#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Quantification of glucose-6-phosphate dehydrogenase activity by spectrophotometry: A systematic review and meta-analysis


Authors: Daniel A. Pfeffer aff001;  Benedikt Ley aff001;  Rosalind E. Howes aff002;  Patrick Adu aff004;  Mohammad Shafiul Alam aff005;  Pooja Bansil aff006;  Yap Boum, II aff007;  Marcelo Brito aff009;  Pimlak Charoenkwan aff010;  Archie Clements aff011;  Liwang Cui aff013;  Zeshuai Deng aff014;  Ochaka Julie Egesie aff015;  Fe Esperanza Espino aff016;  Michael E. von Fricken aff017;  Muzamil Mahdi Abdel Hamid aff018;  Yongshu He aff014;  Gisela Henriques aff019;  Wasif Ali Khan aff005;  Nimol Khim aff020;  Saorin Kim aff020;  Marcus Lacerda aff009;  Chanthap Lon aff021;  Asrat Hailu Mekuria aff022;  Didier Menard aff023;  Wuelton Monteiro aff009;  Francois Nosten aff024;  Nwe Nwe Oo aff026;  Sampa Pal aff006;  Duangdao Palasuwan aff027;  Sunil Parikh aff028;  Ayodhia Pitaloka Pasaribu aff029;  Jeanne Rini Poespoprodjo aff030;  David J. Price aff031;  Arantxa Roca-Feltrer aff033;  Michelle E. Roh aff034;  David L. Saunders aff021;  Michele D. Spring aff021;  Inge Sutanto aff037;  Kamala Ley-Thriemer aff001;  Thomas A. Weppelmann aff038;  Lorenz von Seidlein aff024;  Ari Winasti Satyagraha aff040;  Germana Bancone aff024;  Gonzalo J. Domingo aff006;  Ric N. Price aff001
Authors place of work: Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia aff001;  Malaria Atlas Project, Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom aff002;  Foundation for Innovative New Diagnostics, Geneva, Switzerland aff003;  Department of Medical Laboratory Sciences, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana aff004;  Infectious Diseases Division, International Centre for Diarrheal Diseases Research, Bangladesh, Mohakhali, Dhaka, Bangladesh aff005;  Diagnostics Program, PATH, Seattle, Washington, United States of America aff006;  Médecins sans Frontières Epicentre, Mbarara Research Centre, Mbarara, Uganda aff007;  Mbarara University of Science and Technology, Mbarara, Uganda aff008;  Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brasil aff009;  Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand aff010;  Faculty of Health Sciences, Curtin University, Bentley, Australia aff011;  Telethon Kids Institute, Nedlands, Australia aff012;  Department of Entomology, Pennsylvania State University, University Park, Pennsylvania, United States of America aff013;  Department of Cell Biology and Medical Genetics, Kunming Medical University, Kunming, Yunnan Province, China aff014;  Department of Hematology and Blood Transfusion, Faculty of Medical Sciences, University of Jos and Jos University Teaching Hospital, Jos, Plateau State, Nigeria aff015;  Department of Parasitology, Research Institute for Tropical Medicine, Department of Health, Alabang, Muntinlupa City, Philippines aff016;  Department of Global and Community Health, George Mason University, Fairfax, Virginia, United States of America aff017;  Department of Parasitology and Medical Entomology, Institute of Endemic Diseases, University of Khartoum, Khartoum, Republic of the Sudan aff018;  Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom aff019;  Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia aff020;  Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand aff021;  School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia aff022;  Malaria Genetics and Resistance Group, Institut Pasteur, Paris, France aff023;  Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand aff024;  Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom aff025;  Department of Medical Research, Lower Myanmar, Yangon, Myanmar aff026;  Oxidation in Red Cell Disorders and Health Research Unit, Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand aff027;  Yale School of Public Health, New Haven, Connecticut, United States of America aff028;  Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia aff029;  Yayasan Pengembangan Kesehatan dan Masyarakat Papua (YPKMP), Papua, Indonesia aff030;  Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia aff031;  The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia aff032;  Malaria Consortium, Phnom Penh, Cambodia aff033;  Global Health Group, Malaria Elimination Initiative, University of California, San Francisco, San Francisco, United States of America aff034;  F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America aff035;  US Army Medical Materiel Development Activity, Fort Detrick, Maryland, United States of America aff036;  University of Indonesia, Jakarta, Indonesia aff037;  Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, United States of America aff038;  Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand aff039;  Eijkman Institute for Molecular Biology, Jakarta, Indonesia aff040
Published in the journal: Quantification of glucose-6-phosphate dehydrogenase activity by spectrophotometry: A systematic review and meta-analysis. PLoS Med 17(5): e32767. doi:10.1371/journal.pmed.1003084
Category: Research Article
doi: https://doi.org/10.1371/journal.pmed.1003084

Summary

Background

The radical cure of Plasmodium vivax and P. ovale requires treatment with primaquine or tafenoquine to clear dormant liver stages. Either drug can induce haemolysis in individuals with glucose-6-phosphate dehydrogenase (G6PD) deficiency, necessitating screening. The reference diagnostic method for G6PD activity is ultraviolet (UV) spectrophotometry; however, a universal G6PD activity threshold above which these drugs can be safely administered is not yet defined. Our study aimed to quantify assay-based variation in G6PD spectrophotometry and to explore the diagnostic implications of applying a universal threshold.

