Next Article in Journal
Cannabinoids from C. sativa L.: Systematic Review on Potential Pharmacological Effects against Infectious Diseases Downstream and Multidrug-Resistant Pathogens
Previous Article in Journal / Special Issue
Anticancer Activity of 4-Aryl-1,4-Dihydropyridines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacogenetics and the Blood–Brain Barrier: A Whirlwind Tour of Potential Clinical Utility

1
School of Psychology, Deakin University, Geelong, VIC 3220, Australia
2
Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Melbourne, VIC 3215, Australia
3
Alcohol and Other Drugs Integrated Treatment Team, Mildura Base Public Hospital, Mildura, VIC 3550, Australia
4
Change to Improve Mental Health (CHIME), Mental Health, Drug and Alcohol Services, Barwon Health, Geelong, VIC 3220, Australia
5
Academic Unit of Psychiatry and Addiction Medicine, The Australian National University School of Medicine and Psychology, Canberra Hospital, Canberra, ACT 2600, Australia
*
Author to whom correspondence should be addressed.
Future Pharmacol. 2024, 4(3), 574-589; https://doi.org/10.3390/futurepharmacol4030032
Submission received: 19 July 2024 / Revised: 19 August 2024 / Accepted: 3 September 2024 / Published: 5 September 2024
(This article belongs to the Special Issue Feature Papers in Future Pharmacology 2024)

Abstract

:
Genetic factors influence medication response (pharmacogenetics), affecting the pharmacodynamics and pharmacokinetics of many medicaments used in clinical care. The ability of medications to cross the blood–brain barrier (BBB) represents a critical putative factor in the effectiveness and tolerability of various medications relevant to central nervous system disorders (CNS), cancer, and broader medical conditions at a pharmacokinetic (dosing) level. Pharmacogenetics has the potential to personalise medicine to a greater extent than has been possible, with the potential to help reduce heuristic delays to effective tolerable pharmacotherapy. Here, we critically examine and summarise the evidence, particularly for ABCB1 polymorphisms associated with drug transportation and other clinical relevance. These transporters appear to have a role in BBB pharmacogenetics and may indicate new avenues of research that extend beyond the current paradigm of CYP450 polymorphisms. We identify some of the most promising variants for clinical translation while spotlighting the complexities of the involved systems and limitations of the current empirical literature.

1. Introduction

The blood–brain barrier (BBB) forms the primary physical defence of the central nervous system against potential toxins and pathogens. It does so by actively and passively moderating the diffusion and effusion of substances into and out of the central nervous system (CNS). A combination of tight endothelial junctions, active transporters, and adjacent pericytes and astrocytes comprise the neurovascular unit of the BBB, a complex system whose basic physiology is still being elucidated [1]. Genetic variants are known to influence medicament permeability across the BBB, though rarely consistently across a population. Specific polymorphisms are purported to interfere with BBB permeability and the drug delivery of specific CNS-acting medications and so can influence optimal medication dose based on BBB pharmacokinetics. Regular medication use is extremely common; 47.9% percent of European Union citizens over the age of 15 reported taking at least one medication in the past two weeks and 64.8% percent of adults in the USA reported using at least one medication in the past 12 months (in both estimates, average use was higher for males than females and trended upward over the lifespan [2,3]. Furthermore, the effectiveness of medication can vary wildly from person to person and across drug classes; for example, one analysis suggested that the majority of cancer medications with accelerated FDA failed to demonstrate any benefit [4], and recent estimates suggest that as many as around one-third of psychiatric patients are treatment resistant [5]. The clinical and societal significance of ineffective medications is profound.
Medication molecules in the bloodstream encounter the endothelial BBB and, depending on the drug’s properties and whether it is a substrate for organic anion transporters (OATs) or organic cation transporters (OCTs), it may be actively transported from the bloodstream. This step is facilitated by active transport mechanisms against a concentration gradient. Numerous medications serve as substrates for organic anion or cation transport, benefiting from facilitated or expedited transport across endothelial cells at the BBB. Expressed in brain capillary endothelial cells forming the BBB, human organic anion transporting polypeptide 1A2 (OATP1A2) facilitates the uptake of various organic anions, including drugs, hormones, and toxins, from the bloodstream into the brain [6,7]. Notably, specific OATP1A2 variants, such as rs776374083, rs864309501, rs886050885, and rs141502207, have garnered clinical relevance, as highlighted in research by Wang et al. (2020 [8]). Similarly, organic anion transporter 3 plays a crucial role in converting organic anions from the brain into the bloodstream. Located in the abluminal membrane of brain capillary endothelial cells, OAT3 contributes significantly to maintaining homeostasis in the CNS [9]. Organic cation transporters 1 and 2 (OCT1 and OCT2) are also expressed in the BBB and brain parenchyma. A simplified diagram of the mechanism of action is illustrated in Figure 1.
Once the drug enters the endothelial cells, it faces the tight junction barrier. Tight junction proteins (TJP), such as claudin-5 and occludin, restrict the paracellular movement of the drug between these cells. By sealing the intercellular gaps, TJP prevents the passive diffusion of large and hydrophilic molecules, including medications, from freely traversing between the bloodstream and brain tissue [10]. Claudin-5, the most abundant TJP in the BBB, has undergone extensive clinical investigation due to its critical role in maintaining barrier integrity. As such, polymorphisms in the CLDN5 gene, which encodes Claudin-5, have been scrutinised for their potential to alter BBB permeability and medication response [11]. Notably, four distinct CLDN-5 gene polymorphisms, including rs1042711, rs1042713, rs1800888, and rs1554115894, have emerged as subjects of interest in medication response research (ClinVar, [12]). However, further investigation is warranted to elucidate the precise implications of these genetic variations in clinical practice. More thoroughly investigated are the ATP binding cassette (ABC) transporters at the BBB—a family of active transporters highly expressed at the BBB and relevant to medication bioavailability [1].

1.1. Active Transporters

Transport proteins, including efflux transporters like the ABCB1 gene encoding P-glycoprotein (P-gp or PGP), are embedded in the membranes of brain capillary endothelial cells. They actively pump specific molecules out of the brain and back into the bloodstream. When a drug molecule enters the endothelial cells from the bloodstream (via passive diffusion or transporters), efflux transporters like P-gp can recognise and transport the medication back into the bloodstream. This efflux mechanism helps protect the brain from potentially harmful substances and sometimes contributes to medication resistance. Genetic variations in the P-gp expression can influence the efflux transport function at the BBB, potentially affecting drug transport and permeability with a broad impact on a host of disorders and conditions. Indeed, some of the earliest investigations into P-gp examined cancer cells with multidrug resistance [13], though research quickly extended to substances as diverse as opioids, dexamethasone, and anti-HIV protease inhibitors. Recent estimates suggest that 30% of the most commonly prescribed medications are substrates of P-gp [14]. Of course, pharmacotherapy extends outside of the CNS and the BBB. For example, many large-molecule drugs are completely unable to cross the BBB and so exert pharmacodynamic influence through other means, including indirect CNS effects [15]. Monoclonal antibodies are a useful case study. The alpha4-integrin cell binding agent natalizumab reduces overall lymphocyte transport into the brain as a means to combat multiple sclerosis progression but does not cross the BBB itself despite exerting a clear impact within the CNS [16]. Treatment response to monoclonal antibodies also has demonstrable genetic variation in multiple sclerosis and advanced cancers [17,18]. In that regard, we see that the pharmacogenetic response for anticancer drugs like bevacizumab is associated with genes outside of the ABCB1, such as within the nitrous-oxide receptor-regulating AGAP1 [19]. Interestingly, despite not being directly associated with drug transport, AGAP1 variants have been associated with treatment resistance to dozens of anticancer drugs in mice [20].
ABCB1-deficient animals experience neurological and foetal drug toxicity attributed to a disruption to the area with prominent P-gp encoding genes, such as blood–brain and blood–placenta barriers [21]. Likewise, Abcb1a−/− knockout mice experiencing brain metastases exhibit successful responses to drug treatments that would typically face challenges penetrating the blood–brain barrier, such as paclitaxel. Notably, Abcb1a−/− knockout mice demonstrate brain-serum ratios for both risperidone and its active metabolite, 9-hydroxy risperidone, which are 10 times higher than those observed in control mice. Furthermore, the double knockout mice, lacking both Abcb1a and Abcb1b, show brain-to-plasma ratios ranging from 1.1 to 2.6 times higher for various CNS medications than their wild-type counterparts [22].

1.2. Personalised and Precision Medicine

Pharmacological treatment of disease is a difficult process even for experienced clinicians following standard guidelines of gold-standard treatments since patient factors alter drug pharmacodynamics and pharmacokinetics leading to unexpected issues with efficacy and tolerability. The rise of pharmacogenetics in medicine contributed to the emergence of personalised medicine and the more ambitious precision medicine, two innovative healthcare approaches that focus on customising medical treatments to fit each patient’s unique genetic and other individual characteristics [23]. Broadly, such approaches aim to create individualised treatment plans tailored to maximise drug responsiveness while minimising side effects without lengthy trial-and-error processes [24]. In psychiatry, treatment resistance is common. For example, 25–33% of those with schizophrenia are treatment-resistant, necessitating third-line clozapine, which carries a significant burden of potentially serious side effects and mandates regular monitoring [25]. Likewise, standard targeted cancer treatments are difficult to maintain if effective, and the response reduces over time (for example, [26]). Many such responses are familial; Angst (1961) [27] first identified strong familial responses to antidepressant medications, later corroborated for lithium in bipolar disorder treatment [28] and antipsychotic response in schizophrenia [29,30]. Genes have long memories, and it is unsurprising that our ancestry also influences our ABCB1 genes; at least two genetic studies failed to detect or observe rs1211152 in their samples. one from Africa comprising Xhosa, Afrikaans, and English ancestry [31] and another from the United States [32]. Evidence from Dong et al. in 2011 suggests a minor allele frequency (MAF) of <2% among Mexican-American samples, whereas McMahon et al. (2010) [33] observed an MAF of around 9% in a German sample of European ancestry. Rs2032588 is much more prevalent in African populations (~18%) compared with European populations (~6%) and virtually absent in Asian populations [34]. Guided by the principle of individualised medicine for individual care, researchers have attempted to compare pharmacogenetically-guided treatment to standardised treatment with some success, though few high-quality studies are available (see [35,36]).

