinin-piperaquine in combination with primaquine, either the standard high dose of 0.5 mg base/kg/day for 14 d or a higher dose of 1 mg base/kg/d for 7 d. Patterns of haemolysis were compared between G6PD wild-type and G6PD heterozygous female participants. Between 21 February 2012 and 04 July 2014, 241 female participants were enrolled, of whom 34 were heterozygous for the G6PD Mahidol variant. Haemolysis was substantially greater and a larger proportion of participants reached the threshold of clinically significant haemolysis (fractional haematocrit reduction >25%) in G6PD heterozygotes taking the higher (7 d) primaquine dose (9/17 [53%]) compared with G6PD heterozygotes taking the standard high (14 d) dose (2/16 [13%]; p = 0.022). In heterozygotes, the mean fractional haematocrit reductions were correspondingly greater with the higher primaquine dose (7-d regimen): -20.4% (95% CI -26.0% to -14.8%) (nadir on day 5) compared with the standard high (14 d) dose: -13.1% (95% CI -17.6% to -8.6%) (nadir day 6). Two heterozygotes taking the higher (7 d) primaquine dose required blood transfusion. In wild-type participants, mean haematocrit reductions were clinically insignificant and similar with both doses: -5.8 (95% CI -7.2% to -4.4%) (nadir day 3) compared with -5.5% (95% CI -7.4% to -3.7%) (nadir day 4), respectively. Limitations to this nested cohort study are that the primary objective of the trial was designed to measure efficacy and not haemolysis in relation to G6PD genotype and that the heterozygote groups were small.

Conclusion: Higher daily doses of primaquine have the potential to cause clinically significant haemolysis in G6PD heterozygous females who are reported as phenotypically normal with current point of care tests.

Trial registration: ClinicalTrials.gov NCT01640574.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Trial diagram.
Fig 1. Trial diagram.
CQ, chloroquine 25 mg base/kg divided over 3 d; DP, dihydroartemisinin 7 mg/kg and piperaquine 55 mg/kg divided over 3 d; PMQ-1, primaquine 1 mg base/kg/d x 7 d; PMQ-0.5, primaquine 0.5 mg base/kg/d x 14 d.
Fig 2. Mean fractional haematocrit changes over…
Fig 2. Mean fractional haematocrit changes over time in G6PD heterozygous and wild-type females taking PMQ-1 or PMQ-0.5.
Line graph represents fractional haematocrit plotted as the mean (95% CI). Plotted shapes represent individuals with maximum fractional haematocrit reductions below −25%. Circled shapes represent individuals who received a blood transfusion.
Fig 3. Mean maximum individual fractional haematocrit…
Fig 3. Mean maximum individual fractional haematocrit reductions in G6PD heterozygous and wild-type females taking PMQ-1 or PMQ-0.5.
Het, heterozygote; WT, wild type. Maximum individual fractional haematocrit plotted as mean (95% CI) not adjusted for initial parasitaemia. *The comparator group is the wild-type PMQ-0.5 group. **Comparison between heterozygote PMQ-1 and heterozygote PMQ-0.5 group.
Fig 4. Correlation between maximum individual fractional…
Fig 4. Correlation between maximum individual fractional haematocrit reduction and mean G6PD activity (IU/RBC)*.
*Three G6PD heterozygous females with missing data. The difference between the regression lines for heterozygote PMQ-1 and heterozygote PMQ-0.5 was not statistically significant (p = 0.433). The haemoglobin type was normal unless otherwise indicated. Circled shapes represent participants who received a blood transfusion.

