Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 May 16.
Published in final edited form as: Vox Sang. 2020 Sep 30;116(2):141–154. doi: 10.1111/vox.12999

Pharmacogenomics with red cells: a model to study protein variants of drug transporter genes

Willy Albert Flegel 1, Kshitij Srivastava 1, Tristan Michael Sissung 2, Barry Ronald Goldspiel 3, William Douglas Figg 2
PMCID: PMC9108996  NIHMSID: NIHMS1802179  PMID: 32996603

Abstract

The PharmacoScan pharmacogenomics platform screens for variation in genes that affect drug absorption, distribution, metabolism, elimination, immune adverse reactions and targets. Among the 1,191 genes tested on the platform, 12 genes are expressed in the red cell membrane: ABCC1, ABCC4, ABCC5, ABCG2, CFTR, SLC16A1, SLC19A1, SLC29A1, ATP7A, CYP4F3, EPHX1 and FLOT1. These genes represent 5 ATP-binding cassette proteins, 3 solute carrier proteins, 1 ATP transport protein and 3 genes associated with drug metabolism and adverse drug reactions. Only ABCG2 and SLC29A1 encode blood group systems, JR and AUG, respectively. We propose red cells as an ex vivo model system to study the effect of heritable variants in genes encoding the transport proteins on the pharmacokinetics of drugs. Altered pharmacodynamics in red cells could also cause adverse reactions, such as haemolysis, hitherto unexplained by other mechanisms.

Keywords: pharmacogenomics, pharmacogenetics, drug transporters, erythrocytes, DMET, PharmacoScan

Background

Many proteins of the red cell membrane have been recognized as blood groups. The currently established 36 blood group systems are encoded by 41 genes [1]. They are involved in various cellular functions: transport of substrates (ABCG2, ABCG6, SLC29A1, AQP1, AQP3, SLC14A1, SLC4A1 and XK); cellular adhesion (ACKR1, BCAM, BSG, CD151, CD44, ERMAP, ICAM4, MIC2 and SEMA7A); enzymatic activity (ABO, ACHE, ART4, GBGT1, GCNT2, KEL, FUT1 and FUT3); red cell stability (GYPC, RHAG, RHCE, RHD, SLC4A1 and SMIM1); viral and bacterial attachment (A4GALT, B3GALNT1, FUT3, GYPA, GYPB and GYPE); complement interaction (C4A, C4B, CD55, CD59 and CR1); and unknown function (XG) [14]. Several of these membrane proteins serve as transporters that contribute to the absorption, tissue distribution and elimination of various drugs [1, 2]. Moreover, drug transporters often influence homeostatic expression of a variety of genes that regulate drug metabolism and disposition [5, 6]. The potential for these membrane proteins to influence pharmacology has been poorly studied.

Transporters are classified into 2 superfamilies: ATP-binding cassette (ABC) proteins and solute carrier (SLC) proteins. ABC transporters are involved in the translocation of a wide variety of substrates including amino acids, sugars, vitamins, inorganic ions, peptides, hormones, large polypeptides (>100 kD) and therapeutics [7, 8]. In eukaryotes, ABC proteins contribute only to the ATP-dependent efflux of substrates from cells against a concentration gradient [9, 10]. SLC proteins mediate the cellular uptake of drugs through facilitated diffusion or secondary active transport [11].

Similar to ABC and SLC transporters, the ion pumps (ATPases) [12] and ion channels [13] transport ions, such as Na+, K+, H+, Cl and Ca2+, across the cell membrane, utilizing energy from ATP hydrolysis or electrochemical gradients, respectively. Aquaporins are a special class of bidirectional channel proteins that are involved in the transfer of water across the membrane driven by the osmotic gradient [14].

Inter-individual variation in the human genome due to single-nucleotide variations (SNVs), small-scale insertions and deletions (InDels) and copy number variations (CNVs) may result in altered pharmacokinetic and pharmacodynamic characteristics of drugs leading to a lack of therapeutic efficacy or a risk for drug-induced toxicity [15, 16]. Variations in genes encoding drug transporters have been documented to affect responsiveness to chemotherapeutic agents [15, 17]. Rarely, sensitivity of red cells to the direct toxicity of the drugs can lead to drug-induced haemolytic anaemia [1821]. Some medications bind to the RBC cell surface or alter RBC surface antigens resulting in immune attack [22]. Drug-induced immune complexes can bind to RBCs [22], and alloantibody therapies that react with RBC antigens also cause haemolysis [23]. Lastly, oxidative injury to RBCs results from peroxide formation and subsequent haemolysis, particularly in populations who harbour deleterious variants in G6PD or haemoglobin H [24]. Drug metabolism can alter drug-induced haemolytic anaemia [25, 26]. And drugs bound to red cell proteins, including blood group proteins, can bind drug-dependent antibodies [22, 27]. Such antibodies can cause drug-induced immune haemolytic anaemia [28]. Therefore, the potential for inter-individual variation in drug binding and transport resulting from novel genetic variations should be explored and eventually considered to guide indications and dose recommendations.

The DMET Plus array, launched in 2012, scans 1936 variations (1931 SNVs and 5 CNVs) in 231 absorption, distribution, metabolism and elimination (ADME)-related genes [29, 30]. The PharmacoScan Solution array, an updated version of DMET Plus launched in 2016, scans 4627 variations in 1191 genes of known or suspected pharmacogenomic consequences. PharmacoScan incorporated all 231 genes from DMET Plus and nearly all of its variations but scans many additional variations and genes not present on DMET Plus.

The NIH Clinical Center has implemented a clinical decision support (CDS) for patients who are on medications where SNVs may assist with optimal dose or prediction of adverse events [31, 32]. In this pharmacogenomics approach, we have been screening HLA antigens by nucleotide sequencing to avoid exposure of patient with distinct HLA alleles to drugs associated with severe allergic reactions (e.g. allopurinol and carbamazepine) [31]. Nucleotide variations affecting proteins with transporter and metabolic functions have been determined by the DMET Plus microarray platform to adjust drug dose in patients with variants of high, intermediate or low activity [32].

We review the involvement of blood group proteins and other red cell membrane proteins and their potential applications to provide mechanistic insights in pharmacogenomics. As red cells are easily accessible, we propose an approach of using human red cells with variants of drug transport proteins, naturally occurring among blood donors and other healthy individuals. They can serve as an ex vivo model systems to study the kinetics of drug transport, as it may be affected by the protein variants.

Data search criterion and Methods

The exact number of genes expressed in the red cell membrane with drug transport function is unknown. We examined the blood group genes with drug transport function in red cells and represented on a commercial genotyping platform: the PharmacoScan array (Thermo-Fisher Scientific). The Clinical Pharmacogenetics Implementation Consortium (CPIC) is an international consortium that provides genotype-based drug guidelines to optimize drug therapy [33]. The CPIC drug–gene pairs table includes a total of 363 drug–gene interactions (DGIs), representing 214 unique drugs and 127 unique genes [33]. Among the 1,191 genes present on the 2 arrays, only 113 gene–drug pairs are covered by the CPIC guidelines. One CPIC gene FCGR3A is present on red cell membrane (with low confidence [34]) but not present on any of the arrays. The remaining 13 genes in CPIC (ABL2, ASL, HPRT1, NAGS, SERPINC1, CYP2A7P1, CYB5R1, CYB5R2, CYB5R4, MT-RNR1, PROS1, TMEM43 and YEATS4) are not present on red cell membrane and thus irrelevant for the current approach [34]. By searching the published literature and public databases [34], we retrieved the genes that are present on the PharmacoScan array and also expressed on red cell membranes.

Red cell membrane genes among the PharmacoScan and CPIC drug–gene pairs

We found 12 red cell membrane genes that met our search criterion (Table 1). Apart from the ABC (ABCC1, ABCC4, ABCC5, ABCG2 and CFTR), SLC (SLC16A1, SLC19A1 and SLC29A1) and ATP transporters (ATP7A), 3 additional genes associated with drug metabolism (CYP4F3 and EPHX1) and adverse drug reactions (ADRs; rs3909184 in FLOT1) were identified. Hegedus et al. [34] associated each gene with a confidence level to evaluate the potential validity of its protein’s presence in the red cell membrane: high level if the protein was present in at least two mass spectrometry studies or was an established blood group or CD marker; medium level if the protein was present in at least 1 mass spectrometry study; and low level if the protein was identified only semi-automatically from reviews [34]. We summarized the clinical interpretation of drug-gene pairs, based on the PharmGKB Clinical Annotations tables.

Table 1.

Genes present in the red cell membrane and routinely tested in pharmacogenomics.