Methods and findings

Individual-level data were pooled from studies that used G6PD spectrophotometry. Studies were identified via PubMed search (25 April 2018) and unpublished contributions from contacted authors (PROSPERO: CRD42019121414). Studies were excluded if they assessed only individuals with known haematological conditions, were family studies, or had insufficient details. Studies of malaria patients were included but analysed separately. Included studies were assessed for risk of bias using an adapted form of the Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) tool. Repeatability and intra- and interlaboratory variability in G6PD activity measurements were compared between studies and pooled across the dataset. A universal threshold for G6PD deficiency was derived, and its diagnostic performance was compared to site-specific thresholds. Study participants (n = 15,811) were aged between 0 and 86 years, and 44.4% (7,083) were women. Median (range) activity of G6PD normal (G6PDn) control samples was 10.0 U/g Hb (6.3–14.0) for the Trinity assay and 8.3 U/g Hb (6.8–15.6) for the Randox assay. G6PD activity distributions varied significantly between studies. For the 13 studies that used the Trinity assay, the adjusted male median (AMM; a standardised metric of 100% G6PD activity) varied from 5.7 to 12.6 U/g Hb (p < 0.001). Assay precision varied between laboratories, as assessed by variance in control measurements (from 0.1 to 1.5 U/g Hb; p < 0.001) and study-wise mean coefficient of variation (CV) of replicate measures (from 1.6% to 14.9%; p < 0.001). A universal threshold of 100% G6PD activity was defined as 9.4 U/g Hb, yielding diagnostic thresholds of 6.6 U/g Hb (70% activity) and 2.8 U/g Hb (30% activity). These thresholds diagnosed individuals with less than 30% G6PD activity with study-wise sensitivity from 89% (95% CI: 81%–94%) to 100% (95% CI: 96%–100%) and specificity from 96% (95% CI: 89%–99%) to 100% (100%–100%). However, when considering intermediate deficiency (<70% G6PD activity), sensitivity fell to a minimum of 64% (95% CI: 52%–75%) and specificity to 35% (95% CI: 24%–46%). Our ability to identify underlying factors associated with study-level heterogeneity was limited by the lack of availability of covariate data and diverse study contexts and methodologies.

Conclusions

Our findings indicate that there is substantial variation in G6PD measurements by spectrophotometry between sites. This is likely due to variability in laboratory methods, with possible contribution of unmeasured population factors. While an assay-specific, universal quantitative threshold offers robust diagnosis at the 30% level, inter-study variability impedes performance of universal thresholds at the 70% level. Caution is advised in comparing findings based on absolute G6PD activity measurements across studies. Novel handheld quantitative G6PD diagnostics may allow greater standardisation in the future.

Keywords:

Hematology – Spectrophotometry – Malaria – Plasmodium – Glucose-6-phosphate dehydrogenase deficiency – Primaquine – Antimalarials – quality control

Introduction

Plasmodium vivax and P. ovale both form dormant liver stages (hypnozoites) that can reactivate weeks to months following an initial infection, resulting in relapsing malaria [1]. The complete treatment of either species requires the use of drugs able to clear hypnozoites from the liver (‘radical cure’), alongside standard blood-stage antimalarials. The only licenced antimalarial compounds that can kill hypnozoites and prevent relapses are the 8-aminoquinoline drugs primaquine and tafenoquine. Primaquine is used to treat patients with malaria in two scenarios: either as a single low dose to kill P. falciparum gametocytes and reduce transmission, or at a higher dose administered over 14 days to kill P. vivax hypnozoites. Tafenoquine, is another 8-aminoquinoline drug, which has recently been licensed as a single-dose hypnozoiticidal agent for P. vivax liver stages. While well tolerated in the majority of recipients, standard dosing of either drug can cause severe haemolysis in patients with glucose-6-phosphate dehydrogenase (G6PD) deficiency [2]. Tafenoquine is more slowly eliminated than primaquine; hence, patients are exposed to potentially haemolytic concentrations of the drug for longer. The successful rollout of tafenoquine will therefore require more stringent G6PD screening than is currently available in malaria endemic areas.

G6PD deficiency is a common inherited enzymopathy that is particularly prevalent in malaria-endemic regions [3,4]. Red blood cells (RBCs) of affected individuals are susceptible to haemolysis caused by oxidative stress, induced by a variety of stimuli including drugs (e.g., rasburicase, 8-aminoquinolines, and dapsone), foods (e.g., fava beans), or acute infection [5,6]. The gene encoding G6PD is located on the X chromosome (Xq28). Hence, males inherit a single copy and are either hemizygous G6PD deficient (G6PDd) or G6PD normal (G6PDn), whereas females carry two copies and can be homozygous deficient or normal, or heterozygous for a mutant G6PD allele. Hemizygous and homozygous deficient individuals express >95% deficient RBCs, while heterozygotes harbour two distinct RBC populations, a G6PDd and a G6PDn population. Different patterns of X-inactivation in heterozygotes lead to different proportions of deficient RBCs between individuals and widely varying G6PD phenotypes [7]. Thus, while males are phenotypically either G6PDd or G6PDn, females exhibit a wide range of G6PD levels from very low deficient levels through intermediate to normal. Furthermore, the G6PD gene itself exhibits substantial genetic variability, with over 200 distinct G6PD mutations described [8,9], exhibiting a wide range of enzyme activity levels [10,11].

Ultraviolet (UV) spectrophotometry is the reference standard diagnostic method for quantifying G6PD enzyme activity [12]. Multiple commercial assay kits are available, and although there is strong correlation of measurements between assays, absolute values vary [13,14]. Such absolute values will be important for the universal use of novel point-of-care quantitative diagnostics, such as forthcoming biosensors. The interpretation of quantitative G6PD measurements requires a predetermined definition of ‘normal’ (100%) G6PD activity. However, there is no consensus definition of normal activity. Each laboratory must establish its own reference values and diagnostic thresholds [15]. In 2013, Domingo and colleagues proposed a method of standardising this process [16], deriving normal enzyme activity from the median value of non-deficient males, known as the adjusted male median (AMM). Although this approach is less vulnerable to outliers or the underlying prevalence of G6PDd than the standard mean or median, interpretation of assay results requires derivation of a context-specific local AMM. Once defined, the AMM enables classification of samples based on their relative G6PD activity.