References

    1. Beutler E. The hemolytic effect of primaquine and related compounds: a review. Blood. 1959;14: 103–139.
    1. Beutler E, Mitchell M. Special modifications of the fluorescent screening method for glucose-6-phosphate dehydrogenase deficiency. Blood, J Hematol. 1968;32: 816–818.
    1. Luzzatto L. Glucose 6-phosphate dehydrogenase deficiency: from genotype to phenotype. Hematol J. 2006;91: 1303–1306.
    1. Bancone G, Chu CS, Somsakchaicharoen R, Chowwiwat N, Parker DM, Charunwatthana P, et al. Characterization of G6PD Genotypes and Phenotypes on the Northwestern Thailand–Myanmar Border. PLoS ONE. 2014;9: e116063 10.1371/journal.pone.0116063
    1. Nuchprayoon I, Louicharoen C, Charoenvej W. Glucose-6-phosphate dehydrogenase mutations in Mon and Burmese of southern Myanmar. J Hum Genet. 2008;53: 48–54. 10.1007/s10038-007-0217-3
    1. Beutler E, Blume KG, Kaplan JC, Löhr GW, Ramot B, Valentine WN. International Committee for Standardization in Haematology: Recommended Methods for Red-Cell Enzyme Analysis*. Br J Haematol. 1977;35: 331–340.
    1. WHO Scientific group. World Health Organization Standardization of procedures for the study of glucose-6-phosphate dehydrogenase. World Health Organ Tech Rep Ser. 1967. pp. 1–53.
    1. Bancone G, Chu CS, Chowwiwat N, Somsakchaicharoen R, Wilaisrisak P, Charunwatthana P, et al. Suitability of Capillary Blood for Quantitative Assessment of G6PD Activity and Performances of G6PD Point-of-Care Tests. Am J Trop Med Hyg. 2015;92: 818–824. 10.4269/ajtmh.14-0696
    1. Morris SS, Ruel MT, Cohen RJ, Dewey KG, de la Brière B, Hassan MN. Precision, accuracy, and reliability of hemoglobin assessment with use of capillary blood. Am J Clin Nutr. 1999;69: 1243–1248.
    1. Kheng S, Muth S, Taylor WRJ, Tops N, Kosal K, Sothea K, et al. Tolerability and safety of weekly primaquine against relapse of Plasmodium vivax in Cambodians with glucose-6-phosphate dehydrogenase deficiency. BMC Med. 2015;13: 203 10.1186/s12916-015-0441-1
    1. Hockwald RS, Arnold J, Clayman CB, Alving AS. Toxicitiy of Primaquine in Negroes. J Am Med Assoc. 1952;149: 1568–1570.
    1. Tjitra E, Anstey NM, Sugiarto P, Warikar N, Kenangalem E, Karyana M, et al. Multidrug-resistant Plasmodium vivax associated with severe and fatal malaria: a prospective study in Papua, Indonesia. PLoS Med. 2008;5: e128 10.1371/journal.pmed.0050128
    1. Poespoprodjo JR, Fobia W, Kenangalem E, Lampah DA, Hasanuddin A, Warikar N, et al. Vivax Malaria: A Major Cause of Morbidity in Early Infancy. Clin Infect Dis. 2009;48: 1704–1712. 10.1086/599041
    1. World Health Organization. Guidelines for the treatment of malaria, 2nd Edition Geneva: WHO Press; 2010.
    1. Ganesan S, Tekwani BL, Sahu R, Tripathi LM, Walker LA. Cytochrome P(450)-dependent toxic effects of primaquine on human erythrocytes. Toxicol Appl Pharmacol. 2009;241: 14–22. 10.1016/j.taap.2009.07.012
    1. Fasinu PS, Tekwani BL, Nanayakkara NDPD, Avula B, Herath HBMTB, Wang Y-H, et al. Enantioselective metabolism of primaquine by human CYP2D6. Malar J. 2014;13: 507 10.1186/1475-2875-13-507
    1. World Health Organization. Guidelines for the treatment of malaria, 3rd Edition Geneva: World Health Organization; 2015.
    1. Alving AS, Kellermeyer RW, Tarlov AR, Schrier SL, Carson PE. Biochemical and Genetic Aspects of Primaquine-sensitive Hemolytic Anemia. Ann Intern Med. 1958;49: 240
    1. Tarlov AR, Brewer GJ, Carson PE, Alving AS. Primaquine sensitivity. Glucose-6-phosphate dehydrogenase deficiency: an inborn error of metabolism of medical and biological significance. Arch Intern Med. 1962;109: 209–234.
    1. Beutler E, Dern RJ, Alving AS. The hemolytic effect of primaquine IV. The relationship of cell age to hemolysis. J Lab Clin Med. 1954;44: 439–442.
    1. Ashley EA, Recht J, White NJ. Primaquine: the risks and the benefits. Malar J. 2014;13: 418 10.1186/1475-2875-13-418
    1. Recht J, Ashley EA, White NJ. Safety of 8-Aminoquinoline Antimalarial Medicines. Geneva: World Health Organization; 2014.
    1. Luzzatto L, Seneca E. G6PD deficiency: A classic example of pharmacogenetics with on-going clinical implications. Br J Haematol. 2014;164: 469–480. 10.1111/bjh.12665
    1. Hanboonkunupakarn B, Ashley EA, Jittamala P, Tarning J, Pukrittayakamee S, Hanpithakpong W, et al. Open-Label Crossover Study of Primaquine and Dihydroartemisinin-Piperaquine Pharmacokinetics in Healthy Adult Thai Subjects. Antimicrob Agents Chemother. 2014;58: 7340–7346. 10.1128/AAC.03704-14

Source: PubMed

3
Předplatit