Gene Red cell membrane confidence threshold* Pharmacogenomics platform
DMET PharmacoScan
ABCC1 High Yes Yes
ABCC4 High Yes Yes
ABCC5 High Yes Yes
ABCG2 High Yes Yes
SLC16A1 High Yes Yes
SLC19A1 Medium Yes Yes
SLC29A1 High Yes Yes
CYP4F3 Medium Yes Yes
CFTR High No Yes
FLOT1 High No Yes
ATP7A High Yes Yes
EPHX1 High Yes Yes
*

High = identified in at least 2 mass spectrometry-based studies, an established blood group, or a CD marker for red cells; Medium = identified in only 1 mass spectrometry-based study [34].

Only 2 of the 12 genes define blood group systems

Variations in the proteins of the red cell membrane are the hallmark and requirement for defining blood group systems. However, only 2 of the 12 genes from the present search are defined as blood group systems. The ABCG2 gene encodes the JR (ISBT 032) [35, 36], and the SLC29A1 gene encodes the AUG blood group system (ISBT 036; Table 2) [37].

Table 2.

Genomic characteristics of the 12 genes.

Gene Chromosome location Genomic size GenBank number Exons Length of cDNA (nucleotides) Length of coding sequence (CDS) (nucleotides)
ABCC1 16p13.11 200498 bp NG_028268.1 31 6504 4596
ABCC4 13q32.1 288618 bp NG_050651.1 31 5871 3978
ABCC5 3q27.1 98027 bp NG_047115.1 30 5790 4314
ABCG2 * 4q22.1 68596 bp NG_032067.2 16 4206 1968
SLC16A1 1p13.2 44507 bp NG_015880.2   5 3927 1503
SLC19A1 21q22.3 29905 bp NG_028278.2   6 4982 1776
SLC29A1 6p21.1 10648 bp NG_042893.1 14 2201 1497
CYP4F3 19p13.12 19864 bp NG_007964.1 13 5053 1563
CFTR 7q31.2 188703 bp NG_016465.4 27 6132 4443
FLOT1 6p21.33 15143 bp NC_000006.12 13 1866 1284
ATP7A Xq21.1 139740 bp NG_013224.2 23 8492 4503
EPHX1 1q42.12 35489 bp NG_009776.1   9 1847 1368
*

ABCG2 - Junior blood group system (JR; ISBT 032) [35, 36].

SLC29A1 - Augustine blood group system (AUG; ISBT 036) [37].

For a list of variants in the 12 genes and associated clinical outcomes, see Table S1).

JR blood group system

The high prevalence Jra antigen was first reported in 1970. JR was defined as a blood group system in 2012 [38]. The dbSNP database lists 341 non-synonymous or frame shift variants in the ABCG2 gene. Until today, however, all individuals who developed anti-Jra lack the whole JR protein from their red cell membranes. The antibody can cause haemolytic transfusion reactions and severe haemolytic disease of the foetus and newborn (HDFN) [35, 36, 39].

AUG blood group system

The high prevalence Ata antigen was first identified in 1967. AUG was defined as a blood group system in 2015 [37]. The dbSNP database lists 351 non-synonymous or frame shift variants in the SLC29A1 gene. Only 3 variants encoding 4 antigens in the AUG system are known. Individuals carrying these variants developed alloantibodies, which can cause haemolytic transfusion reactions and mild HDFN [40, 41].

Other blood group systems

In addition to ABCG2 and SLC29A1, the 4 blood group system genes ABO (ABO; ISBT 001), BCAM (LU; ISBT 005), ACKR1 (FY; ISBT 008) and CR1 (KN; ISBT 022) are also represented on the PharmacoScan array. Some resources consider them having impact in pharmacogenomics [42]. We do not review these 4 blood groups because CPIC did not identify a drug-gene pair for them.

Protein structural feature of the 12 genes

As expected for membrane transporters, 9 proteins are multi-pass transmembrane proteins (Table 3). Another 2 proteins, EPHX1 and LTB4H, are single-pass transmembrane proteins. Only 1 protein, FLOT1, is inserted in the inner leaflet of the plasma membrane of the red cell but does not traverse it. None of the 12 proteins identified were GPI-anchored [4345]. The 12 proteins are involved in the transport of a wide variety of drugs in humans (Table 4).

Table 3.

Protein characteristics of the 12 genes.

Gene Protein Length (amino acids) Topology*
Transmembrane segments
Amino-terminal Carboxy-terminal
ABCC1 Multidrug resistance-associated protein 1 (MRP1) 1531 Extracellular Cytoplasm 17
ABCC4 Multidrug resistance-associated protein 4 (MRP4) 1325 Cytoplasm Cytoplasm 12
ABCC5 Multidrug resistance-associated protein 5 (MRP5) 1437 Cytoplasm Cytoplasm 12
ABCG2 ATP-binding cassette sub-family G member 2 (ABCG2)   655 Cytoplasm Cytoplasm   6
SLC16A1 Monocarboxylate transporter 1 (MCT1)   500 Cytoplasm Cytoplasm 12
SLC19A1 Folate transporter 1 (RFC1)   591 Cytoplasm Cytoplasm 12
SLC29A1 Equilibrative nucleoside transporter 1 (ENT1)   498 Cytoplasm Extracellular 11
CYP4F3 Docosahexaenoic acid omega-hydroxylase (LTB4H)   520 Extracellular Cytoplasm   1
CFTR Cystic fibrosis transmembrane conductance regulator (CFTR) 1480 Cytoplasm Cytoplasm 12
FLOT1 Flotillin-1 (FLOT1)   427 Cytoplasm Cytoplasm   0
ATP7A Copper-transporting ATPase 1 (MNK) 1500 Cytoplasm Cytoplasm   8
EPHX1 Epoxide hydrolase 1 (EPHX1)   455 Cytoplasm Extracellular   1
*

Predicted or experimentally proven location of the amino- or carboxy-terminal protein ends at the cytoplasmic or extracellular side of the plasma membrane.

Table 4.

Common substrate drugs of the 12 genes.

Gene Substrate drugs
ABCC1 Doxorubicin, methotrexate
ABCC4 Antivirals: acyclovir, ritonavir, adefovir, tenofovir;
Diuretics: furosemide, hydrochlorothiazide;
Cephalosporins: ceftizoxime, cefazolin;
Cytotoxic drugs: methotrexate, 6-mercaptopurine, 6-thioguanine, topotecan; olmesartan, para-methoxy-N-ethylamphetamine. renal excretion of a wide variety of antiviral, cytostatic, antibiotic and cardiovascular drugs
ABCC5 Methotrexate, 6-thioguanine (anticancer drug), PMEA (anti-HIV drug), 5-fluorouracil, rosuvastatin, atorvastatin, kainic acid, domoic acid, ZJ43
ABCG2 Anthracyclines, daunorubicin, doxorubicin, topotecan, SN-38, irinotecan, methotrexate, imatinib, irinotecan, Mitoxantrone, nucleoside analogs, prazosin, pantoprazole, statins, topotecan, rosuvastatin, teriflunomide, chlorothiazide
SLC16A1 3-bromopyruvate (3-BrPA), a cancer drug candidate that inhibits glycolysis
SLC19A1 5-methyl-tetrahydrofolate, methotrexate
SLC29A1 Cladribine, cytarabine (for AML, ALL etc), fludarabine, gemcitabine capecitabine fialuridine, ribavirin
CYP4F3 Metabolize numerous drug substrates
CFTR Ivacaftor
FLOT1 Carbamazepine
ATP7A Cisplatin, oxaliplatin, carboplatin (overexpression causes sequestration of the drugs into intracellular vesicles)
EPHX1 Carbamazepine

Disease association of the 12 genes

Gene variants (alleles) of any of the 12 genes have been associated with various diseases. Variations can occur at the genetic level, involve changes of the mRNA and protein expression, and affect the localization of the proteins in cellular compartments. The number of such variants is growing, and their tabulation is basic for pharmacogenomics (Table S1).

ABCC1

ABCC1 is the first identified member of the ABCC subgroup and is ubiquitously expressed in almost all human tissues [46]. Increased MRP1 protein or mRNA concentrations or both were found in many haematologic and solid malignancies as predictor of poor chemotherapy response [47]. A number of variations in ABCC1 were associated with therapeutic response, cancer prognosis, drug toxicity and disease susceptibility [48, 49].

ABCC4

Increased MRP4 membrane localization and retention were associated with drug resistance in acute myeloid leukaemia [50]. Expression changes caused by an intronic CNV in ABCC4 correlated with an increased risk for oesophageal squamous cell carcinoma in the Chinese Han population [51]. A large number of SNVs in ABCC4 altered the affinity for the protein’s substrate drugs [49, 52, 53].

ABCC5

ABCC5 variants were associated with tumour response to gemcitabine-based chemoradiotherapy and survival in patients with pancreatic cancer [54]. Increased ABCC5 mRNA concentrations were reported in lung, colon, pancreatic and breast cancer [49].