1.3. Implicated Single Nucleotide Polymorphisms (SNPs)

To assist pharmacogenetic investigators, we have endeavoured to summarise as much of the existing clinical evidence related to pharmacogenetic factors related to BBB permeability and drug delivery. A search of the PubMed National Centre of Biotechnology Information revealed 29 human ABCB1 polymorphisms with potential drug response properties, which we list in Table 1 and organise according to the current clinical significance assigned by the Single Nucleotide Polymorphism Database (dbSNP). For example, extensive investigations have been carried out on the rs2032582 allele. Studies on a humanised mouse model with the 2677 G > T mutation revealed no change in P-glycoprotein expression levels in brain capillary fractions, yet this SNP resulted in increased brain penetration of verapamil, a representative substrate of P-glycoprotein [37]. Moreover, placentas from mothers with the TT/TT genotype exhibited significantly reduced P-glycoprotein expression [38]. A correlation was also observed between the binding potential of verapamil and the dosage of T alleles [39], and Margier et al. (2019) [40] observed an association between rs868755 and increased plasma concentration of 25-hydroxycholecalciferol.
These polymorphisms in the ABCB1 gene are of interest in pharmacogenetics due to their potential function of P-gp, the interaction for drug transport, response, and pharmacokinetics. Furthermore, the presence of likely relevant but under-investigated SNPs suggests potential areas for future research. Given that the specific effects of each polymorphism can vary depending on the drug and the individual’s genetic background, research continues exploring their clinical implications and relevance in optimising drug therapy for individual patients.

2. Results

Given the breadth of research into these SNPs and the heterogeneity of the samples, we have grouped our findings according to the conditions investigated.

2.1. Alzheimer’s

Rs2032582 has been investigated in two studies examining Swiss and Italians with Alzheimer’s disease, though the authors observed no association between the SNP and donepezil efficacy [41,42].

2.2. Cancer and Non-Cancerous Tumours

Investigations into pharmacogenetic factors and cancer are relatively common. For example, in multiple myeloma patients, rs2235013 carriers present with increased drug response and decreased mortality [43] and with improved chemotherapy response and recurrence-free survival in late-stage lung cancer patients with the same [44]. A similar treatment response in chemotherapy-treated stage III lung cancer has been seen for rs2235046. Furthermore, Caronia et al. (2011) [45] found a strong association between the genotype rs10276036 and increased five-year survival rates in osteosarcoma patients treated with a combination of cisplatin, adriamycin, methotrexate, vincristine, and cyclophosphamide. Similarly, Weissfeld et al. (2014) [44] reported an association between the genotype and increased survival and disease-free status at four years post-diagnosis in stage III-IV lung cancer patients treated with chemotherapy, suggesting that the genetic marker might have some prognostic value in cancer treatment. While rs3213619 has been associated with grade two neurotoxicity in oesophageal squamous cell carcinoma [46], it was not associated with survival or prognosis in adriamycin cancer patients treated with taxane-containing regimens [47]. The variant is also potentially protective against paclitaxel-induced peripheral neuropathy [48], particularly in breast cancer [49]. Carriers of rs1045642 report less fatigue and fewer sleep disorders in prolactin adenoma patients treated with cabergoline [50], but higher rates of seizures but a lower incidence of posterior reversible encephalopathy syndrome (PRES) in children with lymphatic leukaemia undergoing treatment [51]. Rs3842 has been associated with improved survival rates in chronic myeloid leukaemia patients treated with dasatinib [52].
The presence of other SNPs might signal a more difficult treatment and recovery for patients. Carriers of rs1202168 appear to have an increased risk of colorectal cancers in two predominantly Caucasian samples (German and Czech; [53]) but might be protective against colorectal cancers in German menopausal hormone replacement therapy patients [54]. Just as concerning is that women homozygous for the rs2214102 allele were found to be at a significantly increased breast cancer risk when taking combined oestrogen–progestogen contraceptive pills [55]. Likewise, patients with breast cancer carrying the ancestral homozygous genotype (GG) had significantly worse progression-free survival than carriers of the non-ancestral allele [56]. Similarly, Sági et al. (2021) [51] found an association between rs1128503 and higher rates of seizures and CNS relapse in children with lymphatic leukaemia, particularly with the CC allele, which trends toward worse outcomes at six months compared to other variants, as reported by Cousar et al. (2013) [57]. Still, more research suggests that rs2235074 could potentially be related by haploblock to xenobiotic efflux in myeloma [58]. Furthermore, Burgueño-Rodríguez et al. (2023) [59] identified an association between rs9282564 and increased risk of neurotoxicity in paediatric acute lymphoblastic leukaemia patients treated with prednisone, vincristine, L-asparaginase, daunorubicin, and methotrexate, indicating potential limitations or risks associated with this treatment regimen. The rs2235035 allele has been observed as associated with increased daunorubicinol clearance in a paediatric oncology sample [60].
Other findings have been less straightforward. Rs2235033 was not found to be associated with methotrexate response in halo-hematopoietic stem cell transplantation in paediatric patients with malignant haematological diseases [61]. Rs13237132 has no available evidence directly addressing drug response in this SNP, though, despite some earlier findings, it does not appear to be associated with ovarian cancer outcomes [62], particularly when treated with taxane [63]. Even less is known about the associated function of rs2235015, which has been found to be unrelated to the side effects of treatment with cabergoline in patients with prolactinomas [50].

2.3. Depression

Depression, particularly major depressive disorder (MDD), is another popular target for pharmacogenetic investigation. Rs1045642 appears to act as a protective factor for depression in a Chinese sample (Xie et al., 2015) [64], though this contrasts observation with a Japanese sample, which demonstrated increased depression rates in carriers [65]. The ABCB1 genotypes of rs1922242 exhibited a significant association with the severity of depressive symptoms in MDD patients who underwent continuous escitalopram treatment for 8 weeks [66]. Elsewhere, Chang et al. (2015) [67] observed that MDD patients with the rs2032582 polymorphism exhibited worse responses to antidepressant treatment, though peri-treatment cortisol levels were unchanged regardless. Conversely, Shan et al. (2019) [68] found no correlation between ABCB1 polymorphisms rs1045642, rs2032582, rs2235040, rs1128503, or rs2235015 and response to SSRIs and SNRIs in the Chinese population, though carriers of rs2032583 reported improved SSRI treatment response, particularly if carrying the TT genotype. Likewise, carriers of rs1128503 (particularly TT genotypes) displayed significantly improved treatment response and escitalopram, venlafaxine [69], or paroxetine [70] efficacy. Ray et al. (2015) [71] also observed an association between rs9282564 and a greater likelihood and faster remission of MDD with sertraline treatment.
Though Menu et al. (2010) [72] found no impact on antidepressant therapy efficacy or tolerance, others found that a specific polymorphism of rs1045642-TT predicted lower effective doses of escitalopram and increased response from venlafaxine [66,69]. However, Kato et al. (2008) [70] suggested that carriers might have poorer responses to paroxetine, suggesting differential treatment responses based on genetic factors. The rs2032582 polymorphism significantly affects both citalopram plasma and cerebrospinal fluid concentrations [73]. The results are more conflicting for paroxetine, where it was associated with paroxetine response in a Japanese cohort [70] but not Swiss [74] or Slovakian cohorts [75]. By comparison, rs2235020 has been associated with improved rates of remission of MDD in a sample of Mexican Americans treated with fluoxetine [76].
Inpatients with MDD who were homozygous rs2235015 GG/TT carriers had an inverse association between medication dosage and hospital stay [77]. However, no association with the polymorphism was observed for symptom severity, and this finding was corroborated in a larger naturalised cohort study of Dutch participants with depressive or anxious disorders [78]. Recent examinations suggest a negative association with depressive symptoms after at least two weeks of antidepressant treatment [79]. The mixed evidence is summarised best by the systematic review and meta-analysis by Magarbeh et al. (2023) [80], who found little robust evidence of an association in antidepressant treatment efficacy for outpatients. Otherwise, Silberbauer et al. (2022) [81] observed that the presence of the minor allele A (or AC) is associated with lower serotonin transporter occupancy than those with the major allele. Though investigated, it further appears to be unrelated to sexual dysfunction in women taking SSRIs for depression [82]. Elsewhere, and despite extensive research, most studies have not demonstrated an association between rs2032588 and psychotropic response or side effect profile. A large (n = 789) naturalistic Dutch cohort study demonstrated a reduction in PGP-dependent antidepressant (citalopram, venlafaxine, paroxetine, and fluvoxamine) side effects [83]. Though rs58898486 has been associated with an increased risk of MDD in Mexican Americans, the researchers do not appear to have included the SNP in their investigation of antidepressant response [76].

2.4. Diabetes Mellitus

Some evidence suggests that the SNP rs4148727 is associated with the lipid profile in type II diabetes mellitus, most strongly for apolipoprotein-A and triglycerides, with likely implications for statin management in the condition [84].

2.5. Epilepsy and Other Convulsant Disorders

Zimprich et al. (2004) [85] reported a significant link between drug resistance and the rs2032582 allele in epilepsy patients, later confirmed by Hung et al. (2005) [86]. Though Vahab et al. (2009) [87] and Sun et al. (2016) [88] found no significant association between these genetic polymorphisms and antiepileptic drug resistance, Lovric et al. (2012) [89] observed higher lamotrigine concentrations in Croatian epilepsy patients with this polymorphism and Smolarz et al. (2017) [90] identified a significant association with drug-resistant epilepsy in the Polish population. Others separately reported associations between genetic variations and drug-resistant epilepsy in Mexican, Tunisian, Han Chinese, and Uygur populations [91,92,93,94]. Additionally, a Jordanian cross-sectional study found an association between rs2032588 and anticonvulsant drug resistance in a cohort of 86 Jordanian men, but not women, with epilepsy [95].
No association with rs868755 was found with drug resistance or sensitivity among children with refractory epilepsy [96]. Meanwhile, the rs1202168 variant is potentially related to the development of abnormal haline dispositions in mesial temporal lobe epilepsy [97], and Louis et al. (2022) [98] have reported an associated decreased seizure occurrence after surgical resections for drug-resistant temporal epilepsy in people with rs10276036.

2.6. HIV

Rs3842 was not associated with plasma concentrations of dolutegravir in HIV-positive patients [99]. Though there is some evidence that the gene is associated with efavirenz plasma concentration in Tanzanian and Ethiopian patients [100], there was no association observed in a Brazilian study of predominantly African ancestry [101]. However, the rs10276036 genotype TT is strongly associated with an increased risk of nevirapine drug hypersensitivity in people with HIV infections [102,103].

2.7. Influenza

Research suggests increased neuropsychiatric adverse reactions in rs1045642 or rs2032582 carrier children with oseltamivir-treated influenza H1N1/09 [104]. Children with heterozygous diplotype SNPs had neuropsychiatric adverse reactions at three times the rate of wild-type homozygous children, though small sample sizes prevented statistical significance.

2.8. Ischaemic Events and Stroke

The presence of rs4148727 was not associated with an increased risk of ischaemic stroke in a South Korean sample but was associated with reduced severity [105]. No significant association was observed for an improvement in clopidogrel efficacy compared to aspirin for stroke prevention in a large Chinese sample of high-risk patients [106].

2.9. Nephrotic Syndrome

Zaorska et al. (2021) [107] established a link between rs1922240 and nephrotic syndrome, where the rare G allele is associated with the occurrence of childhood nephrotic syndrome, steroid dependence (for the AAC haplotype), and mesangial proliferative glomerulonephritis (wild A and AA alleles).