Treatment decisions for primaquine or tafenoquine are currently based on two important thresholds: 30% and 70% normal enzyme activity, respectively. The former is the approximate cutoff activity for most qualitative tests [17]. The latter is designed to exclude heterozygous females with intermediate enzyme activity who are also at risk of haemolysis [18,19]. Setting the threshold too low risks falsely categorising patients as G6PDn and exposing individuals to primaquine-induced haemolysis. Setting the threshold too high potentially excludes G6PDn patients from receiving radical cure, putting them at risk of repeated episodes of vivax malaria and associated morbidity.

The aim of the current study was to pool spectrophotometric data from diverse laboratory and geographic contexts to quantify the degree to which assay-based variability influences inter- and intra-study comparability of G6PD spectrophotometry, and to explore the implications of this variability on diagnosing severe and intermediate G6PD deficiency.

Methods

Data collation

The data for this prospectively planned meta-analysis were pooled from a systematic literature review and unpublished contributions from collaborating investigators (PROSPERO: CRD42019121414) according to PRISMA guidelines (S1 File). Studies involving individual-level spectrophotometric measurements of G6PD activity were identified via a PubMed search (25 April 2018) using the following terms: G6PD OR “glucose-6-phosphate dehydrogenase” OR “glucose 6 phosphate dehydrogenase”) AND (quantitative OR spectrophot*). Additional studies were identified via reference lists and correspondence with authors. In view of the wide range of assay manufacturers and laboratory methodologies in the published literature, only papers published between January 2005 and April 2018 were screened for inclusion to ensure comparability of diagnostic protocols and quality control procedures. The title, abstract, and full text of studies between these dates were then screened to identify those that used UV spectrophotometry to define G6PD activity. Only studies that measured NADPH formation at a wavelength of 340 nm were included. Authors were contacted via email and invited to contribute individual patient and quality control data. A minimum of two attempts was made to contact authors before excluding studies. Studies targeting specific ethnic groups (e.g., African-American blood donors) were included, but those that only included individuals with known haematological conditions, family studies, or studies for which insufficient details were available were excluded. Studies of patients with malaria were included but analysed separately from individuals without malaria. One paper included studies performed in two different countries and was considered as two separate data sources ([20]; S1 Table).

Demographic data and available haematological parameters were collated and stored in a standardised database along with metadata of the study characteristics. Samples missing data on either G6PD activity (U/g Hb) or sex were excluded from the analysis, as were those for which measured G6PD activity was extreme (>30 U/g Hb). Quality of included studies was assessed using an adapted form of the QUADAS-2 tool ([21]; S2 File).

Data analysis

Intra-laboratory assay repeatability was investigated from replicate measures. For each sample, the coefficient of variation (CV) and absolute difference between replicate measures were calculated, along with a mean CV and mean difference for each study. CV values and proportion of samples with high inter-replicate variability were compared between studies using the Kruskal-Wallis and chi-squared tests.

To assess inter- and intra-laboratory variability in G6PD spectrophotometry, data from the measurement of manufacturer-provided quality control samples were used to quantify the magnitude and variance of control measurements (deficient, intermediate, and normal) and how these differed between assays and studies.

World Health Organization (WHO) prequalification for in vitro diagnostics recently defined G6PD deficiency for males and females at below 30% and intermediate activity between 30% and 80% G6PD activity [22]. The analysis performed here uses the 70% threshold used for the tafenoquine clinical trials [18,19]. For population studies, the AMM [16] was calculated for each study separately and defined as 100% G6PD activity. G6PD deficiency was defined as ‘activity below 30% of the respective study AMM’ and intermediate G6PD deficiency as ‘activity between 30% and 70% of the respective study AMM’. Variation between different study populations was assessed by assay and whether the participant had malaria. Studies with a high risk of bias due to patient selection were excluded from the analysis (S2 Table). To preclude the influence of heterozygosity on the observed variability, only male samples were compared. G6PD activity distributions from control and participant samples were compared using the Kruskal-Wallis test with pairwise Mann-Whitney-Wilcoxon tests using Bonferroni correction (magnitude) and Levene’s test (variance).

A universal AMM was calculated by applying the standard AMM formula to a comparable subset of all included samples. To control for differences due to assay variability or malaria status, the universal AMM was only derived from samples tested using the Trinity assay in patients without malaria. For each study, separately and across the pooled subset of data, the diagnostic performance (sensitivity, specificity, and negative predictive value [NPV]) of this universal AMM was then evaluated at both the 30% and 70% thresholds, considering diagnoses derived from study-specific AMMs as the reference. Exact binomial confidence limits were estimated for both sensitivity and specificity. Performance was compared to a conservative universal threshold, where 100% G6PD activity was determined by the upper limit of the 95% CI of the mean G6PD activity for G6PDn males in the included subset. Due to the limited availability of relevant covariate data from included studies, the universal AMM, conservative threshold, and threshold performances were not adjusted for study-level covariates. Furthermore, such adjustment for study-level covariates would impose an impractical level of complexity in clinical practice.

All data management, visualisation, and statistical analyses were performed using the tidyverse [23], ggplot2 [24], cowplot [25], epiR [26], and base R packages in R version 3.5.2 [27]. Prior ethical approval was obtained for all primary studies included in the analysis.

Results

In total, 312 studies were identified from the literature published between January 2005 and April 2018, as well as 18 unpublished studies. Of these, 243 studies were excluded based on title, abstract, or full text review, and 55 studies were excluded because the corresponding author did not respond or the relevant data were not available (Fig 1). A further two studies were excluded due to methodological criteria (incomparable measurement units or spectrophotometry wavelength). Data from 231 individuals were excluded for reasons stated in Fig 1.

