U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Pratt VM, Scott SA, Pirmohamed M, et al., editors. Medical Genetics Summaries [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2012-.

Cover of Medical Genetics Summaries

Medical Genetics Summaries [Internet].

Show details

Primaquine Therapy and G6PD and CYP2D6 Genotype

, PhD.

Author Information and Affiliations

Created: .

Estimated reading time: 37 minutes

Introduction

Primaquine is a potent antimalarial medication indicated for the radical cure of malaria caused by Plasmodium vivax (P. vivax) and Plasmodium ovale (P. ovale) species (1, 2). Malaria is a blood borne infection caused by infection of Plasmodium parasites that is spread by mosquitos. The P. vivax and P. ovale species present a particular challenge to treat because the parasitic life cycle includes a dormant, liver-specific stage that is not susceptible to other antimalarial medications. Thus, primaquine is often used with other therapies such as chloroquine or artemisinin-based medicines that target the reproductive, active forms of the parasite. Primaquine is also used to prevent transmission of malaria caused by Plasmodium falciparum (P. falciparum) species. A single, low dose (SLD) of primaquine has gametocidal activity, which does not cure the individual but does provide malaria transmission control.

Primaquine is a pro-drug that must be activated by the cytochrome P450 (CYP) enzyme system. Metabolism by the cytochrome P450 member 2D6 (CYP2D6) and cytochrome P450 nicotinamide adenine dinucleotide phosphate (NADPH):oxidoreductase (CPR) generates 2 hydroxylated active metabolites that generate hydrogen peroxide (H2O2). This causes significant oxidative stress to the malarial parasite and the host human cells. Individuals who are glucose-6-phosphate dehydrogenase (G6PD) deficient are particularly susceptible to oxidative stress and may experience acute hemolytic anemia (AHA). Before starting a course of primaquine, individuals should be tested for G6PD deficiency to ensure safe administration (1, 2). According to the FDA-approved drug label, individuals with severe G6PD deficiency should not take primaquine (Table 1) (1).

The World Health Organization (WHO) recommends that individuals with G6PD deficiency should be treated with a modified course of primaquine therapy. The recommended course for individuals with G6PD deficiency is a single dose once per week for 8 weeks, while the standard course is daily administration for 14 days (Table 2) (2). The Clinical Pharmacogenetics Implementation Consortium (CPIC) reports that the risk of adverse effects of primaquine therapy for G6PD-deficient individuals is dose-dependent, with the SLD regimen presenting the least risk (Table 3) (3).

Primaquine is contraindicated during pregnancy and is not recommended for breastfeeding individuals when the G6PD status of the baby is unknown (1, 2). Primaquine is not approved for individuals under 6 months of age. Individuals with acute illness that are prone to granulocytopenia or individuals taking another hemolytic medication are also contraindicated from taking primaquine. (1)

Table 1.

The FDA Drug Label for Primaquine Phosphate (2021)

G6PD statusRiskRecommendation
DeficientHemolytic anemiaG6PD testing has to be performed before using primaquine.
Due to the limitations of G6PD tests, physicians need to be aware of residual risk of hemolysis
Severe deficiencyHemolytic anemiaPrimaquine should not be prescribed
Mild to moderate deficiencyHemolytic anemiaA decision to prescribe primaquine must be based on an assessment of the risks and benefits of using primaquine. If primaquine administration is considered, baseline hematocrit and hemoglobin must be checked before treatment and close hematological monitoring (for example, at day 3 and 8) is required
Unknown, testing unavailableHemolytic anemiaA decision to prescribe primaquine must be based on an assessment of the risks and benefits of using primaquine. Risk factors for G6PD deficiency or favism must be assessed. Baseline hematocrit and hemoglobin must be checked before treatment and close hematological monitoring (for example, at day 3 and 8) is required

This table is adapted from (1). G6PD - glucose-6-phosphate dehydrogenase

Table 3.

The CPIC Guidelines for Primaquine based on G6PD Phenotype

G6PD status (predicted from genotype)aDosing recommendationRiskClassification of recommendation
NormalNo reason to avoid primaquine based on G6PD statusLowStrong
DeficientAvoid primaquine at ≥ standard dose (0.25–0.5 mg/kg daily for 14 days)HighStrong
DeficientMedium dose (0.75 mg/kg or 45 mg, once weekly for 8 weeks) for P. vivax malaria; monitor individuals closely for hemolysisMediumStrong
DeficientSingle low dose (0.25 mg/kg) for P. falciparum malariaLow to noStrong
Deficient with CNSHAAvoid primaquineHighStrong
Variable or indeterminantAscertain G6PD status by enzyme activity; drug use should be guided by activity-based phenotype.bVariable or unknownModerate

This table is adapted from (3). Mg/kg - milligram per kilogram of the individual’s body weight. CNSHA - chronic non-spherocytic hemolytic anemia, G6PD - glucose-6-phosephate dehydrogenase, P. vivax - Plasmodium vivax, P. falciparum - Plasmodium falciparum

a

Definition of G6PD status based on data from the World Health Organization and US Centers for Disease Control.

b

X-linked mosaicism in individuals with more than one X chromosome (who are heterozygous for G6PD alleles of different functional status) can lead to variable G6PD function, enzyme-based assays should be used to determine G6PD activity and guide dosing. An enzyme activity-based test is also recommended for individuals with any allele of unknown function.

Drug: Primaquine

Primaquine is an 8-aminoquinoline antimalarial medication, indicated for the radical cure of P. vivax and P. ovale caused malaria (1, 4). Primaquine is approved by the WHO to prevent relapse of P. vivax and P. ovale malaria, often with chloroquine (in areas with chloroquine-sensitive P. vivax) (2). Though once not recommended in high-transmission settings, the WHO 2022 Malaria Guidelines state that “given the benefits of preventing relapse and in the light of changing epidemiology worldwide and more aggressive targets for malaria control and elimination, the [WHO Global Malaria Programme] group now recommends that primaquine be used in all settings” (2). In severe P. vivax malaria, primaquine therapy should be administered after the completion of other antimalarial therapies that target the active parasites, such as artesunate or chloroquine (2).

The standard administration of primaquine is a daily dose of 0.25 mg base per kg of body weight (mg/kg bw) for 14 days (1, 2). However, some sources recommend a standard adult dose of 15 mg base daily for 14 days, with increased dosing based on either weight over 70 kg or known infection with a frequently relapsing strain of Plasmodium (5, 6). In individuals with G6PD deficiency, either standard dosage presents a significant risk of life-threatening hemolysis, so an adjusted regimen of 0.75 mg/kg bw once a week for 8 weeks with close medical supervision is conditionally recommended by the WHO (2). Primaquine may also be administered as a SLD (0.25 mg/kg bw) in addition to artemisinin combination therapy (ACT) to eliminate malaria caused by P. falciparum in low-transmission areas (2). The benefit of SLD primaquine is primarily targeted to the community level as a means to reduce transmission, as this dose causes sterilization of the mature P. falciparum parasite, rather than curing the infected individual (2, 7, 8).

Primaquine is a pro-drug that needs to be metabolized to exert the desired antimalarial effect. Primaquine is metabolized by 2 different pathways: monoamine oxidase-A (MAO-A) generates the inactive metabolite carboxyprimaquine, while CYP2D6 and CPR generates the 2 active metabolites 5-hydroxyprimaquine and 5-hydroxy-6-desmethylprimaquine (9, 10). Spontaneous oxidation of the active metabolites generates quinoneimine and H2O2 that contribute to the antiparasitic activity of primaquine. The enzyme CPR then mediates redox cycling of quinoneimine to the primaquine active metabolites (9, 10). Primaquine has an estimated systemic half-life of 6 hours, necessitating multiple doses for effective radical cure of P. vivax malaria (11).

Primaquine can cause significant oxidative stress due to the accumulation of H2O2 in red blood cells, leading to AHA. Under normal homeostatic conditions, NADPH protects cells from oxidative stress. The enzyme G6PD generates NADPH and is particularly critical in red blood cells where it is the only source of NADPH. Individuals who G6PD deficient are especially sensitive to oxidative stress, whether due to endogenous or exogenous sources. As a result, primaquine is contraindicated at standard doses in individuals with G6PD deficiency (1). As discussed below, the residual amount of G6PD activity can vary based on the specific underlying genotype of an individual, so the risk of AHA varies based on the degree of deficiency. The FDA-approved drug label for primaquine advises monitoring blood cell counts and hemoglobin routinely during therapy even in individuals with normal levels of G6PD activity (1).

Contraindications for primaquine treatment include pregnancy, acute illness with a predisposition to granulocytopenia (often seen in rheumatoid arthritis or systemic lupus erythematosus), and medication with other potentially hemolytic drugs (1). The WHO further advise to avoid primaquine therapy in breastfeeding women unless the G6PD status of the breastfed infant is known to be within the normal range (2). One small study found that the estimated primaquine dose that a nursing infant receives following maternal dosing with 0.5 mg/kg/day primaquine was estimated to be 0.6% of the infant daily dose (0.5 mg/day) (12). The amount of primaquine excreted into breastmilk is low, and some sources suggest that G6PD-deficient infants over 28 days of age have a low risk of hemolysis due to primaquine exposure in breastmilk (13); however avoidance of primaquine by nursing mothers when the infants G6PD status is unknown is recommended by WHO, FDA, the US Centers for Disease Control and Prevention, as well as the United Kingdom malaria treatment guidelines (1, 2, 6, 14).

Primaquine is not approved for use in children younger than 6 months of age (2), though one study of infants in Indonesia found that severe clinical outcomes following primaquine treatment in infants under 12 months of age were rare (15). On-going studies suggest that younger children (14 years of age and younger) may require a higher weight-adjusted dose due to lower exposure primaquine and its metabolites (16, 17, 18). The FDA-approved label recommends caution with dose selection for geriatric individuals, as this population has a higher frequency of decreased hepatic, renal, and cardiac function. Initiating therapy at the low end of recommended dosing range is recommended (1). However, both the FDA and the Health Canada approved drug labels clearly state that efficacy and safety of primaquine has not been assessed in individuals over age 65 (1, 5).