2.10. Opioid Response and Dependence

Rs2032582 has been associated with a higher likelihood of requiring increased methadone doses [108] and improved methadone plasma concentrations and treatment outcomes [109] in Taiwanese patients, thus influencing both the kinetics of methadone-P-glycoprotein interaction and methadone’s potency [110,111]. The need for higher doses of methadone was also reported in Han Chinese [112]. These data suggest the rs2032582 allele increases methadone potency via increasing BBB permeability, which is of considerable clinical significance given the high inter-individual variability in methadone response coupled with the significant toxicity of methadone in terms of cardiac and CNS effects, as well as potential drug–drug interactions via being a substrate for various CYP450 enzymes relevant to psychotropics (i.e., 2B6, 3A4, and 2D6) [113]. Similar impacts on plasma concentration and toxicity have been seen with rs1045642 although there are conflicting results. Higher methadone levels were highlighted as a potential risk factor during opioid replacement therapy by Iwersen-Bergmann et al. (2021) [114], linked to higher pain sensitivity by Zahari et al. (2017) [115] in a Malay sample. Though investigated in several other settings, rs1128503 has not been linked to methadone concentration [114].
A series of alleles, rs1211152, rs2214103, rs2235018, rs2235020, rs2235074, rs2888599, and rs55852620 have been investigated for tramadol response by the Bruce Budowle Laboratory (University of North Texas Health Science Center; [116]), though the results do not appear to have been published yet. This is not dissimilar to rs9282564, which has had several investigations, though few corroborating findings from the clinical literature. Indirect evidence suggests that the SNP might be protective against opioid overdose ([117]). However, children undergoing tonsillectomy using intravenous morphine with GG and GA genotypes of ABCB1 polymorphism rs9282564 had higher risks of opioid-related respiratory depression, [118].

2.11. Psychotic Disorders

In studies of antipsychotic medications and rs2032582, Geers et al. (2020) [119] found a protective effect against antipsychotic-induced hyperprolactinemia in a subgroup of schizophrenia patients treated with risperidone or paliperidone. Cho et al. (2010) [120] reported significantly higher pharmacokinetic parameters following levosulpiride administration, increased sleep duration was observed in Russian adolescents during episodes of psychosis [121], and Kuzman et al. (2008) [122] reported that women with schizophrenia were less prone to significant weight gain during risperidone treatment. Interestingly, male rs1045642 carriers are associated with higher clozapine serum concentrations and with an associated body mass index increase [123].
Though Skogh et al. (2011) [124] demonstrated elevated serum and CSF olanzapine concentrations, Consoli et al. (2009) [125] noted lower clozapine concentrations and Rafaniello et al. (2018) [126] found a lower aripiprazole concentration-to-dose ratio. These findings were corroborated by Belmonte et al. (2018) [127], Hattori et al. (2018) [128], and Koller et al. (2018) who identified that the variants rs1045642, rs2235048, rs1128503, and rs2032582 as especially relevant [129] for aripiprazole and by Xing et al. (2006) [130] and Yasui-Furukori et al. (2007) [131] for risperidone. In contrast, additional research carried out by Suzuki et al. (2014) [132] observed that within a Japanese sample, CYP2D6-variant carriers but not ABCB1-variant carriers had an increased aripiprazole concentration and dose ratios compared to non-carriers.

2.12. Post-Surgical Recovery

Sánchez-Lázaro et al. (2015) [133] reported an association between rs9282564 and significantly lower renal function after heart transplantation when treated with calcineurin inhibitors (tacrolimus or cyclosporine), which could be a potential risk factor for adverse outcomes in transplant patients. Other work suggests that the SNP rs2235013 is associated with increased cyclosporine levels in the months following heart transplant [134], and should be similarly monitored.

2.13. Rheumatoid Arthritis

Numerous associations for methotrexate treatment in rheumatoid arthritis and ACBB1-variants have been observed; rs868755, rs10280623, and rs1858923 were associated with methotrexate toxicity, with the latter also associated with toxicity. Carriers of homogeneous TT and G genotypes appear particularly susceptible [135].

2.14. Traumatic Brain Injury

Patients with severe traumatic brain injuries possessing homogeneous T-allele rs1045642 had more favorable six-month recovery compared to G allele carriers [57].

3. Discussion

Our review has attempted to summarise a considerable body of work into the pharmacogenetics of the BBB and drug delivery in hopes of assisting future research into personalised medicine. Tailoring medicine to the individual by accounting for genetic variation in the permeability of the BBB holds a great deal of promise, particularly within oncology and psychiatry. A slim majority of the identified SNPs (15/29) have been investigated for treatment response or survival in various cancer populations, with more than a handful of clinically relevant findings. Certainly, the severity and impact of cancer have contributed to such research impetus and such work has yielded good fruit for medicine; the observation that rs2235046 was associated with improved treatment response and disease-free survival in stage III lung cancer patients and rs10276036 for stage IV could provide hope for those affected [44]. The promise of pharmacogenetics in psychiatry appears comparably strong. We observed substantial evidence of clinical utility for improved treatment response for MDD, schizophrenia, and epilepsy. Similarly to our findings with oncology, our observation that rs2032583-TT and rs1128503-TT are strongly associated with superior SSRI and venlafaxine efficacy in treating MDD compared to other genotypes could lead to more precise psychiatric treatment.

Implications and Next Steps

ABCB1 is not the only target for pharmacogenetically-guided medicine. A recent meta-analysis into the benefits of pharmacogenetically-guided medicine over traditional approaches for MDD examined twelve randomised controlled trials that instead used the hepatic CYP450 CYP2D6 and CYP2C19 SNPs. The authors observed strong benefits in pharmacogenetically guided conditions for improvement over time, remission of symptoms, and recovery, though they noted that the effects appeared to diminish in more recent studies [136]. Slightly earlier meta-analytic work in MDD but without a specific focus on any particular gene suggested slightly less substantial but still favourable benefits over traditional treatment approaches. Overall, the findings suggest that pharmacogenetics has the potential to significantly improve patient quality of life through targeted approaches and that this benefit might be improved further as additional research identifies more and better targets. Crucially, most existing work that compares pharmacogenetic treatment to traditional approaches focuses on psychiatric medicine, and almost exclusively MDD. However, there are some indications for optimism. At least one early meta-analysis suggests that patients who were administered warfarin at a pharmacogenetically guided dosage experienced substantially fewer major bleeding and thromboembolic events compared to patients treated under standard conditions, and that this benefit increased for longer treatment durations [137]. However, updated analyses have dampened enthusiasm somewhat as earlier significant effects have not been replicated (as is the case for thromboembolic events) and costs remain prohibitive [138,139]. As with MDD, CYP-variants appear to have been the main focus.
As the field of hepatic pharmacogenetics, which explores the genetic variations influencing drug metabolism and response in the liver, gains wider clinical adoption in psychiatry, we are of the considered view that the addition of ABC transporter variants has the scope to enhance dose optimisation. As many psychotropic medications are substrates for active efflux by ABC transporters such as ABCB1, the addition of such variants to phase I hepatic metabolism variants stands to enhance the accuracy of psychotropic pharmacogenetics for dose finding.
Further basic and clinical studies will help pave the way to the future enhancement of psychotropic pharmacogenetics.

Author Contributions

Conceptualization, D.R.S. and A.B.S.; methodology, investigation, writing—original draft preparation, review, and editing, D.R.S., T.T.T.T., R.M.L., R.B., F.A.W. and A.B.S.; visualization, R.M.L. All authors have read and agreed to the published version of the manuscript.

Funding

The authors have received no funding relevant to this review. DRS is supported by the NHRMC Medical Research Future Fund (APP1200214; 2021609).

Conflicts of Interest

A.S. has equity in CNSDose, an Australian DNA lab that performs pharmacogenetic analysis. CNSDose had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results All other authors declare that they have no conflicts of interest.