ABCG2

Increased ABCG2 protein concentrations were associated with poor outcome in large B-cell lymphoma [55] and acute myeloid leukaemia [56]. Increased ABCG2 protein expression correlated with reduced survival of patients with small cell and non-small cell lung cancers [57]. A genome-wide association study (GWAS)-associated ABCG2 alleles with hyperuricaemia and gout [5860]. ABCG2 variations were associated with various malignancies including colorectal cancer, lymphoma and leukaemia [61]. The ABCG2 variant (rs2231142, Gln141Ly) causes reduction of transport activity [62] and increased drug concentrations leading to drug-induced toxicity [63]. Alloimmunizations occurred, complicated transfusions and caused HDFN disease (see JR blood group).

SLC16A1

MCT1 protein was overexpressed in cancer cells and involved in pH regulation [64]. The SLC16A1 variant (rs1049434, Asp490Glu) correlated with survival rates in patients with non-small cell lung [65] and colorectal cancers [66]. SLC16A1 promoter mutations were implicated in hereditary exercise-induced hyperinsulinism and hypoglycaemia [67] and ketoacidosis [68].

SLC19A1

SLC19A1 variants affected methotrexate toxicity and outcome in leukaemia [69]. A recent meta-analysis suggested a role of SLC19A1 rs1051266 variant in haematopoietic malignancies [70].

SLC29A1

Decreased ENT1 protein expression correlated with recurrence and poor outcome in patients with hepatocellular carcinoma after surgery [71]. Expression of SLC29A1 mRNA and ENT1 protein in tumour tissues was a predictive marker of outcome in cancer patients receiving gemcitabine [72]. SLC29A1 promoter region variants altered gene expression and gemcitabine chemosensitivity [73]. The SLC29A1 variant (rs45573936, Ile216Thr) may increase the risk for seizures during alcohol withdrawal [74]. Alloimmunizations occurred, complicated transfusions and caused HDFN disease (see AUG blood group).

CYP4F3

CYP4F3 variants were associated with the risk of ulcerative colitis [75] and lung cancer [76].

CFTR

Absence, reduced concentration, or malfunction of the CFTR protein resulted in cystic fibrosis [77, 78] and cystic fibrosis-associated diseases, including bronchiectasis [79], chronic pancreatitis [80] and congenital bilateral absence of the vas deferens [81].

FLOT1

The FLOT1 gene is located 620 kb upstream of the HLA-B gene on the short arm of chromosome 6. A FLOT1 variant (rs3909184) was identified as a tagging SNV for the HLA-B* 15:02 allele, associated with carbamazepine-induced Stevens–Jonson syndrome and toxic epidermal necrolysis in the Asian population [31, 82, 83]. A recent study identified FLOT1 variants affecting FLOT1 mRNA expression as susceptibility risk factor for major depressive disorder [84]. Upregulation of FLOT1 mRNA or FLOT1 protein expression may promote oesophageal squamous cell [85], colorectal [86], breast [87] and hepatocellular cancer [88].

ATP7A

ATP7A variants caused various copper transport disorders, such as Menkes disease [89], occipital horn syndrome [90] and the ATP7A-related distal motor neuropathy [91].

EPHX1

The low-activity genotype of the EPHX1 exon 3 variant (rs1051740, Tyr113His) was associated with a decreased risk for lung cancer in Caucasians [92]. Functional variants were also associated with susceptibility to various cancers, such as lung [93], upper aerodigestive tract [9496], colorectal [97], bladder [98] and breast cancer [99].

Advantages of red cells in pharmacologic studies

Previous studies, using site-directed mutagenesis, have been applied in cell cultures, such as human embryonic kidney-293 [100] and Madin–Darby canine kidney cells [101] or oocytes from Xenopus laevis [102]. However, these methods and cellular assays can be artificial, expensive, laborious and time-consuming. Proteomic analysis of the red cells, the most abundant cells in human body [103], has identified multiple transporter proteins in their membrane. Several of these proteins are known to be involved in the influx or efflux of clinically important drugs [34].

The membrane structure of the red cell is arguably the best studied of all human cell types [104], which enables us to draw worthwhile conclusions [105]. Red cells can be haemolysed and later resealed to regain limited permeability [106]. This technical feature is rather unique for red cells. No wonder that several studies utilized resealed human erythrocyte membranes, dubbed ghosts, as model system for drug transport studies [107, 108]. Use of ghosts circumvented the interference from proteins and enzymes present in the erythrocyte cytoplasm [109].

Study topics for pharmacogenomics with red cells

Clinical syndromes: haemolysis

The SLC28A3, a drug transporter gene not expressed on the red cell membrane, is tested on both the DMET and PharmacoScan arrays. A SLC28A3 variant (rs10838138) was associated with a lower incidence of severe haemolytic anaemia in patients with chronic hepatitis C receiving pegylated interferon and ribavirin [110]. Haemolytic events may however remain undetected until the haemolysis becomes rather severe. Haemolysis by drugs can be caused by 2 mechanisms: (1) non-immune mediated, and (2) immune mediated.

Haemolysis, non-immune mediated

Non-immune-mediated drug-induced haemolytic anaemia is due to direct toxicity through irreversible damage of red cells [1821, 25, 26, 28]. Various other factors such as red cell enzymopathy, infections, uraemia, diabetic ketoacidosis, deficient of vitamin E and low levels of glucose can increase the haemolytic effect of a drug [28]. Drugs, such as phenylhydrazine [111] cause haemolysis in all subjects in relatively low concentrations; while primaquine, acetanilid, nitrofurantoin, p-aminosalicylic acid, naphthalene, phenylsemicarbazide, sulphonamides and sulphones cause haemolysis in normal subjects only in high concentrations [28, 112, 113]. Genetic variants in drug transport or drug metabolism genes may determine the intracellular concentration of the drug and its impact on haemolysis.

Haemolysis, immune mediated

Although underdiagnosed, an incidence of approximately 1 per million per year [114, 115] has been proposed for drug-induced immune haemolytic anaemia, a rare but severe hypersensitivity reaction to drug administration [116, 117]. It is caused by warm autoantibodies against red cells induced by many antibiotic, anti-inflammatory and chemotherapy drugs [118, 119]. A large and growing list of drugs have been associated with drug-induced immune haemolytic anaemia, and the most common are piperacillin, cefotetan and ceftriaxone [118]. Platinum-based chemotherapeutic agents such as oxaliplatin, cisplatin and carboplatin are also known to induce drug-induced immune haemolytic anaemia in rare cases [25, 118, 120, 121]. While drug-induced immune haemolytic anaemia is often diagnosed by excluding alternative causes rather than by direct evidence, genetic variants of red cell membrane proteins, other than blood group proteins, are not routinely considered.

We wonder how many clinical haemolytic events are not properly attributed to be caused by variants of membrane proteins? Each protein variant is rare, but a large fraction of patients may carry one of the host of such variants.

Reservoir or sink for a drug

Red cells may function as a reservoir or sink. Their effectiveness can vary if protein variants are involved. Drug transporter proteins can bind drugs to the red cell surface or transport the drug into the red cell cytoplasm. Either way, the drug’s plasma concentration may be reduced, delaying or preventing efficient delivery of therapeutics to target tissues. The role of red cell membrane proteins has been studied extensively in drug transport or drug binding [122]. The effect of these proteins’ variants has not been systematically evaluated so far.

Drug delivery

Resealed red cells have been manufactured for in vivo drug delivery [123]. They have a long life span, excellent biocompatibility, complete biodegradability and low immunogenicity [124]. Protein variants may be a lesser concern when allogeneic red cells are manufactured. In an autologous setting, the variant of a red cell membrane protein in the patient would matter.

Drugs can be targeted to red cells in two ways, such as encapsulation and conjugation. The drugs are encapsulated inside the ghosts, which reduces the possibility of an immune reaction and protects the drug from inactivation [125]. Molecular variants of transport proteins may alter the entrapment and eventual release of the drug. By chemical or genetic means, drugs can be physically conjugated to lectins and other ligands that bind to distinct red cell membrane proteins [126]. For example, single-chain variable region fragment (scFv) of TER-119, a monoclonal antibody to the mouse analogue of human glycophorin A (GPA), was genetically attached to complement-regulating proteins including decay-accelerating factor (DAF) which protected the mouse red cells against lysis by complement [127]. Of course, molecular variants of red cell surface proteins can alter the binding affinity of the drug-ligand conjugates and affect the bioavailability of the drug.

Limitations

Red cells recapitulate the in vivo condition where the expression of a transporter protein and presence of multiple transporters for same drug are accounted for. Studying the kinetics of drug transport using red cells harbouring naturally occurring variants of drug transport proteins may allow direct insight in pharmacokinetics for red cells. Such results may be carefully extrapolated to other cell types that express any of the 12 genes in their cell membranes. However, using ghosts as model systems has its limitations: the protein isoforms and the amount of protein expressed may differ between red cells and other tissues; also, the membrane lipid composition, cytoskeleton proteins and interacting proteins differ among cell types.