PRISMA flow diagram depicting the literature search and data-screening procedures.
Fig. 1. PRISMA flow diagram depicting the literature search and data-screening procedures.
G6PD, glucose-6-phosphate dehydrogenase; RBC, red blood cell.

The final dataset comprised spectrophotometric measurements from 15,811 individuals collected from 30 studies (20 in Asia, 5 in Africa, and 5 in the Americas; Table 1). The age of participants ranged from 0 to 86 years, and 44.4% of participants (7,083 individuals) were female. G6PD spectrophotometry results were generated from six different manufacturer’s assays: Trinity Biotech, Ireland (21 studies, 12,222 participants); Randox Laboratories, United Kingdom (4 studies, 1,476 participants); Pointe Scientific, United States (2 studies, 883 participants); Sigma-Aldrich, US (2 studies, 691 participants); BIOLABO, France (1 study, 320 participants), and Spinreact, Spain (1 study, 319 participants).

Tab. 1. Characteristics of the final pooled database by malaria status.
Characteristics of the final pooled database by malaria status.

Assay repeatability

Replicate measurements of G6PD activity were available from five studies, including 14.4% (2,204/15,811) of samples (Table 2 and Fig 2). One study (n = 609) performed triplicate measures on all samples, while four studies (n = 1,595) performed duplicate measurements on all samples and a third test on a subset of samples with high inter-replicate variability (n = 53). The magnitude of inter-replicate variability differed between studies, with the study-specific maximum difference ranging from 0.65 U/g Hb to 8.64 U/g Hb and the mean CV ranging from 1.6% to 14.9% (Table 2). The percentage of samples that showed a high inter-replicate difference (>2 U/g Hb) differed significantly across all studies (p < 0.001), ranging from 0% to 32% of each study’s samples. Similar inter-study differences were evident when considering relative variability, with the percentage of samples exhibiting a high CV (>10%) ranging from 1.53% to 62%.

Repeatability of G6PD spectrophotometry.
Fig. 2. Repeatability of G6PD spectrophotometry.
Inter-replicate agreement in five studies enrolling 2,204 participants. (A) Absolute G6PD activity (U/g Hb) of replicate measures. Each point represents a single G6PD activity measurement, with measurements from the same individual connected by vertical lines for clarity. (B) Relative difference (CV; %) between replicate measures of G6PD activity. Five points with CV greater than 50% are shown as ‘50+’ for clarity (CV = 51, 61, 98, 110, 140). For both panels, the assay used is labelled for each study and the study-wise mean (range) inter-replicate difference is shown. Note: One study, Ley et al., 2018, provided measurements from the same samples using both Randox (n = 92) and Trinity (n = 100). CV, coefficient of variation; G6PD, glucose-6-phosphate dehydrogenase.
Tab. 2. Inter-replicate differences in G6PD spectrophotometry.
Inter-replicate differences in G6PD spectrophotometry.

Inter- and intra-laboratory variability

Quality control data were available from nine studies: seven that used the Trinity assay (using normal, intermediate, and deficient controls) and three studies that used the Randox assay (normal and deficient controls; Fig 3). Results differed significantly across studies for all control categories (p < 0.001), when considering both the Trinity and Randox assays. Smaller inter-study differences were observed between the studies using Randox, all of which were conducted in the same laboratory. Intra-study variability (study-level variance of control measurements) differed significantly for all control categories for studies using both the Trinity (normal, p < 0.001; intermediate, p < 0.001; deficient, p < 0.001), and Randox (normal, p = 0.002; deficient, p = 0.02) assays.

Inter-study variation in control sample measurements.
Fig. 3. Inter-study variation in control sample measurements.
Quality control data from nine studies are shown (n = 1,509). The three leftmost panels contain measurements of deficient, intermediate, and normal controls from studies using the Trinity assay; panels on the right depict deficient and normal control measurements using the Randox assay. The median and interquartile range for each category are superimposed in black. Points are coloured to indicate studies conducted in the same laboratory. A square-root transformation is applied to the y-axis to depict variation in deficient samples more clearly. Note: One study, Ley et al., 2018, provided measurements from the same samples using both Randox (n = 92) and Trinity (n = 100). G6PD, glucose-6-phosphate dehydrogenase.

To control for differences attributable to assay method, the analysis of the variability in spectrophotometry data between study populations was first addressed in the 18 studies (6,245 individuals) using the Trinity assay. There was considerable inter-study variation in the distribution of G6PD activity and the derived AMM values across studies (Fig 4). The AMM ranged from 5.7 to 12.6 across studies consisting of participants without malaria and 7.8 U/g Hb to 12.4 U/g Hb for those with malaria. The inter-study differences in G6PD activity were statistically significant across all studies (p < 0.05). In the studies that used a spectrophotometry assay other than Trinity, there was significant inter-study variation in G6PDn males for Pointe Scientific (p < 0.001), Randox (p = 0.04), and Sigma-Aldrich (p < 0.001) (S1 Fig).

Inter-study variation in G6PD activity measured using the Trinity assay.
Fig. 4. Inter-study variation in G6PD activity measured using the Trinity assay.
The distribution of G6PD activity measurements is shown for males enrolled in representative studies using the Trinity G6PD assay. Participants with (n = 2,694) and without malaria (n = 3,516) are shown separately. The AMM (black text), 70% threshold (blue), and 30% threshold (red) are labelled for each study (U/g Hb). Point colours indicate the country of origin of each study. Filled points represent G6PDn individuals (>30% study AMM) and hollow points indicate G6PDd individuals (<30% study AMM). Note: Roh et al., 2016, included 63 males <1 year of age, who may have had elevated G6PD activity [31,32]. AMM, adjusted male median; G6PD, glucose-6-phosphate dehydrogenase; G6PDd, G6PD deficient; G6PDn, G6PD normal.

