Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (695)

Search Parameters:
Keywords = pharmaco-genetics

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 978 KiB  
Review
NUDT15 Pharmacogenetics in Acute Lymphoblastic Leukemia: Synthesizing Progress for Personalized Thiopurine Therapy
by Isfahan Shah Lubis, Kusnandar Anggadiredja, Aluicia Anita Artarini, Nur Melani Sari, Nur Suryawan and Zulfan Zazuli
Med. Sci. 2025, 13(3), 112; https://doi.org/10.3390/medsci13030112 - 5 Aug 2025
Abstract
The management of acute lymphoblastic leukemia (ALL), the most common pediatric malignancy, critically relies on thiopurine therapy, such as 6-mercaptopurine (6-MP), during the maintenance phase. However, significant inter-individual response variety and high risk of myelosuppression often disrupt therapy efficacy. Pharmacogenetics offer crucial strategies [...] Read more.
The management of acute lymphoblastic leukemia (ALL), the most common pediatric malignancy, critically relies on thiopurine therapy, such as 6-mercaptopurine (6-MP), during the maintenance phase. However, significant inter-individual response variety and high risk of myelosuppression often disrupt therapy efficacy. Pharmacogenetics offer crucial strategies to personalized therapy. While thiopurine methyltransferase (TPMT) was initially the primary focus, the discovery of nudix hydrolase 15 (NUDT15) appears as a more comprehensive determinant of thiopurine intolerance. This review aims to consolidate and critically evaluate the advancement achieved in unraveling the biological mechanism and clinical significance of NUDT15 pharmacogenetics in thiopurine therapy. Foundational studies showed the vital role of NUDT15 in the detoxification of active thiopurines, with common genetic variants (for instance, p. Arg139Cys) significantly disrupting its activity, leading to the accumulation of toxic metabolites. Observational studies consistently associated NUDT15 variants with severe myelosuppression, notably in Asian populations. Recent randomized controlled trials (RCTs) confirmed that NUDT15 genotype-guided dosing effectively reduces thiopurine-induced toxicity without interfering with the therapeutic outcome. Despite these advancements, challenges remain present, including the incomplete characterization of rare variants, limited data in the diverse Asian populations, and the need for standardized integration with metabolite monitoring. In conclusion, NUDT15 pharmacogenetics is essential for improving patient safety and thiopurine dosage optimization in the treatment of ALL. For thiopurine tailored medicine to be widely and fairly implemented, future research should focus on increasing genetic data across different populations, improving the dose adjustment algorithm, and harmonizing therapeutic guidelines. Full article
Show Figures

Figure 1

42 pages, 1287 KiB  
Review
A Comprehensive Review of the Latest Approaches to Managing Hypercholesterolemia: A Comparative Analysis of Conventional and Novel Treatments: Part II
by Narcisa Jianu, Ema-Teodora Nițu, Cristina Merlan, Adina Nour, Simona Buda, Maria Suciu, Silvia Ana Luca, Laura Sbârcea, Minodora Andor and Valentina Buda
Pharmaceuticals 2025, 18(8), 1150; https://doi.org/10.3390/ph18081150 - 1 Aug 2025
Viewed by 529
Abstract
Cardiovascular disease (CVD) remains the leading cause of mortality worldwide, with hypercholesterolemia identified as a major, but modifiable risk factor. This review serves as the second part of a comprehensive analysis of dyslipidemia management. The first installment laid the groundwork by detailing the [...] Read more.
Cardiovascular disease (CVD) remains the leading cause of mortality worldwide, with hypercholesterolemia identified as a major, but modifiable risk factor. This review serves as the second part of a comprehensive analysis of dyslipidemia management. The first installment laid the groundwork by detailing the key pathophysiological mechanisms of lipid metabolism, the development of atherosclerosis, major complications of hyperlipidemia, and the importance of cardiovascular risk assessment in therapeutic decision-making. It also examined non-pharmacological interventions and conventional therapies, with a detailed focus on statins and ezetimibe. Building upon that foundation, the present article focuses exclusively on emerging pharmacological therapies designed to overcome limitations of standard treatment. It explores the mechanisms, clinical applications, safety profiles, and pharmacogenetic aspects of novel agents such as proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors (alirocumab, evolocumab), small interfering RNA (siRNA) therapy (inclisiran), adenosine triphosphate–citrate lyase (ACL) inhibitor (bempedoic acid), microsomal triglyceride transfer protein (MTP) inhibitor (lomitapide), and angiopoietin-like protein 3 (ANGPTL3) inhibitor (evinacumab). These agents offer targeted strategies for patients with high residual cardiovascular risk, familial hypercholesterolemia (FH), or statin intolerance. By integrating the latest advances in precision medicine, this review underscores the expanding therapeutic landscape in dyslipidemia management and the evolving potential for individualized care. Full article
(This article belongs to the Special Issue Pharmacotherapy of Dyslipidemias, 2nd Edition)
Show Figures