In addition to the hemolysis risks, primaquine therapy may also trigger QT prolongation. As such, it should be avoided in conjunction with other medications that prolong the QT interval, in individuals with cardiac conditions such as long QT syndrome, ventricular arrhythmias or bradycardia (1). Other adverse reactions to primaquine can include nausea, vomiting, epigastric distress, abdominal cramps, dizziness, rash, and pruritus. Overdosage of primaquine phosphate can cause these adverse reactions as well as central nervous system and cardiovascular disturbances, cyanosis, granulocytopenia and AHA, among others (1).

Disease: Malaria

Malaria is a serious tropical disease caused by a parasite (Plasmodium) that spreads to humans by infected mosquitos. The only available vaccine is moderately effective and acts only against P. falciparum species (19). Widely recommended antimalarial drugs such as mefloquine or atovaquone-proguanil can be used for prevention –– this is known as chemoprophylaxis. The type of chemoprophylaxis recommended depends upon the individual taking the prophylaxis (namely, age, pregnancy status, and medical comorbidities) and the nature of their exposure –– specifically, the country of residence or traveled to, the length of stay, the species of Plasmodium that are most prevalent, and the level of drug resistance. For individuals residing in malaria-endemic regions, the WHO recommends a variety of preventative chemotherapies that can be used in infants, children, during pregnancy or collectively for the population of endemic areas (2).

Despite chemoprophylaxis, travel to malaria-endemic areas is not without risk. Individuals at elevated risk for malaria complications include pregnant women (20) and adults who have had their spleen removed (21). If travel cannot be avoided, chemoprophylaxis should be combined with additional precautions to avoid mosquito bites, such as bed nets and repellents. In 2021, the WHO estimated 247 million cases of malaria occurred worldwide, and malaria was responsible for 619,000 deaths. (22)

Malaria is found in over 100 countries and occurs throughout most tropical regions in the world. These regions include large parts of Africa, Asia, Central and South America, and parts of the Middle East and Pacific islands (22, 23). Individuals who are heterozygous carriers for sickle cell disease and G6PD deficiency have a protective advantage against malaria, and as a result, the frequency of such genetic conditions is higher in countries where malaria is endemic (24).

Malaria is transmitted to humans by the bite of an infected Anopheles mosquito. Only female mosquitos spread the infection (females feed on human blood, males feed on nectar). Although malaria can also be spread by sharing contaminated needles or via a contaminated blood transfusion, these are rare means of transmission.

There are several different Plasmodium species, but only a few species cause the most malaria cases:

P. falciparum

  • The most common cause of malaria, and death from malaria
  • Predominates in sub-Saharan Africa
  • Also found in regions of Australasia (Papua New Guinea, Southeast Asia), and the Caribbean (Haiti and the Dominican Republic)

P. vivax

  • A common cause of malaria outside of Africa
  • Most frequent species found in Central and South America
  • Parasite has a dormant, hypnozoite stage
  • Early gametocytes that infect mosquitos

Plasmodium malariae

  • Less common
  • Found in most areas where malaria is endemic

P. ovale

  • Less common
  • Parasite has a dormant, hypnozoite stage

Plasmodium knowlesi

  • Less common
  • Found in some Southeast Asia areas

The first stage of malaria infection begins when an infected mosquito bites the human host. Typically, mosquitos bite at dusk, or during the night. As the mosquito feeds, infective parasite sporozoites (the motile spore-like stage in the life cycle of this parasitic sporozoan, which is the infective agent) are inoculated into humans. The sporozoites travel to the liver, where they invade liver cells and asexually reproduce to form schizonts. The liver schizonts contain daughter merozoites. This process is asymptomatic, and because it occurs outside of the red blood cell (erythrocyte), it is known as the exoerythrocytic stage.

Some species of the parasite (P. vivax and P. ovale) have an additional dormant stage in the liver. The parasite exists as hypnozoites, which can stay in the liver for weeks or months without causing any clinical symptoms.

The second stage of malaria infection is the erythrocytic stage. It begins when the liver schizonts rupture and release the daughter merozoites into the bloodstream. The merozoites invade red blood cells, digest hemoglobin, produce a toxic metabolite (hemozoin), and damage red blood cell membranes. Infected, brittle red blood cells are rapidly broken down (hemolysis) and if too many damaged red blood cells get trapped in the spleen, the spleen can rapidly enlarge (splenic sequestration).

Some of the daughter merozoites differentiate into male or female gametocytes (sexual forms). When they are ingested by a mosquito, they mature, fertilize, reproduce, and develop into sporozoites. When the mosquito feeds again, the sporozoites are inoculated into another human host and the cycle of malaria transmission is complete.

The erythrocytic stage of malaria is usually associated with fever, and malaria should always be suspected in anyone with a fever who has recently returned from a malaria-endemic region, even if antimalarial chemoprophylaxis was correctly followed. Other symptoms and signs include nausea, vomiting, abdominal pain, tachycardia (fast heart rate), diaphoresis (sweating), chills, and myalgia (muscle pain). The complications of malaria infection include severe anemia, cerebral malaria, and multi-organ failure. Without correct diagnosis and prompt treatment, malaria can be fatal.

Gene: G6PD

The G6PD enzyme is encoded by the G6PD gene, which is located on the long arm of X chromosome (Xq28). Variants in the G6PD gene that result in a complete loss of enzymatic activity are not viable; variants observed in living humans generally impact the stability of the enzyme. As such, males can only be hemizygous (have one G6PD allele) while females randomly inactivate one X chromosome during development, resulting in a mosaic expression of either one X chromosome or the other in their somatic cells. This mosaicism can occur in the hematopoietic progenitor cells that give rise to red blood cells, resulting in mixed expression of G6PD alleles. Females with Turner syndrome (45, X) have only one X chromosome and thus are also hemizygous for the G6PD gene. Males with Klinefelter syndrome have an additional X chromosome (47, XXY) and thus 2 G6PD alleles. Thus, it is important to consider the number of X chromosomes for an individual when determining G6PD genotype or phenotype.

Glucose-6-phosphate dehydrogenase deficiency affects 400 million people worldwide, with a worldwide prevalence of approximately 5% (25). Glucose-6-phosphate dehydrogenase deficiency appears to be protective against malaria infection leading to a higher prevalence (more than 25%) in countries where malaria is, or once was, endemic; for example, tropical Africa, tropical and subtropical Asia, the Middle East, and some parts of the Mediterranean (26, 27, 28). In the US, G6PD deficiency is more common among African Americans, affecting approximately 12% (29).

The G6PD enzyme catalyzes the first step in the hexose monophosphate shunt (HMP) pathway, which converts glucose to pentose sugars for nucleic acid synthesis. In this step NADP+ is reduced to NADPH, which protects cells from oxidative stress. In mature red blood cells, the HMP pathway is the only source of NADPH. This promotes the generation of reduced glutathione that protects the sulfhydryl groups of hemoglobin, which are susceptible to oxidation by H2O2 and oxygen radicals. Red blood cells that lack G6PD also have a deficiency of NADPH. (30)

Red blood cells that are G6PD deficient have a normal function but are more susceptible to increased oxidative stress (for example, by reactive oxygen species and H2O2). Oxidative stress occurs naturally, but is increased during illnesses, such as infections and diabetic ketoacidosis. Oxidative stress can also follow the ingestion of fresh fava beans (favism), and is an adverse effect of several drugs, for example, the antimalarial drugs primaquine and tafenoquine, the antibacterials dapsone and sulfamethoxazole, the skin cancer drug dabrafenib, and the uric acid lowering drugs pegloticase and rasburicase.

While some reports estimate the frequency of G6PD deficiency to be <0.3% in individuals of European, Finish, or Amish descent, other more targeted population analyses have estimated the frequency of G6PD deficiency to be 0.5% in Portuguese males, 6.4% in males from Cyprus, and 8.3% in newborn males in Greece (31, 32, 33, 34). Among Asian populations, estimates broadly range from 2.7–3.5% of individuals will be G6PD deficient (31). However, a study in Cambodia observed 16% of their male study participants were G6PD deficient (<30% activity) while 32% of their female study participants demonstrated an intermediate level of G6PD activity (30–80%) and 4% were deficient (35). Other studies in Asia report the frequency of G6PD deficiency to be approximately 9–31% in Thailand, almost 30% among the Kachin ethnic group from Myanmar and China, 8% in Lao PDR, 9% in Vietnam, and 15.8% in Myanmar (36, 37, 38, 39, 40). Thus, it is difficult to predict the likelihood of an individual being G6PD deficient based solely on their geographic ancestry, as it can vary significantly within commonly used ancestral designations.

Most individuals with G6PD deficiency are asymptomatic –– they have a normal lifespan and may not know they have G6PD deficiency. However, at birth, they may be predisposed to neonatal jaundice, and throughout life, they will be sensitive to oxidizing agents. All individuals with G6PD deficiency should avoid exposure to oxidizing agents when possible, including drugs such as primaquine.

Symptomatic individuals with G6PD deficiency may suffer from episodes of AHA or, the more severe condition, chronic non-spherocytic hemolytic anemia (CNSHA). The management of hemolytic episodes depends on the severity of hemolysis, with more severe cases requiring blood transfusions. Folic acid may be given to prevent the worsening of anemia in individuals with folate deficiency and to induce formation of new red blood cells.

More than 200 genetic variants of the G6PD gene have been identified so far (41), with approximately 400 biochemical and enzyme variants (42). Most known G6PD variants are missense, which can also be inherited as haplotypes that are comprised of more than one variant allele (43). Large deletions are rare, and a complete lack of G6PD activity is thought to be fatal in utero.