Universal thresholds

Participants were categorised as being either severely deficient (<30% AMM; ineligible for primaquine) or severely/intermediately deficient (<70% AMM; ineligible for tafenoquine) using the study-specific AMMs. In patients without malaria assessed using the Trinity assay, the pooled AMM (i.e., 100% G6PD activity threshold) was 9.4 U/g Hb, resulting in a 70% threshold of 6.6 U/g Hb and a 30% threshold of 2.8 U/g Hb. The conservative universal threshold (calculated as the upper limit of the 95% CI of the mean G6PD activity, instead of the median used for the AMM) yielded a 100% threshold of 9.7 U/g Hb; 70% threshold of 6.8 U/g Hb, and 30% threshold of 2.9 U/g Hb.

Using the site-specific AMM as the reference, the pooled AMM correctly categorised 89% to 100% of severely deficient patients (both males and females) at the 30% threshold with a pooled sensitivity of 97% (95% CI: 95%–98%). At this threshold, the study-wise specificity was greater than 96% for all studies with a pooled specificity of 100% (95% CI: 100%–100%) (Table 3, S3 File: S7–S12 Figs). Seventeen individuals (12 females and 5 males, out of a total of 7,520) were falsely classified as G6PDn, with a mean (range) G6PD activity of 26.8% (23%–29.6%) of the local AMM (S4 Table, Fig 5, S3 File: S2 and S3 Figs).

Universal diagnostic classifications by G6PD activity—both males and females, using pooled AMM.
Fig. 5. Universal diagnostic classifications by G6PD activity—both males and females, using pooled AMM.
Relative G6PD activity is shown (x-axis, normalised to site-specific AMM) for individuals falling into each diagnostic category, as depicted by coloured bars (false normal [FN], red; false deficient [FD], orange; true normal [TN], light grey; true deficient [TD], black). Diagnoses are shown at both the 30% threshold (left; 17 FN, 21 FD, 6,926 TN, 556 TD) and 70% threshold (right; 139 FN, 258 FD, 5,983 TN, 1,140 TD). All individuals tested with Trinity without malaria infection are shown, except 69 individuals with G6PD activity >200% local AMM (n = 7,451). AMM, adjusted male median; G6PD, glucose-6-phosphate dehydrogenase.
Tab. 3. Performance of pooled universal AMM diagnostic thresholds.
Performance of pooled universal AMM diagnostic thresholds.

At the 70% threshold, the study-wise sensitivity of the universal AMM ranged between 64% and 100% with a pooled sensitivity of 89% (95% CI: 87%–91%). At this threshold, the specificity ranged from 35% to 100%, with a pooled specificity of 96% (95% CI: 95%–96%) (Table 3 & S3 File: S7–S12 Figs). One hundred and thirty-nine individuals, 107 females and 32 males, were falsely classified as G6PDn at the 70% level, with a mean (range) G6PD activity of 56.5% (52.4%–59.5%) of the local AMM (S4 Table, Fig 5, S3 File: S2 and S3 Figs). At both the 30% and 70% threshold, performance was slightly improved when considering the conservative universal thresholds (S3 and S4 Tables, S3 File: S4–S6, S13–S19 Figs). Diagnostic performance of universal thresholds did not differ substantially in patients with or without malaria. At all thresholds, when there was a difference in diagnostic performance of universal thresholds, the performance in females was worse than in males, across all studies and within individual studies (Table 3 and S3 File: S8–S13 Figs).

Discussion

Safe implementation of radical cure of malaria with primaquine or tafenoquine will be critical for the timely elimination of P. vivax. In view of the risk of drug-induced haemolysis, patients should be tested for G6PD deficiency prior to treatment to avoid exposing vulnerable individuals to oxidative stress. Widespread application of radical cure will thus require robust and easily interpretable diagnostic thresholds for G6PD enzyme activity. Our pooled analysis of G6PD activity across a diverse range of studies and geographical locations highlights significant inter-study and intra-study differences in absolute G6PD measurements derived using spectrophotometry. This variability has strong potential to confound reliable diagnosis of G6PD deficiency.

We observed considerable variability in G6PD activity measurements between sites, even when considering the same spectrophotometric assay. The differences in the quantification of control samples suggest substantial contribution of laboratory- or assay-based factors, although these may be exacerbated by unmeasured genetic or environmental differences between studies. Despite the large sample size of this meta-analysis, the data lacked the granular information necessary to isolate specific laboratory or procedural factors at play. The presence of differences between research laboratories illustrates the likely pervasiveness of interlaboratory differences in G6PD spectrophotometry, with fundamental implications for comparing absolute G6PD activity measurements between studies.

Despite this variability, our findings demonstrate strong performance of a universal threshold for identifying G6PD activity below 30% of the local AMM. At this level, the universal diagnostic threshold (2.8 U/g Hb) demonstrated robust diagnostic performance with sensitivity and specificity exceeding 97%. In these cohorts, 17 out of 7,520 individuals would receive primaquine despite having a G6PD activity less than 30% the local AMM; however, all of these misdiagnoses occurred around the diagnostic cutoff, with a minimum G6PD activity of 23% (S4 Table, Fig 5). The majority of the 17 misdiagnoses were in females, suggesting that a portion of these may come from heterozygous females with G6PD activities spanning the 30% threshold. Hence, this threshold may have utility in certain contexts where a local AMM is unavailable (e.g., validation studies of qualitative assays).