Figure 1

28 pages, 2004 KiB  
Review
Opioid Use in Cancer Pain Management: Navigating the Line Between Relief and Addiction
by Maite Trullols and Vicenç Ruiz de Porras
Int. J. Mol. Sci. 2025, 26(15), 7459; https://doi.org/10.3390/ijms26157459 - 1 Aug 2025
Viewed by 149
Abstract
The use of opioids for cancer-related pain is essential but poses significant challenges due to the risk of misuse and the development of opioid use disorder (OUD). This review takes a multidisciplinary perspective based on the current scientific literature to analyze the pharmacological [...] Read more.
The use of opioids for cancer-related pain is essential but poses significant challenges due to the risk of misuse and the development of opioid use disorder (OUD). This review takes a multidisciplinary perspective based on the current scientific literature to analyze the pharmacological mechanisms, classification, and therapeutic roles of opioids in oncology. Key risk factors for opioid misuse—including psychiatric comorbidities, prior substance use, and insufficient clinical monitoring—are discussed in conjunction with validated tools for pain assessment and international guidelines. The review emphasizes the importance of integrating toxicological, pharmacological, physiological, and public health perspectives to promote rational opioid use. Pharmacogenetic variability is explored as a determinant of treatment response and addiction risk, underscoring the value of personalized medicine. Evidence-based strategies such as early screening, psychosocial interventions, and the use of buprenorphine-naloxone are presented as effective measures for managing OUD in cancer patients. Ultimately, this work advocates for safe, patient-centered opioid prescribing practices that ensure effective pain relief without compromising safety or quality of life. Full article
(This article belongs to the Special Issue Recent Progress of Opioid Research, 2nd Edition)
Show Figures

Figure 1

16 pages, 1138 KiB  
Review
Cardiac Myosin Inhibitors in the Treatment of Hypertrophic Cardiomyopathy: Clinical Trials and Future Challenges
by Arnold Kukowka and Marek Droździk
Biomolecules 2025, 15(8), 1098; https://doi.org/10.3390/biom15081098 - 29 Jul 2025
Viewed by 372
Abstract
Hypertrophic cardiomyopathy (HCM) is a prevalent and often underdiagnosed genetic cardiac disorder characterized by left ventricular hypertrophy and, in many cases, dynamic left ventricular outflow tract obstruction (LVOTO). The development of cardiac myosin inhibitors (CMIs) represents an emerging therapeutic approach in the pharmacological [...] Read more.
Hypertrophic cardiomyopathy (HCM) is a prevalent and often underdiagnosed genetic cardiac disorder characterized by left ventricular hypertrophy and, in many cases, dynamic left ventricular outflow tract obstruction (LVOTO). The development of cardiac myosin inhibitors (CMIs) represents an emerging therapeutic approach in the pharmacological management of obstructive HCM (oHCM). This review offers an integrated and up-to-date synthesis of the cardiac myosin inhibitor class, with a focus on mavacamten, aficamten, and the broader landscape of emerging agents. It also highlights recent clinical trial outcomes, pharmacokinetic and pharmacogenetic considerations, and potential future directions in therapy. Furthermore, we incorporate the most recent data up to May 2025, including late-breaking trial results and real-world safety findings, aiming to provide clinicians with a practical and comprehensive perspective on this evolving drug class. A narrative review was conducted by systematically searching PubMed, Scopus, Google Scholar, and ClinicalTrials.gov for English-language articles and trials published between January 2016 and May 2025. Keywords included “cardiac myosin inhibitor”, mavacamten”, “aficamten”, “MYK-224”, and “hypertrophic cardiomyopathy.” Inclusion criteria encompassed clinical trials and comprehensive reviews specifically addressing CMIs in cardiac applications. CMIs such as mavacamten and aficamten have demonstrated significant clinical benefits in reducing LVOT gradients, improving exercise capacity, and alleviating symptoms in patients with oHCM. Mavacamten is currently approved for clinical use, while aficamten is in advanced regulatory review. Comparative data suggest potential advantages of aficamten in the onset of action, pharmacokinetic profile, and tolerability. Emerging evidence supports the exploration of CMIs in pediatric populations, heart failure with preserved ejection fraction (HFpEF), and non-obstructive HCM (nHCM), although results are still preliminary. Cardiac myosin inhibitors offer a novel, pathophysiology-targeted approach to managing oHCM. While mavacamten has established efficacy, next-generation agents like aficamten may offer improved safety and versatility. Further long-term studies are needed to clarify their role across broader patient populations. Full article
Show Figures