Transplant and iatrogenic chimeras

Peripheral blood, routinely used for pharmacogenetic analysis, would reflect the genotype of the donor after a hematopoietic stem cell transplantation. Chronic transfused patients and patients with solid organ transplants are known to accept donor granulocytes and lymphocytes even with leucoreduced donor blood [128, 129]. Being an emerging field, there is a dearth of information on the relevance of donor or recipient genotype to pharmacologic outcome, and both the donor and recipient genetic backgrounds and their discrepancies should be taken into account.

Therapeutics with potentially important RBC pharmacogenomics relationships

Methotrexate

Methotrexate polyglutamates accumulate within erythrocytes in a dose-dependent fashion, significantly influencing long-term methotrexate plasma concentrations [130]. One study evaluated the relationship between ABCC1 variants and methotrexate concentrations in erythrocytes, finding that rs35592 was associated with lower methotrexate polyglutamate concentrations and rs3784862 was associated with higher concentrations [131]. Other studies have identified genetic variants in the folate transporter (SLC19A1, FOLT and RFC1) that are associated with erythrocyte folate concentrations [132,133]. Although controversial [134], RBC methotrexate polyglutamate concentrations are associated with genetic variants in SLC19A1 [135]. SLC19A1 loss results in reduced methotrexate uptake and methotrexate resistance in erythroleukaemia cells [136]. Variants in RBC transporters have also been associated with methotrexate plasma concentrations [137], and RBC folate concentrations have been associated with methotrexate outcomes [138]. Although methotrexate likely targets white cells, methotrexate polyglutamates in circulating RBCs may be associated with clinical efficacy of methotrexate, determining both dose and therapeutic selection [139]. Such relationships may underlie the association between variants in ABCC1, SLC19A1 and other polymorphisms with methotrexate efficacy. Thus, understanding how allelic variants in RBC transporters influence this relationship may increase the likelihood of developing precision use of methotrexate. This field remains in its infancy.

Mercaptopurines

Located in a variety of tissues, including erythrocytes, thiopurine methyl transferase (TPMT) is the major metabolic detoxification route for mercaptopurines. Red blood cells may act as a reservoir for mercaptopurine metabolites, and low erythrocyte TPMT activity is a marker for mercaptopurine toxicity [140] and lower risk of relapse [141]. Both MRP4 and MRP5 transport mercaptopurine out of red blood cells, whereas ENT1 is a mercaptopurine uptake transporter associated with mercaptopurine sensitivity [142144]. The rs3765534 polymorphism in ABCC4 impairs membrane localization and is associated with significant mercaptopurine sensitivity [145, 146]. One study determined that variants in SLC29A1 were associated with erythrocyte concentrations of thiopurines in patients receiving azathioprine for neuromyelitis optica spectrum disorders [147]; however, the genetic influences of erythrocyte transport and its implications on the pharmacology of mercaptopurines are rather poorly studied.

Antiretrovirals

Low erythrocyte inosine triphosphatase (ITPA) activity is associated with the development of adverse events during antiretroviral therapy [148, 149] and metabolizes purine analogues used in HIV treatment [149]. Since ITPA activity is decreased in individuals infected with HIV [150], factors influencing ITPA metabolism in erythrocytes may be of significant importance. Several studies have identified variants in transporters that are associated with the pharmacokinetics or clinical outcome of antiretrovirals [151158]. However, to our knowledge, no study has yet determined whether these variants are associated with intra-erythrocyte concentration of these medications, and therefore, the availability of antiretroviral substrates to erythrocyte ITPA.

Nucleoside analogues

SLC29A1 (encoding ENT1) is involved in the pharmacology of many nucleoside analogues (e.g. cytarabine, gemcitabine, 5FU, pentostatin, zidovudine, ribavirin, dipyridamole and draflazine) [159]. Interestingly, we did not find a single study that has evaluated whether RBC ENT1 uptake effects the pharmacology of these medications. Since ribavirin is known to cause dose-limiting haemolytic anaemia [160], variants in this transporter should be studied to determine whether a population of individuals is at particular risk of haemolytic anaemia during ribavirin therapy.

ABCG2 substrates

ABCG2 transports a very wide variety of medications from different classes, and genetic variants in ABCG2 have been associated with the pharmacokinetics and outcomes of numerous therapeutics (Table S1). The implications of erythrocyte ABCG2 expression remain poorly characterized. Yet, changes in the expression of ABCG2 resulting from genetic variation are reflected in the red cell membrane [161]. One study discovered a novel ABCG2 variant (ABCG2-M71V; rs148475733) after noting that certain patients had very low (50% of average) ABCG2 erythrocyte membrane expression levels [162]. Thus, RBC transporter expression can be used to identify potentially important variants affecting the expression or function of transporters. Further study is warranted on ABCG2 expression in red cell membranes and the implications of such expression in pharmacology.

CFTR potentiators

Erythrocytes are representative of the CFTR status of patients [163]. Membrane preparations from erythrocytes are already used to study CFTR structure, function and density [164166]. Numerous genetic variants are associated with CFTR potentiators (Table S1). Thus, erythrocyte membrane preparations may be useful for non-invasive diagnostic purposes, developing novel CFTR potentiators, or understanding unusual clinical outcomes [167, 168]. Such approaches do not appear to be prevalent in the literature.

Summary

Red cells are easily accessible for pharmacologic studies. The DMET and more recently the PharmacoScan arrays are increasingly used worldwide for clinical pharmacogenetic decision-making. A thorough search of literature identified 12 genes that are scanned by the arrays and also expressed in the red cell membrane. We propose red cells as an ex vivo model system to study the effect of variants of these 12 membrane proteins on the pharmacokinetics of drugs.

Supplementary Material

vox12999-sup-0001-tables1

Table S1: Pharmacogenomic variants and associated clinical outcomes.

Acknowledgements

This work was supported in part by the Intramural Research Program (project ID Z99 CL999999) of the NIH Clinical Center and (grant ID ZIA BC 010627) of the National Cancer Institute at the National Institutes of Health.

Footnotes

Conflict of interest

The authors declared having no competing financial interest relevant to this article.

Publisher's Disclaimer: Statement of disclaimer

The views expressed do not necessarily represent the view of the National Institutes of Health, the Department of Health and Human Services, or the U.S. Federal Government.

Supporting Information

Additional Supporting Information may be found in the online version of this article:

References

  • 1.Storry JR, Clausen FB, Castilho L, et al. : International society of blood transfusion working party on red cell immunogenetics and blood group terminology: report of the Dubai, Copenhagen and Toronto meetings. Vox Sang 2019; 114:95–102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Pourazar A: Red cell antigens: Structure and function. Asian J Transfus Sci 2007; 1:24–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Cartron JP, Bailly P, Le Van KC, et al. : Insights into the structure and function of membrane polypeptides carrying blood group antigens. Vox Sang 1998; 74(Suppl 2):29–64 [DOI] [PubMed] [Google Scholar]
  • 4.Cooling L: Blood groups in infection and host susceptibility. Clin Microbiol Rev 2015; 28:801–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Giacomini KM, Huang SM, Tweedie DJ, et al. : Membrane transporters in drug development. Nat Rev Drug Discov 2010; 9:215–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Fisel P, Nies AT, Schaeffeler E, et al. : The importance of drug transporter characterization to precision medicine. Expert Opin Drug Metab Toxicol 2017; 13:361–5 [DOI] [PubMed] [Google Scholar]
  • 7.Higgins CF: ABC transporters: from microorganisms to man. Annu Rev Cell Biol 1992; 8:67–113 [DOI] [PubMed] [Google Scholar]
  • 8.Blight MA, Holland IB: Structure and function of haemolysin B, P-glycoprotein and other members of a novel family of membrane translocators. Mol Microbiol 1990; 4:873–80 [DOI] [PubMed] [Google Scholar]
  • 9.Vasiliou V, Vasiliou K, Nebert DW: Human ATP-binding cassette (ABC) transporter family. Hum Genomics 2009; 3:281–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Licht A, Schneider E: ATP binding cassette systems: structures, mechanisms, and functions. Central Eur J Biol 2011; 6:785 [Google Scholar]
  • 11.He L, Vasiliou K, Nebert DW: Analysis and update of the human solute carrier (SLC) gene superfamily. Hum Genomics 2009; 3:195–206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Muller V, Gruber G: ATP synthases: structure, function and evolution of unique energy converters. Cell Mol Life Sci 2003; 60:474–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Camerino DC, Desaphy JF, Tricarico D, et al. : Therapeutic approaches to ion channel diseases. Adv Genet 2008; 64:81–145 [DOI] [PubMed] [Google Scholar]
  • 14.Benga G: Water channel proteins (later called aquaporins) and relatives: past, present, and future. IUBMB Life 2009; 61:112–33 [DOI] [PubMed] [Google Scholar]
  • 15.McLean C, Wilson A, Kim RB: Impact of transporter polymorphisms on drug development: is it clinically significant? J Clin Pharmacol 2016; 56 (Suppl 7):S40–58 [DOI] [PubMed] [Google Scholar]
  • 16.Yee SW, Chen L, Giacomini KM: Pharmacogenomics of membrane transporters: past, present and future. Pharmacogenomics 2010; 11: 475–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.He Y, Hoskins JM, McLeod HL: Copy number variants in pharmacogenetic genes. Trends Mol Med 2011; 17:244–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hitomi Y, Cirulli ET, Fellay J, et al. : Inosine triphosphate protects against ribavirin-induced adenosine triphosphate loss by adenylosuccinate synthase function. Gastroenterology 2011; 140:1314–21 [DOI] [PubMed] [Google Scholar]
  • 19.Tanaka Y, Tamura Y, Yokomori H, et al. : Rapidity and severity of hemoglobin decreasing associated with erythrocyte inosine triphosphatase activity and ATP concentration during chronic hepatitis C treatment. Biol Pharm Bull 2016; 39:615–9 [DOI] [PubMed] [Google Scholar]
  • 20.Kleinegris MC, Koek GH, Mast K, et al. : Ribavirin-induced externalization of phosphatidylserine in erythrocytes is predominantly caused by inhibition of aminophospholipid translocase activity. Eur J Pharmacol 2012; 693:1–6 [DOI] [PubMed] [Google Scholar]
  • 21.Salehi M, Masoumi-Asl H, Assarian M, et al. : Delayed hemolytic anemia after treatment with artesunate: case report and literature review. Curr Drug Saf 2019; 14:60–6 [DOI] [PubMed] [Google Scholar]
  • 22.Arndt PA: Drug-induced immune hemolytic anemia: the last 30 years of changes. Immunohematology 2014; 30:44–54 [PubMed] [Google Scholar]
  • 23.Tormey CA, Hendrickson JE: Transfusion-related red blood cell alloantibodies: induction and consequences. Blood 2019; 133:1821–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Harcke SJ, Rizzolo D, Harcke HT: G6PD deficiency: An update. JAAPA 2019; 32:21–6 [DOI] [PubMed] [Google Scholar]
  • 25.Johnson ST, Fueger JT, Gottschall JL: One center’s experience: the serology and drugs associated with drug-induced immune hemolytic anemia–a new paradigm. Transfusion 2007; 47:697–702 [DOI] [PubMed] [Google Scholar]
  • 26.Dhaliwal G, Cornett PA, Tierney LM Jr: Hemolytic anemia. Am Fam Physician 2004; 69:2599–606 [PubMed] [Google Scholar]
  • 27.Garratty G: Drug-induced immune hemolytic anemia. Hematol Am Soc Hematol Educ Program 2009; 2009:73–9 [DOI] [PubMed] [Google Scholar]
  • 28.Dausset J, Contu L: Drug-induced hemolysis. Annu Rev Med 1967; 18:55–70 [DOI] [PubMed] [Google Scholar]
  • 29.Deeken J: The Affymetrix DMET platform and pharmacogenetics in drug development. Curr Opin Mol Ther 2009; 11:260–8 [PubMed] [Google Scholar]
  • 30.Arbitrio M, Di Martino MT, Scionti F, et al.: DMET (Drug Metabolism Enzymes and Transporters): a pharmacogenomic platform for precision medicine. Oncotarget 2016; 7:54028–50 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Goldspiel BR, Flegel WA, DiPatrizio G, et al.: Integrating pharmacogenetic information and clinical decision support into the electronic health record. J Am Med Inform Assoc 2014; 21:522–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Sissung TM, McKeeby JW, Patel J, et al. : Pharmacogenomics Implementation at the National Institutes of Health Clinical Center. J Clin Pharmacol 2017; 57(Suppl 10):S67–s77 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.https://cpicpgx.org/genes-drugs/. Updated February 3, 2020.
  • 34.Hegedus T, Chaubey PM, Varady G, et al. : Inconsistencies in the red blood cell membrane proteome analysis: generation of a database for research and diagnostic applications. Database (Oxford) 2015; 2015: bav056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zelinski T, Coghlan G, Liu XQ, et al. : ABCG2 null alleles define the Jr(a-) blood group phenotype. Nat Genet 2012; 44:131–2 [DOI] [PubMed] [Google Scholar]
  • 36.Saison C, Helias V, Ballif BA, et al. : Null alleles of ABCG2 encoding the breast cancer resistance protein define the new blood group system Junior. Nat Genet 2012; 44:174–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Daniels G, Ballif BA, Helias V, et al. : Lack of the nucleoside transporter ENT1 results in the Augustine-null blood type and ectopic mineralization. Blood 2015; 125:3651–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Castilho L, Reid ME: A review of the JR blood group system. Immunohematology 2013; 29:63–8 [PubMed] [Google Scholar]
  • 39.Castilho L: An update on the JR blood group system. Immunohematology 2019; 35:43–4 [PubMed] [Google Scholar]
  • 40.Daniels G: The Augustine blood group system, 48 years in the making. Immunohematology 2016; 32:100–3 [PubMed] [Google Scholar]
  • 41.Daniels G: An update on the Augustine blood group system. Immunohematology 2019; 35:1–2 [PubMed] [Google Scholar]
  • 42.Barbarino JM, Whirl-Carrillo M, Altman RB, et al. : PharmGKB: A worldwide resource for pharmacogenomics information. Wiley Interdiscip Rev Syst Biol Med 2018; 10:e1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Rojewski MT, Schrezenmeier H, Flegel WA: Tissue distribution of blood group membrane proteins beyond red cells: evidence from cDNA libraries. Transfus Apher Sci 2006; 35:71–82 [DOI] [PubMed] [Google Scholar]
  • 44.Weinstock C, Anliker M, von Zabern I: CD59: A long-known complement inhibitor has advanced to a blood group system. Immunohematology 2015; 31:145–51 [PubMed] [Google Scholar]
  • 45.Weinstock C, Anliker M, von Zabern I: An update on the CD59 blood group system. Immunohematology 2019; 35:7–8 [PubMed] [Google Scholar]
  • 46.Cole SP, Bhardwaj G, Gerlach JH, et al. : Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line. Science 1992; 258:1650–4 [DOI] [PubMed] [Google Scholar]
  • 47.Cole SP: Targeting multidrug resistance protein 1 (MRP1, ABCC1): past, present, and future. Annu Rev Pharmacol Toxicol 2014; 54:95–117 [DOI] [PubMed] [Google Scholar]