References

  1. Wu, D.; Chen, Q.; Chen, X.; Han, F.; Chen, Z.; Wang, Y. The blood-brain barrier: Structure, regulation, and drug delivery. Signal Transduct. Target. Ther. 2023, 8, 217. [Google Scholar] [CrossRef] [PubMed]
  2. Cohen, R.A.; Mykyta, L. QuickStats: Percentage of Adults Aged ≥18 Years Who Took Prescription Medication During the Past 12 Months, by Sex and Age Group—National Health Interview Survey, United States, 2021. Morb. Mortal. Wkly. Rep. 2023, 72, 450. [Google Scholar]
  3. Eurostat Medication Use Statistics. Available online: https://ec.europa.eu/eurostat/statistics-explained/index.php?title=Medicine_use_statistics (accessed on 12 August 2024).
  4. Gyawali, B.; Hey, S.P.; Kesselheim, A.S. Assessment of the clinical benefit of cancer drugs receiving accelerated approval. JAMA Intern. Med. 2019, 179, 906–913. [Google Scholar] [CrossRef] [PubMed]
  5. Howes, O.D.; Thase, M.E.; Pillinger, T. Treatment resistance in psychiatry: State of the art and new directions. Mol. Psychiatry 2022, 27, 58–72. [Google Scholar] [CrossRef]
  6. Kalliokoski, A.; Niemi, M. Impact of OATP transporters on pharmacokinetics. Br. J. Pharmacol. 2009, 158, 693–705. [Google Scholar] [CrossRef]
  7. Zhou, Y.; Yuan, J.; Li, Z.; Wang, Z.; Cheng, D.; Du, Y.; Li, W.; Kan, Q.; Zhang, W. Genetic Polymorphisms and Function of the Organic Anion-Transporting Polypeptide 1A2 and Its Clinical Relevance in Drug Disposition. Pharmacology 2015, 95, 201–208. [Google Scholar] [CrossRef]
  8. Wang, J.; Yin, J.; Li, W.; Xiao, C.; Han, J.; Zhou, F. Association between SLCO1A2 genetic variation and methotrexate toxicity in human rheumatoid arthritis treatment. J. Biochem. Mol. Toxicol. 2020, 34, e22513. [Google Scholar] [CrossRef]
  9. Erdman, A.R.; Mangravite, L.M.; Urban, T.J.; Lagpacan, L.L.; Castro, R.A.; de la Cruz, M.; Chan, W.; Huang, C.C.; Johns, S.J.; Kawamoto, M.; et al. The human organic anion transporter 3 (OAT3;SLC22A8): Genetic variation and functional genomics. Am. J. Physiol.-Ren. Physiol. 2006, 290, F905–F912. [Google Scholar] [CrossRef]
  10. Liu, W.-Y.; Wang, Z.-B.; Zhang, L.-C.; Wei, X.; Li, L. Tight junction in blood-brain barrier: An overview of structure, regulation, and regulator substances. CNS Neurosci. Ther. 2012, 18, 609–615. [Google Scholar] [CrossRef]
  11. Greene, C.; Hanley, N.; Campbell, M. Claudin-5: Gatekeeper of neurological function. Fluids Barriers CNS 2019, 16, 3. [Google Scholar] [CrossRef]
  12. National Center for Biotechnology Information. ClinVar. [VCV000828556.2]. Available online: https://www.ncbi.nlm.nih.gov/clinvar/variation/VCV000828556.2 (accessed on 12 August 2024).
  13. Demeule, M.; Régina, A.; Jodoin, J.; Laplante, A.; Dagenais, C.; Berthelet, F.; Moghrabi, A.; Béliveau, R. Drug transport to the brain: Key roles for the efflux pump P-glycoprotein in the blood–brain barrier. Vasc. Pharmacol. 2002, 38, 339–348. [Google Scholar] [CrossRef]
  14. Iversen, D.B.; Andersen, N.E.; Dalgård Dunvald, A.C.; Pottegård, A.; Stage, T.B. Drug metabolism and drug transport of the 100 most prescribed oral drugs. Basic Clin. Pharmacol. Toxicol. 2022, 131, 311–324. [Google Scholar] [CrossRef] [PubMed]
  15. Pardridge, W.M. Blood-brain barrier and delivery of protein and gene therapeutics to brain. Front. Aging Neurosci. 2020, 11, 373. [Google Scholar] [CrossRef]
  16. Zarzuelo Romero, M.J.; Pérez Ramírez, C.; Carrasco Campos, M.I.; Sánchez Martín, A.; Calleja Hernández, M.Á.; Ramírez Tortosa, M.C.; Jiménez Morales, A. Therapeutic value of single nucleotide polymorphisms on the efficacy of new therapies in patients with multiple sclerosis. J. Pers. Med. 2021, 11, 335. [Google Scholar] [CrossRef] [PubMed]
  17. Ahmed, F.T.; Ali, S.H.; Al Gawwam, G.A. Integrin alpha-4 gene polymorphism in relation to natalizumab response in multiple sclerosis patients. Neurol. Asia 2023, 28, 349. [Google Scholar] [CrossRef]
  18. Quintanilha, J.C.; Wang, J.; Sibley, A.B.; Xu, W.; Espin-Garcia, O.; Jiang, C.; Etheridge, A.S.; Ratain, M.J.; Lenz, H.J.; Bertagnolli, M. Genome-wide association studies of survival in 1520 cancer patients treated with bevacizumab-containing regimens. Int. J. Cancer 2022, 150, 279–289. [Google Scholar] [CrossRef]
  19. Meurer, S.; Pioch, S.; Wagner, K.; Muller-Esterl, W.; Gross, S. AGAP1, a novel binding partner of nitric oxide-sensitive guanylyl cyclase. J. Biol. Chem. 2004, 279, 49346–49354. [Google Scholar] [CrossRef] [PubMed]
  20. Catalogue of Somatic Mutations in Cancer. AGAP1 Gene View. Available online: https://cancer.sanger.ac.uk/cosmic/gene/analysis?ln=AGAP1 (accessed on 12 August 2024).
  21. Hodges, L.M.; Markova, S.M.; Chinn, L.W.; Gow, J.M.; Kroetz, D.L.; Klein, T.E.; Altman, R.B. Very important pharmacogene summary. Pharmacogenetics Genom. 2011, 21, 152–161. [Google Scholar] [CrossRef]
  22. Sissung, T.M.; Baum, C.E.; Kirkland, C.T.; Gao, R.; Gardner, E.R.; Figg, W.D. Pharmacogenetics of membrane transporters: An update on current approaches. Mol. Biotechnol. 2010, 44, 152–167. [Google Scholar] [CrossRef]
  23. Ashley, E.A. Towards precision medicine. Nat. Rev. Genet. 2016, 17, 507–522. [Google Scholar] [CrossRef]
  24. Vicente, A.M.; Ballensiefen, W.; Jönsson, J.-I. How personalised medicine will transform healthcare by 2030: The ICPerMed vision. J. Transl. Med. 2020, 18, 180. [Google Scholar] [CrossRef] [PubMed]
  25. Siskind, D.; Siskind, V.; Kisely, S. Clozapine Response Rates among People with Treatment-Resistant Schizophrenia: Data from a Systematic Review and Meta-Analysis. Can. J. Psychiatry 2017, 62, 772–777. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, M.; Herbst, R.S.; Boshoff, C. Toward personalized treatment approaches for non-small-cell lung cancer. Nat. Med. 2021, 27, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
  27. Angst, J. A clinical analysis of the effects of tofranil in depression. Psychopharmacologia 1961, 2, 381–407. [Google Scholar] [CrossRef]
  28. Grof, P.; Duffy, A.; Cavazzoni, P.; Grof, E.; Garnham, J.; MacDougall, M.; O’Donovan, C.; Alda, M. Is Response to Prophylactic Lithium a Familial Trait? J. Clin. Psychiatry 2002, 63, 942–947. [Google Scholar] [CrossRef]
  29. Arranz, M.J.; de Leon, J. Pharmacogenetics and pharmacogenomics of schizophrenia: A review of last decade of research. Mol. Psychiatry 2007, 12, 707–747. [Google Scholar] [CrossRef] [PubMed]
  30. Arranz, M.J.; Rivera, M.; Munro, J.C. Pharmacogenetics of Response to Antipsychotics in Patients with Schizophrenia. CNS Drugs 2011, 25, 933–969. [Google Scholar] [CrossRef] [PubMed]
  31. Ikediobi, O.; Aouizerat, B.; Xiao, Y.; Gandhi, M.; Gebhardt, S.; Warnich, L. Analysis of pharmacogenetic traits in two distinct South African populations. Hum. Genom. 2011, 5, 265–282. [Google Scholar] [CrossRef]
  32. Ma, J.; Divers, J.; Palmer, N.D.; Julian, B.A.; Israni, A.K.; Schladt, D.; Pastan, S.O.; Chattrabhuti, K.; Gautreaux, M.D.; Hauptfeld, V.; et al. Deceased donor multidrug resistance protein 1 and caveolin 1 gene variants may influence allograft survival in kidney transplantation. Kidney Int. 2015, 88, 584–592. [Google Scholar] [CrossRef] [PubMed]
  33. McMahon, F.J.; Akula, N.; Schulze, T.G.; Muglia, P.; Tozzi, F.; Detera-Wadleigh, S.D.; Steele, C.J.M.; Breuer, R.; Strohmaier, J.; Wendland, J.R.; et al. Meta-analysis of genome-wide association data identifies a risk locus for major mood disorders on 3p21.1. Nat. Genet. 2010, 42, 128–131. [Google Scholar]
  34. Consortium, G.P.; Auton, A.; Brooks, L.; Durbin, R.; Garrison, E.; Kang, H. A global reference for human genetic variation. Nature 2015, 526, 68–74. [Google Scholar] [CrossRef] [PubMed]
  35. David, V.; Fylan, B.; Bryant, E.; Smith, H.; Sagoo, G.S.; Rattray, M. An Analysis of Pharmacogenomic-Guided Pathways and Their Effect on Medication Changes and Hospital Admissions: A Systematic Review and Meta-Analysis. Front. Genet. 2021, 12, 698148. [Google Scholar] [CrossRef] [PubMed]
  36. O’Shea, J.; Ledwidge, M.; Gallagher, J.; Keenan, C.; Ryan, C. Pharmacogenetic interventions to improve outcomes in patients with multimorbidity or prescribed polypharmacy: A systematic review. Pharmacogenomics J. 2022, 22, 89–99. [Google Scholar] [CrossRef] [PubMed]
  37. Yamasaki, Y.; Moriwaki, T.; Ogata, S.; Ito, S.; Ohtsuki, S.; Minegishi, G.; Abe, S.; Ohta, Y.; Kazuki, K.; Kobayashi, K.; et al. Influence of MDR1 gene polymorphism (2677G>T) on expression and function of P-glycoprotein at the blood-brain barrier: Utilizing novel P-glycoprotein humanized mice with mutation. Pharmacogenetics Genom. 2022, 32, 288–292. [Google Scholar] [CrossRef]
  38. Hitzl, M.; Schaeffeler, E.; Hocher, B.; Slowinski, T.; Halle, H.; Eichelbaum, M.; Kaufmann, P.; Fritz, P.; Fromm, M.F.; Schwab, M. Variable expression of P-glycoprotein in the human placenta and its association with mutations of the multidrug resistance 1 gene (MDR1, ABCB1). Pharmacogenetics 2004, 14, 309–318. [Google Scholar] [CrossRef]