Figure 1

24 pages, 1681 KiB  
Review
Molecular Insight into the Role of HLA Genotypes in Immunogenicity and Secondary Refractoriness to Anti-TNF Therapy in IBD Patients
by Mladen Maksic, Irfan Corovic, Tijana Maksic, Jelena Zivic, Milos Zivic, Natasa Zdravkovic, Aleksa Begovic, Marija Medovic, Djordje Kralj, Zeljko Todorovic, Milica Cekerevac, Rasa Medovic and Milos Nikolic
Int. J. Mol. Sci. 2025, 26(15), 7274; https://doi.org/10.3390/ijms26157274 - 28 Jul 2025
Viewed by 338
Abstract
The emergence of anti-TNF agents has revolutionized the management of inflammatory bowel disease, yet a significant proportion of patients experience primary non-response or secondary loss of response due to immunogenicity. As the field of precision medicine advances, genetic predictors such as human leukocyte [...] Read more.
The emergence of anti-TNF agents has revolutionized the management of inflammatory bowel disease, yet a significant proportion of patients experience primary non-response or secondary loss of response due to immunogenicity. As the field of precision medicine advances, genetic predictors such as human leukocyte antigen (HLA) variants are gaining increasing attention. This review provides a comprehensive synthesis of current evidence on the role of HLA genotypes in inflammatory bowel disease susceptibility and disease behavior, with a focus on their mechanistic and clinical relevance in anti-TNF therapy. Special emphasis is placed on HLA-DQA1*05, a validated predictor of anti-drug antibody formation and reduced therapeutic durability. We explore the immunological basis of HLA-mediated immunogenicity, summarize pharmacogenetic and biomarker findings, and discuss how HLA typing may be integrated into treatment algorithms to improve patient stratification and long-term outcomes. As immunogenetics continues to inform clinical decision-making, understanding the interplay between HLA polymorphisms and therapeutic response offers new opportunities for biomarker-guided, personalized care in inflammatory bowel disease. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

33 pages, 1268 KiB  
Review
A Comprehensive Review of the Latest Approaches to Managing Hypercholesterolemia: A Comparative Analysis of Conventional and Novel Treatments: Part I
by Ema-Teodora Nițu, Narcisa Jianu, Cristina Merlan, Darius Foica, Laura Sbârcea, Valentina Buda, Maria Suciu, Adelina Lombrea and Dana Emilia Movilă
Life 2025, 15(8), 1185; https://doi.org/10.3390/life15081185 - 25 Jul 2025
Cited by 1 | Viewed by 956
Abstract
Hypercholesterolemia is a major modifiable risk factor for atherosclerotic cardiovascular disease (ASCVD), affecting a significant proportion of the adult population worldwide. This narrative review provides a comprehensive and up-to-date overview of hyperlipidemia management, spanning from epidemiological trends and underlying pathophysiological mechanisms to the [...] Read more.
Hypercholesterolemia is a major modifiable risk factor for atherosclerotic cardiovascular disease (ASCVD), affecting a significant proportion of the adult population worldwide. This narrative review provides a comprehensive and up-to-date overview of hyperlipidemia management, spanning from epidemiological trends and underlying pathophysiological mechanisms to the limitations of conventional therapies such as statins and ezetimibe. Particular emphasis is placed on cardiovascular risk assessment, current stratification tools, and international guideline-based interventions. The present paper, focusing primarily on the biological mechanisms of dyslipidemia and the clinical use of traditional lipid-lowering agents, serves as the first part of a two-part series, preceding a forthcoming review of novel pharmacological approaches. Our data synthesis is based on a structured literature search conducted across Google Scholar, PubMed, and Scopus, including studies published up to June 2025. The review also includes aspects related to non-pharmacological strategies, pharmacoeconomic considerations, and pharmacogenetic influences on treatment response. Ultimately, this work aims to equip clinicians with evidence-based, nuanced insights essential for optimizing lipid management and reducing cardiovascular risk, while setting the foundation for understanding how emerging therapies may overcome current therapeutic limitations. Full article
Show Figures