The normal (wild-type) copy of the G6PD gene is known as G6PD B, and is found in most Caucasians, Asians, and Africans. Common G6PD variants include:

G6PD A+ (p.Asn126Asp) has normal enzyme activity and is not associated with hemolysis, and is found in up to 30% of individuals of African descent and approximately 1.5% of Latinos (44, 45)

G6PD A- (p.Asn126Asp with p.Val68Met) is associated with mild to moderate hemolysis and is found in up to 15% of African Americans (46). Additional A- haplotypes have also been identified, both with the A+ variant with a second variant (p.Asn126Asp with p.Arg227Leu; and p.Asn126Asp with p.Leu323Pro. See Nomenclature table below for additional information) (47)

G6PD Mediterranean (p.Ser218Phe) can cause severe hemolysis, and is a common pathogenic variant in Caucasians (48)

G6PD Canton (p.Arg489Leu) can cause severe hemolysis, and is found in Asians (49)

G6PD Viangchan (p.Val291Met) is the most common G6PD variant among Thais, Laotians, Cambodians, and Malaysians (50, 51)

The WHO recently updated its categorization of G6PD variants into 4 classes based on the median residual enzyme activity in males (expressed as a percentage of normal activity) (52). Class A variants have <20% activity and are associated with CNSHA.

Most individuals with G6PD deficiency have variants that belong to class B (enzyme activity less than 45%). Class B variants are associated with intermittent hemolysis, usually triggered by infection or drugs, but most of the time, affected individuals are asymptomatic. Class C variants show median G6PD activity from 60–150% and are not associated with hemolysis. In class U are all the variants with unknown clinical significance, regardless of activity level. The CPIC has assigned G6PD phenotypes based on G6PD genotypes; the updated WHO categories are provided in Table 4 for completeness (3).

Table 4.

Assignment of likely G6PD Phenotype based on Genotype/Diplotype (CPIC, 2022)

Likely phenotypeDefinitionaGenotypeWHO class for G6PD variantsbExample of diplotypec
NormalVery mild or no enzyme deficiency (no less than 60% of normal enzyme levels) (60–150% of normal activity)An X chromosome hemizygote who has a nondeficient (class IV) alleleIV
(C)
B, Sao Borja
An individual who has 2 nondeficient (class IV) allelesIV/IV
(C)
B/B, B/Sao Borja
DeficientLess than 10–60% of normal enzyme activity
(20–45% of normal activity)
An X chromosome hemizygote who has a deficient (class II–III) alleleII, III
(B)
A–, Orissa, Kalyan-Kerala, Mediterranean, Canton, Chatham
An individual who has 2 deficient (class II–III variants) allelesII/II, II/III, III/III
(B)
A–/A–, A–/Orissa, Orissa/Kalyan-Kerala, Mediterranean/Mediterranean, Chatham/ Mediterranean, Canton/Viangchan
Deficient with CNSHASevere enzyme deficiency (<10% activity) and associated with CNSHA
(<20% of normal activity)
An X chromosome hemizygote who has a class I alleleI
(A)
Bangkok, Villeurbanne
An individual who has 2 deficient (class I variants) allelesI/I
(A)
Bangkok/Bangkok, Bangkok/Villeurbanne
VariabledNormal or deficient enzyme activitycAn individual who has one nondeficient (class IV) and one deficient (class I–III variants) alleleIV/I, IV/II, IV/III
(U)
B/A–, B/Mediterranean, B/Bangkok
IndeterminantUncertain(U)

CNSHA - chronic non-spherocytic hemolytic anemia, WHO - World Health Organization, G6PD - glucose-6-phosphate dehydrogenase

a

The traditional (Class I–IV) and updated (A, B, C, and U) activity levels are both provided, with the updated activity ranges provided in parentheses where relevant.

b

WHO classifications were under revision at the time of Clinical Pharmacogenetics Implementation Consortium publication, updated classifications (using A, B, C, and U designations) have been proposed based on enzyme activity levels and are provided in parenthesis here (52).
Class I alleles are extremely rare; the distinction between class II and III alleles is not clear. Almost all individuals will have class II, III, or IV alleles. It should be noted that the class of a variant may have been assigned only by the clinical manifestations of an individual in which the variant was subsequently identified.

c

Due to the large number of G6PD variants, many other diplotypes may be possible besides those given as examples here; see Supplementary data from(3) for a more comprehensive list of alleles with their assigned WHO class. For Human Genome Variation Society terms, please see the Nomenclature table below. The alleles and diplotypes provided here are based upon the historic class I-IV definitions and may not fit the updated WHO classification.

d

Due to X-linked mosaicism, females heterozygous for one nondeficient (class IV) and one deficient (class I–III variants) allele may display a normal or a deficient phenotype. It is, therefore, difficult to predict the phenotype of these individuals (Supplementary Material online (G6PD heterozygotes)) (3).

This table is adapted from (3).

Phenoconversion of G6PD Phenotype

Increased turnover of red blood cells may lead to a temporary increase in G6PD activity as measured by enzyme activity assays. One study of 335 individuals with acute malaria infection (either P. vivax or P. falciparum) found that, on average, G6PD enzyme activity was 10.4% lower in the convalescent, post-infection state than during acute malarial infection. Furthermore, 66–87% of individuals who, following resolution of their malarial infection, had intermediate to severe G6PD deficiency yet they had presented with normal levels of G6PD activity during the acute infection stage (53).

Gene: CYP2D6

The CYP450 superfamily is a large and diverse group of enzymes that form the major system for metabolizing lipids, hormones, toxins, and drugs in the liver. The CYP450 genes are very polymorphic and can result in decreased, absent, or increased enzyme activity. One prominent member, CYP2D6, is responsible for the metabolism of many commonly prescribed drugs, including antidepressants, antipsychotics, analgesics, and beta-blockers (54).

The CYP2D6 Alleles

The CYP2D6 gene is highly polymorphic, as over 100 star (*) alleles have been described and cataloged at the Pharmacogene Variation (PharmVar) Consortium, and each allele is associated with either normal, decreased, or absent enzyme function (Table 5). (55) Star alleles are defined by the variants detected on one chromosome (haplotype).

The combination of CYP2D6 haplotypes that a person has is used to determine their diplotype (for example, CYP2D6 *4/*4). Based on their impact on enzyme function, each allele can be assigned an activity score from 0 to 1, which in turn is then used to assign a phenotype (for example, CYP2D6 PM). However, the activity score system is not standardized across all clinical laboratories or CYP2D6 genotyping platforms. The CPIC revised their activity scoring guidelines in October 2019 to promote harmonization. The CYP2D6 phenotype is predicted from the diplotype activity score defined by the sum of the allele score values, which usually ranges from 0 to 3.0: (56)

An ultrarapid metabolizer (UM) has an activity score greater than 2.25

A normal metabolizer phenotype (NM) has an activity score of 1.25–2.25

An intermediate metabolizer (IM) has an activity score of >0–<1.25

A poor metabolizer (PM) has an activity score of 0

Table 5.

Activity Status of Selected CYP2D6 Alleles

Allele typeCYP2D6 allelesActivity score
Normal function *1, *2, *27, *33 1
Decreased function *17, *41, *49 0.5
Strongly decreased function *10 0.25
No function *3, *4, *5, *6, *36 0

For a comprehensive list of CYP2D6 alleles, please See the Pharmacogene Variation Consortium . Activity scores from (56).

The CYP2D6*1 allele is the wild-type allele when no variants are detected and is associated with normal enzyme activity and the NM phenotype. The CYP2D6*2, *27, and *33 alleles are also considered to have near-normal activity.

Other CYP2D6 alleles include variants that produce a non-functioning enzyme (for example, *3, *4, *5, and *6) (57, 58, 59, 60) or an enzyme with decreased activity (for example, *10, *17, and *41) (61, 62, 63) (see Table 5). There are large inter-ethnic differences in the frequency of these alleles, with *3, *4, *5, *6, and *41 being more common in individuals with European ancestry, *17 more common in Africans, and *10 more common in Asians. (64)

Larger structural variants at the CYP2D6 locus have also been described, including gene duplications, deletions, tandem alleles, and gene conversions. As one might expect, deletions result in a no-function allele (for example, the *5 allele is a deletion). Duplications have been reported for alleles with normal function and decreased function, as well. In the case of allele duplications, the activity scores for the full complement of CYP2D6 alleles are summed to determine the predicted metabolizer phenotype. Additional details on structural variants are available from PharmVar (65).

The frequency of the CYP2D6 star alleles with altered function varies across global populations, resulting in different frequencies of the resulting metabolizer phenotype(s). Given CYP2D6’s role in the metabolism of many drugs, the literature on allele and phenotype frequency is expansive. Most populations have a high frequency for normal-function star alleles, and thus a high proportion of the population are NMs. However, reduced-function alleles like CYP2D6*10 are highly prevalent in East Asian populations, leading to a higher proportion of IM phenotype individuals in this ancestral group. Many groups in sub-Saharan Africa have higher frequencies of decreased-function alleles like CYP2D6*17 and *29, which can correlate with lower metabolizer scores in these individuals. More details regarding published allele and phenotype frequencies are available in the CYP2D6 supplemental chapter.

Phenoconversion of CYP2D6 Phenotype

Factors other than genotype can affect CYP2D6 enzyme activity and, thus the metabolizer phenotype of any individual. Administration of multiple drugs, sometimes called polypharmacy or co-medications, can lead to a phenomenon called phenoconversion, whereby an individual with one metabolizer genotype can have the enzymatic activity of a different metabolizer group (higher or lower, depending on the medications). The enzymatic activity of CYP2D6 can be inhibited or reduced by medications including duloxetine, paroxetine, fluoxetine, bupropion, and quinidine (66, 67, 68, 69). This can result in NMs or IMs responding to medications as if they were PMs. Thus, co-medication with multiple CYP2D6 strong or moderate inhibitors may result in reduced metabolism of drug substrates. In contrast, discontinuing a concomitant CYP2D6 inhibitor can then revert the individual's CYP2D6 activity back to genetically predicted phenotype baseline. Both chloroquine and primaquine are used to treat malaria in regions with chloroquine-sensitive Plasmodium species, however both inhibit CYP2D6 enzyme activity and co-administration was found to inhibit CYP2D6-mediated hydroxylation (70).