  39. van Assema, D.M.; Lubberink, M.; Rizzu, P.; van Swieten, J.C.; Schuit, R.C.; Eriksson, J.; Scheltens, P.; Koepp, M.; Lammertsma, A.A.; van Berckel, B.N. Blood-brain barrier P-glycoprotein function in healthy subjects and Alzheimer’s disease patients: Effect of polymorphisms in the ABCB1 gene. EJNMMI Res. 2012, 2, 57. [Google Scholar] [CrossRef]
  40. Margier, M.; Collet, X.; May, C.; Desmarchelier, C.; André, F.; Lebrun, C.; Defoort, C.; Bluteau, A.; Borel, P.; Lespine, A.; et al. ABCB1 (P-glycoprotein) regulates vitamin D absorption and contributes to its transintestinal efflux. FASEB J. 2018, 33, 2084–2094. [Google Scholar] [CrossRef] [PubMed]
  41. Magliulo, L.; Dahl, M.-L.; Lombardi, G.; Fallarini, S.; Villa, L.M.; Biolcati, A.; Scordo, M.G. Do CYP3A and ABCB1 genotypes influence the plasma concentration and clinical outcome of donepezil treatment? Eur. J. Clin. Pharmacol. 2010, 67, 47–54. [Google Scholar] [CrossRef]
  42. Noetzli, M.; Guidi, M.; Ebbing, K.; Eyer, S.; Wilhelm, L.; Michon, A.; Thomazic, V.; Stancu, I.; Alnawaqil, A.-M.; Bula, C.; et al. Population pharmacokinetic approach to evaluate the effect of CYP2D6, CYP3A, ABCB1, POR and NR1I2 genotypes on donepezil clearance. Br. J. Clin. Pharmacol. 2014, 78, 135–144. [Google Scholar] [CrossRef]
  43. Macauda, A.; Castelli, E.; Buda, G.; Pelosini, M.; Butrym, A.; Watek, M.; Kruszewski, M.; Vangsted, A.J.; Rymko, M.; Jamroziak, K.; et al. Inherited variation in the xenobiotic transporter pathway and survival of multiple myeloma patients. Br. J. Haematol. 2018, 183, 375–384. [Google Scholar] [CrossRef]
  44. Weissfeld, J.L.; Diergaarde, B.; Nukui, T.; Buch, S.; Pennathur, A.; Socinski, M.A.; Siegfried, J.M.; Romkes, M. Inherited variation in the ATP-binding cassette transporter ABCB1 and survival after chemotherapy for stage III-IV lung cancer. J. Thorac. Oncol. 2014, 9, 1264–1271. [Google Scholar] [CrossRef]
  45. Caronia, D.; Patiño-Garcia, A.; Peréz-Martínez, A.; Pita, G.; Moreno, L.T.; Zalacain-Díez, M.; Molina, B.; Colmenero, I.; Sierrasesúmaga, L.; Benítez, J.; et al. Effect of ABCB1 and ABCC3 polymorphisms on osteosarcoma survival after chemotherapy: A pharmacogenetic study. PLoS ONE 2011, 6, e26091. [Google Scholar] [CrossRef] [PubMed]
  46. Yang, Y.; Jia, J.; Sun, Z.; Liu, C.; Li, Z.; Xiao, Y.; Yu, J.; Du, F.; Shi, Y.; Sun, J.; et al. Polymorphism of FGD4 and Myelosuppression in Patients with Esophageal Squamous Cell Carcinoma. Future Oncol. 2021, 17, 2351–2363. [Google Scholar] [CrossRef] [PubMed]
  47. Sun, Z.; Yang, X.; Du, F.; Shi, Y.; Sun, J.; Jia, J.; Liu, C.; Xiao, Y.; Yu, J.; Zhang, X.; et al. Association between polymorphisms of ABCB1 and prognosis in esophageal squamous cell carcinoma patients treated with taxane. J. Gene Med. 2022, 24, e3434. [Google Scholar] [CrossRef] [PubMed]
  48. Boora, G.K.; Kanwar, R.; Kulkarni, A.A.; Abyzov, A.; Sloan, J.; Ruddy, K.J.; Banck, M.S.; Loprinzi, C.L.; Beutler, A.S. Testing of candidate single nucleotide variants associated with paclitaxel neuropathy in the trial NCCTG N08C1 (Alliance). Cancer Med. 2016, 5, 631–639. [Google Scholar] [CrossRef] [PubMed]
  49. Abraham, J.E.; Guo, Q.; Dorling, L.; Tyrer, J.; Ingle, S.; Hardy, R.; Vallier, A.-L.; Hiller, L.; Burns, R.; Jones, L.; et al. Replication of Genetic Polymorphisms Reported to Be Associated with Taxane-Related Sensory Neuropathy in Patients with Early Breast Cancer Treated with Paclitaxel. Clin. Cancer Res. 2014, 20, 2466–2475. [Google Scholar] [CrossRef] [PubMed]
  50. Athanasoulia, A.P.; Sievers, C.; Ising, M.; Brockhaus, A.C.; Yassouridis, A.; Stalla, G.K.; Uhr, M. Polymorphisms of the drug transporter gene ABCB1 predict side effects of treatment with cabergoline in patients with PRL adenomas. Eur. J. Endocrinol. 2012, 167, 327–335. [Google Scholar] [CrossRef]
  51. Sági, J.C.; Gézsi, A.; Egyed, B.; Jakab, Z.; Benedek, N.; Attarbaschi, A.; Köhrer, S.; Sipek, J.; Winkowska, L.; Zaliova, M.; et al. Pharmacogenetics of the Central Nervous System-Toxicity and Relapse Affecting the CNS in Pediatric Acute Lymphoblastic Leukemia. Cancers 2021, 13, 2333. [Google Scholar] [CrossRef] [PubMed]
  52. Madejczyk, A.M.; Canzian, F.; Góra-Tybor, J.; Campa, D.; Sacha, T.; Link-Lenczowska, D.; Florek, I.; Prejzner, W.; Całbecka, M.; Rymko, M.; et al. Impact of genetic polymorphisms of drug transporters ABCB1 and ABCG2 and regulators of xenobiotic transport and metabolism PXR and CAR on clinical efficacy of dasatinib in chronic myeloid leukemia. Front. Oncol. 2022, 12, 952640. [Google Scholar] [CrossRef]
  53. Campa, D.; Sainz, J.; Pardini, B.; Vodickova, L.; Naccarati, A.; Rudolph, A.; Novotny, J.; Försti, A.; Buch, S.; von Schönfels, W.; et al. A comprehensive investigation on common polymorphisms in the MDR1/ABCB1 transporter gene and susceptibility to colorectal cancer. PLoS ONE 2012, 7, e32784. [Google Scholar] [CrossRef]
  54. Rudolph, A.; Sainz, J.; Hein, R.; Hoffmeister, M.; Frank, B.; Forsti, A.; Brenner, H.; Hemminki, K.; Chang-Claude, J. Modification of menopausal hormone therapy-associated colorectal cancer risk by polymorphisms in sex steroid signaling, metabolism and transport related genes. Endocr. Relat. Cancer 2011, 18, 371–384. [Google Scholar] [CrossRef]
  55. MARIE-GENICA Consortium on Genetic Susceptibility for Menopausal Hormone Therapy Related Breast Cancer Risk. Polymorphisms in the BRCA1 and ABCB1 genes modulate menopausal hormone therapy associated breast cancer risk in postmenopausal women. Breast Cancer Res. Treat. 2009, 120, 727–736. [Google Scholar]
  56. Vaclavikova, R.; Ehrlichova, M.; Hlavata, I.; Pecha, V.; Kozevnikovova, R.; Trnkova, M.; Adamek, J.; Edvardsen, H.; Kristensen, V.N.; Gut, I.; et al. Detection of frequent ABCB1 polymorphisms by high-resolution melting curve analysis and their effect on breast carcinoma prognosis. Clin. Chem. Lab. Med. 2012, 50, 1999–2007. [Google Scholar] [CrossRef] [PubMed]
  57. Cousar, J.m.L.; Conley, Y.P.; Willyerd, F.A.; Sarnaik, A.A.; Puccio, A.M.; Empey, P.E.; Kochanek, P.M.; Bell, M.J.; Okonkwo, D.O.; Clark, R.S.B. Influence of ATP-binding cassette polymorphisms on neurological outcome after traumatic brain injury. Neurocrit. Care 2013, 19, 192–198. [Google Scholar] [CrossRef] [PubMed]
  58. Martino, A.; Campa, D.; Buda, G.; Sainz, J.; García-Sanz, R.; Jamroziak, K.; Reis, R.M.; Weinhold, N.; Jurado, M.; Ríos, R.; et al. Polymorphisms in xenobiotic transporters ABCB1, ABCG2, ABCC2, ABCC1, ABCC3 and multiple myeloma risk: A case–control study in the context of the International Multiple Myeloma rESEarch (IMMEnSE) consortium. Leukemia 2011, 26, 1419–1422. [Google Scholar] [CrossRef] [PubMed]
  59. Burgueño-Rodríguez, G.; Méndez, Y.; Olano, N.; Schelotto, M.; Castillo, L.; Soler, A.M.; da Luz, J. Pharmacogenetics of pediatric acute lymphoblastic leukemia in Uruguay: Adverse events related to induction phase drugs. Front. Pharmacol. 2023, 14, 1278769. [Google Scholar] [CrossRef]
  60. Thompson, P.; Wheeler, H.E.; Delaney, S.M.; Lorier, R.; Broeckel, U.; Devidas, M.; Reaman, G.H.; Scorsone, K.; Sung, L.; Dolan, M.E.; et al. Pharmacokinetics and pharmacogenomics of daunorubicin in children: A report from the Children’s Oncology Group. Cancer Chemother. Pharmacol. 2014, 74, 831–838. [Google Scholar] [CrossRef]
  61. Ji, Q.; Zhang, Y.; Hu, Y.; Liu, L.; Cao, S.; Gao, L.; Li, B.; Tian, Y.; Kong, L.; Wu, S.; et al. The Influence of Methotrexate-Related Transporter and Metabolizing Enzyme Gene Polymorphisms on Peri-Engraftment Syndrome and Graft-Versus-Host Disease after Haplo-Hematopoietic Stem Cell Transplantation in Pediatric Patients with Malignant Hematological Diseases. Blood 2023, 142 (Suppl. S1), 6960. [Google Scholar]
  62. White, K.L.; Vierkant, R.A.; Fogarty, Z.C.; Charbonneau, B.; Block, M.S.; Pharoah, P.D.P.; Chenevix-Trench, G.; for AOCS/ACS group; Rossing, M.A.; Cramer, D.W.; et al. Analysis of over 10,000 Cases finds no association between previously reported candidate polymorphisms and ovarian cancer outcome. Cancer Epidemiol. Biomark. Prev. 2013, 22, 987–992. [Google Scholar] [CrossRef]
  63. Peethambaram, P.; Fridley, B.L.; Vierkant, R.A.; Larson, M.C.; Kalli, K.R.; Elliott, E.A.; Oberg, A.L.; White, K.L.; Rider, D.N.; Keeney, G.L. Polymorphisms in ABCB1 and ERCC2 associated with ovarian cancer outcome. Int. J. Mol. Epidemiol. Genet. 2011, 2, 185. [Google Scholar]
  64. Xie, W.-W.; Zhang, L.; Wu, R.-R.; Yu, Y.; Zhao, J.-P.; Li, L.-H. Case-control association study of ABCB1 gene and major depressive disorder in a local Chinese Han population. Neuropsychiatr. Dis. Treat. 2015, 11, 1967–1971. [Google Scholar]