Figure 1

16 pages, 654 KiB  
Article
Effect of Pharmacogenetics on Renal Outcomes of Heart Failure Patients with Reduced Ejection Fraction (HFrEF) in Response to Dapagliflozin
by Neven Sarhan, Mona F. Schaalan, Azza A. K. El-Sheikh and Bassem Zarif
Pharmaceutics 2025, 17(8), 959; https://doi.org/10.3390/pharmaceutics17080959 - 24 Jul 2025
Viewed by 356
Abstract
Background/Objectives: Heart failure with reduced ejection fraction (HFrEF) is associated with significant renal complications, affecting disease progression and patient outcomes. Sodium-glucose co-transporter-2 (SGLT2) inhibitors have emerged as a key therapeutic strategy, offering cardiovascular and renal benefits in these patients. However, interindividual variability [...] Read more.
Background/Objectives: Heart failure with reduced ejection fraction (HFrEF) is associated with significant renal complications, affecting disease progression and patient outcomes. Sodium-glucose co-transporter-2 (SGLT2) inhibitors have emerged as a key therapeutic strategy, offering cardiovascular and renal benefits in these patients. However, interindividual variability in response to dapagliflozin underscores the role of pharmacogenetics in optimizing treatment efficacy. This study investigates the influence of genetic polymorphisms on renal outcomes in HFrEF patients treated with dapagliflozin, focusing on variations in genes such as SLC5A2, UMOD, KCNJ11, and ACE. Methods: This prospective, observational cohort study was conducted at the National Heart Institute, Cairo, Egypt, enrolling 200 patients with HFrEF. Genotyping of selected single nucleotide polymorphisms (SNPs) was performed using TaqMan™ assays. Renal function, including estimated glomerular filtration rate (eGFR), Kidney Injury Molecule-1 (KIM-1), and Neutrophil Gelatinase-Associated Lipocalin (NGAL) levels, was assessed at baseline and after six months of dapagliflozin therapy. Results: Significant associations were found between genetic variants and renal outcomes. Patients with AA genotype of rs3813008 (SLC5A2) exhibited the greatest improvement in eGFR (+7.2 mL ± 6.5, p = 0.004) and reductions in KIM-1 (−0.13 pg/mL ± 0.49, p < 0.0001) and NGAL (−6.1 pg/mL ± 15.4, p < 0.0001). Similarly, rs12917707 (UMOD) TT genotypes showed improved renal function. However, rs5219 (KCNJ11) showed no significant impact on renal outcomes. Conclusions: Pharmacogenetic variations influenced renal response to dapagliflozin in HFrEF patients, particularly in SLC5A2 and UMOD genes. These findings highlighted the potential of personalized medicine in optimizing therapy for HFrEF patients with renal complications. Full article
(This article belongs to the Section Clinical Pharmaceutics)
Show Figures

Figure 1

25 pages, 912 KiB  
Article
Association of SLCO1B3 and SLCO1B1 Polymorphisms with Methotrexate Efficacy and Toxicity in Saudi Rheumatoid Arthritis Patients
by Rania Magadmi, Ahlam M. Alharthi, Lina A. Alqurashi, Ibtisam M. Jali, Zeina W. Sharawi, Maha H. Jamal, Yasser Bawazir, Mohammad Mustafa, Sami M. Bahlas, Basma T. Jamal, Hassan Daghasi, Abdulrahman S. Altowairqi and Dalal Sameer Al Shaer
Pharmaceuticals 2025, 18(7), 1069; https://doi.org/10.3390/ph18071069 - 20 Jul 2025
Viewed by 372
Abstract
Background: Methotrexate (MTX) remains the most commonly prescribed drug used to treat rheumatoid arthritis (RA). Polymorphisms in solute carrier organic anion transporter family member 1B3 (SLCO1B3) and SLCO1B1 may play a critical role in MTX pharmacokinetics and patient outcomes. However, research [...] Read more.
Background: Methotrexate (MTX) remains the most commonly prescribed drug used to treat rheumatoid arthritis (RA). Polymorphisms in solute carrier organic anion transporter family member 1B3 (SLCO1B3) and SLCO1B1 may play a critical role in MTX pharmacokinetics and patient outcomes. However, research on these polymorphisms in Saudi Arabia remains limited. We evaluated the association of SLCO1B3 (rs4149117, rs7311358) and SLCO1B1 (rs2306283, rs4149056) polymorphisms with MTX efficacy and safety in Saudi patients with RA. Methods: This multicenter, case-control study included patients diagnosed with RA in Jeddah and Taif. Demographic and clinical data were collected and analyzed. Genotyping of SLCO1B3 (rs4149117, rs7311358) and SLCO1B1 (rs2306283, rs4149056) polymorphisms was performed using Sanger sequencing. Statistical analyses, including logistic regression and haplotype analysis, were conducted to evaluate associations between these polymorphisms, MTX efficacy, and toxicity. Results: The study cohort comprised 100 patients with RA, with 46 showing a good response to MTX and 54 showing a poor response. Clinical predictors of MTX response were significantly higher in patients with poor response. Both SLCO1B3 polymorphisms (rs4149117, rs7311358) were significantly associated with anemia. Significant associations were found between SLCO1B1 (rs2306283) and gastrointestinal disturbances and anemia. The GAAT haplotype was significantly more prevalent among good responders, while the TGGT haplotype was significantly associated with poor responders. Conclusions: These results highlight the importance of genetic testing in predicting MTX treatment outcomes and tailoring personalized treatment plans for patients with RA to improve efficacy and minimize adverse effects. Full article
Show Figures

