Preview

Pharmacogenetics and Pharmacogenomics

Advanced search
No 1 (2025)
View or download the full issue PDF (Russian)

FROM EDITOR

What is already known on this topic?

  • Pharmacogenetics is not a new science; it has deep historical roots dating back to the early 20th century (initial descriptions of pharmacogenetic phenomena, discovery of cytochrome P450).
  • It has evolved from studying isolated adverse drug reactions to a systemic understanding of the polygenic nature of the response to most drugs.
  • Associations have been established between polymorphisms in specific genes (CYP2C19, CYP2D6, TPMT, DPYD, etc.) and drug efficacy/safety.
  • Implementation into clinical practice in the Russian Federation faces significant barriers: insufficient integration into clinical guidelines, economic constraints, regulatory gaps, and a lack of knowledge among physicians.

What does this article add new?

  • The article presents a strategic program and vision for the development of pharmacogenetics in Russia for the next decade (until 2035).
  • It announces the creation of new large-scale infrastructure projects: a National DNA Biobank, a National Pharmacogenetic Knowledge Base, and a clinical decision support system (CDSS).
  • It defines the specific technological trends the discipline will focus on: a shift towards multiomics approaches (genomics, proteomics, metabolomics), active use of artificial intelligence for risk prediction, and reducing the cost of whole-genome sequencing.
  • It outlines plans to establish a world-class Center for Predictive Genetics and Pharmacogenetics.

How might this influence clinical practice in the foreseeable future?

  • A paradigm shift in prescribing: Moving from the concept of a "standard dose" to personalized prescription based on a genetic passport, sensor data, and AI prediction.
  • Increased accessibility: Reduced testing costs will make it a routine procedure not only in major centers but also in primary healthcare.
  • Integration into clinical guidelines: The developed algorithms and models will be directly embedded into treatment standards and drug labels.
  • Emergence of new services: Creation of drug information centers and the development of telemedicine consultations with clinical pharmacologists for test interpretation.
  • Proactive medicine: In oncology, the concept of "pharmacogenomic prophylaxis" for carriers of high-risk mutations may emerge.
  • Mandatory testing: In fields like psychiatry, pharmacogenetic testing may become mandatory prior to prescribing therapy.
3-8 95
Abstract

This article is dedicated to the 10th anniversary of the journal “Pharmacogenetics and Pharmacogenomics” and presents a comprehensive analysis of the evolution of the discipline. The article covers historical milestones: from the description of the first pharmacogenetic phenomena and the discovery of cytochrome P450 in the mid-20th century to modern multiomic approaches integrating genomics, transcriptomics and proteomics. Particular attention is paid to the contribution of the journal, which published 242 scientific papers that transformed data on polymorphisms of key genes (CYP2D6, VKORC1, DPYD) into clinical algorithms. Systemic barriers to the implementation of pharmacogenetics in Russia are analyzed: limited use in clinical practice, persistently high cost of NGS sequencing, and the lack of uniform regulatory standards. Much attention in the articles is paid to "ethnopharmacogenetics" — the study of the frequencies of pharmacogenetic markers in the indigenous peoples of Russia, which is important for prioritizing the implementation of certain pharmacogenetic tests in different regions. The prospects for the development of pharmacogenetics include the creation of a National DNA Biobank and a register of patients with an “inadequate” pharmacological response, the development of models for predicting and personalizing pharmacotherapy taking into account pharmacogenetic markers, as well as their clinical validation and “packaging” in a system for supporting medical decision-making, which should be constantly updated based on new research results collected in the National Pharmacogenetic Research Knowledge Base. All this would be impossible without the development of pharmacogenetic test systems available for use in medical organizations in the country, as well as the formation of relevant competencies in doctors and other healthcare professionals. The future lies in integrating pharmacogenetics into comprehensive and large-scale clinical and pharmacological counseling of patients, including using telemedicine.

PHARMACOGENETICS STUDY

What is already known on this topic?

Genetics influences antidepressant efficacy. It is known that the response to drugs, particularly Selective Serotonin Reuptake Inhibitors (SSRIs), varies among patients, and some of this variability is due to genetic factors.