  65. Fujii, T.; Ota, M.; Hori, H.; Sasayama, D.; Hattori, K.; Teraishi, T.; Yamamoto, N.; Hashikura, M.; Tatsumi, M.; Higuchi, T.; et al. Association between the functional polymorphism (C3435T) of the gene encoding P-glycoprotein (ABCB1) and major depressive disorder in the Japanese population. J. Psychiatr. Res. 2012, 46, 555–559. [Google Scholar] [CrossRef] [PubMed]
  66. Lin, K.-M.; Chiu, Y.-F.; Tsai, I.J.; Chen, C.-H.; Shen, W.W.; Liu, S.C.; Lu, S.-C.; Liu, C.-Y.; Hsiao, M.-C.; Tang, H.-S.; et al. ABCB1 gene polymorphisms are associated with the severity of major depressive disorder and its response to escitalopram treatment. Pharmacogenetics Genom. 2011, 21, 163–170. [Google Scholar] [CrossRef] [PubMed]
  67. Chang, H.H.; Chou, C.-H.; Yang, Y.K.; Lee, I.H.; Chen, P.S. Association between ABCB1 Polymorphisms and Antidepressant Treatment Response in Taiwanese Major Depressive Patients. Clin. Psychopharmacol. Neurosci. 2015, 13, 250–255. [Google Scholar] [CrossRef] [PubMed]
  68. Shan, X.-X.; Qiu, Y.; Xie, W.-W.; Wu, R.-R.; Yu, Y.; Wu, H.-S.; Li, L.-H. ABCB1 Gene Is Associated With Clinical Response to SNRIs in a Local Chinese Han Population. Front. Pharmacol. 2019, 10, 761. [Google Scholar] [CrossRef] [PubMed]
  69. Singh, A.B.; Bousman, C.A.; Ng, C.H.; Byron, K.; Berk, M. ABCB1 polymorphism predicts escitalopram dose needed for remission in major depression. Transl. Psychiatry 2012, 2, e198. [Google Scholar] [CrossRef] [PubMed]
  70. Kato, M.; Fukuda, T.; Serretti, A.; Wakeno, M.; Okugawa, G.; Ikenaga, Y.; Hosoi, Y.; Takekita, Y.; Mandelli, L.; Azuma, J.; et al. ABCB1 (MDR1) gene polymorphisms are associated with the clinical response to paroxetine in patients with major depressive disorder. Progress Neuro-Psychopharmacol. Biol. Psychiatry 2008, 32, 398–404. [Google Scholar] [CrossRef]
  71. Ray, A.; Tennakoon, L.; Keller, J.; Sarginson, J.E.; Ryan, H.S.; Murphy, G.M.; Lazzeroni, L.C.; Trivedi, M.H.; Kocsis, J.H.; DeBattista, C.; et al. ABCB1 (MDR1) predicts remission on P-gp substrates in chronic depression. Pharmacogenomics J. 2014, 15, 332–339. [Google Scholar] [CrossRef]
  72. Menu, P.; Gressier, F.; Verstuyft, C.; Hardy, P.; Becquemont, L.; Corruble, E. Antidepressants and ABCB1 Gene C3435T Functional Polymorphism: A Naturalistic Study. Neuropsychobiology 2010, 62, 193–197. [Google Scholar] [CrossRef]
  73. Nikisch, G.; Eap, C.; Baumann, P. Citalopram enantiomers in plasma and cerebrospinal fluid of ABCB1 genotyped depressive patients and clinical response: A pilot study. Pharmacol. Res. 2008, 58, 344–347. [Google Scholar] [CrossRef]
  74. Gex-Fabry, M.; Eap, C.B.; Oneda, B.; Gervasoni, N.; Aubry, J.-M.; Bondolfi, G.; Bertschy, G. CYP2D6 and ABCB1 Genetic Variability: Influence on Paroxetine Plasma Level and Therapeutic Response. Ther. Drug Monit. 2008, 30, 474–482. [Google Scholar] [CrossRef]
  75. Vancova, Z.; Cizmarikova, M.; Dragasek, J.; Zofcakova, S.; Kolarcik, P.; Mojzis, J. Does G2677T Polymorphism of the MDR1 Gene Make a Difference in the Therapeutic Response to Paroxetine in Depressed Patients in a Slovakian Population? Med. Sci. Monit. 2018, 24, 3136–3145. [Google Scholar] [CrossRef] [PubMed]
  76. Dong, C.; Wong, M.L.; Licinio, J. Sequence variations of ABCB1, SLC6A2, SLC6A3, SLC6A4, CREB1, CRHR1 and NTRK2: Association with major depression and antidepressant response in Mexican-Americans. Mol. Psychiatry 2009, 14, 1105–1118. [Google Scholar] [CrossRef]
  77. Breitenstein, B.; Scheuer, S.; Pfister, H.; Uhr, M.; Lucae, S.; Holsboer, F.; Ising, M.; Brückl, T.M. The clinical application of ABCB1 genotyping in antidepressant treatment: A pilot study. CNS Spectr. 2013, 19, 165–175. [Google Scholar] [CrossRef] [PubMed]
  78. de Klerk, O.L.; Nolte, I.M.; Bet, P.M.; Bosker, F.J.; Snieder, H.; den Boer, J.A.; Bruggeman, R.; Hoogendijk, W.J.; Penninx, B.W. ABCB1 gene variants influence tolerance to selective serotonin reuptake inhibitors in a large sample of Dutch cases with major depressive disorder. Pharmacogenomics J. 2012, 13, 349–353. [Google Scholar] [CrossRef]
  79. Geers, L.M.; Ochi, T.; Vyalova, N.M.; Losenkov, I.S.; Paderina, D.Z.; Pozhidaev, I.V.; Simutkin, G.G.; Bokhan, N.A.; Wilffert, B.; Touw, D.J.; et al. Influence of eight ABCB1 polymorphisms on antidepressant response in a prospective cohort of treatment-free Russian patients with moderate or severe depression: An explorative psychopharmacological study with naturalistic design. Hum. Psychopharmacol. 2022, 37, e2826. [Google Scholar] [CrossRef] [PubMed]
  80. Magarbeh, L.; Hassel, C.; Choi, M.; Islam, F.; Marshe, V.S.; Zai, C.C.; Zuberi, R.; Gammal, R.S.; Men, X.; Scherf-Clavel, M.; et al. ABCB1 Gene Variants and Antidepressant Treatment Outcomes: A Systematic Review and Meta-Analysis Including Results from the CAN-BIND-1 Study. Clin. Pharmacol. Ther. 2023, 114, 88–117. [Google Scholar] [CrossRef]
  81. Silberbauer, L.R.; Rischka, L.; Vraka, C.; Hartmann, A.M.; Godbersen, G.M.; Philippe, C.; Pacher, D.; Nics, L.; Klöbl, M.; Unterholzner, J.; et al. ABCB1 variants and sex affect serotonin transporter occupancy in the brain. Mol. Psychiatry 2022, 27, 4502–4509. [Google Scholar] [CrossRef]
  82. Bly, M.J.; Bishop, J.R.; Thomas, K.L.H.; Ellingrod, V.L. P-glycoprotein (PGP) polymorphisms and sexual dysfunction in female patients with depression and SSRI-associated sexual side effects. J. Sex. Marital. Ther. 2013, 39, 280–288. [Google Scholar] [CrossRef]
  83. Bet, P.M.; Verbeek, E.C.; Milaneschi, Y.; Straver, D.B.; Uithuisje, T.; Bevova, M.R.; Hugtenburg, J.G.; Heutink, P.; Penninx, B.W.; Hoogendijk, W.J. A common polymorphism in the ABCB1 gene is associated with side effects of PGP-dependent antidepressants in a large naturalistic Dutch cohort. Pharmacogenomics J. 2016, 16, 202–208. [Google Scholar] [CrossRef] [PubMed]
  84. Wu, J.; Wang, X.; Chen, H.; Yang, R.; Yu, H.; Wu, Y.; Hu, Y. Type 2 Diabetes Risk and Lipid Metabolism Related to the Pleiotropic Effects of an ABCB1 Variant: A Chinese Family-Based Cohort Study. Metabolites 2022, 12, 875. [Google Scholar] [CrossRef] [PubMed]
  85. Zimprich, F.; Sunder-Plassmann, R.; Stogmann, E.; Gleiss, A.; Dal-Bianco, A.; Zimprich, A.; Plumer, S.; Baumgartner, C.; Mannhalter, C. Association of an ABCB1 gene haplotype with pharmacoresistance in temporal lobe epilepsy. Neurology 2004, 63, 1087–1089. [Google Scholar] [CrossRef] [PubMed]
  86. Hung, C.-C.; Tai, J.J.; Lin, C.-J.; Lee, M.-J.; Liou, H.-H. Complex Haplotypic Effects of the ABCB1 Gene on Epilepsy Treatment Response. Pharmacogenomics 2005, 6, 411–417. [Google Scholar] [CrossRef]
  87. Vahab, S.A.; Sen, S.; Ravindran, N.; Mony, S.; Mathew, A.; Vijayan, N.; Nayak, G.; Bhaskaranand, N.; Banerjee, M.; Satyamoorthy, K. Analysis of Genotype and Haplotype Effects of ABCB1 (MDR1) Polymorphisms in the Risk of Medically Refractory Epilepsy in an Indian Population. Drug Metab. Pharmacokinet. 2009, 24, 255–260. [Google Scholar] [CrossRef]
  88. Sun, F.; Cao, B.-Q.; Wang, B.; Wu, S.-Q.; Jiang, D.-H. Association between ABCB1 genetic polymorphism and the effect on epilepsy following phenytoin treatment. Exp. Ther. Med. 2016, 12, 1780–1784. [Google Scholar] [CrossRef]
  89. Lovrić, M.; Božina, N.; Hajnšek, S.; Kuzman, M.R.; Sporiš, D.; Lalić, Z.; Božina, T.; Granić, P. Association Between Lamotrigine Concentrations and ABCB1 Polymorphisms in Patients With Epilepsy. Ther. Drug Monit. 2012, 34, 518–525. [Google Scholar] [CrossRef]
  90. Smolarz, B.; Skalski, D.; Rysz, A.; Marchel, A.; Romanowicz, H.; Makowska, M. Polymorphism of the multidrug resistance 1 gene MDR1 G2677T/A (rs2032582) and the risk of drug-resistant epilepsy in the Polish adult population. Acta Neurol. Belg. 2017, 117, 849–855. [Google Scholar] [CrossRef] [PubMed]
  91. Chouchi, M.; Klaa, H.; Ben-Youssef Turki, I.; Hila, L. ABCB1 Polymorphisms and Drug-Resistant Epilepsy in a Tunisian Population. Dis. Markers 2019, 2019, 1343650. [Google Scholar] [CrossRef]
  92. Escalante-Santiago, D.; Feria-Romero, I.A.; Ribas-Aparicio, R.M.; Rayo-Mares, D.; Fagiolino, P.; Vázquez, M.; Escamilla-Núñez, C.; Grijalva-Otero, I.; López-García, M.A.; Orozco-Suárez, S. MDR-1 and MRP2 Gene Polymorphisms in Mexican Epileptic Pediatric Patients with Complex Partial Seizures. Front. Neurol. 2014, 5, 184. [Google Scholar] [CrossRef]
  93. Kwan, P.; Wong, V.; Ng, P.W.; Lui, C.H.T.; Sin, N.C.; Poon, W.S.; Ng, H.K.; Wong, K.S.; Baum, L. Gene-Wide Tagging Study of Association between ABCB1 Polymorphisms and Multidrug Resistance in Epilepsy in Han Chinese. Pharmacogenomics 2009, 10, 723–732. [Google Scholar] [CrossRef] [PubMed]