Graphical abstract

20 pages, 763 KiB  
Review
Therapeutic Potential of Calcium Channel Blockers in Neuropsychiatric, Endocrine and Pain Disorders
by Aarish Manzar, Aleksandar Sic, Crystal Banh and Nebojsa Nick Knezevic
Cells 2025, 14(14), 1114; https://doi.org/10.3390/cells14141114 - 20 Jul 2025
Viewed by 738
Abstract
Calcium channel blockers (CCBs), originally developed for cardiovascular indications, have gained attention for their therapeutic potential in neuropsychiatric, endocrine, and pain-related disorders. In neuropsychiatry, nimodipine and isradipine, both L-type CCBs, show mood-stabilizing and neuroprotective effects, with possible benefits in depression, bipolar disorder, and [...] Read more.
Calcium channel blockers (CCBs), originally developed for cardiovascular indications, have gained attention for their therapeutic potential in neuropsychiatric, endocrine, and pain-related disorders. In neuropsychiatry, nimodipine and isradipine, both L-type CCBs, show mood-stabilizing and neuroprotective effects, with possible benefits in depression, bipolar disorder, and schizophrenia. In endocrinology, verapamil, a non-dihydropyridine L-type blocker, has been associated with the preservation of pancreatic β-cell function and reduced insulin dependence in diabetes. CCBs may also aid in managing primary aldosteronism and pheochromocytoma, particularly in patients with calcium signaling mutations. In pain medicine, α2δ ligands and selective blockers of N-type and T-type channels demonstrate efficacy in neuropathic and inflammatory pain. However, their broader use is limited by challenges in central nervous system (CNS) penetration, off-target effects, and heterogeneous trial outcomes. Future research should focus on pharmacogenetic stratification, novel delivery platforms, and combination strategies to optimize repurposing of CCBs across disciplines. Full article
Show Figures

Graphical abstract

16 pages, 670 KiB  
Systematic Review
Pharmacogenetic Implications for Antidepressant Therapy in Major Depression: A Systematic Review Covering 2019–2024
by Anna Fornaguera and Marta Miarons
J. Clin. Med. 2025, 14(14), 5102; https://doi.org/10.3390/jcm14145102 - 18 Jul 2025
Viewed by 550
Abstract
Background/Objectives: Major depressive disorder (MDD), including late-onset forms, is a prevalent and disabling condition. Despite multiple pharmacological treatment options, over half of patients fail to achieve full remission. This systematic review aims to assess current evidence on the influence of pharmacogenetic factors [...] Read more.
Background/Objectives: Major depressive disorder (MDD), including late-onset forms, is a prevalent and disabling condition. Despite multiple pharmacological treatment options, over half of patients fail to achieve full remission. This systematic review aims to assess current evidence on the influence of pharmacogenetic factors on antidepressant response and safety, with a focus on patients with major and late-life depression. Methods: We conducted a systematic review following PRISMA guidelines (PROSPERO: CRD42020212345). Studies published in the past five years involving adult patients with MDD or late-onset depression and pharmacogenetic data were included. Results: From 793 abstracts screened, 29 studies with 39,975 participants were included. CYP2C19 and CYP2D6 were the most frequently analyzed genes (41% and 17% of studies, respectively). Poor metabolizers for CYP2C19 showed higher plasma levels of SSRIs, leading to increased adverse effects. In contrast, ultrarapid metabolizers had significantly lower response rates. Variants in SLC6A4 and other genes (e.g., HTR2A, ABCB1) were also associated with treatment outcomes. Combinatorial pharmacogenetic testing showed superior predictive value compared to single-gene approaches. Conclusions: Genetic variants in CYP2C19, CYP2D6, and SLC6A4 may affect the efficacy and tolerability of antidepressant therapy. Integrating this information into clinical practice may allow more personalized prescribing and improved outcomes. Full article
Show Figures