The use of tafenoquine requires a more stringent threshold to reduce the risk of haemolysis in heterozygous females with intermediate deficiency. At the 70% enzyme activity level, the diagnostic performance of pooled universal cutoffs was worse than that for the 30% threshold. This is likely a consequence of both inter-study variation in G6PD activity, as well as natural variation (noise) in G6PD activity levels around this 70% limit. Of the 139 individuals misclassified as G6PDn at this level, only 36 (25.4%) exhibited a G6PD activity less than 60% of the local AMM, and all had a G6PD activity greater than 53%. Again, similar to the 30% threshold, the majority of these were females that are in the 60%–70% activity range. The exact relationship between G6PD activity and the haemolytic risk associated with tafenoquine is unknown, although the risk is thought to be inversely correlated with enzyme activity [33]. Reassuringly, false normal diagnoses at the 70% enzyme activity level using the universal thresholds occurred in a minority of individuals with G6PD activity mostly near the 70% mark.

The universal thresholds defined in our study are based on the Trinity assay kit, which is now discontinued. While Alam and colleagues have shown good correlation between Trinity measurements and results from a Pointe Scientific assay, with little difference in absolute measurements (n = 50, [14]), a study by Pal and colleagues found a more modest correlation in absolute measurements (n = 183; [13]). Consequently, demonstration of suitability of a proposed universal threshold will need to be demonstrated with each specific assay prior to its widespread endorsement and application. Promising point-of-care quantitative biosensors reduce the need for sample storage and complex processing [12,34]. As such, these new assays may exhibit superior interlaboratory comparability and in future may provide the basis for a universal definition of G6PD deficiency. Until there is consensus regarding safe and robust universal thresholds, site- and assay-specific definitions of G6PD deficiency will still be required. Currently, laboratories either use the product insert ranges or determine normal ranges based on normal samples tested in the laboratory.

We identified notable differences in assay repeatability and variance of control measurements between sites. Such assay-based variability may arise from inconsistencies in assay procedures, or sample handling [16,34,35]. This variation demonstrates the importance of performing and monitoring replicate measurements and control sample measurements in order to minimise assay-based variability and maximise comparability of results.

It is worth noting that diagnostic definitions of ‘intermediate’ G6PD activity have varied over time and contexts. The early WHO classifications of G6PD variants considered anything above 60% to be normal G6PD activity [36], current WHO guidelines place the cutoff at 80% normal activity [17], and prescription requirements for tafenoquine in the US and Australia require a G6PD activity of 70% or higher [37]. Regardless of the definition, our study highlights the significant challenges in establishing an estimate of 100% G6PD activity (in clinically relevant units of U/g Hb) from which patients’ relative enzyme activity can be calculated.

Our study has a number of limitations. First, although we excluded studies in which haematological conditions may have influenced observed G6PD activity levels, the final dataset consisted of samples from diverse clinical and community survey contexts. This may have led to unequal influence of undiagnosed health conditions (e.g., other haematological conditions) upon measured G6PD activity. However, there was no clear pattern in G6PD activity by study type. Furthermore, while it has been suggested that neonates have elevated G6PD activity [31,32], introducing possible bias, few of the included studies enrolled newborns ([38,39]). Second, it is common practice to exclude replicate measures that differ by more than a certain amount (e.g., CV > 10%) as well as control measurements falling outside of an expected range. Included datasets may have already excluded these values in some, but not all, cases, meaning that the current study would overestimate true assay performance. Third, the study did not consider lot-to-lot variability in control isolates (which consist of lyophilised blood specimens) and assay reagents; these may have contributed to some of the inter-study variability observed for both the controls and AMM values. Nevertheless, the current findings represent an indication of inter- and intra-study variability in ‘valid’ results of G6PD spectrophotometry.

Conclusions

Interlaboratory variability hinders the definition of universal cutoff values for the classification of G6PD activity using spectrophotometry, particularly at the 70% G6PD activity level. Caution is advised in comparing research findings based on absolute G6PD activity measurements across studies, such as those characterising novel variants or assessing clinical safety in patients exposed to 8-aminoquinolines. In these cases, the derivation of relative G6PD activity using the AMM remains a more appropriate approach. Because assay precision varies considerably between laboratories, the use of replicate measures and control sample measurements is crucial to ensure quality control. Clinical laboratories typically provide the patients’ G6PD value and a normal G6PD range for the laboratory, which is often determined such that it discriminates at or above the 70% threshold required to prescribe tafenoquine. Novel point-of-care assays, such as recently developed quantitative biosensors, are currently being evaluated in field trials. These assays are designed to require less sample preparation and offer more robust performance across diverse temperature ranges and clinical contexts than spectrophotometry. As such, they may provide superior inter-site comparability than the current reference standard of spectrophotometry; however, until this has been shown, routine quantitative diagnosis of G6PDd will require site- and assay-specific local definitions of G6PD activity to ensure that tafenoquine can be administered safely. In any case, no diagnostic assay is perfect, meaning that radical cure treatment policies must be accompanied by patient and health worker training on the warning signs and risks of haemolysis, along with access to transfusion services when needed.

Supporting information

S1 File [pdf]
PRISMA checklist.

S2 File [pdf]
Adapted QUADAS-2 [] tool used for quality appraisal.

S3 File [pdf]
Additional figures indicating diagnostic performance for alternative universal thresholds.

S4 File [pdf]
List of data contacts for each included study.

S1 Fig [c]
Inter-study variability for assays other than Trinity.

S1 Table [xlsx]
Characteristics of data sources included in the pooled database.

S2 Table [xlsx]
Quality appraisal of included studies using adapted QUADAS-2 [] tool.

S3 Table [xlsx]
Performance of a conservative universal diagnostic threshold.

S4 Table [xlsx]
Frequency and G6PD activity of false normal individuals using universal diagnostic thresholds.