Other Genes of Interest

The P450 oxidoreductase (POR) gene encodes the CPR enzyme that is involved in primaquine metabolism. Deficiency of POR presents with a variety of phenotypes; potential clinical presentations include 21-hydroxylase deficiency, polycystic ovary syndrome, and Antley-Bixler syndrome, as well as a distinct disorder of sexual development (71, 72). Genetic variants in POR have been shown to affect the enzymatic activity of many members of the CYP450 family, including 3A4 and 2D6 (73).

Genetic variation in MAO-A (rs6323, NM_000240.4:c.891G>T) has been found to be associated with reduced metabolism of primaquine to carboxyprimaquine in healthy volunteers (74). Variation in CYP2C19 was also shown to influence primaquine metabolism in healthy volunteers, though the clinical impact of the variation was unclear (74).

Drug transport proteins can also impact the efficacy of various medications. Notably, variations in transport proteins encoded by solute carrier organic anion transporter (SLCO)1A2 and SLCO1B1 (75), as well as CYP2C8 (76) variants associated with decreased enzymatic activity, have been associated with altered clearance of P. vivax parasites and a higher frequency of relapse after primaquine and chloroquine therapy. It is possible that the apparent decrease in therapeutic effect associated with these genetic variations is due to altered chloroquine transport or metabolism, particularly as chloroquine is known to be transported by SLCOs and metabolized by CYP2C8.

Linking G6PD and CYP2D6 Genetic Variation with Treatment Response

Individuals with G6PD deficiency (<20% activity) or intermediate deficiency (20–45% activity) (2) are at a significant risk of hemolysis when treated with the standard primaquine course (daily for 14 days) but have shown tolerance for an extended course with a single dose of primaquine per week for 8 weeks (2). One study in Cambodia found that 95% of the individuals with reduced G6PD activity were able to complete the 8-week course of primaquine and no severe adverse events were recorded (35). A meta-analysis of 20 different trials, based in Africa or Asia, of SLD primaquine found that the proposed WHO regimen (0.25 mg/kg) was, indeed, safe, even in the context of G6PD deficiency (7). While individuals with G6PD deficiency demonstrated a more significant drop in hemoglobin concentration in the 2–3 days immediately following treatment and were more likely to experience hemoglobinuria within 72 hours of primaquine treatment, these effects were transitory and only 2 (out of 194) individuals required further intervention (7). Other risk factors for anemia following SLD primaquine, aside from G6PD deficiency, were high parasite density, young age, and the primary anemia risk factor: low baseline hemoglobin levels (7).

Based on non-clinical metabolism data and limited clinical data, the Health Canada approved drug label for primaquine advises that CYP2D6 polymorphism may be associated with variable clinical response and suggests it may be useful to consider drug-drug interaction or CYP2D6 metabolizer status; it further states that for CYP2D6 PMs, alternative treatment should be considered (5). Several studies in the literature suggest that individuals who are CYP2D6 IM or PMs may not respond well to standard primaquine therapy for the radical cure of P. vivax malaria, which may result in a relapse of malaria symptoms and positive malaria tests weeks to months later (77). There are several case reports that link CYP2D6 reduced enzymatic activity or IM/PM genotypes with malaria relapse after primaquine therapy (78, 79, 80, 81, 82, 83). A study of 25 individuals found that IM or PM phenotypes were associated with malaria relapse, while individuals with CYP2D6 NM phenotype did not experience relapse following treatment with chloroquine and primaquine (84). Additionally, a case-control study of 57 individuals found that CYP2D6 IM or PM phenotype (determined either by genotype or inferred based on reduced dextromethorphan metabolism) strongly correlated with increased frequency of malaria relapse (85). A prospective cohort study with 190 individuals found P. vivax malaria relapse was more common among individuals with reduced CYP2D6 activity alleles (86). A study of 260 individuals living in a P. vivax endemic region of the Amazon found a significant correlation between CYP2D6 reduced-function genotype (AS<1) and risk of malaria recurrence (87). In Korea, individuals with CYP2D6 IM phenotype were more likely (an odds ratio of 2.33) to have P. vivax malaria relapse even after treatment with primaquine (82). Similar results were observed in a study with 120 individuals in the Yunnan Province of China, where the c.886C>T and CYP2D6*2 variants were associated with relapse of P. vivax malaria after treatment with chloroquine and an 8-day course of primaquine (88). A meta-analysis of 9 studies that included a total of 970 individuals from Asia, Brazil, and Oceania found that CYP2D6 IM and PMs were nearly twice as likely to experience malaria relapse following primaquine therapy as compared to NM or UM individuals (82). In contrast, a small study of 51 individuals treated with chloroquine and primaquine combination therapy did not observe any significant enrichment for CYP2D6 IM or PMs among the relapse group, though the limited number of relapses and sample size may have left this study underpowered (89). A larger study with 157 subjects from Australia also found no association between CYP2D6 activity and malaria relapse (90).

Reduced CYP2D6 activity was found to be associated with reduced clearance of P. falciparum gametocytes in SLD primaquine therapy in a study of 774 individuals from Africa; however, even with reduced CYP2D6 activity, the addition of SLD primaquine was more effective to clear gametocytes than ACT alone (91). This same study found no significant impact of CYP2D6 activity on the frequency or degree of anemia following SLD primaquine in G6PD deficient individuals (91). Similarly, a study of 157 children, aged 1–10 years, found no difference in the incidence or severity of acute hemolysis nor in efficacy against the Plasmodium parasites among individuals with reduced CYP2D6 or G6PD activity treated with ACT with SLD primaquine, leading the authors to conclude that this treatment regimen is both safe and sufficient to reduce P. falciparum transmission (7). The WHO guidelines do not require G6PD testing when administering SLD primaquine (2).

The G6PD and CYP2D6 Gene Interactions with Medications Used for Additional Indications

Medications that can induce oxidative stress in red blood cells can trigger hemolysis readily in individuals with G6PD enzyme deficiency. Many of these medications are antimalarials (tafenoquine, for example) but many more medications pose a hazard for G6PD deficient individuals.

Urate-lowering medications: both refractory gout and tumor lysis syndrome can cause systemic elevation of urate levels, medications such as rasburicase and pegloticase are uricase enzymes that aid in the breakdown of uric acid into more soluble metabolites. These reactions produce H2O2 as a byproduct, thus increasing oxidative stress in the body.

Kinase inhibitors: anticancer medications such as dabrafenib may also increase oxidative stress.

Antimicrobial medications: nitrofurantoin, often used for urinary tract infections, was determined to be a medication of moderate risk for AHA in G6PD-deficient individuals by CPIC and may call for additional monitoring. In contrast, CPIC found sulfamethoxazole to be a medication with low-to-no risk in G6PD deficient individuals. (3)

Additional information on gene-drug interactions for G6PD are available from PharmGKBCPIC and the FDA (search for “G6PD”).

The CYP family of enzymes is involved in the metabolism of many substances and CYP2D6 especially has been implicated in altered pharmacologic responses for many compounds. The drugs can be categorized into many different classes:

Antipsychotics—for example, aripiprazole, risperidone, thioridazine and—to a lesser extent—clozapine is metabolized by CYP2D6. According to the FDA, aripiprazole dosage should be reduced for PMs and thioridazine is contraindicated for individuals who are known to have reduced CYP2D6 activity due to increased risk of potentially fatal side effects. The UMs may have a decreased plasma concentration of risperidone.

Tricyclic antidepressants—for example, amitriptyline, and imipramine may require dosage adjustments, potentially guided by therapeutic drug monitoring, to achieve the desired therapeutic range in UMs or PMs. Ultimately, tricyclic antidepressants may be ineffective in CYP2D6 UMs.

Serotonin and norepinephrine reuptake inhibitors—for example atomoxetine and venlafaxine may have reduced efficacy in UMs at standard doses while PMs are at risk of elevated plasma concentrations for both medications. The Dutch Pharmacogenetics Working Group advises against the use of venlafaxine in CYP2D6 PMs and IMs.

Cardiovascular dysfunction—for example, carvedilol, metoprolol, and propafenone are all metabolized by CYP2D6, and PMs will have higher plasma concentrations of these medications compared with NMs resulting in potentially undesired side effects or (in the case of metoprolol) extensive slowing of the heart rate.

Anticancer medications—for example, tamoxifen is activated by CYP2D6, and IMs or PMs may have reduced benefit from tamoxifen therapy.

Pain management—for example, codeine and tramadol are pro-drugs that require activation by CYP2D6 to achieve the desired analgesic effect.

Various therapies for genetic disorders—for example eliglustat used in the treatment of Gaucher disease, and deutetrabenazine used in the treatment of Huntington disease—have reduced dose recommendations for CYP2D6 PMs. The CYP2D6 UMs may not achieve adequate concentrations of eliglustat and therefore CYP2D6 genotyping is required before initiation of eliglustat therapy.

It is important to note that CYP2D6 is the most common biomarker in drug responses for FDA drug labels, the list provided here is by no means exhaustive. Additional information on gene-drug interactions for CYP2D6 are available from PharmGKBCPIC and the FDA (search for “CYP2D6”).

Genetic Testing

The NIH Genetic Testing Registry (GTR) displays genetic tests that are available for primaquine response, the G6PD gene, and the CYP2D6 gene. Molecular genetic testing can be used to confirm the diagnosis of G6PD deficiency, and testing may also be used to screen females with a family history of G6PD deficiency to see if they are carriers.

While many biochemical G6PD variants are known, the genetic underpinnings of some of these variants may still be unknown. Additionally, quantitative, or semi-quantitative tests for G6PD enzyme activity may be more readily available in some settings. Whether the clinical test is biochemical or molecular, assessment of G6PD enzyme activity is required before administering primaquine for the radical cure of P. vivax or P. ovale malaria, per the FDA (1). A number of point-of-care tests have been developed and tested (92, 93, 94) to improve the accessibility of G6PD genetic testing before administration of medications like primaquine, though the availability of such testing in areas with the highest malarial burden is still lacking (95).