Specific genes are being studied. Two key genes of the serotonergic system—SLC6A4 (encodes the serotonin transporter protein) and TPH1 (encodes an enzyme involved in serotonin synthesis)—are targets of pharmacogenetic research.

The results are contradictory. Data on the influence of polymorphisms in these genes (such as rs4795541 in SLC6A4 and rs1800532 in TPH1) on SSRI efficacy are inconsistent and often depend on the patients' ethnicity, the class of antidepressant, and the study design.

What does this study add?

  • New data for a specific population. This is the first study conducted on patients with mixed anxiety-depressive disorder from the Republic of Bashkortostan.
  • New markers of inefficacy. It was found that in the studied population, the *L/L* genotype of the SLC6A4 gene (rs4795541) and the *A/A* genotype of the TPH1 gene (rs1800532) are associated with lower efficacy of sertraline therapy (evidenced by higher scores on psychometric scales for depression and anxiety throughout the study).
  • Confirmation of the hypothesis. The results confirm the hypothesis of a significant influence of these polymorphic variants on the pharmacogenetics of sertraline, but highlight the ethnic specificity of the conclusions.

How might this affect clinical practice in the foreseeable future?

  • Potential for personalized medicine. In the future, the results of such studies could form the basis for genetic tests to predict sertraline efficacy before starting treatment.
  • Optimizing drug selection. If a patient is found to have the L/L (SLC6A4*) or A/A (TPH1*) genotypes prior to therapy, a physician could consider prescribing a different antidepressant (e.g., from a different pharmacological class) with a higher likelihood of success. This would help reduce the time needed to find an effective therapy and lower the risk of side effects from ineffective treatment.
  • Need for further research. For widespread implementation into clinical practice, larger studies on diverse populations across Russia are needed to refine and verify the obtained data, followed by the development of clinical guidelines based on them.
9-16 68
Abstract

In an attempt to improve the efficacy of antidepressants, pharmacogenetic studies of polymorphic variants of genes involved in the pharmacodynamics of selective serotonin reuptake inhibitors are being conducted worldwide. This study examined the relationship between polymorphic variants of the serotonergic neurotransmission genes rs4795541 of the SLC6A4 gene and rs1800532 of the TPH1 gene and the response to sertraline therapy in patients with mixed anxiety-depressive disorder (F 41.2) from the Republic of Bashkortostan. Patients took sertraline for 6 weeks. The effectiveness of therapy was assessed at baseline and after 2, 4 and 6 weeks using standard psychometric scales. All patients underwent blood sampling with subsequent genotyping. Statistical processing was performed in the SPSS Statistics v. 13.0 program. It was revealed that the rs4795541*L/L and rs7997012*A/A genotypes can be considered as markers of low efficacy of sertraline therapy. The obtained results confirm the hypothesis about the influence of polymorphisms of the studied genes on the pharmacogenetics of sertraline and emphasize the need for further research.

What is already known on this topic?

  • Sensitivity to warfarin is individual and highly dependent on genetic polymorphisms, primarily of the CYP2C9 gene (responsible for drug metabolism) and the VKORC1 gene (its target).
  • By blocking VKORC1, warfarin inhibits not only the synthesis of clotting factors but also the activation of matrix Gla protein (MGP)—a powerful inhibitor of vascular calcification.
  • This links warfarin therapy to the risk of vascular calcification.
  • Chronic rheumatic heart disease (CRHD) itself is characterized by progressive calcification of the heart valves.
  • The frequency of VKORC1 and CYP2C9 alleles has significant ethnic variations, but data on the Chuvash population were limited.

What does the study add?

  • This is the first report of allele frequencies of key pharmacogenetic markers (VKORC1 −1639G>A, CYP2C9*2, CYP2C9*3) specifically in Chuvash patients with CRHD.
  • VKORC1 A allele: 48.6%
  • T allele (CYP2C9*2): 10.15%
  • C allele (CYP2C9*3): 4.05%
  • A novel association was established: in this patient cohort, homozygous carriage of the A allele (AA genotype) of the VKORC1 gene was significantly associated with the development of aortic valve calcification (p=0.023).
  • The very high prevalence of valve calcification (75.7%) in patients with CRHD was confirmed, with the aortic (50%) and mitral (45.9%) valves most commonly affected.