  • 48.Yin J, Zhang J: Multidrug resistance-associated protein 1 (MRP1/ABCC1) polymorphism: from discovery to clinical application. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2011; 36:927–38 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Chen ZS, Tiwari AK: Multidrug resistance proteins (MRPs/ABCCs) in cancer chemotherapy and genetic diseases. FEBS J 2011; 278:3226–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Pitre A, Ge Y, Lin W, et al. : An unexpected protein interaction promotes drug resistance in leukemia. Nat Commun 2017; 8:1547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Sun Y, Shi N, Lu H, et al. : ABCC4 copy number variation is associated with susceptibility to esophageal squamous cell carcinoma. Carcinogenesis 2014; 35:1941–50 [DOI] [PubMed] [Google Scholar]
  • 52.Tsukamoto M, Sato S, Satake K, et al. : Quantitative evaluation of drug resistance profile of cells expressing wild-type or genetic polymorphic variants of the human ABC transporter ABCC4. Int J Mol Sci 2017; 18:1435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Tsukamoto M, Yamashita M, Nishi T, et al. : A human ABC transporter ABCC4 gene SNP (rs11568658, 559 G > T, G187W) reduces ABCC4-dependent drug resistance. Cells 2019; 8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tanaka M, Okazaki T, Suzuki H, et al. : Association of multi-drug resistance gene polymorphisms with pancreatic cancer outcome. Cancer 2011; 117:744–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kim JE, Singh RR, Cho-Vega JH, et al. : Sonic hedgehog signaling proteins and ATP-binding cassette G2 are aberrantly expressed in diffuse large B-cell lymphoma. Mod Pathol 2009; 22:1312–20 [DOI] [PubMed] [Google Scholar]
  • 56.van den Heuvel-Eibrink MM, Wiemer EA, Prins A, et al. : Increased expression of the breast cancer resistance protein (BCRP) in relapsed or refractory acute myeloid leukemia (AML). Leukemia 2002; 16:833–9 [DOI] [PubMed] [Google Scholar]
  • 57.Horsey AJ, Cox MH, Sarwat S, et al. : The multidrug transporter ABCG2: still more questions than answers. Biochem Soc Trans 2016; 44:824–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Dehghan A, Kottgen A, Yang Q, et al. : Association of three genetic loci with uric acid concentration and risk of gout: a genome-wide association study. Lancet 2008; 372:1953–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Matsuo H, Takada T, Ichida K, et al. : Common defects of ABCG2, a high-capacity urate exporter, cause gout: a function-based genetic analysis in a Japanese population. Sci Transl Med 2009; 1:5ra11. [DOI] [PubMed] [Google Scholar]
  • 60.Woodward OM, Kottgen A, Kottgen M: ABCG transporters and disease. FEBS J 2011; 278:3215–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Chen P, Zhao L, Zou P, et al. : The contribution of the ABCG2 C421A polymorphism to cancer susceptibility: a meta-analysis of the current literature. BMC Cancer 2012; 12:383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Imai Y, Nakane M, Kage K, et al. : C421A polymorphism in the human breast cancer resistance protein gene is associated with low expression of Q141K protein and low-level drug resistance. Mol Cancer Ther 2002; 1:611–6 [PubMed] [Google Scholar]
  • 63.Mizuno T, Fukudo M, Terada T, et al. : Impact of genetic variation in breast cancer resistance protein (BCRP/ABCG2) on sunitinib pharmacokinetics. Drug Metab Pharmacokinet 2012; 27:631–9 [DOI] [PubMed] [Google Scholar]
  • 64.Pinheiro C, Longatto-Filho A, Azevedo-Silva J, et al. : Role of monocarboxylate transporters in human cancers: state of the art. J Bioenerg Biomembr 2012; 44:127–39 [DOI] [PubMed] [Google Scholar]
  • 65.Guo X, Chen C, Liu B, et al. : Genetic variations in monocarboxylate transporter genes as predictors of clinical outcomes in non-small cell lung cancer. Tumour Biol 2015; 36:3931–9 [DOI] [PubMed] [Google Scholar]
  • 66.Fei F, Guo X, Chen Y, et al. : Polymorphisms of monocarboxylate transporter genes are associated with clinical outcomes in patients with colorectal cancer. J Cancer Res Clin Oncol 2015; 141:1095–102 [DOI] [PubMed] [Google Scholar]
  • 67.Otonkoski T, Jiao H, Kaminen-Ahola N, et al. : Physical exercise-induced hypoglycemia caused by failed silencing of monocarboxylate transporter 1 in pancreatic beta cells. Am J Hum Genet 2007; 81:467–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Balasubramaniam S, Lewis B, Greed L, et al. : Heterozygous monocarboxylate transporter 1 (MCT1, SLC16A1) deficiency as a cause of recurrent ketoacidosis. JIMD Rep 2016; 29:33–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gregers J, Christensen IJ, Dalhoff K, et al. : The association of reduced folate carrier 80G>A polymorphism to outcome in childhood acute lymphoblastic leukemia interacts with chromosome 21 copy number. Blood 2010;115:4671–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Huang X, Gao Y, He J, et al. : The association between RFC1 G80A polymorphism and cancer susceptibility: Evidence from 33 studies. J Cancer 2016; 7:144–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Gao P-T, Cheng J-W, Gong Z-J, et al. : Low SLC29A1 expression is associated with poor prognosis in patients with hepatocellular carcinoma. Am J Cancer Res 2017; 7:2465–77 [PMC free article] [PubMed] [Google Scholar]
  • 72.Spratlin J, Sangha R, Glubrecht D, et al. : The absence of human equilibrative nucleoside transporter 1 is associated with reduced survival in patients with gemcitabine-treated pancreas adenocarcinoma. Clin Cancer Res 2004; 10:6956–61 [DOI] [PubMed] [Google Scholar]
  • 73.Myers SN, Goyal RK, Roy JD, et al. : Functional single nucleotide polymorphism haplotypes in the human equilibrative nucleoside transporter 1. Pharmacogenet Genom 2006; 16:315–20 [DOI] [PubMed] [Google Scholar]
  • 74.Kim JH, Karpyak VM, Biernacka JM, et al. : Functional role of the polymorphic 647 T/C variant of ENT1 (SLC29A1) and its association with alcohol withdrawal seizures. PLoS One 2011; 6:e16331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ananthakrishnan AN, Khalili H, Song M, et al. : Genetic polymorphisms in fatty acid metabolism modify the association between dietary n3: n6 intake and risk of ulcerative colitis: a prospective cohort study. Inflamm Bowel Dis 2017; 23:1898–904 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Yin J, Liu H, Liu Z, et al. : Pathway-analysis of published genome-wide association studies of lung cancer: A potential role for the CYP4F3 locus. Mol Carcinog 2017; 56:1663–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Watson MS, Cutting GR, Desnick RJ, et al. : Cystic fibrosis population carrier screening: 2004 revision of American College of Medical Genetics mutation panel. Genet Med 2004; 6:387–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Rommens JM, Iannuzzi MC, Kerem B, et al. : Identification of the cystic fibrosis gene: chromosome walking and jumping. Science 1989; 245:1059–65 [DOI] [PubMed] [Google Scholar]
  • 79.Pignatti PF, Bombieri C, Marigo C, et al. : Increased incidence of cystic fibrosis gene mutations in adults with disseminated bronchiectasis. Hum Mol Genet 1995; 4:635–9 [DOI] [PubMed] [Google Scholar]
  • 80.Sharer N, Schwarz M, Malone G, et al. : Mutations of the cystic fibrosis gene in patients with chronic pancreatitis. N Engl J Med 1998; 339:645–52 [DOI] [PubMed] [Google Scholar]
  • 81.Chillon M, Casals T, Mercier B, et al. : Mutations in the cystic fibrosis gene in patients with congenital absence of the vas deferens. N Engl J Med 1995; 332:1475–80 [DOI] [PubMed] [Google Scholar]
  • 82.He Y, Hoskins JM, Clark S, et al. : Accuracy of SNPs to predict risk of HLA alleles associated with drug-induced hypersensitivity events across racial groups. Pharmacogenomics 2015; 16:817–24 [DOI] [PubMed] [Google Scholar]
  • 83.de Bakker PI, McVean G, Sabeti PC, et al. : A high-resolution HLA and SNP haplotype map for disease association studies in the extended human MHC. Nat Genet 2006; 38:1166–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Zhong J, Li S, Zeng W, et al. : Integration of GWAS and brain eQTL identifies FLOT1 as a risk gene for major depressive disorder. Neuropsychopharmacology 2019; 44:1542–1551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Song L, Gong H, Lin C, et al. : Flotillin-1 promotes tumor necrosis factor-alpha receptor signaling and activation of NF-kappaB in esophageal squamous cell carcinoma cells. Gastroenterology 2012; 143(995–1005): e12. [DOI] [PubMed] [Google Scholar]
  • 86.Thorn CC, Freeman TC, Scott N, et al. : Laser microdissection expression profiling of marginal edges of colorectal tumours reveals evidence of increased lactate metabolism in the aggressive phenotype. Gut 2009; 58:404–12 [DOI] [PubMed] [Google Scholar]
  • 87.Lin C, Wu Z, Lin X, et al. : Knockdown of FLOT1 impairs cell proliferation and tumorigenicity in breast cancer through upregulation of FOXO3a. Clin Cancer Res 2011; 17:3089–99 [DOI] [PubMed] [Google Scholar]