The available CYP2D6 tests include targeted single-gene tests as well as multi-gene panels or genome-wide sequencing tests. In addition, variant CYP2D6 alleles to be included in clinical genotyping assays have been recommended by the Association for Molecular Pathology (96). The test results may include an interpretation of the individual’s predicted metabolizer phenotype, which can be confirmed by checking the diplotype and calculating the CYP2D6 activity score, as described in the “CYP2D6 Alleles” section above. When individuals have more than 2 copies of the CYP2D6, the copies of the allele are denoted by an “xN”, for example, CYP2D6*1/*2x2. Some laboratories also use the notation of DUP to indicate an increase in copy number, but the report does not always specify the number of duplications nor the allele that has been duplicated due to technical limitations.

Therapeutic Recommendations based on Genotype

This section contains excerpted 1 information on gene-based dosing recommendations. Neither this section nor other parts of this review contain the complete recommendations from the sources.

2021 Statement from the US Food and Drug Administration (FDA):

Due to the risk of hemolytic anemia in patients with G6PD deficiency, G6PD testing has to be performed before using primaquine. Due to the limitations of G6PD tests, physicians need to be aware of residual risk of hemolysis and adequate medical support and follow-up to manage hemolytic risk should be available.

Primaquine should not be prescribed for patients with severe G6PD deficiency…

In case of mild to moderate G6PD deficiency, a decision to prescribe primaquine must be based on an assessment of the risks and benefits of using primaquine. If primaquine administration is considered, baseline hematocrit and hemoglobin must be checked before treatment and close hematological monitoring (e.g. at day 3 and 8) is required. Adequate medical support to manage hemolytic risk should be available.

When the G6PD status is unknown and G6PD testing is not available, a decision to prescribe primaquine must be based on an assessment of the risks and benefits of using primaquine. Risk factors for G6PD deficiency or favism must be assessed. Baseline hematocrit and hemoglobin must be checked before treatment and close hematological monitoring (e.g. at day 3 and 8) is required. Adequate medical support to manage hemolytic risk should be available.

Please review the complete therapeutic recommendations that are located here: (1).

2022 Statement from the Clinical Pharmacogenetics Implementation Consortium (CPIC)

It is recommended to avoid primaquine at standard (or higher) anti-relapse dosages of 0.25–0.5 mg/kg daily for 14 days for the treatment of P. vivax or P. ovale in G6PD deficiency. For the anti-gametocyte treatment of Plasmodium falciparum malaria, the single-dose regimen of 0.25 mg/kg is considered safe and effective (low to no risk in G6PD deficiency). For the treatment of P. vivax or P. ovale malaria for radical cure of liver-stage infections, 0.75 mg/kg once weekly for 8 weeks (WHO) or 45 mg for adults once weekly for 8 weeks (CDC) is considered in the medium risk category, and patients should be monitored closely for hemolysis… No dose of primaquine is considered safe in patients who are G6PD deficient with CNSHA and thus should be avoided.

Please review the complete therapeutic recommendations that are located here: (3).

2020 Statement from Health Canada:

Hemolytic anemia and G6PD deficiency

Due to the risk of hemolytic anemia in G6PD deficient patients, G6PD testing has to be performed before using primaquine. […] Due to the limitations of G6PD tests, physicians need to be aware of residual risk of hemolysis and adequate medical support and follow-up to manage hemolytic risk should be available. Observe particular caution in individuals with a personal or family history of hemolytic anemia.

In case of mild to moderate G6PD deficiency, a decision to prescribe primaquine must be based on an assessment of the risks and benefits of using primaquine; if primaquine administration is considered, the dosage regimen should be adapted accordingly (see DOSAGE AND ADMINISTRATION) and close hematological monitoring is required.

[….]

CYP2D6 genotype:

Based on non-clinical data, primaquine activity probably depends on the formation CYP2D6 metabolite(s). Therefore, CYP2D6 polymorphism may be associated with variability in clinical response to primaquine.

Limited clinical data reported more elevated treatment failure rates in patients with CYP2D6 poor or intermediate metabolizer status than in patients with normal/extensive metabolizer status (see ACTION AND CLINICAL PHARMACOLOGY).

In case of treatment failure, after checking patient’s compliance to treatment, it may be useful to reconsider potential concomitant use of CYP2D6 inhibitors (see DRUG INTERACTIONS) and to assess the patient’s CYP2D6 status if feasible. For poor CYP2D6 metabolizers, alternative treatment should be considered.

Please review the complete therapeutic recommendations that are located here: (5).

Nomenclature for Selected G6PD and CYP2D6 Alleles

Nomenclature of Selected G6PD Alleles


Common allele name / condition
Alternative names / conditionHGVS reference sequenceWHO Classification*dbSNP reference identifier for allele location
CodingProtein
G6PD BWTNM_001042351.3NP_001035810.1IV/ Normal --
G6PD A+p.Asn126AspNM_001042351.3:c.376A>GNP_001035810.1:p.Asn126AspIV/ Normal rs1050828
G6PD Sao Borjap.Asp113AsnNM_001042351.3:c.337G>ANP_001035810.1:p.Asp113AsnIV/ Normal rs5030870
G6PD A-A-202A/376GNM_001042351.3:c.376A>GNP_001035810.1:p.Asn126AspIII/ Deficient rs1050828 rs1050829
NM_001042351.3:c.202G>ANP_001035810.1:p.Val68Met
G6PD A-A-680T/376GNM_001042351.3:c.376A>GNP_001035810.1:p.Asn126AspIII/ Deficient rs1050828 rs137852328
NM_001042351.3:c.680G>TNP_001035810.1:p.Arg227Leu
G6PD A-A-968C/376GNM_001042351.3:c.376A>GNP_001035810.1:p.Asn126AspIII/ Deficient rs1050828 rs76723693
NM_001042351.3:c.968T>CNP_001035810.1:p.Leu323Pro
G6PD Bangkokp.Lys275AsnNM_001042351.3:c.202G>ANP_001035810.1:p.Val68MetIII/ Deficient
G6PD Kalyan-Keralap.Glu317LysNM_001042351.3:c.949G>ANP_001035810.1:p.Glu317LysIII/ Deficient rs137852339
G6PD Orissap.Ala44GlyNM_001042351.3:c.131C>GNP_001035810.1:p.Ala44GlyIII/ Deficient rs78478128
GP6D Cantonp.Arg459LeuNM_001042351.3:c.1376G>TNP_001035810.1:p.Arg459LeuII/ Deficient rs72554665
G6PD Chathamp.Ala335ThrNM_001042351.3:c.1003G>ANP_001035810.1:p.Ala335ThrII/ Deficient rs5030869
G6PD Mediterraneanp.Ser188PheNM_001042351.3:c.563C>TNP_001035810.1:pSer188PheII/ Deficient rs5030868
G6PD Viangchanp.Val291MetNM_001042351.3:c.871G>ANP_001035810.1:p.Val291MetII/ Deficient rs137852327
G6PD Villeurbannep.Thr334delNM_001042351.3:c.1000_1002delACCNP_001035810.1:pThr334delI/Deficient with CNSHAn/a

Additional allele information available from PharmGKB and CPIC’s G6PD Allele Definition Table (revised 2018).

Guidelines for the description and nomenclature of gene variations are available from the Human Genome Variation Society (HGVS).

*

WHO classifications based on (97) WHO - World Health Organization; PharmGKB - Pharmacogenomics Knowledgebase; CPIC - Clinical Pharmacogenetics Implementation Consortium; CNSHA - chronic non-spherocytic hemolytic anemia, G6PD - glucose-6-phosphate dehydrogenase

Nomenclature of Selected CYP2D6 Alleles

Common allele nameAlternative namesHGVS reference sequencedbSNP reference identifier for allele location
CodingProtein
CYP2D6*22851C>T NM_000106.6:c.457G>C NP_000097.3:p.Arg296Cys rs16947
4181G>C NM_000106.6:c.1457G>C NP_000097.3:p.Ser486Thr rs1135840
CYP2D6*32550delANM_000106.6:c.775delNP_000097.3:p.Arg259fs rs35742686
CYP2D6*41846G>A NM_000106.6:c.506-1G>A Variant occurs in a non-coding region (splice variant causes a frameshift) rs3892097
CYP2D6*5Gene deletion
CYP2D6*61707 del T NM_000106.6:c.454delT NP_000097.3:p.Trp152Glyfs rs5030655
CYP2D6*10100C>T NM_000106.6:c.100C>T NP_000097.3:p.Pro34Ser rs1065852
4181G>C NM_000106.6:c.1457G>C NP_000097.3:p.Ser486Thr rs1135840
CYP2D6*171022C>T NM_000106.6:c.320C>T NP_000097.3:p.Thr107Ile rs28371706
2851C>T NM_000106.6:c.886T>C NP_000097.3:p.Cys296Arg rs16947
4181G>C NM_000106.6:c.1457G>C NP_000097.3:p.Ser486Thr rs1135840
CYP2D6*273854G>ANM_000106.6:c.1228G>ANP_000097.3:p.Glu410Lys rs769157652
CYP2D6*312851C>T NM_000106.6:c.886C>T NP_000097.3:p.Arg296Cys rs16947
4043G>A NM_000106.6:c.1319G>A NP_000097.3:p.Arg440His rs267608319
4181G>C NM_000106.6:c.1457G>C NP_000097.3:p.Ser486Thr rs1135840
CYP2D6*36[1]100C>T NM_000106.6:c.100C>T NP_000097.3:p.Pro34Ser rs1065852
4129C>GNM_000106.6:c.1405C>GNP_000097.3:p.Pro469Ala rs1135833
4132A>GNM_000106.6:c.1408A>GNP_000097.3:p.Thr470Ala rs1135835
4156C>T+4157A>CNM_000106.6:c.1432C>T+ NM_000106.6:c.1433A>CNP_000097.3:p.His47Serrs28371735+ rs766507177
4159G>CNM_000106.6:c.1435G>CNP_00097.3:p.Gly479Arg
4165T>GNM_000106.6:c.1441T>GNP_00097.3:p.Phe481Val
4168G>A+4169C>GNM_000106.6:c.1444G>A+ NM_000106.6:c.1445C>GNP_000097.3:p.Ala482Serrs74478221+ rs75467367
4181G>C NM_000106.6:c.1457G>C NP_000097.3:p.Ser486Thr rs1135840
CYP2D6*412851C>T NM_000106.6:c.886T>C NP_000097.3:p.Cys296Arg rs16947
2989G>A NM_000106.6:c.985+39G>A Variant occurs in a non-coding region (impacts slicing). rs28371725
4181G>C NM_000106.6:c.1457G>C NP_000097.3:p.Ser486Thr rs1135840
CYP2D6*49100C>T NM_000106.6:c.100C>T NP_000097.3:p.Pro34Ser rs1065852
1612T>ANM_00106.6:c.358T>ANP_000097.3:p.Phe120Ile rs1135822
4181G> NM_000106.6:c.1457G>C NP_000097.3:p.Ser486Thr rs1135840
[1]

CYP2D6*36 is a gene conversion with CYP2D7; variants provided here are from the Pharmacogene Variation Consortium.