Figure 1

16 pages, 1511 KiB  
Article
Genotype–Drug–Diet Interactions in Metabolic Regulation: CYP1A2 rs762551 Modulates the Effect of Caffeine on Lipid and Glucose Profiles in the Context of Pharmacotherapy
by Laura Claudia Popa, Ahmed Abu-Awwad, Simona Sorina Farcas, Simona-Alina Abu-Awwad and Nicoleta Ioana Andreescu
Nutrients 2025, 17(14), 2288; https://doi.org/10.3390/nu17142288 - 10 Jul 2025
Viewed by 744
Abstract
Background/Objectives: Inter-individual metabolic responses to caffeine are shaped by CYP1A2 clearance rate and by concurrent lipid- or glucose-lowering drugs. We investigated how habitual caffeine intake relates to serum cholesterol and fasting glucose under different CYP1A2 rs762551 genotypes and statin or oral antidiabetic [...] Read more.
Background/Objectives: Inter-individual metabolic responses to caffeine are shaped by CYP1A2 clearance rate and by concurrent lipid- or glucose-lowering drugs. We investigated how habitual caffeine intake relates to serum cholesterol and fasting glucose under different CYP1A2 rs762551 genotypes and statin or oral antidiabetic (OAD) use. Methods: A prospective cross-sectional analysis was performed on 358 adults (AA = 65, AC = 163, CC = 130) with recorded genotype, daily caffeine intake, total cholesterol, fasting glucose, and medication status. Multivariable linear regression tested the main and interaction effects of caffeine (mg day−1), genotype, and therapy. Results: Caffeine intake was positively associated with cholesterol levels (β = 0.30; p < 0.001). A significant genotype × caffeine interaction (β = 0.27; p < 0.001) revealed the steepest rise in fast metabolisers (AA) not on statins, an effect largely blunted by statin therapy. For glucose, the genotype × caffeine term was also significant (β = 0.30; p < 0.001). Among slow metabolisers (CC) without OADs, caffeine correlated positively with glycaemia (r = 0.34; p = 0.028), whereas in fast metabolisers on OADs the association reversed (r = −0.36; p = 0.015). No meaningful associations occurred in AC carriers. Conclusions: Caffeine’s metabolic impact depends jointly on CYP1A2 genotype and medication. It raises cholesterol in fast metabolisers lacking statins and elevates glucose in slow metabolisers without OADs, yet may lower glucose when rapid metabolisation coincides with antidiabetic therapy. Accounting for this gene–diet–drug interplay could refine caffeine guidance within precision nutrition frameworks. Full article
(This article belongs to the Section Nutrition and Metabolism)
Show Figures

Figure 1

20 pages, 636 KiB  
Opinion
Clinician Experiences at the Frontier of Pharmacogenomics and Future Directions
by Stefan Thottunkal, Claire Spahn, Benjamin Wang, Nidhi Rohatgi, Jison Hong, Abha Khandelwal and Latha Palaniappan
J. Pers. Med. 2025, 15(7), 294; https://doi.org/10.3390/jpm15070294 - 7 Jul 2025
Viewed by 1170
Abstract
Pharmacogenomics (PGx) has emerged as a powerful tool to personalize drug selection and dosing based on a patient’s genetic profile. However, there are a range of challenges that impede uptake in current clinical practice. For example, clinicians often express frustration with commercially available [...] Read more.
Pharmacogenomics (PGx) has emerged as a powerful tool to personalize drug selection and dosing based on a patient’s genetic profile. However, there are a range of challenges that impede uptake in current clinical practice. For example, clinicians often express frustration with commercially available PGx panel tests, which fail to consistently include all key actionable PGx genes (according to the Clinical Pharmacogenetics Implementation Consortium (CPIC), Food and Drug Administration (FDA) PGx guidelines, or The Dutch Pharmacogenetics Working Group (DPWG) guidelines) and instead are too long with clinically unimportant information (unvalidated genotypes). Additionally, the lack of EMR integration, clinician education and awareness of the benefits of PGx impedes uptake. This paper examines key challenges identified in clinical practice and proposes future directions, focusing on limiting PGx reports to essential data, providing point-of-prescription alerts, and establishing reimbursement pathways that encourage adoption. Future directions include leveraging large language models, integrating point-of-prescription alerts and phenoconversion calculators into the electronic medical record, increasing the genomic diversity of PGx study populations, and streamlining coverage by payers. Full article
(This article belongs to the Section Pharmacogenetics)
Show Figures