How might this affect clinical practice in the foreseeable future?

  • The obtained data support the feasibility of pharmacogenetic testing in Chuvash patients with CRHD before prescribing warfarin.
  • The test results could help personalize anticoagulant therapy. For carriers of the VKORC1 AA genotype, when choosing therapy, a physician would consider not only the increased risk of bleeding due to hypersensitivity to warfarin but also a potentially higher risk of progression of aortic valve calcification.
  • This may tip the scales in favor of considering warfarin-alternative anticoagulants (DOACs), if there are no other contraindications (e.g., the presence of a mechanical valve prosthesis), especially in patients with already established calcification.
  • For patients who continue warfarin therapy, knowing the genotype will allow for more precise dose titration and enhanced monitoring of the valvular apparatus using echocardiography.
17-23 71
Abstract

Relevance. Patients with chronic rheumatic heart disease (CRHD) often require warfarin, individual sensitivity to which is determined by polymorphisms of the CYP2C9 and VKORC1 genes. Warfarin use is associated with arterial calcification, which is explained by inhibition of the matrix protein MGP. This may be significant in CRHD, a characteristic feature of which is calcification of the heart valves.

Objective. To study the polymorphisms of CYP2C9 (C430T and A1075C), VKORC1 (−1639G>A), and their association with calcification of the heart valves in Chuvash patients with CRHD receiving warfarin.

Methods. The study included patients meeting the following criteria: 18–80 years old, confirmed CRHD, warfarin therapy, self-identification as an ethnic
Chuvash. Calcification of the valves was confirmed by echocardiography. Genotyping was performed by PCR.

Results. The frequency of the A allele of the VKORC1 gene (−1639G>A) was 48.60 %, the T allele (CYP2C9*2) — 10.15%, and the C allele (CYP2C9*3) — 4.05 %. Calcification of the heart valves was detected in 75.7 % of the examined patients: the aortic (50 %) and mitral (45.9 %) valves were most often affected, while the tricuspid valve was involved less often (4.1 %). Statistical analysis demonstrated a significant association between homozygous carriage of the VKORC1 (−1639G>A) allele A and the development of aortic valve calcification (p = 0.023), while the difference between the AA and GA groups also reached statistical significance (p = 0.021).

Conclusion. Identification of VKORC1 genetic variants, as well as the association of carriage of the A allele in homozygous form with valve calcification in patients with CRHD receiving warfarin therapy, can help in personalizing anticoagulant therapy, taking into account both the risks of hypocoagulation and the possible progression of heart valve calcification.

What is already known on this topic?

  • Arterial hypertension (AH) is a global problem and a major modifiable risk factor for cardiovascular diseases.
  • Genetic variants (polymorphisms) of the CYP2C9 gene (specifically, *2 and *3) significantly affect the metabolism of many drugs by reducing enzyme activity.
  • It is known that these polymorphisms affect the pharmacokinetics and efficacy of losartan (another ARB): in carriers of "slow" alleles, its effect is diminished.
  • For irbesartan, data were conflicting: some studies showed higher plasma concentrations in carriers of the *3 allele, but this did not always clearly translate into a greater clinical effect.
  • Valsartan is considered metabolically independent of the CYP450 system, and no significant influence of CYP2C9 genetics on its efficacy had been previously described.

What does the new study add?

  • Direct comparison of two ARBs: This is the first study to directly compare the influence of CYP2C9 polymorphisms on the efficacy of irbesartan and valsartan within a single trial.
  • Identification of a short-term effect: The study shows that carrying the *2 and *3 alleles is associated with a more pronounced reduction in blood pressure (both SBP and DBP) after just 3 weeks of therapy with both drugs.
  • New data on valsartan: Surprisingly, the study found that the efficacy of valsartan (which is barely metabolized by CYP2C9) is also influenced by these genetic variants, especially in the short term. This is a novel finding that contradicts established views.
  • Effect "leveling off" by 3 months: By the end of the 3-month treatment course, no statistically significant difference in efficacy between carriers of different genotypes remained. This suggests that the influence of genetics is most important at the start of treatment.