CYP2D6 - cytochrome P450 member 2D6, dbSNP - database of single nucleotide polymorphisms

Alleles described in this table are selected based on discussion in the text above. This is not intended to be an exhaustive description of known alleles.

Pharmacogenetic Allele Nomenclature: International Workgroup Recommendations for Test Result Reporting (98).

Guidelines for the description and nomenclature of gene variations are available from the Human Genome Variation Society (HGVS).

Nomenclature for Cytochrome P450 enzymes is available from the Pharmacogene Variation (PharmVar) Consortium.

Acknowledgments

The author would like to thank Amit Sharma, PhD, Former Director of the National Institute for Malaria Research, Group Leader, Molecular Medicine Group, ICGEB, New Delhi, India, and Katherine Riden, PharmD, AccessDx Laboratory, Houston, TX, USA for reviewing this summary.

References

1.
Primaquine Phosphate. Sanofi-Aventis U.S. LLC; 2021. Available from: https://dailymed​.nlm​.nih.gov/dailymed/drugInfo​.cfm?setid=1bfbf4ae-81b8-4160-a00d-6322aadd4b59.
2.
WHO Guidelines for Malaria, 3 June 2022. Geneva, Switzerland, WHO Global Malaria Programme; [Cited Available from: https://app​.magicapp​.org/#/guideline/6287.
3.
Gammal R.S., Pirmohamed M., Somogyi A.A., Morris S.A., et al. Expanded Clinical Pharmacogenetics Implementation Consortium Guideline for Medication Use in the Context of G6PD Genotype. Clin Pharmacol Ther. 2022 [PMC free article: PMC10281211] [PubMed: 36049896]
4.
Baird J.K. 8-Aminoquinoline Therapy for Latent Malaria. Clin Microbiol Rev. 2019;32(4) [PMC free article: PMC6750137] [PubMed: 31366609]
5.
Product Monograph Primaquine phosphate Laval, Quebec, Canada: Sanofi-Aventis Canada Inc; 2020. Available from: https://pdf​.hres.ca/dpd_pm/00057221.PDF.
6.
Lalloo D.G., Shingadia D., Bell D.J., Beeching N.J., et al. UK malaria treatment guidelines 2016. J Infect. 2016;72(6):635–649. [PMC free article: PMC7132403] [PubMed: 26880088]
7.
Stepniewska K., Allen E.N., Humphreys G.S., Poirot E., et al. Safety of single-dose primaquine as a Plasmodium falciparum gametocytocide: a systematic review and meta-analysis of individual patient data. BMC Med. 2022;20(1):350. [PMC free article: PMC9479278] [PubMed: 36109733]
8.
Ashley E.A., Recht J., White N.J. Primaquine: the risks and the benefits. Malar J. 2014;13:418. [PMC free article: PMC4230503] [PubMed: 25363455]
9.
Nain M., Mohan M., Sharma A. Effects of Host Genetic Polymorphisms on the Efficacy of the Radical Cure Malaria Drug Primaquine. Am J Trop Med Hyg. 2022;106(3):764–767. [PMC free article: PMC8922494] [PubMed: 35008050]
10.
Camarda G., Jirawatcharadech P., Priestley R.S., Saif A., et al. Antimalarial activity of primaquine operates via a two-step biochemical relay. Nat Commun. 2019;10(1):3226. [PMC free article: PMC6642103] [PubMed: 31324806]
11.
Suarez-Kurtz G. Impact of CYP2D6 Genetic Variation on Radical Cure of Plasmodium vivax Malaria. Clin Pharmacol Ther. 2021;110(3):595–598. [PubMed: 34042179]
12.
Gilder M.E., Hanpithakphong W., Hoglund R.M., Tarning J., et al. Primaquine Pharmacokinetics in Lactating Women and Breastfed Infant Exposures. Clin Infect Dis. 2018;67(7):1000–1007. [PMC free article: PMC6137118] [PubMed: 29590311]
13.
Primaquine, in Drugs and Lactation Database (LactMed(R)). 2006: Bethesda (MD).
14.
Malaria, G.H.D.o.P.D.a. Treatment of Malaria: Guidelines for Clinicians (United States). 2023 14 Feb 2023 7 March 2023; Available from: https://www​.cdc.gov/malaria​/diagnosis_treatment/clinicians1​.html.
15.
Setyadi A., Arguni E., Kenangalem E., Hasanuddin A., et al. Safety of primaquine in infants with Plasmodium vivax malaria in Papua, Indonesia. Malar J. 2019;18(1):111. [PMC free article: PMC6444676] [PubMed: 30940140]
16.
Chu C.S., Watson J.A., Phyo A.P., Win H.H., et al. Determinants of Primaquine and Carboxyprimaquine Exposures in Children and Adults with Plasmodium vivax Malaria. Antimicrob Agents Chemother. 2021;65(11):e0130221. p. [PMC free article: PMC8522776] [PubMed: 34398667]
17.
Vieira M., Matos Lopes T.R., Mello A., de Sena L.W.P., et al. Doses of primaquine administered to children with Plasmodium vivax according to an age-based dose regimen. Pathog Glob Health. 2020;114(7):388–392. [PMC free article: PMC7580727] [PubMed: 32705964]
18.
Goncalves B.P., Pett H., Tiono A.B., Murry D., et al. Age, Weight, and CYP2D6 Genotype Are Major Determinants of Primaquine Pharmacokinetics in African Children. Antimicrob Agents Chemother. 2017;61(5) [PMC free article: PMC5404566] [PubMed: 28289025]
19.
Dobano C., Ubillos I., Jairoce C., Gyan B., et al. RTS,S/AS01E immunization increases antibody responses to vaccine-unrelated Plasmodium falciparum antigens associated with protection against clinical malaria in African children: a case-control study. BMC Med. 2019;17(1):157. [PMC free article: PMC6693200] [PubMed: 31409398]
20.
CDC. CDC- Malaria- Travelers- Risk Assessment. 2018 23 July 2020 14 August 2020; Available from: https://www​.cdc.gov/malaria​/travelers/risk_assessment.html.
21.
Chiodini, P., D. Patel and C. Whitty, Guidelines for malaria prevention in travellers from the UK 2019. 2019, Public Health England Advisory Committee on Malaria Prevention: London.
22.
World malaria report 2022, Geneva, [Cited 12 Jan 2023]. Available from: https://www​.who.int/teams​/global-malaria-programme​/reports/world-malaria-report-2022.
23.
Tse E.G., Korsik M., Todd M.H. The past, present and future of anti-malarial medicines. Malar J. 2019;18(1):93. [PMC free article: PMC6431062] [PubMed: 30902052]
24.
Luzzatto L. Sickle cell anaemia and malaria. Mediterr J Hematol Infect Dis. 2012;4(1):e2012065. p. [PMC free article: PMC3499995] [PubMed: 23170194]
25.
Ruwende C., Khoo S.C., Snow R.W., Yates S.N., et al. Natural selection of hemi- and heterozygotes for G6PD deficiency in Africa by resistance to severe malaria. Nature. 1995;376(6537):246–9. [PubMed: 7617034]
26.
Ruwende C., Hill A. Glucose-6-phosphate dehydrogenase deficiency and malaria. J Mol Med (Berl). 1998;76(8):581–8. [PubMed: 9694435]
27.
Glucose-6-phosphate dehydrogenase deficiency. WHO Working Group. Bull World Health Organ. 1989;67(6):601–11. [PMC free article: PMC2491315] [PubMed: 2633878]
28.
Chinevere T.D., Murray C.K., Grant E. Jr, Johnson G.A., et al. Prevalence of glucose-6-phosphate dehydrogenase deficiency in U.S. Army personnel. Mil Med. 2006;171(9):905–7. [PubMed: 17036616]
29.
Kaplan M., Herschel M., Hammerman C., Hoyer J.D., et al. Hyperbilirubinemia among African American, glucose-6-phosphate dehydrogenase-deficient neonates. Pediatrics. 2004;114(2):e213–9. [PubMed: 15286259]
30.
Cappellini M.D., Fiorelli G. Glucose-6-phosphate dehydrogenase deficiency. Lancet. 2008;371(9606):64–74. [PubMed: 18177777]
31.
Koromina M., Pandi M.T., van der Spek P.J., Patrinos G.P., et al. The ethnogeographic variability of genetic factors underlying G6PD deficiency. Pharmacol Res. 2021;173:105904. p. [PubMed: 34551338]
32.
Manco, L., C. Bento, L. Relvas, T. Maia, et al., Molecular Heterogeneity of Glucose-6-Phosphate Dehydrogenase (G6PD) Deficiency in the Portuguese Population. Acta Med Port, 2022. [PubMed: 36150187]
33.
Drousiotou A., Touma E.H., Andreou N., Loiselet J., et al. Molecular characterization of G6PD deficiency in Cyprus. Blood Cells Mol Dis. 2004;33(1):25–30. [PubMed: 15223006]
34.
Molou E., Schulpis K.H., Thodi G., Georgiou V., et al. Glucose-6-Phosphate Dehydrogenase (G6PD) deficiency in Greek newborns: the Mediterranean C563T mutation screening. Scand J Clin Lab Invest. 2014;74(3):259–63. [PubMed: 24460025]
35.
Kheang S.T., Ridley R., Ngeth E., Ir P., et al. G6PD testing and radical cure for Plasmodium vivax in Cambodia: A mixed methods implementation study. PLoS One. 2022;17(10):e0275822. p. [PMC free article: PMC9584508] [PubMed: 36264996]
36.