Zdroje

1. Lover AA, Baird JK, Gosling R, Price R. Malaria elimination: Time to target all species. Am J Trop Med Hyg. 2018. Epub 2018/05/16. doi: 10.4269/ajtmh.17-0869 29761762.

2. Baird JK, Battle KE, Howes RE. Primaquine ineligibility in anti-relapse therapy of Plasmodium vivax malaria: the problem of G6PD deficiency and cytochrome P-450 2D6 polymorphisms. Malar J. 2018;17:42. doi: 10.1186/s12936-018-2190-z PMC5778616. 29357870

3. Howes RE, Piel FB, Patil AP, Nyangiri OA, Gething PW, Dewi M, et al. G6PD deficiency prevalence and estimates of affected populations in malaria endemic countries: a geostatistical model-based map. PLoS Med. 2012;9(11):e1001339. doi: 10.1371/journal.pmed.1001339 23152723

4. Nkhoma ET, Poole C, Vannappagari V, Hall SA, Beutler E. The global prevalence of glucose-6-phosphate dehydrogenase deficiency: A systematic review and meta-analysis. Blood Cells, Molecules and Diseases. 2009;42(3):267–78. doi: 10.1016/j.bcmd.2008.12.005 19233695

5. Luzzatto L, Arese P. Favism and glucose-6-phosphate dehydrogenase deficiency. N Engl J Med. 2018;378(1):60–71. doi: 10.1056/nejmra1708111 29298156

6. Belfield KD, Tichy EM. Review and drug therapy implications of glucose-6-phosphate dehydrogenase deficiency. Am J Health Syst Pharm. 2018;75(3):97–104. doi: 10.2146/ajhp160961 29305344

7. Domingo GJ, Advani N, Satyagraha AW, Sibley CH, Rowley E, Kalnoky M, et al. Addressing the gender-knowledge gap in glucose-6-phosphate dehydrogenase deficiency: challenges and opportunities. Int Health. 2018;11(1):7–14. doi: doi.org/10.1093/inthealth/ihy060

8. Gómez-Manzo S, Marcial-Quino J, Vanoye-Carlo A, Serrano-Posada H, Ortega-Cuellar D, González-Valdez A, et al. Glucose-6-phosphate dehydrogenase: Update and analysis of new mutations around the world. Int J Mol Sci. 2016;17(12):2069. doi: 10.3390/ijms17122069 PMC5187869. 27941691

9. Minucci A, Moradkhani K, Hwang MJ, Zuppi C, Giardina B, Capoluongo E. Glucose-6-phosphate dehydrogenase (G6PD) mutations database: Review of the “old” and update of the new mutations. Blood Cells Mol Dis. 2012;48(3):154–65. doi: 10.1016/j.bcmd.2012.01.001 22293322

10. Luzzatto L. Glucose 6-phosphate dehydrogenase deficiency: from genotype to phenotype. Haematologica. 2006;91(10):1303. 17018377

11. Cunningham AD, Colavin A, Huang KC, Mochly-Rosen D. Coupling between protein stability and catalytic activity determines pathogenicity of G6PD variants. Cell Rep. 2017;18(11):2592–9. doi: 10.1016/j.celrep.2017.02.048 28297664

12. Ley B, Bancone G, von Seidlein L, Thriemer K, Richards JS, Domingo GJ, et al. Methods for the field evaluation of quantitative G6PD diagnostics: a review. Malar J. 2017;16(1):361. doi: 10.1186/s12936-017-2017-3 28893237

13. Pal S, Bansil P, Bancone G, Hrutkay S, Kahn M, Gornsawun G, et al. Evaluation of a novel quantitative test for glucose-6-phosphate dehydrogenase deficiency: bringing quantitative testing for glucose-6-phosphate dehydrogenase deficiency closer to the patient. Am J Trop Med Hyg. 2018:-. doi: 10.4269/ajtmh.18-0612 30350771

14. Alam MS, Kibria MG, Jahan N, Price RN, Ley B. Spectrophotometry assays to determine G6PD activity from Trinity Biotech and Pointe Scientific G6PD show good correlation. BMC Res Notes. 2018;11(1):855. doi: 10.1186/s13104-018-3964-7 30514365

15. 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(2):331–40. doi: 10.1111/j.1365-2141.1977.tb00589.x 857853

16. Domingo GJ, Satyagraha AW, Anvikar A, Baird K, Bancone G, Bansil P, et al. G6PD testing in support of treatment and elimination of malaria: recommendations for evaluation of G6PD tests. Malar J. 2013;12(1):391. doi: 10.1186/1475-2875-12-391 24188096

17. World Health Organization. Testing for G6PD deficiency for safe use of primaquine in radical cure of P. vivax and P. ovale malaria. Geneva: WHO, 2016.

18. Lacerda MVG, Llanos-Cuentas A, Krudsood S, Lon C, Saunders DL, Mohammed R, et al. Single-dose tafenoquine to prevent relapse of Plasmodium vivax malaria. N Engl J Med. 2019;380(3):215–28. doi: 10.1056/NEJMoa1710775 30650322

19. Llanos-Cuentas A, Lacerda MVG, Hien TT, Vélez ID, Namaik-larp C, Chu CS, et al. Tafenoquine versus primaquine to prevent relapse of Plasmodium vivax malaria. N Engl J Med. 2019;380(3):229–41. doi: 10.1056/NEJMoa1802537 30650326

20. Henriques G, Phommasone K, Tripura R, Peto TJ, Raut S, Snethlage C, et al. Comparison of glucose-6 phosphate dehydrogenase status by fluorescent spot test and rapid diagnostic test in Lao PDR and Cambodia. Malar J. 2018;17(1):243. doi: 10.1186/s12936-018-2390-6 29929514

21. Whiting PF, Rutjes AWS, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, et al. QUADAS-2: A revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med. 2011;155(8):529–36. doi: 10.7326/0003-4819-155-8-201110180-00009 22007046

22. World Health Organization. Technical specifications series for submission to WHO prequalification–diagnostic assessment: in vitro diagnostic medical devices to identify glucose-6-phosphate dehydrogenase (G6PD) activity. Geneva: World Health Organization, 2016.