How might this affect clinical practice in the foreseeable future?

  • Rationale for pharmacogenetic testing at therapy initiation: The results indicate the potential benefit of genetic testing (CYP2C9) at the very beginning of AH therapy selection to predict the early treatment response.
  • Personalized drug and dose selection: Knowing a patient's genotype could help a physician:
  • For irbesartan: In carriers of "slow" alleles, a more powerful initial effect can be expected, potentially allowing for earlier achievement of target BP and less frequent need for therapy intensification (dose increase).
  • For valsartan: The discovered association requires further study, but if confirmed, it would also open possibilities for more accurate response prediction.
  • Improving treatment adherence: Rapid achievement of target BP levels in the first weeks of treatment can motivate the patient and increase their long-term adherence to therapy.
  • Need for further research: For implementation into routine practice, larger-scale studies and an assessment of the cost-effectiveness of this approach are necessary.
24-35 79
Abstract

Arterial hypertension (AH) is one of the most significant modifiable risk factors for cardiovascular diseases, affecting approximately 1.5 billion people worldwide. The study of genetic polymorphisms involved in blood pressure regulation is a promising direction for elucidating the molecular and biological mechanisms underlying the pathogenesis of hypertension. Analyzing associations between gene variants and the response to antihypertensive therapy offers opportunities to develop personalized treatment strategies aimed at improving the efficacy and safety of pharmacotherapy.

Objective. To evaluate the pharmacodynamic efficacy of angiotensin II receptor blockers (ARBs), used as monotherapy or in combination with hydrochlorothiazide, in patients with newly diagnosed AH depending on their genetic background, specifically the CYP2C9 gene polymorphisms Arg144Cys
(rs1799853, CYP2C9*2) and Ile359Leu (rs1057910, CYP2C9*3).

Materials and methods. The study included 179 patients from the Moscow region with newly diagnosed grade 1–2 arterial hypertension, comprising 141 (78.8 %) women and 38 (21.2 %) men aged 32 to 69 years. Participants were randomly assigned to receive either irbesartan or valsartan as monotherapy or in combination with hydrochlorothiazide using simple randomization. Venous blood samples for genotyping CYP2C9*2 and *3 polymorphisms were collected three weeks after enrollment. Office blood pressure was measured at baseline, at 3 weeks, and at 3 months of therapy.

Results. In patients with newly diagnosed AH who had not previously received antihypertensive treatment, a comparative analysis of the effectiveness of irbesartan and valsartan was performed based on CYP2C9*2 (Arg144Cys) and CYP2C9*3 (Ile359Leu) genotypes. Carriers of the *2 and *3 alleles showed a more pronounced reduction in office systolic and diastolic blood pressure after three weeks of therapy with both irbesartan and valsartan. However, by the end of the 3-month follow-up, no statistically significant association was observed between genotype and the magnitude of the antihypertensive response. The influence of CYP2C9 polymorphisms on heart rate was limited and mostly did not reach statistical significance.

Conclusion. The findings suggest a potential role for pharmacogenetic testing in the initiation of angiotensin II receptor blocker therapy in patients with newly diagnosed arterial hypertension.

36-40 66
Abstract

Tamoxifen is the gold standard of endocrine therapy in patients with ER+ breast cancer. However, the efficacy of tamoxifen directly depends on adherence to treatment. Adherence may decrease due to adverse drug reactions and individual differences in drug metabolism. The article presents data from a study of adherence to tamoxifen therapy after 5 years of follow-up in relation to pharmacogenetic associations and adverse drug reactions. Pharmacogenetic testing can improve adherence to treatment by personalizing therapy and reducing the incidence of adverse drug events.

What is already known on this topic?