Nuinoon M., Krithong R., Pramtong S., Sasuk P., et al. Prevalence of G6PD deficiency and G6PD variants amongst the southern Thai population. PeerJ. 2022;10:e14208. p. [PMC free article: PMC9559062] [PubMed: 36248708]
37.
Li Q., Yang F., Liu R., Luo L., et al. Prevalence and Molecular Characterization of Glucose-6-Phosphate Dehydrogenase Deficiency at the China-Myanmar Border. PLoS One. 2015;10(7):e0134593. p. [PMC free article: PMC4520570] [PubMed: 26226515]
38.
Bancone G., Menard D., Khim N., Kim S., et al. Molecular characterization and mapping of glucose-6-phosphate dehydrogenase (G6PD) mutations in the Greater Mekong Subregion. Malar J. 2019;18(1):20. [PMC free article: PMC6343352] [PubMed: 30674319]
39.
Sathupak S., Leecharoenkiat K., Kampuansai J. Prevalence and molecular characterization of glucose-6-phosphate dehydrogenase deficiency in the Lue ethnic group of northern Thailand. Sci Rep. 2021;11(1):2956. [PMC free article: PMC7858617] [PubMed: 33536585]
40.
Thedsawad A., Wanachiwanawin W., Taka O., Hantaweepant C. Cut-off values for diagnosis of G6PD deficiency by flow cytometry in Thai population. Ann Hematol. 2022;101(10):2149–2157. [PMC free article: PMC9463333] [PubMed: 35840819]
41.
Gomez-Manzo S., Marcial-Quino J., Vanoye-Carlo A., Serrano-Posada H., et al. Glucose-6-Phosphate Dehydrogenase: Update and Analysis of New Mutations around the World. Int J Mol Sci. 2016;17(12) [PMC free article: PMC5187869] [PubMed: 27941691]
42.
Valencia S.H., Ocampo I.D., Arce-Plata M.I., Recht J., et al. Glucose-6-phosphate dehydrogenase deficiency prevalence and genetic variants in malaria endemic areas of Colombia. Malar J. 2016;15(1):291. [PMC free article: PMC4880879] [PubMed: 27225440]
43.
Miwa S., Fujii H. Molecular basis of erythroenzymopathies associated with hereditary hemolytic anemia: tabulation of mutant enzymes. Am J Hematol. 1996;51(2):122–32. [PubMed: 8579052]
44.
Boyer S.H., Porter I.H., Weilbacher R.G. Electrophoretic heterogeneity of glucose-6-phosphate dehydrogenase and its relationship to enzyme deficiency in man. Proc Natl Acad Sci U S A. 1962;48:1868–76. [PMC free article: PMC221054] [PubMed: 14014720]
45.
G6PD frequency table, Clinical Pharmacogenetics Implementation Consortium; [Cited 15 Oct 2022]. Available from: https://files​.cpicpgx​.org/data/report/current​/frequency/G6PD_frequency_table​.xlsx.
46.
Reys L., Manso C., Stamatoyannopoulos G. Genetic studies on southeastern Bantu of Mozambique. I. Variants of glucose-6-phosphate dehydrogenase. Am J Hum Genet. 1970;22(2):203–15. [PMC free article: PMC1706530] [PubMed: 5435642]
47.
McDonagh E.M., Thorn C.F., Bautista J.M., Youngster I., et al. PharmGKB summary: very important pharmacogene information for G6PD. Pharmacogenet Genomics. 2012;22(3):219–28. [PMC free article: PMC3382019] [PubMed: 22237549]
48.
Oppenheim A., Jury C.L., Rund D., Vulliamy T.J., et al. G6PD Mediterranean accounts for the high prevalence of G6PD deficiency in Kurdish Jews. Hum Genet. 1993;91(3):293–4. [PubMed: 8478015]
49.
McCurdy P.R., Kirkman H.N., Naiman J.L., Jim R.T., et al. A Chinese variant of glucose-6-phosphate dehydrogenase. J Lab Clin Med. 1966;67(3):374–85. [PubMed: 4379606]
50.
Louicharoen C., Nuchprayoon I. G6PD Viangchan (871G>A) is the most common G6PD-deficient variant in the Cambodian population. J Hum Genet. 2005;50(9):448–452. [PubMed: 16155737]
51.
Yusoff N.M., Shirakawa T., Nishiyama K., Ee C.K., et al. G6PD Viangchan and G6PD Mediterranean are the main variants in G6PD deficiency in the Malay population of Malaysia. Southeast Asian J Trop Med Public Health. 2003;34 Suppl 3:135–7. [PubMed: 15906717]
52.
Meeting report of the technical consultation to review the classification of glucose-6-phosphate dehydrogenase (G6PD), Global Malaria Programme Malaria Policy Advisory Group; [Cited 7 Oct 2022]. Available from: https://www​.who.int/publications​/m/item/WHO-UCN-GMP-MPAG-2022.01.
53.
Ley B., Alam M.S., Satyagraha A.W., Phru C.S., et al. Variation in Glucose-6-Phosphate Dehydrogenase activity following acute malaria. PLoS Negl Trop Dis. 2022;16(5):e0010406. p. [PMC free article: PMC9094517] [PubMed: 35544453]
54.
Nofziger C., Turner A.J., Sangkuhl K., Whirl-Carrillo M., et al. PharmVar GeneFocus: CYP2D6. Clin Pharmacol Ther. 2020;107(1):154–170. [PMC free article: PMC6925641] [PubMed: 31544239]
55.
Gaedigk A., Ingelman-Sundberg M., Miller N.A., Leeder J.S., et al. The Pharmacogene Variation. (PharmVar) Consortium: Incorporation of the Human Cytochrome P450 (CYP) Allele Nomenclature Database. Clin Pharmacol Ther. 2018;103(3):399–401. [PMC free article: PMC5836850] [PubMed: 29134625]
56.
CPIC. CPIC® Guideline for Codeine and CYP2D6. 2019 October 2019 2020 June Available from: https://cpicpgx​.org/guidelines​/guideline-for-codeine-and-cyp2d6/.
57.
Yokota H., Tamura S., Furuya H., Kimura S., et al. Evidence for a new variant CYP2D6 allele CYP2D6J in a Japanese population associated with lower in vivo rates of sparteine metabolism. Pharmacogenetics. 1993;3(5):256–63. [PubMed: 8287064]
58.
Codeine and Morphine Pathway, Pharmacokinetics Palo Alto (CA): Stanford University, [Cited 2012 July 24]. Available from: http://www​.pharmgkb.org​/pathway/PA146123006.
59.
Ingelman-Sundberg M. Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity. Pharmacogenomics J. 2005;5(1):6–13. [PubMed: 15492763]
60.
Haplotype CYP2D6*1, Palo Alto (CA): Stanford University, [Cited 2020 June 11]. Available from: http://www​.pharmgkb.org​/haplotype/PA165816576.
61.
Haplotype CYP2D6*4, Palo Alto (CA): Stanford University, [Cited 8 October 2015]. Available from: http://www​.pharmgkb.org​/haplotype/PA165816579.
62.
Haplotype CYP2D6*6, Palo Alto (CA): Stanford University, [Cited 8 October 2015]. Available from: http://www​.pharmgkb.org​/haplotype/PA165816581.
63.
Haplotype CYP2D6*10, Palo Alto (CA): Stanford University, [Cited 8 October 2015]. Available from: http://www​.pharmgkb.org​/haplotype/PA165816582.
64.
Bradford L.D. CYP2D6 allele frequency in European Caucasians, Asians, Africans and their descendants. Pharmacogenomics. 2002;3(2):229–43. [PubMed: 11972444]
65.
Consortium, P.V. Structural Variation for CYP2D6. 2022 14 March 2022; Available from: https://a​.storyblok.com​/f/70677/x/d842ef4108​/cyp2d6_structural-variation_v3-1​.pdf.
66.
FDA. Drug Development and Drug Interactions: Table of Substrates, Inhibitors and Inducers. 2020; Available from: https://www​.fda.gov/drugs​/drug-interactions-labeling​/drug-development-and-drug-interactions-table-substrates-inhibitors-and-inducers.
67.
Smith D.M., Weitzel K.W., Elsey A.R., Langaee T., et al. CYP2D6-guided opioid therapy improves pain control in CYP2D6 intermediate and poor metabolizers: a pragmatic clinical trial. Genet Med. 2019;21(8):1842–1850. [PMC free article: PMC6650382] [PubMed: 30670877]
68.
Codeine sulfate tablets for oral use [package insert]. Philadelphia, PA: Lannett Company, I.; 2019. Available from: https://dailymed​.nlm​.nih.gov/dailymed/drugInfo​.cfm?setid=5819bdf7-300e-45b8-8f3a-447b53656293.
69.
Monte A.A., West K., McDaniel K.T., Flaten H.K., et al. CYP2D6 Genotype Phenotype Discordance Due to Drug-Drug Interaction. Clin Pharmacol Ther. 2018;104(5):933–939. [PMC free article: PMC6197912] [PubMed: 29882961]
70.
Fasinu P.S., Tekwani B.L., Avula B., Chaurasiya N.D., et al. Pathway-specific inhibition of primaquine metabolism by chloroquine/quinine. Malar J. 2016;15:466. [PMC free article: PMC5020452] [PubMed: 27618912]
71.
Pandey A.V., Sproll P. Pharmacogenomics of human P450 oxidoreductase. Front Pharmacol. 2014;5:103. [PMC free article: PMC4023047] [PubMed: 24847272]
72.
Bai Y., Li J., Wang X. Cytochrome P450 oxidoreductase deficiency caused by R457H mutation in POR gene in Chinese: case report and literature review. J Ovarian Res. 2017;10(1):16. [PMC free article: PMC5348910] [PubMed: 28288674]
73.