Figure 1

16 pages, 679 KiB  
Article
Pharmacogenetic Biomarkers of Ibrutinib Response and Toxicity in Chronic Lymphocytic Leukemia: Insights from an Observational Study
by Noelia Pérez-Gómez, Antonio Sanz-Solas, Beatriz Cuevas, María Victoria Cuevas, Cristina Alonso-Madrigal, Javier Loscertales, Rodolfo Álvarez-Nuño, Covadonga García, Pablo Zubiaur, Gonzalo Villapalos-García, Raúl Miguel Parra-Garcés, Gina Mejía-Abril, Raquel Alcaraz, Raquel Vinuesa, Francisco Javier Díaz-Gálvez, María González-Oter, Natalia García-Sancha, Raúl Azibeiro-Melchor, Tomás José González-López, Francisco Abad-Santos, Jorge Labrador and Miriam Saiz-Rodríguezadd Show full author list remove Hide full author list
Pharmaceuticals 2025, 18(7), 996; https://doi.org/10.3390/ph18070996 - 2 Jul 2025
Viewed by 425
Abstract
Background/Objectives: Ibrutinib is a selective Bruton’s tyrosine kinase inhibitor approved for the treatment of chronic lymphocytic leukemia (CLL). This drug exhibits significant variability in response and toxicity profile, possibly due to genetic polymorphisms in drug-metabolizing enzymes and transporters. The aim of this observational [...] Read more.
Background/Objectives: Ibrutinib is a selective Bruton’s tyrosine kinase inhibitor approved for the treatment of chronic lymphocytic leukemia (CLL). This drug exhibits significant variability in response and toxicity profile, possibly due to genetic polymorphisms in drug-metabolizing enzymes and transporters. The aim of this observational study is to address interindividual variability in the efficacy and safety of ibrutinib treatment in 49 CLL patients. Methods: Genotyping of nine polymorphisms was performed by quantitative polymerase chain reaction (qPCR) using a ViiA7® PCR Instrument and TaqMan assays, and ibrutinib plasma concentrations were determined using high-performance liquid chromatography coupled to a tandem mass spectrometry detector (HPLC-MS/MS). Results: Our study confirmed a high response rate, with 62% of patients achieving complete remission (CR), 9% CR with incomplete hematologic recovery (CRi), and 24% partial remission (PR). The impact of genetic polymorphisms on the CR rate was evaluated, revealing no statistically significant associations for CYP3A4, CYP3A5, ABCB1, ABCG2, and SLCO1B1 variants. However, a tendency was observed for patients carrying ABCB1 rs1128503, rs1045642 T/T, or rs2032582 A/A genotypes to achieve a higher CR rate. Adverse drug reactions (ADRs) were frequent, with vascular disorders (39%) and infections (27%) being the most common. Genetic polymorphisms influenced ibrutinib toxicity, with CYP3A4 *1/*22 appearing to be protective against overall ADRs. Conclusions: The unexpected association between CYP3A4 *1/*22 genotype and lower ADR incidence, as well as the trend toward improved treatment response in patients carrying ABCB1 genotypes, suggests compensatory metabolic mechanisms. However, given the small sample size, larger studies are needed to confirm these findings and their clinical implications, while also aiming to uncover other non-genetic factors that may contribute to a better understanding of the variability in treatment response and toxicity. Full article
(This article belongs to the Special Issue Therapeutic Drug Monitoring and Adverse Drug Reactions: 2nd Edition)
Show Figures