  • Falls are a global problem: they are the second leading cause of death from unintentional injuries in the elderly.
  • The causes are multifactorial: they include internal (age, diseases) and external (environment) factors, as well as modifiable and non-modifiable ones.
  • Medications are a key modifiable risk factor: certain drug classes (psychotropic, cardiovascular, hypoglycemic) significantly increase the risk of falls.
  • There is a hypothesis about the role of genetics: it is assumed that individual genetic characteristics (pharmacogenetics), which affect drug metabolism, may indirectly influence the risk of falls, but empirical data is very scarce.

What does this study add?

  • A specific study was conducted: this is the first analysis on a Russian cohort (172 patients) of the association between cytochrome P450 gene polymorphisms (*CYP2D6, CYP2C19, CYP3A4/5*) and falls in elderly patients with cardiovascular pathology.
  • Main result - no direct link: no statistically significant differences were found in the distribution of the studied genetic variants between the fall group and the control group as a whole.
  • But associations were found in subgroups: it was discovered that in patients already taking certain medications (beta-blockers, aspirin, inhaled glucocorticoids, alpha-blockers) and those with comorbid diabetes, carrying heterozygous ("intermediate metabolism") and minor genotypes was associated with an increased risk of falls.
  • Conclusion: genetic status by itself does not predict falls, but can be an important modifying risk factor when taking specific medications or having certain diseases.

How might this affect clinical practice in the foreseeable future?

  • Risk personalization: in the future, with more data, pharmacogenetic testing could help identify at-risk patients (taking specific medications) who have a higher probability of falls due to their metabolic profile.
  • More cautious prescribing: for patients with identified "unfavorable" genotypes, doctors could more carefully adjust doses, avoid polypharmacy, or choose alternative drugs not metabolized by the problematic enzymes.
  • Enhanced monitoring: such patients could be placed under closer observation for more active monitoring, education, and fall prevention measures.
  • Basis for further research: this work sets the direction for larger and more targeted studies to more accurately determine for which specific "drug-gene" pairs the risk of falls is highest.
41-48 80
Abstract

Objective. To evaluate the possibility of preventing falls in elderly patients with comorbid cardiovascular pathology by optimizing pharmacotherapy based on pharmacogenetic parameters.

Materials and methods. We studied 172 medical records of people over 65 years of age with comorbid cardiovascular pathologies undergoing treatment in a multidisciplinary hospital in Moscow for the period from 2017 to 2020. All of them underwent blood sampling for genotyping by polymorphisms CYP2D6*4, CYP2C19*2, CYP2C19*3, CYP2C19*17, CYP3A5*3, CYP3A4*22 by polymerase chain reaction — PCR (real-time PCR). All the data obtained were analyzed in pairs using conjugacy tables. To determine statistical reliability, Pearson's -2 methods and Fisher's exact criterion were used. The genotype distribution was checked for compliance with the Hardy–Weinberg equation. To confirm the calculations obtained, mathematical models were additionally constructed using logistic regressions with step-by-step filter selection. The values of p < 0.05 were considered reliable.

Results. The genotyping data was analyzed in comparison groups: 40 patients who had a fall in the last year, and 132 patients without falls. According to the results of the pharmacogenetic study, there was no significant (statistically) difference between patients from our sample with and without falls in the history of CYP2D6*4, CYP2C19*2, CYP2C19*3, CYP2C19*17, CYP3A5*3, CYP3A4*22 polymorphisms and CYP2C19 gene phenotypes. However, a wider range of data was obtained when assessing genotypes in subgroups of patients according to the therapy taken and the presence of concomitant pathology. The predominance of heterozygous and minor genotypes (CT/TT) according to CYP2C19*17 was noted in collapsed patients taking beta-blockers in ophthalmic and oral forms simultaneously (p = 0.05). A predominance of the heterozygous genotype, characterized as an "intermediate" metabolizer, was found among the fallen patients in the subgroups taking enteric acetylsalicylic acid according to the CYP2C19*2 (GA/AA) polymorphism (p = 0.044), in those taking inhaled glucocorticoids of the CYP2C19*17 (CT) polymorphism (p = 0.047) and phenotypic variants of the gene CYP2C19 (p = 0.029), in patients taking alpha-blockers of the CYP3A5*3 (GA) polymorphism (p = 0.026). Patients with falls and diabetes mellitus (as a concomitant disease) have a history of heterozygous genotype (GA) for CYP3A5*3 (p = 0.006), which correlates (p = 0.01) with data from a subgroup of patients taking hypoglycemic therapy.