23. Wickham H. tidyverse: Easily install and load the ‘tidyverse’ 2017. R package version 1.2.1. Available from: https://CRAN.R-project.org/package=tidyverse

24. Wickham H. ggplot2: Elegant graphics for data analysis. New York: Springer-Verlag; 2009.

25. Wilke CO. cowplot: Streamlined plot theme and plot annotations for 'ggplot2' 2019. R package version 0.9.4. Available from: https://CRAN.R-project.org/package=cowplot

26. Stevenson M. epiR: Tools for the analysis of epidemiological data 2018. R package version 0.9–99. Available from: https://CRAN.R-project.org/package=epiR

27. R Core Team. R: A Language and Environment for Statistical Computing Vienna, Austria: R Foundation for Statistical Computing; 2018. Available from: https://www.R-project.org.

28. 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(4):818–24. Epub 2015/02/04. doi: 10.4269/ajtmh.14-0696 25646252; PubMed Central PMCID: PMC4385780.

29. Oo NN, Bancone G, Maw LZ, Chowwiwat N, Bansil P, Domingo GJ, et al. Validation of G6PD point-of-care rests among healthy volunteers in Yangon, Myanmar. PLoS ONE. 2016;11(4):e0152304–e. doi: 10.1371/journal.pone.0152304 27035821.

30. Satyagraha AW, Sadhewa A, Elvira R, Elyazar I, Feriandika D, Antonjaya U, et al. Assessment of point-of-care diagnostics for G6PD deficiency in malaria endemic rural Eastern Indonesia. PLoS Negl Trop Dis. 2016;10(2):e0004457. doi: 10.1371/journal.pntd.0004457 26894297

31. Travis SF, Kumar SP, Paez PC, Delivoria-Papadopoulos M. Red cell metabolic alterations in postnatal life in term infants. Glycolytic enzymes and glucose-6-phosphate dehydrogenase. Pediatr Res. 1980;14(12):1349. doi: 10.1203/00006450-198012000-00016 6451861

32. Yang W-C, Tai S, Hsu C-L, Fu C-M, Chou A-K, Shao P-L, et al. Reference levels for glucose-6-phosphate dehydrogenase enzyme activity in infants 7–90 days old in Taiwan. J Formos Med Assoc. 2019. https://doi.org/10.1016/j.jfma.2019.03.010.

33. Rueangweerayut R, Bancone G, Harrell EJ, Beelen AP, Kongpatanakul S, Mohrle JJ, et al. Hemolytic potential of tafenoquine in female volunteers heterozygous for glucose-6-phosphate dehydrogenase (G6PD) deficiency (G6PD Mahidol variant) versus G6PD-normal volunteers. Am J Trop Med Hyg. 2017;97(3):702–11. Epub 2017/07/28. doi: 10.4269/ajtmh.16-0779 28749773.

34. Alam MS, Kibria MG, Jahan N, Thriemer K, Hossain MS, Douglas NM, et al. Field evaluation of quantitative point of care diagnostics to measure glucose-6-phosphate dehydrogenase activity. PLoS ONE. 2018;13(11):e0206331. doi: 10.1371/journal.pone.0206331 30388146

35. Major CA, Pretlow L, Fincher E, Koziatek V. Validating the assessment of glucose-6-phosphate dehydrogenase (G6PD). Clin Lab Sci. 2006;19(3):134–9. 16910228

36. WHO Working Group. Glucose-6-phosphate dehydrogenase deficiency. Bull World Health Organ. 1989;67(6):601–11. PMC2491315. 2633878

37. Commons RJ, McCarthy JS, Price RN. Tafenoquine for the radical cure and prevention of malaria: the importance of testing for G6PD deficiency. Med J Aust. 2020;212(4):152–3.e1.

38. Charoenkwan P, Tantiprabha W, Sirichotiyakul S, Phusua A, Sanguansermsri T. Prevalence and molecular characterization of glucose-6-phosphate dehydrogenase deficiency in northern Thailand. Southeast Asian J of Trop Med Public Health. 2014;45(1):187–93. Epub 2014/06/27. 24964669.

39. Roh ME, Oyet C, Orikiriza P, Wade M, Mwanga-Amumpaire J, Boum Y 2nd, et al. Screening for glucose-6-phosphate dehydrogenase deficiency using three detection methods: A cross-sectional survey in Southwestern Uganda. Am J Trop Med Hyg. 2016;95(5):1094–9. doi: 10.4269/ajtmh.16-0552 27672207.


Článek vyšel v časopise

PLOS Medicine


2020 Číslo 5
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Hypertenze a hypercholesterolémie – synergický efekt léčby
nový kurz
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Multidisciplinární zkušenosti u pacientů s diabetem
Autoři: Prof. MUDr. Martin Haluzík, DrSc., prof. MUDr. Vojtěch Melenovský, CSc., prof. MUDr. Vladimír Tesař, DrSc.

Úloha kombinovaných preparátů v léčbě arteriální hypertenze
Autoři: prof. MUDr. Martin Haluzík, DrSc.

Halitóza
Autoři: MUDr. Ladislav Korábek, CSc., MBA

Terapie roztroušené sklerózy v kostce
Autoři: MUDr. Dominika Šťastná, Ph.D.

Všechny kurzy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#