Figure 1

16 pages, 604 KiB  
Article
The Role of GST Gene Polymorphic Variants in Antipsychotic-Induced Metabolic Disorders in Schizophrenia: A Pilot Study
by Irina A. Mednova, Ekaterina V. Mikhalitskaya, Natalia M. Vyalova, Diana Z. Paderina, Dmitry A. Petkun, Vladimir V. Tiguntsev, Elena G. Kornetova, Nikolay A. Bokhan and Svetlana A. Ivanova
Pharmaceuticals 2025, 18(7), 941; https://doi.org/10.3390/ph18070941 - 21 Jun 2025
Viewed by 467
Abstract
The life expectancy of patients with psychotic disorders is significantly shorter than that of the general population; antipsychotic-induced metabolic disorders play a significant role in reducing life expectancy. Both metabolic syndrome (MetS) and schizophrenia are multifactorial conditions. One area where the two conditions [...] Read more.
The life expectancy of patients with psychotic disorders is significantly shorter than that of the general population; antipsychotic-induced metabolic disorders play a significant role in reducing life expectancy. Both metabolic syndrome (MetS) and schizophrenia are multifactorial conditions. One area where the two conditions overlap is oxidative stress, which is present in both diseases. The glutathione-S-transferase (GST) system is a major line of defense against exogenous toxicants and oxidative damage to cells. The aim of our study was to perform an association analysis of gene polymorphisms with metabolic disorders in patients with schizophrenia treated with antipsychotic therapy. Methods: A total of 639 white patients with schizophrenia (ICD-10) from Siberia (Russia) were included in the study. Genotyping was carried out using real-time polymerase chain reaction for two single-nucleotide polymorphisms (SNPs) in the GSTP1 (rs614080 and rs1695) and one SNP in the GSTO1 (rs49252). Results: We found that rs1695*GG genotype of GSTP1 is a risk factor for the development of overweight (OR 2.36; 95% CI: 1.3–4.29; p = 0.0054). In the subgroup of patients receiving first-generation antipsychotics as basic therapy, the risk of overweight was associated with carriage of the rs1695*GG (OR 5.43; 95% CI: 2.24–13.16; p < 0.001) genotype of GSTP1 in a recessive model of inheritance. In contrast, an association of rs1695*G GSTP1 with obesity (OR: 0.42; 95% CI: 0.20–0.87; p = 0.018) was shown in the dominant model of inheritance in patients receiving second-generation antipsychotics. Conclusions: The pilot results obtained confirm the hypothesis of a violation of the antioxidant status, in particular the involvement of GSTP1, in the development of antipsychotic-induced metabolic disorders in schizophrenia. Further studies with larger samples and different ethnic groups are needed to confirm the obtained results. Full article
(This article belongs to the Special Issue Therapeutic Drug Monitoring and Adverse Drug Reactions: 2nd Edition)
Show Figures

Figure 1

42 pages, 643 KiB  
Review
Systematic Review of Pharmacogenetics of Immunosuppressants in Heart Transplantation
by Juan Eduardo Megías-Vericat, Tomás Palanques-Pastor, Mireya Fernández-Sánchez, Eduardo Guerrero-Hurtado, Mayte Gil-Candel, Antonio Solana-Altabella, Octavio Ballesta-López, María Centelles-Oria, Javier García-Pellicer and José Luis Poveda-Andrés
Cardiogenetics 2025, 15(2), 18; https://doi.org/10.3390/cardiogenetics15020018 - 17 Jun 2025
Viewed by 454
Abstract
The standard immunosuppressive treatments in heart transplantation are calcineurin inhibitors, corticosteroids, and antimetabolite agents or inhibitors of the mammalian target of rapamycin. Pharmacogenetic studies show the impact on clinical course of genetic variability in genes that encode transporters, metabolizers, or molecular targets of [...] Read more.
The standard immunosuppressive treatments in heart transplantation are calcineurin inhibitors, corticosteroids, and antimetabolite agents or inhibitors of the mammalian target of rapamycin. Pharmacogenetic studies show the impact on clinical course of genetic variability in genes that encode transporters, metabolizers, or molecular targets of immunosuppressants. The aim of this systematic review is to elucidate the role that pharmacogenetics of immunosuppressant drugs plays in clinical outcomes upon heart transplantation. PubMed, EMBASE, the Cochrane Central Register, and the Database of Abstracts of Reviews of Effects were searched without restrictions. The 64 studies analyzed followed these criteria: (1) were based on clinical data on heart transplantation patients; (2) analyzed the associations between polymorphisms and clinical response; (3) analyzed the impact of polymorphisms on immunosuppressant safety. CYP3A4/5 variants were associated with higher doses of tacrolimus, whereas POR*28 variants with lower doses—ABCB1, ABCC2, SLCO1B1, and SLC13A1—contribute to interindividual variability in drug absorption, distribution, and toxicity. An ABCC2 polymorphism (rs717620) was related to higher risk of graft rejection in pediatrics. Variations in HLA-G, TNF-α and TGF-β genes influence transplant rejection risk and immune response. Implementing pharmacogenetic screening of polymorphisms could enhance therapeutic outcomes by improving drug efficacy, reducing toxicity, and ultimately increasing heart graft survival rates. Strong evidence supports genotyping for CYP3A5 and TPMT, but further research is required for transporter genes and cytokine polymorphisms. Full article
(This article belongs to the Section Molecular Genetics)
Show Figures

Figure 1

Back to TopTop