  94. Zhao, T.; Li, H.-J.; Feng, J.; Zhang, H.-L.; Ting-Ting, W.; Ma, L.; Yu, J.; Zhao, W.-B.; Sun, L.; Yu, L.-H.; et al. Impact of ABCB1 Polymorphisms on Lacosamide Serum Concentrations in Uygur Pediatric Patients With Epilepsy in China. Ther. Drug Monit. 2022, 44, 455–464. [Google Scholar] [CrossRef] [PubMed]
  95. Abduljabbar, R.; Eid, T.D.; Yousef, A.-M.; Mukred, S.R.; Zawiah, M. SCN1A polymorphisms influence the antiepileptic drugs responsiveness in Jordanian epileptic patients. J. Med. Biochem. 2023, 42, 214. [Google Scholar] [CrossRef]
  96. Gao, L.; Yin, X.; Li, Y.; Xiao, H.; Yang, L.; Fan, H.; Qi, H.; Zhang, J.; Feng, J.; Zheng, F. Association of MDR1 gene polymorphisms with refractory epilepsy in children. Zhonghua Yi Xue Yi Chuan Xue Za Zhi = Zhonghua Yixue Yichuanxue Zazhi = Chin. J. Med. Genet. 2019, 36, 1073–1076. [Google Scholar]
  97. Das, A.; Balan, S.; Mathew, A.; Radhakrishnan, V.; Banerjee, M.; Radhakrishnan, K. Corpora amylacea deposition in the hippocampus of patients with mesial temporal lobe epilepsy: A new role for an old gene? Indian. J. Hum. Genet. 2011, 17 (Suppl. S1), S41–S47. [Google Scholar] [PubMed]
  98. Louis, S.; Busch, R.M.; Lal, D.; Hockings, J.; Hogue, O.; Morita-Sherman, M.; Vegh, D.; Najm, I.; Ghosh, C.; Bazeley, P.; et al. Genetic and molecular features of seizure-freedom following surgical resections for focal epilepsy: A pilot study. Front. Neurol. 2022, 13, 942643. [Google Scholar] [CrossRef]
  99. Tsuchiya, K.; Hayashida, T.; Hamada, A.; Oki, S.; Oka, S.; Gatanaga, H. High plasma concentrations of dolutegravir in patients with ABCG2 genetic variants. Pharmacogenetics Genom. 2017, 27, 416–419. [Google Scholar] [CrossRef] [PubMed]
  100. Ngaimisi, E.; Habtewold, A.; Minzi, O.; Makonnen, E.; Mugusi, S.; Amogne, W.; Yimer, G.; Riedel, K.-D.; Janabi, M.; Aderaye, G.; et al. Importance of ethnicity, CYP2B6 and ABCB1 genotype for efavirenz pharmacokinetics and treatment outcomes: A parallel-group prospective cohort study in two sub-Saharan Africa populations. PLoS ONE 2013, 8, e67946. [Google Scholar] [CrossRef]
  101. Coelho, A.V.C.; Silva, S.P.S.; de Alencar, L.C.A.; Stocco, G.; Crovella, S.; Brandão, L.A.C.; Guimarães, R.L. ABCB1 and ABCC1 variants associated with virological failure of first-line protease inhibitors antiretroviral regimens in Northeast Brazil patients. J. Clin. Pharmacol. 2013, 53, 1286–1293. [Google Scholar] [CrossRef]
  102. Dhoro, M.; Ngara, B.; Kadzirange, G.; Nhachi, C.; Masimirembwa, C. Genetic Variants of Drug Metabolizing Enzymes and Drug Transporter (ABCB1) as Possible Biomarkers for Adverse Drug Reactions in an HIV/AIDS Cohort in Zimbabwe. Curr. HIV Res. 2014, 11, 481–490. [Google Scholar] [CrossRef]
  103. Dhoro, M.; Zvada, S.; Ngara, B.; Nhachi, C.; Kadzirange, G.; Chonzi, P.; Masimirembwa, C. CYP2B6*6, CYP2B6*18, Body weight and sex are predictors of efavirenz pharmacokinetics and treatment response: Population pharmacokinetic modeling in an HIV/AIDS and TB cohort in Zimbabwe. BMC Pharmacol. Toxicol. 2015, 16, 4. [Google Scholar] [CrossRef]
  104. L‘Huillier, A.G.; Ing Lorenzini, K.; Crisinel, P.-A.; Rebsamen, M.C.; Fluss, J.; Korff, C.M.; Barbe, R.P.; Siegrist, C.-A.; Dayer, P.; Posfay-Barbe, K.M.; et al. ABCB1 Polymorphisms and Neuropsychiatric Adverse Events in Oseltamivir-Treated Children During Influenza H1N1/09 Pandemia. Pharmacogenomics 2011, 12, 1493–1501. [Google Scholar] [CrossRef] [PubMed]
  105. Kim, Y.-O.; Kim, S.-Y.; Yun, D.H.; Lee, S.-W. Association between ABCB1 Polymorphisms and Ischemic Stroke in Korean Population. Exp. Neurobiol. 2012, 21, 164–171. [Google Scholar] [CrossRef] [PubMed]
  106. Pan, Y.; Chen, W.; Wang, Y.; Li, H.; Johnston, S.C.; Simon, T.; Zhao, X.; Liu, L.; Wang, D.; Meng, X.; et al. Clopidogrel in High-Risk Patients With Acute Nondisabling Cerebrovascular Events, I. Association Between ABCB1 Polymorphisms and Outcomes of Clopidogrel Treatment in Patients With Minor Stroke or Transient Ischemic Attack: Secondary Analysis of a Randomized Clinical Trial. JAMA Neurol. 2019, 76, 552–560. [Google Scholar]
  107. Zaorska, K.; Zawierucha, P.; Świerczewska, M.; Ostalska-Nowicka, D.; Zachwieja, J.; Nowicki, M. Prediction of steroid resistance and steroid dependence in nephrotic syndrome children. J. Transl. Med. 2021, 19, 130. [Google Scholar] [CrossRef]
  108. Levran, O.; O’Hara, K.; Peles, E.; Li, D.; Barral, S.; Ray, B.; Borg, L.; Ott, J.; Adelson, M.; Kreek, M.J. ABCB1 (MDR1) genetic variants are associated with methadone doses required for effective treatment of heroin dependence. Hum. Mol. Genet. 2008, 17, 2219–2227. [Google Scholar] [CrossRef]
  109. Lee, H.-Y.; Li, J.-H.; Sheu, Y.-L.; Tang, H.-P.; Chang, W.-C.; Tang, T.-C.; Yeh, Y.-C.; Wang, S.-Y.; Liu, R.-H. Moving toward personalized medicine in the methadone maintenance treatment program: A pilot study on the evaluation of treatment responses in Taiwan. Biomed. Res. Int. 2013, 2013, 741403. [Google Scholar] [CrossRef] [PubMed]
  110. Coller, J.; Barratt, D.; Dahlen, K.; Loennechen, M.; Somogyi, A. ABCB1 genetic variability and methadone dosage requirements in opioid-dependent individuals. Clin. Pharmacol. Ther. 2006, 80, 682–690. [Google Scholar] [CrossRef]
  111. Hung, C.-C.; Chiou, M.-H.; Teng, Y.-N.; Hsieh, Y.-W.; Huang, C.-L.; Lane, H.-Y. Functional impact of ABCB1 variants on interactions between P-glycoprotein and methadone. PLoS ONE 2013, 8, e59419. [Google Scholar] [CrossRef]
  112. Hung, C.-C.; Chiou, M.-H.; Huang, B.-H.; Hsieh, Y.-W.; Hsieh, T.-J.; Huang, C.-L.; Lane, H.-Y. Impact of Genetic Polymorphisms in ABCB1, CYP2B6, OPRM1, ANKK1 and DRD2 Genes on Methadone Therapy in Han Chinese Patients. Pharmacogenomics 2011, 12, 1525–1533. [Google Scholar] [CrossRef]
  113. Kreutzwiser, D.; Tawfic, Q.A. Methadone for pain management: A pharmacotherapeutic review. CNS Drugs 2020, 34, 827–839. [Google Scholar] [CrossRef]
  114. Iwersen-Bergmann, S.; Plattner, S.; Hischke, S.; Müller, A.; Andresen-Streichert, H.; Jungen, H.; Erb, R.; Beer-Sandner, B. Brain/blood ratios of methadone and ABCB1 polymorphisms in methadone-related deaths. Int. J. Leg. Med. 2021, 135, 473–482. [Google Scholar] [CrossRef] [PubMed]
  115. Zahari, Z.; Lee, C.S.; Ibrahim, M.A.; Musa, N.; Mohd Yasin, M.A.; Lee, Y.Y.; Tan, S.C.; Mohamad, N.; Ismail, R. ABCB1 Polymorphisms and Cold Pressor Pain Responses. Nurs. Res. 2017, 66, 134–144. [Google Scholar] [CrossRef]
  116. Bruce Budowle Laboratory (University of North Texas Health Science Center). Available online: https://www.ncbi.nlm.nih.gov/clinvar/submitters/506790/ (accessed on 12 August 2024).
  117. Christoffersen, D.J.; Damkier, P.; Feddersen, S.; Möller, S.; Thomsen, J.L.; Brasch-Andersen, C.; Brøsen, K. The ABCB1, rs9282564, AG and TT Genotypes and the COMT, rs4680, AA Genotype are Less Frequent in Deceased Patients with Opioid Addiction than in Living Patients with Opioid Addiction. Basic Clin. Pharmacol. Toxicol. 2016, 119, 381–388. [Google Scholar] [CrossRef]
  118. Sadhasivam, S.; Chidambaran, V.; Zhang, X.; Meller, J.; Esslinger, H.; Zhang, K.; Martin, L.J.; McAuliffe, J. Opioid-induced respiratory depression: ABCB1 transporter pharmacogenetics. Pharmacogenomics J. 2014, 15, 119–126. [Google Scholar] [CrossRef]
  119. Geers, L.M.; Pozhidaev, I.V.; Ivanova, S.A.; Freidin, M.B.; Schmidt, A.F.; Cohen, D.; Boiko, A.S.; Paderina, D.Z.; Fedorenko, O.Y.; Semke, A.V.; et al. Association between 8 P-glycoprotein (MDR1/ABCB1) gene polymorphisms and antipsychotic drug-induced hyperprolactinaemia. Br. J. Clin. Pharmacol. 2020, 86, 1827–1835. [Google Scholar] [CrossRef]
  120. Cho, H.Y.; Yoo, H.D.; Lee, Y.B. Influence of ABCB1 genetic polymorphisms on the pharmacokinetics of levosulpiride in healthy subjects. Neuroscience 2010, 169, 378–387. [Google Scholar] [CrossRef] [PubMed]
  121. Ivashchenko, D.V.; Khoang, S.Z.; Makhmudova, B.V.; Buromskaya, N.I.; Shimanov, P.V.; Deitch, R.V.; Akmalova, K.A.; Shuev, G.N.; Dorina, I.V.; Nastovich, M.I.; et al. Pharmacogenetics of antipsychotics in adolescents with acute psychotic episode during first 14 days after admission: Effectiveness and safety evaluation. Drug Metab. Drug Interact. 2020, 35, 20200102. [Google Scholar] [CrossRef] [PubMed]
  122. Kuzman, M.R.; Medved, V.; Bozina, N.; Hotujac, L.; Sain, I.; Bilusic, H. The influence of 5-HT2C and MDR1 genetic polymorphisms on antipsychotic-induced weight gain in female schizophrenic patients. Psychiatry Res. 2008, 160, 308–315. [Google Scholar] [CrossRef]
  123. Piatkov, I.; Caetano, D.; Assur, Y.; Lau, S.L.; Jones, T.; Boyages, S.C.; McLean, M. ABCB1 and ABCC1 single-nucleotide polymorphisms in patients treated with clozapine. Pharmgenomics Pers. Med. 2017, 10, 235–242. [Google Scholar] [CrossRef]
  124. Skogh, E.; Sjödin, I.; Josefsson, M.; Dahl, M.-L. High Correlation Between Serum and Cerebrospinal Fluid Olanzapine Concentrations in Patients With Schizophrenia or Schizoaffective Disorder Medicating With Oral Olanzapine as the Only Antipsychotic Drug. J. Clin. Psychopharmacol. 2011, 31, 4–9. [Google Scholar] [CrossRef]