  • 88.Zhang SH, Wang CJ, Shi L, et al. : High expression of FLOT1 Is associated with progression and poor prognosis in hepatocellular carcinoma. PLoS One 2013; 8:e64709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Skjorringe T, Amstrup Pedersen P, Salling Thorborg S, et al. : Characterization of ATP7A missense mutants suggests a correlation between intracellular trafficking and severity of Menkes disease. Sci Rep 2017; 7:757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Das S, Levinson B, Vulpe C, et al. : Similar splicing mutations of the Menkes/mottled copper-transporting ATPase gene in occipital horn syndrome and the blotchy mouse. Am J Hum Genet 1995; 56:570–6 [PMC free article] [PubMed] [Google Scholar]
  • 91.Kennerson ML, Nicholson GA, Kaler SG, et al. : Missense mutations in the copper transporter gene ATP7A cause X-linked distal hereditary motor neuropathy. Am J Hum Genet 2010; 86:343–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kiyohara C, Yoshimasu K, Takayama K, et al. : EPHX1 polymorphisms and the risk of lung cancer: a HuGE review. Epidemiology 2006; 17:89–99 [DOI] [PubMed] [Google Scholar]
  • 93.Gsur A, Zidek T, Schnattinger K, et al. : Association of microsomal epoxide hydrolase polymorphisms and lung cancer risk. Br J Cancer 2003; 89:702–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Jourenkova-Mironova N, Mitrunen K, Bouchardy C, et al. : High-activity microsomal epoxide hydrolase genotypes and the risk of oral, pharynx, and larynx cancers. Cancer Res 2000; 60:534–6 [PubMed] [Google Scholar]
  • 95.Park JY, Schantz SP, Lazarus P: Epoxide hydrolase genotype and orolaryngeal cancer risk: interaction with GSTM1 genotype. Oral Oncol 2003; 39:483–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Muir C, Weiland L: Upper aerodigestive tract cancers. Cancer 1995; 75:147–53 [DOI] [PubMed] [Google Scholar]
  • 97.Sachse C, Smith G, Wilkie MJ, et al. : A pharmacogenetic study to investigate the role of dietary carcinogens in the etiology of colorectal cancer. Carcinogenesis 2002; 23:1839–49 [DOI] [PubMed] [Google Scholar]
  • 98.Srivastava DS, Mandhani A, Mittal RD: Genetic polymorphisms of cytochrome P450 CYP1A1 (*2A) and microsomal epoxide hydrolase gene, interactions with tobacco-users, and susceptibility to bladder cancer: a study from North India. Arch Toxicol 2008; 82:633–9 [DOI] [PubMed] [Google Scholar]
  • 99.Spurdle AB, Chang JH, Byrnes GB, et al. : A systematic approach to analysing gene-gene interactions: polymorphisms at the microsomal epoxide hydrolase EPHX and glutathione S-transferase GSTM1, GSTT1, and GSTP1 loci and breast cancer risk. Cancer Epidemiol Biomarkers Prev 2007; 16:769–74 [DOI] [PubMed] [Google Scholar]
  • 100.Dessilly G, Elens L, Panin N, et al. : ABCB1 1199G>A genetic polymorphism (Rs2229109) influences the intracellular accumulation of tacrolimus in HEK293 and K562 recombinant cell lines. PLoS One 2014; 9: e91555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kagawa T, Hirose S, Arase Y, et al. : No contribution of the ABCB11 p. 444A polymorphism in Japanese patients with drug-induced cholestasis. Drug Metab Dispos 2015; 43:691–7 [DOI] [PubMed] [Google Scholar]
  • 102.Urban TJ, Sebro R, Hurowitz EH, et al. : Functional genomics of membrane transporters in human populations. Genome Res 2006; 16:223–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Sender R, Fuchs S, Milo R: Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell 2016; 164:337–40 [DOI] [PubMed] [Google Scholar]
  • 104.Kakhniashvili DG, Bulla LA Jr, Goodman SR: The human erythrocyte proteome: analysis by ion trap mass spectrometry. Mol Cell Proteomics 2004; 3:501–9 [DOI] [PubMed] [Google Scholar]
  • 105.Mohandas N, Gallagher PG: Red cell membrane: past, present, and future. Blood 2008; 112:3939–48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Bodemann H, Passow H: Factors controlling the resealing of the membrane of human erythrocyte ghosts after hypotonic hemolysis. J Membr Biol 1972; 8:1–26 [DOI] [PubMed] [Google Scholar]
  • 107.Cundall RB, Dyer A, McHugh JO: Diffusion of drugs from resealed human erythrocyte membrane. J Chem Soc Faraday Trans 1 1981; 77:1039–50 [Google Scholar]
  • 108.Bojesen IN, Hansen HS: Membrane transport of anandamide through resealed human red blood cell membranes. J Lipid Res 2005; 46: 1652–9 [DOI] [PubMed] [Google Scholar]
  • 109.Fye HKS, Mrosso P, Bruce L, et al. : A robust mass spectrometry method for rapid profiling of erythrocyte ghost membrane proteomes. Clin Proteomics 2018; 15:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Doehring A, Hofmann WP, Schlecker C, et al. : Role of nucleoside transporters SLC28A2/3 and SLC29A1/2 genetics in ribavirin therapy: protection against anemia in patients with chronic hepatitis C. Pharmacogenet Genom 2011; 21:289–96 [DOI] [PubMed] [Google Scholar]
  • 111.Beutler E: Drug-induced hemolytic anemia. Pharmacol Rev 1969; 21:73–103 [PubMed] [Google Scholar]
  • 112.Hayes DM, Felts JH: Sulfonamide methemoglobinemia and hemolytic anemia during remal failure. Am J Med Sci 1964; 247:552–7 [DOI] [PubMed] [Google Scholar]
  • 113.De Leeuw N, Shapiro L, Lowenstein L: Drug-induced hemolytic anemia. Ann Intern Med 1963; 58:592–607 [DOI] [PubMed] [Google Scholar]
  • 114.Garbe E, Andersohn F, Bronder E, et al. : Drug induced immune haemolytic anaemia in the Berlin Case-Control Surveillance Study. Br J Haematol 2011; 154:644–53 [DOI] [PubMed] [Google Scholar]
  • 115.Renard D, Rosselet A: Drug-induced hemolytic anemia: Pharmacological aspects. Transfus Clin Biol 2017; 24:110–4 [DOI] [PubMed] [Google Scholar]
  • 116.Petz LD, Garratty G: Immune hemolytic anemias, Philadelphia PA: Gulf Professional Publishing, 2004 [Google Scholar]
  • 117.Garratty G: Immune hemolytic anemia caused by drugs. Expert Opin Drug Saf 2012; 11:635–42 [DOI] [PubMed] [Google Scholar]
  • 118.Garratty G, Arndt PA: Drugs that have been shown to cause drug-induced immune hemolytic anemia or positive direct antiglobulin tests: some interesting findings since 2007. Immunohematology 2014; 30:66–79 [PubMed] [Google Scholar]
  • 119.Arndt PA, Garratty G: The changing spectrum of drug-induced immune hemolytic anemia. Semin Hematol 2005; 42:137–44 [DOI] [PubMed] [Google Scholar]
  • 120.Maloisel F, Kurtz JE, Andres E, et al. : Platin salts-induced hemolytic anemia: cisplatin- and the first case of carboplatin-induced hemolysis. Anticancer Drugs 1995; 6:324–6 [PubMed] [Google Scholar]
  • 121.Arndt P, Garratty G, Isaak E, et al. : Positive direct and indirect antiglobulin tests associated with oxaliplatin can be due to drug antibody and/or drug-induced nonimmunologic protein adsorption. Transfusion 2009; 49:711–8 [DOI] [PubMed] [Google Scholar]
  • 122.Villa CH, Pan DC, Zaitsev S, et al. : Delivery of drugs bound to erythrocytes: new avenues for an old intravascular carrier. Ther Deliv 2015; 6:795–826 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Pierige F, Serafini S, Rossi L, et al. : Cell-based drug delivery. Adv Drug Deliv Rev 2008; 60:286–95 [DOI] [PubMed] [Google Scholar]
  • 124.Hamidi M, Zarrin A, Foroozesh M, et al. : Applications of carrier erythrocytes in delivery of biopharmaceuticals. J Control Release 2007; 118:145–60 [DOI] [PubMed] [Google Scholar]
  • 125.Villa CH, Anselmo AC, Mitragotri S, et al. : Red blood cells: Supercarriers for drugs, biologicals, and nanoparticles and inspiration for advanced delivery systems. Adv Drug Deliv Rev 2016; 106:88–103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Villa CH, Cines DB, Siegel DL, et al. : Erythrocytes as carriers for drug delivery in blood transfusion and beyond. Transfus Med Rev 2017; 31:26–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Spitzer D, Unsinger J, Bessler M, et al. : ScFv-mediated in vivo targeting of DAF to erythrocytes inhibits lysis by complement. Mol Immunol 2004; 40:911–9 [DOI] [PubMed] [Google Scholar]
  • 128.Bloch EM, Jackman RP, Lee TH, et al. : Transfusion-associated microchimerism: the hybrid within. Transfus Med Rev 2013; 27:10–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Peck JR, Elkhammas EA, Li F, et al. : Passenger lymphocyte syndrome: a forgotten cause of postliver transplant jaundice and anemia. Exp Clin Transplant 2015; 13:200–2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Mohamed HJ, Sorich MJ, Kowalski SM, et al. : The role and utility of measuring red blood cell methotrexate polyglutamate concentrations in inflammatory arthropathies–a systematic review. Eur J Clin Pharmacol 2015; 71:411–23 [DOI] [PubMed] [Google Scholar]
  • 131.den Boer E, de Rotte MC, Pluijm SM, et al. : Determinants of erythrocyte methotrexate polyglutamate levels in rheumatoid arthritis. J Rheumatol 2014; 41:2167–78 [DOI] [PubMed] [Google Scholar]