Burkhard, F.Z., S. Parween, S.S. Udhane, C.E. Fluck, et al., P450 Oxidoreductase deficiency: Analysis of mutations and polymorphisms. J Steroid Biochem Mol Biol, 2017. 165(Pt A): p. 38-50. [PubMed: 27068427]
74.
Ariffin N.M., Islahudin F., Kumolosasi E., Makmor-Bakry M. Effects of MAO-A and CYP450 on primaquine metabolism in healthy volunteers. Parasitol Res. 2019;118(3):1011–1018. [PubMed: 30706164]
75.
Sortica V.A., Lindenau J.D., Cunha M.G. SLCO1A2, SLCO1B1 and SLCO2B1 polymorphisms influences chloroquine and primaquine treatment in Plasmodium vivax malaria. Pharmacogenomics. 2017;18(15):1393–1400. O.O. MD, et al. p. [PMC free article: PMC7099631] [PubMed: 28975866]
76.
Silvino A.C., Costa G.L., Araujo F.C., Ascher D.B., et al. Variation in Human Cytochrome P-450 Drug-Metabolism Genes: A Gateway to the Understanding of Plasmodium vivax Relapses. PLoS One. 2016;11(7):e0160172. p. [PMC free article: PMC4965052] [PubMed: 27467145]
77.
Stewart A.G.A., Zimmerman P.A., McCarthy J.S. Genetic Variation of G6PD and CYP2D6: Clinical Implications on the Use of Primaquine for Elimination of Plasmodium vivax. Front Pharmacol. 2021;12:784909. p. [PMC free article: PMC8661410] [PubMed: 34899347]
78.
Ingram R.J., Crenna-Darusallam C., Soebianto S., Noviyanti R., et al. The clinical and public health problem of relapse despite primaquine therapy: case review of repeated relapses of Plasmodium vivax acquired in Papua New Guinea. Malar J. 2014;13:488. [PMC free article: PMC4295472] [PubMed: 25495607]
79.
He X., Pan M., Zeng W., Zou C., et al. Multiple relapses of Plasmodium vivax malaria acquired from West Africa and association with poor metabolizer CYP2D6 variant: a case report. BMC Infect Dis. 2019;19(1):704. [PMC free article: PMC6688248] [PubMed: 31399061]
80.
Mat Salleh N.H., Rahman M.F.A., Samsusah S., De Silva J.R., et al. Case report: recurrence of Plasmodium vivax malaria due to defective cytochrome P450 2D6 function in Pos Lenjang, Pahang, Malaysia. Trans R Soc Trop Med Hyg. 2020;114(9):700–703. [PubMed: 32511702]
81.
Martin Ramirez A., Lombardia Gonzalez C., Soler Maniega T., Gutierrez Liarte A., et al. Several Plasmodium vivax relapses after correct primaquine treatment in a patient with impaired cytochrome P450 2D6 function. Malar J. 2020;19(1):259. [PMC free article: PMC7368755] [PubMed: 32680522]
82.
Choi S., Choi H., Park S.Y., Kwak Y.G., et al. Four Times of Relapse of Plasmodium vivax Malaria Despite Primaquine Treatment in a Patient with Impaired Cytochrome P450 2D6 Function. Korean J Parasitol. 2022;60(1):39–43. [PMC free article: PMC8898651] [PubMed: 35247953]
83.
de Pina-Costa A., Silvino A.C.R., Dos Santos E.M., Pedro R.S., et al. Increased primaquine total dose prevents Plasmodium vivax relapses in patients with impaired CYP2D6 activity: report of three cases. Malar J. 2021;20(1):341. [PMC free article: PMC8364036] [PubMed: 34391426]
84.
Bennett J.W., Pybus B.S., Yadava A., Tosh D., et al. Primaquine failure and cytochrome P-450 2D6 in Plasmodium vivax malaria. N Engl J Med. 2013;369(14):1381–2. [PubMed: 24088113]
85.
Baird J.K., Louisa M., Noviyanti R., Ekawati L., et al. Association of Impaired Cytochrome P450 2D6 Activity Genotype and Phenotype With Therapeutic Efficacy of Primaquine Treatment for Latent Plasmodium vivax Malaria. JAMA Netw Open. 2018;1(4):e181449. p. [PMC free article: PMC6324265] [PubMed: 30646129]
86.
Brasil L.W., Rodrigues-Soares F., Santoro A.B., Almeida A.C.G., et al. CYP2D6 activity and the risk of recurrence of Plasmodium vivax malaria in the Brazilian Amazon: a prospective cohort study. Malar J. 2018;17(1):57. [PMC free article: PMC5795836] [PubMed: 29390987]
87.
Silvino A.C.R., Kano F.S., Costa M.A., Fontes C.J.F., et al. Novel Insights into Plasmodium vivax Therapeutic Failure: CYP2D6 Activity and Time of Exposure to Malaria Modulate the Risk of Recurrence. Antimicrob Agents Chemother. 2020;64(5) [PMC free article: PMC7179649] [PubMed: 32122891]
88.
Huang H., Dong Y., Xu Y., Deng Y., et al. The association of CYP2D6 gene polymorphisms in the full-length coding region with higher recurrence rate of vivax malaria in Yunnan Province, China. Malar J. 2021;20(1):160. [PMC free article: PMC7981985] [PubMed: 33743705]
89.
Chamnanphon M., Gaedigk A., Puangpetch A., Pasomsub E., et al. Pharmacogene Variation in Thai Plasmodium vivax Relapse Patients Treated with a Combination of Primaquine and Chloroquine. Pharmgenomics Pers Med. 2020;13:1–12. [PMC free article: PMC6966953] [PubMed: 32021383]
90.
Chen N., Dowd S., Gatton M.L., Auliff A., et al. Cytochrome P450 2D6 profiles and their relationship with outcomes of primaquine anti-relapse therapy in Australian Defence Force personnel deployed to Papua New Guinea and East Timor. Malar J. 2019;18(1):140. [PMC free article: PMC6471761] [PubMed: 30999967]
91.
Pett H., Bradley J., Okebe J., Dicko A., et al. CYP2D6 Polymorphisms and the Safety and Gametocytocidal Activity of Single-Dose Primaquine for Plasmodium falciparum. Antimicrob Agents Chemother. 2019;63(10) [PMC free article: PMC6761544] [PubMed: 31383656]
92.
Bahk Y.Y., Ahn S.K., Jeon H.J., Na B.K., et al. An Evaluation of a New Quantitative Point-of Care Diagnostic to Measure Glucose-6-phosphate Dehydrogenase Activity. Korean J Parasitol. 2022;60(4):281–288. [PMC free article: PMC9441449] [PubMed: 36041490]
93.
Anderle A., Bancone G., Domingo G.J., Gerth-Guyette E., et al. Point-of-Care Testing for G6PD Deficiency: Opportunities for Screening. Int J Neonatal Screen. 2018;4(4):34. [PMC free article: PMC6832607] [PubMed: 31709308]
94.
Djigo O.K.M., Ould Khalef Y., Ould Ahmedou Salem M.S., Gomez N., et al. Assessment of CareStart G6PD rapid diagnostic test and CareStart G6PD biosensor in Mauritania. Infect Dis Poverty. 2021;10(1):105. [PMC free article: PMC8340529] [PubMed: 34353361]
95.
Grobusch M.P., Rodriguez-Morales A.J., Schlagenhauf P. The Primaquine Problem-and the Solution? Point-of-care Diagnostics for Glucose 6-Phosphate Dehydrogenase Deficiency. Clin Infect Dis. 2019;69(8):1443–1445. [PubMed: 30783651]
96.
Pratt V.M., Cavallari L.H., Del Tredici A.L., Gaedigk A., et al. Recommendations for Clinical CYP2D6 Genotyping Allele Selection: A Joint Consensus Recommendation of the Association for Molecular Pathology, College of American Pathologists, Dutch Pharmacogenetics Working Group of the Royal Dutch Pharmacists Association, and European Society for Pharmacogenomics and Personalized Therapy. J Mol Diagn. 2021 [PMC free article: PMC8579245] [PubMed: 34118403]
97.
Yoshida A., Beutler E., Motulsky A.G. Human glucose-6-phosphate dehydrogenase variants. Bull World Health Organ. 1971;45(2):243–53. [PMC free article: PMC2427914] [PubMed: 5316621]
98.
Kalman L.V., Agundez J., Appell M.L., Black J.L., et al. Pharmacogenetic allele nomenclature: International workgroup recommendations for test result reporting. Clin Pharmacol Ther. 2016;99(2):172–85. [PMC free article: PMC4724253] [PubMed: 26479518]

Footnotes

1

The FDA labels specific drug formulations. We have substituted the generic names for any drug labels in this excerpt. The FDA may not have labeled all formulations containing the generic drug. Certain terms, genes and genetic variants may be corrected in accordance with nomenclature standards, where necessary. We have given the full name of abbreviations, shown in square brackets, where necessary.

Copyright Notice

All Medical Genetics Summaries content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 International (CC BY 4.0) license which permits copying, distribution, and adaptation of the work, provided the original work is properly cited and any changes from the original work are properly indicated. Any altered, transformed, or adapted form of the work may only be distributed under the same or similar license to this one.

Bookshelf ID: NBK592855PMID: 37428853

Views

Tests in GTR by Condition

Tests in GTR by Gene

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...