  125. Consoli, G.; Lastella, M.; Ciapparelli, A.; Dell‘Osso, M.C.; Ciofi, L.; Guidotti, E.; Danesi, R.; Dell‘Osso, L.; Del Tacca, M.; Di Paolo, A. ABCB1 polymorphisms are Associated with Clozapine Plasma Levels in Psychotic Patients. Pharmacogenomics 2009, 10, 1267–1276. [Google Scholar] [CrossRef]
  126. Rafaniello, C.; Sessa, M.; Bernardi, F.F.; Pozzi, M.; Cheli, S.; Cattaneo, D.; Baldelli, S.; Molteni, M.; Bernardini, R.; Rossi, F.; et al. The predictive value of ABCB1, ABCG2, CYP3A4/5 and CYP2D6 polymorphisms for risperidone and aripiprazole plasma concentrations and the occurrence of adverse drug reactions. Pharmacogenomics J. 2017, 18, 422–430. [Google Scholar] [CrossRef]
  127. Belmonte, C.; Ochoa, D.; Román, M.; Saiz-Rodríguez, M.; Wojnicz, A.; Gómez-Sánchez, C.I.; Martín-Vílchez, S.; Abad-Santos, F. Influence of CYP2D6, CYP3A4, CYP3A5 and ABCB1 Polymorphisms on Pharmacokinetics and Safety of Aripiprazole in Healthy Volunteers. Basic Clin. Pharmacol. Toxicol. 2018, 122, 596–605. [Google Scholar] [CrossRef] [PubMed]
  128. Hattori, S.; Suda, A.; Kishida, I.; Miyauchi, M.; Shiraishi, Y.; Fujibayashi, M.; Tsujita, N.; Ishii, C.; Ishii, N.; Moritani, T.; et al. Effects of ABCB1 gene polymorphisms on autonomic nervous system activity during atypical antipsychotic treatment in schizophrenia. BMC Psychiatry 2018, 18, 231. [Google Scholar] [CrossRef] [PubMed]
  129. Koller, D.; Belmonte, C.; Lubomirov, R.; Saiz-Rodríguez, M.; Zubiaur, P.; Román, M.; Ochoa, D.; Carcas, A.; Wojnicz, A.; Abad-Santos, F. Effects of aripiprazole on pupillometric parameters related to pharmacokinetics and pharmacogenetics after single oral administration to healthy subjects. J. Psychopharmacol. 2018, 32, 1212–1222. [Google Scholar] [CrossRef]
  130. Xing, Q.; Gao, R.; Li, H.; Feng, G.; Xu, M.; Duan, S.; Meng, J.; Zhang, A.; Qin, S.; He, L. Polymorphisms of the ABCB1 Gene are Associated with the Therapeutic Response to Risperidone in Chinese Schizophrenia Patients. Pharmacogenomics 2006, 7, 987–993. [Google Scholar] [CrossRef] [PubMed]
  131. Yasui-Furukori, N.; Tsuchimine, S.; Saito, M.; Nakagami, T.; Sato, Y.; Kaneko, S. Association between major Multidrug Resistance 1 (MDR1) gene polymorphisms and plasma concentration of prolactin during risperidone treatment in schizophrenic patients. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2007, 31, 1230–1234. [Google Scholar] [CrossRef]
  132. Suzuki, T.; Mihara, K.; Nakamura, A.; Kagawa, S.; Nagai, G.; Nemoto, K.; Kondo, T. Effects of Genetic Polymorphisms of CYP2D6, CYP3A5, and ABCB1 on the Steady-State Plasma Concentrations of Aripiprazole and Its Active Metabolite, Dehydroaripiprazole, in Japanese Patients With Schizophrenia. Ther. Drug Monit. 2014, 36, 651–655. [Google Scholar] [CrossRef] [PubMed]
  133. Sánchez-Lázaro, I.; Herrero, M.J.; Jordán-De Luna, C.; Bosó, V.; Almenar, L.; Rojas, L.; Martínez-Dolz, L.; Megías-Vericat, J.E.; Sendra, L.; Miguel, A.; et al. Association of SNPs with the Efficacy and Safety of Immunosuppressant Therapy after Heart Transplantation. Pharmacogenomics 2015, 16, 971–979. [Google Scholar] [CrossRef]
  134. de Luna, C.J.; Cervera, M.H.; Lázaro, I.S.; Bonet, L.A.; Andrés, J.P.; Pellicer, S.A. Pharmacogenetic study of ABCB1 and CYP3A5 genes during the first year following heart transplantation regarding tacrolimus or cyclosporine levels. Transplant. Proc. 2011, 43, 2241–2243. [Google Scholar] [CrossRef]
  135. Moya, P.; Salazar, J.; Arranz, M.J.; Díaz-Torné, C.; Del Río, E.; Casademont, J.; Corominas, H.; Baiget, M. Methotrexate pharmacokinetic genetic variants are associated with outcome in rheumatoid arthritis patients. Pharmacogenomics 2016, 17, 25–29. [Google Scholar] [CrossRef]
  136. Arnone, D.; Omar, O.; Arora, T.; Östlundh, L.; Ramaraj, R.; Javaid, S.; Govender, R.D.; Ali, B.R.; Patrinos, G.P.; Young, A.H. Effectiveness of pharmacogenomic tests including CYP2D6 and CYP2C19 genomic variants for guiding the treatment of depressive disorders: Systematic review and meta-analysis of randomised controlled trials. Neurosci. Biobehav. Rev. 2023, 144, 104965. [Google Scholar] [CrossRef] [PubMed]
  137. Li, X.; Yang, J.; Wang, X.; Xu, Q.; Zhang, Y.; Yin, T. Clinical benefits of pharmacogenetic algorithm-based warfarin dosing: Meta-analysis of randomized controlled trials. Thromb. Res. 2015, 135, 621–629. [Google Scholar] [CrossRef] [PubMed]
  138. Tse, G.; Gong, M.; Li, G.; Wong, S.H.; Wu, W.K.; Wong, W.T.; Roever, L.; Lee, A.P.W.; Lip, G.Y.; Wong, M.C. Genotype-guided warfarin dosing vs. conventional dosing strategies: A systematic review and meta-analysis of randomized controlled trials. Br. J. Clin. Pharmacol. 2018, 84, 1868–1882. [Google Scholar] [CrossRef] [PubMed]
  139. Yang, T.; Zhou, Y.; Chen, C.; Lu, M.; Ma, L.; Cui, Y. Genotype-guided dosing versus conventional dosing of warfarin: A meta-analysis of 15 randomized controlled trials. J. Clin. Pharm. Ther. 2019, 44, 197–208. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of action of ABCB1 across the luminal membrane (LM) and abluminal membrane (AM) of the blood–brain barrier using nutrient transporters (yellow and orange), peptides transporters, (purple), and ABC transporters (green) ion transporters (red). The direction(s) of transport is indicated by the arrows. Pictured transporters include Monocarboxylate Transporter 8 (MCT8), Glucose transporter 1 (GLUT1), Monocarboxylate Transporter 1 (MCT1), Human L-type amino acid transporter 1 (L1/LAT1), Cationic Amino Acid Transporter (Y+), insulin receptor (IR), transferring receptor (TFR), Peroxisomal Targeting Signal 1 (PTS1), ATP-binding cassette B1 (ABCB1), ATP-binding cassette G2 (ABCG2), sodium-dependent concentrative transporters A (A), Alanine/serine/cysteine transporter (ASC), Excitatory Amino Acid Transporters (EAATs), Low density lipoprotein receptor-related protein 1 (LRP1), Organo Anion Transporter P1C1 (OATP1C1) and Sodium-Potassium ATPase (Na+,K+,ATPase/NKA).
Figure 1. Mechanisms of action of ABCB1 across the luminal membrane (LM) and abluminal membrane (AM) of the blood–brain barrier using nutrient transporters (yellow and orange), peptides transporters, (purple), and ABC transporters (green) ion transporters (red). The direction(s) of transport is indicated by the arrows. Pictured transporters include Monocarboxylate Transporter 8 (MCT8), Glucose transporter 1 (GLUT1), Monocarboxylate Transporter 1 (MCT1), Human L-type amino acid transporter 1 (L1/LAT1), Cationic Amino Acid Transporter (Y+), insulin receptor (IR), transferring receptor (TFR), Peroxisomal Targeting Signal 1 (PTS1), ATP-binding cassette B1 (ABCB1), ATP-binding cassette G2 (ABCG2), sodium-dependent concentrative transporters A (A), Alanine/serine/cysteine transporter (ASC), Excitatory Amino Acid Transporters (EAATs), Low density lipoprotein receptor-related protein 1 (LRP1), Organo Anion Transporter P1C1 (OATP1C1) and Sodium-Potassium ATPase (Na+,K+,ATPase/NKA).
Futurepharmacol 04 00032 g001
Table 1. Clinical investigations of pharmacogenetically informed drug response of SNPs in the ABCB1 gene. Alternative names are listed in parentheses.
Table 1. Clinical investigations of pharmacogenetically informed drug response of SNPs in the ABCB1 gene. Alternative names are listed in parentheses.
Drug Response and PharmacokineticsLimited Information
Rs IDrs3842, rs1045642 (C3435T), rs1922240, rs2032582 (G2677T/A), rs2235013, rs2235033, rs2235046, rs2888599, rs4148727, rs9282564, rs13237132, rs17064, rs868755, rs1128503, rs1202168, rs1211152, rs1922242, rs2032588, rs2214102, rs2214103, rs2235018, rs2235020, rs2235035, rs2235074, rs3213619, rs10276036rs2235015 (DRD2 Taq1A), rs55852620, rs58898486
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Skvarc, D.R.; Truong, T.T.T.; Lundin, R.M.; Barnes, R.; Wilkes, F.A.; Singh, A.B. Pharmacogenetics and the Blood–Brain Barrier: A Whirlwind Tour of Potential Clinical Utility. Future Pharmacol. 2024, 4, 574-589. https://doi.org/10.3390/futurepharmacol4030032

AMA Style

Skvarc DR, Truong TTT, Lundin RM, Barnes R, Wilkes FA, Singh AB. Pharmacogenetics and the Blood–Brain Barrier: A Whirlwind Tour of Potential Clinical Utility. Future Pharmacology. 2024; 4(3):574-589. https://doi.org/10.3390/futurepharmacol4030032

Chicago/Turabian Style

Skvarc, David R., Trang T. T. Truong, Robert M. Lundin, Russell Barnes, Fiona A. Wilkes, and Ajeet B. Singh. 2024. "Pharmacogenetics and the Blood–Brain Barrier: A Whirlwind Tour of Potential Clinical Utility" Future Pharmacology 4, no. 3: 574-589. https://doi.org/10.3390/futurepharmacol4030032

Article Metrics

Back to TopTop