  • 132.Stanislawska-Sachadyn A, Mitchell LE, Woodside JV, et al. : The reduced folate carrier (SLC19A1) c.80G>A polymorphism is associated with red cell folate concentrations among women. Ann Hum Genet 2009; 73:484–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chatzikyriakidou A, Vakalis KV, Kolaitis N, et al. : Distinct association of SLC19A1 polymorphism −43T>C with red cell folate levels and of MTHFR polymorphism 677C>T with plasma folate levels. Clin Biochem 2008; 41:174–6 [DOI] [PubMed] [Google Scholar]
  • 134.Yamamoto T, Shikano K, Nanki T, et al. : Folylpolyglutamate synthase is a major determinant of intracellular methotrexate polyglutamates in patients with rheumatoid arthritis. Sci Rep 2016; 6:35615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Dervieux T, Furst D, Lein DO, et al. : Polyglutamation of methotrexate with common polymorphisms in reduced folate carrier, aminoimidazole carboxamide ribonucleotide transformylase, and thymidylate synthase are associated with methotrexate effects in rheumatoid arthritis. Arthritis Rheum 2004; 50:2766–74 [DOI] [PubMed] [Google Scholar]
  • 136.Ding BC, Witt TL, Hukku B, et al. : Association of deletions and translocation of the reduced folate carrier gene with profound loss of gene expression in methotrexate-resistant K562 human erythroleukemia cells. Biochem Pharmacol 2001; 61:665–75 [DOI] [PubMed] [Google Scholar]
  • 137.Lopez-Lopez E, Ballesteros J, Pinan MA, et al. : Polymorphisms in the methotrexate transport pathway: a new tool for MTX plasma level prediction in pediatric acute lymphoblastic leukemia. Pharmacogenet Genom 2013; 23:53–61 [DOI] [PubMed] [Google Scholar]
  • 138.den Hoed MA, Lopez-Lopez E, te Winkel ML, et al. : Genetic and metabolic determinants of methotrexate-induced mucositis in pediatric acute lymphoblastic leukemia. Pharmacogenomics J 2015; 15:248–54 [DOI] [PubMed] [Google Scholar]
  • 139.Angelis-Stoforidis P, Vajda FJ, Christophidis N: Methotrexate polyglutamate levels in circulating erythrocytes and polymorphs correlate with clinical efficacy in rheumatoid arthritis. Clin Exp Rheumatol 1999; 17:313–20 [PubMed] [Google Scholar]
  • 140.McLeod HL, Krynetski EY, Relling MV, et al. : Genetic polymorphism of thiopurine methyltransferase and its clinical relevance for childhood acute lymphoblastic leukemia. Leukemia 2000; 14:567–72 [DOI] [PubMed] [Google Scholar]
  • 141.Bostrom B, Erdmann G: Cellular pharmacology of 6-mercaptopurine in acute lymphoblastic leukemia. Am J Pediatr Hematol Oncol 1993; 15:80−6 [PubMed] [Google Scholar]
  • 142.Lee MN, Kang B, Choi SY, et al. : Impact of genetic polymorphisms on 6-thioguanine nucleotide levels and toxicity in pediatric patients with IBD treated with azathioprine. Inflamm Bowel Dis 2015; 21:2897908. [DOI] [PubMed] [Google Scholar]
  • 143.Zaza G, Cheok M, Yang W, et al. : Gene expression and thioguanine nucleotide disposition in acute lymphoblastic leukemia after in vivo mercaptopurine treatment. Blood 2005; 106:1778–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Matimba A, Li F, Livshits A, et al. : Thiopurine pharmacogenomics: association of SNPs with clinical response and functional validation of candidate genes. Pharmacogenomics 2014; 15:433–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Krishnamurthy P, Schwab M, Takenaka K, et al. : Transporter-mediated protection against thiopurine-induced hematopoietic toxicity. Cancer Res 2008; 68:4983–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Ban H, Andoh A, Imaeda H, et al. : The multidrug-resistance protein 4 polymorphism is a new factor accounting for thiopurine sensitivity in Japanese patients with inflammatory bowel disease. J Gastroenterol 2010; 45:1014–21 [DOI] [PubMed] [Google Scholar]
  • 147.Mei S, Li X, Gong X, et al. : LC-MS/MS analysis of erythrocyte thiopurine nucleotides and their association with genetic variants in patients with neuromyelitis optica spectrum disorders taking azathioprine. Ther Drug Monit 2017; 39:5–12 [DOI] [PubMed] [Google Scholar]
  • 148.Peltenburg NC, Bierau J, Bakker JA, et al. : Erythrocyte Inosine triphosphatase activity: A potential biomarker for adverse events during combination antiretroviral therapy for HIV. PLoS One 2018; 13: e0191069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Peltenburg NC, Bierau J, Schippers JA, et al. : Metabolic events in HIV-infected patients using abacavir are associated with erythrocyte inosine triphosphatase activity. J Antimicrob Chemother 2019; 74:157–64 [DOI] [PubMed] [Google Scholar]
  • 150.Peltenburg NC, Leers MP, Bakker JA, et al. : Inosine triphosphate pyrophosphohydrolase expression: decreased in leukocytes of HIV-infected patients using combination antiretroviral therapy. J Acquir Immune Defic Syndr 2016; 73:390–5 [DOI] [PubMed] [Google Scholar]
  • 151.Coelho AV, Silva SP, de Alencar LC, et al. : ABCB1 and ABCC1 variants associated with virological failure of first-line protease inhibitors antiretroviral regimens in Northeast Brazil patients. J Clin Pharmacol 2013; 53:1286–93 [DOI] [PubMed] [Google Scholar]
  • 152.Kiser JJ, Carten ML, Aquilante CL, et al. : The effect of lopinavir/ritonavir on the renal clearance of tenofovir in HIV-infected patients. Clin Pharmacol Ther 2008; 83:265–72 [DOI] [PubMed] [Google Scholar]
  • 153.Kiser JJ, Aquilante CL, Anderson PL, et al. : Clinical and genetic determinants of intracellular tenofovir diphosphate concentrations in HIV-infected patients. J Acquir Immune Defic Syndr 2008; 47:298–303 [DOI] [PubMed] [Google Scholar]
  • 154.Rungtivasuwan K, Avihingsanon A, Thammajaruk N, et al. : Influence of ABCC2 and ABCC4 polymorphisms on tenofovir plasma concentrations in Thai HIV-infected patients. Antimicrob Agents Chemother 2015; 59:3240–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Likanonsakul S, Suntisuklappon B, Nitiyanontakij R, et al. : A single-nucleotide polymorphism in ABCC4 is associated with tenofovir-related Beta2-microglobulinuria in Thai patients with HIV-1 infection. PLoS One 2016; 11:e0147724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Anderson PL, Lamba J, Aquilante CL, et al. : Pharmacogenetic characteristics of indinavir, zidovudine, and lamivudine therapy in HIV-infected adults: a pilot study. J Acquir Immune Defic Syndr 2006; 42:441–9 [DOI] [PubMed] [Google Scholar]
  • 157.Tsuchiya K, Hayashida T, Hamada A, et al. : High plasma concentrations of dolutegravir in patients with ABCG2 genetic variants. Pharmacogenet Genomics 2017; 27:416–9 [DOI] [PubMed] [Google Scholar]
  • 158.Baxi SM, Greenblatt RM, Bacchetti P, et al. : Evaluating the association of single-nucleotide polymorphisms with tenofovir exposure in a diverse prospective cohort of women living with HIV. Pharmacogenomics J 2018; 18:245–50 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Boswell-Casteel RC, Hays FA: Equilibrative nucleoside transporters-A review. Nucleosides Nucleotides Nucleic Acids 2017; 36:7–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Endres CJ, Moss AM, Ke B, et al. : The role of the equilibrative nucleoside transporter 1 (ENT1) in transport and metabolism of ribavirin by human and wild-type or Ent1−/−mouse erythrocytes. J Pharmacol Exp Ther 2009; 329:387–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Kasza I, Varady G, Andrikovics H, et al. : Expression levels of the ABCG2 multidrug transporter in human erythrocytes correspond to pharmacologically relevant genetic variations. PLoS One 2012; 7:e48423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Zambo B, Bartos Z, Mozner O, et al. : Clinically relevant mutations in the ABCG2 transporter uncovered by genetic analysis linked to erythrocyte membrane protein expression. Sci Rep 2018; 8:7487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Lange T, Jungmann P, Haberle J, et al. : Reduced number of CFTR molecules in erythrocyte plasma membrane of cystic fibrosis patients. Mol Membr Biol 2006; 23:317–23 [DOI] [PubMed] [Google Scholar]
  • 164.Schillers H: Imaging CFTR in its native environment. Pflugers Arch 2008; 456:163–77 [DOI] [PubMed] [Google Scholar]
  • 165.Ebner A, Nikova D, Lange T, et al. : Determination of CFTR densities in erythrocyte plasma membranes using recognition imaging. Nanotechnology 2008; 19:384017. [DOI] [PubMed] [Google Scholar]
  • 166.Decherf G, Bouyer G, Egee S, et al. : Chloride channels in normal and cystic fibrosis human erythrocyte membrane. Blood Cells Mol Dis 2007; 39:24–34 [DOI] [PubMed] [Google Scholar]
  • 167.De Boeck K, Derichs N, Fajac I, et al. : New clinical diagnostic procedures for cystic fibrosis in Europe. J Cyst Fibros 2011; 10(Suppl 2):S53–66 [DOI] [PubMed] [Google Scholar]
  • 168.Stumpf A, Wenners-Epping K, Walte M, et al. : Physiological concept for a blood based CFTR test. Cell Physiol Biochem 2006; 17:29–36 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

vox12999-sup-0001-tables1

Table S1: Pharmacogenomic variants and associated clinical outcomes.

RESOURCES