Conclusion. Thus, closer attention to patients with heterozygous and minor genotype variants in order to prevent falls is highly justified. It is necessary to consider the possibility of analyzing the likelihood of falls, to ensure that drugs are prescribed in lower doses, if possible, and to monitor the state of health of elderly patients more closely.

PERSONALIZED THERAPY

What is already known on this topic?

  • The combination of CAD and AF requires combined antithrombotic therapy (clopidogrel + anticoagulant) to prevent both thromboembolism and stent thrombosis.
  • The main problem with this therapy is the increased risk of hemorrhagic complications, which reduces treatment adherence and worsens the prognosis.
  • The efficacy and safety of a key drug (clopidogrel) varies significantly among patients.
  • Some of this variability is genetically determined (polymorphism of the CYP2C19 gene), which affects the rate of drug metabolism.
  • It was previously shown that carriers of the CYP2C19*17 allele (ultra-rapid metabolizers) may have an increased risk of bleeding.

What does this study add?

  • The article provides a visual clinical case of a patient with a high risk of bleeding on standard therapy, for whom genetic testing revealed an "ultra-rapid metabolizer" status (CYP2C19*17/*17).
  • In addition to CYP2C19, other polymorphisms (genes ABCB1 and CYP3A5) were found in the patient, which, according to literature, are also associated with an increased risk of bleeding when taking anticoagulants.
  • Using a single case, it demonstrates a complex genetic risk profile, not just the influence of a single gene.
  • It presents the authors' own research data from 150 patients, confirming that there are statistically significantly more ultra-rapid metabolizers among patients with bleeding (19.2% vs. 3.4%).

How might this affect clinical practice in the foreseeable future?

  • It substantiates the feasibility of implementing pharmacogenetic testing (particularly for the CYP2C19*17 polymorphism) into the routine practice of cardiologists for bleeding risk stratification.
  • This will allow for therapy personalization at the stage of its prescription: in patients with an identified genetically high risk of bleeding, a shorter course of triple therapy or alternative regimens can be considered immediately.
  • Timely therapy adjustment based on genetic data (de-escalation) will help prevent the development of severe bleeding, while maintaining treatment effectiveness and improving patient adherence.
  • The "treat the patient, not the disease" approach will become more concrete and technological, which in perspective could reduce the number of hospitalizations and improve patient prognosis.
49-56 84
Abstract

The use of antithrombotic drugs is a fundamental principle in the treatment of patients with coronary artery disease and atrial fibrillation. However, when prescribing combined treatment regimens, there is a natural increase in the risk of hemorrhagic complications, both clinically significant, life-threatening, and minor annoying ones that reduce patient compliance, which steadily leads to thromboembolic complications. The interindividual variability in the efficacy and safety of clopidogrel and direct oral anticoagulants may be due, among other factors, to the genetic characteristics of the patient. The results of modern research are ambiguous and dictate further study of this issue. The article presents a clinical case of a 61-year-old man with a combination of coronary artery disease and atrial fibrillation, who was prescribed clopidogrel, acetylsalicylic acid and rivaroxaban after stenting. Against the background of the development of extensive hematomas up to 15 cm in diameter and nosebleeds, treatment was replaced first with double antithrombotic therapy (acetylsalicylic acid was discontinued), and then with clopidogrel and apixaban. According to the results of an additional pharmacogenetic study, it was revealed that the patient was a carrier of CYP2C19*17/*17 (the CYP2C19 ultra-rapid metabolizer genotype), CYP3A5*3/*3, of the TT genotype according to the polymorphic variant rs2032582 of the ABCB1 gene. The bleeding was assessed retrospectively and then over the follow-up period (16 weeks). Understanding the genetic factors influencing the therapeutic response may allow doctors to optimize commonly accepted treatment regimens and minimize unwanted drug reactions.



Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 License.


ISSN 2588-0527 (Print)
ISSN 2686-8849 (Online)