Advances in Clinical Pharmacogenetics

A special issue of Pharmaceutics (ISSN 1999-4923). This special issue belongs to the section "Clinical Pharmaceutics".

Deadline for manuscript submissions: closed (30 April 2023) | Viewed by 17308

Special Issue Editors


E-Mail Website
Guest Editor
Human Genotyping Unit, CeGen (Spanish National Genotyping Centre), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Alamagro, 3, 28029 Madrid, Spain
Interests: pharmacogenetics; pharmacogenetics clinical implementation; cancer genomics; genetic susceptibility factors

E-Mail Website
Guest Editor
Human Genotyping Unit, CeGen (Spanish National Genotyping Centre), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Alamagro, 3, 28029 Madrid, Spain
Interests: genetic susceptibility factors; precision medicine; pharmacogenomics; cancer genomics

Special Issue Information

Dear Colleagues,

We are pleased to invite you to contribute to this Special Issue of Phamaceutics (2020 impact factor: 6.321) entitled “Advances in Clinical Pharmacogenetics”.

Pharmacogenetics aims to optimize the selection of drugs and correct doses to achieve a greater safety and effectiveness of therapies through the use of patients’ genetic data. The increased understanding of the effect of genetic variants in drug responses has allowed implementing pharmacogenetics in the clinical setting, showing the benefits of personalized medicine.

This Special Issue aims to collect articles to increase the knowledge of any field in the pharmacogenetics area and its implementation in the clinical setting.

In this Special Issue, original research articles and reviews are welcome. Research areas may include (but are not limited to) the following: pharmacogenetics, clinical implementation of pharmacogenetics, identification of genetic factors associated with drug responses, data analysis in the clinical setting, barriers and opportunities, population diversity in pharmacogenetics.

We look forward to receiving your contributions.

Dr. Rocio Nuñez-Torres
Dr. Anna González-Neira
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Pharmaceutics is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • pharmacogenetics
  • clinical implementation
  • precision medicine
  • personalized medicine
  • adverse drug reactions
  • genetic susceptibility factors
  • pharmacogenetic cost effectiveness
  • pharmacogenetic diagnostic techniques
  • genomics
  • biomarkers
  • data analysis and interpretation
  • artificial intelligence
  • bioinformatic tools
  • population diversity in pharmacogenetics

Published Papers (10 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

14 pages, 2338 KiB  
Article
Race, Ethnicity, and Pharmacogenomic Variation in the United States and the United Kingdom
by Shivam Sharma, Leonardo Mariño-Ramírez and I. King Jordan
Pharmaceutics 2023, 15(7), 1923; https://doi.org/10.3390/pharmaceutics15071923 - 11 Jul 2023
Cited by 3 | Viewed by 1684
Abstract
The relevance of race and ethnicity to genetics and medicine has long been a matter of debate. An emerging consensus holds that race and ethnicity are social constructs and thus poor proxies for genetic diversity. The goal of this study was to evaluate [...] Read more.
The relevance of race and ethnicity to genetics and medicine has long been a matter of debate. An emerging consensus holds that race and ethnicity are social constructs and thus poor proxies for genetic diversity. The goal of this study was to evaluate the relationship between race, ethnicity, and clinically relevant pharmacogenomic variation in cosmopolitan populations. We studied racially and ethnically diverse cohorts of 65,120 participants from the United States All of Us Research Program (All of Us) and 31,396 participants from the United Kingdom Biobank (UKB). Genome-wide patterns of pharmacogenomic variation—6311 drug response-associated variants for All of Us and 5966 variants for UKB—were analyzed with machine learning classifiers to predict participants’ self-identified race and ethnicity. Pharmacogenomic variation predicts race/ethnicity with averages of 92.1% accuracy for All of Us and 94.3% accuracy for UKB. Group-specific prediction accuracies range from 99.0% for the White group in UKB to 92.9% for the Hispanic group in All of Us. Prediction accuracies are substantially lower for individuals who identified with more than one group in All of Us (16.7%) or as Mixed in UKB (70.7%). There are numerous individual pharmacogenomic variants with large allele frequency differences between race/ethnicity groups in both cohorts. Frequency differences for toxicity-associated variants predict hundreds of adverse drug reactions per 1000 treated participants for minority groups in All of Us. Our results indicate that race and ethnicity can be used to stratify pharmacogenomic risk in the US and UK populations and should not be discounted when making treatment decisions. We resolve the contradiction between the results reported here and the orthodoxy of race and ethnicity as non-genetic, social constructs by emphasizing the distinction between global and local patterns of human genetic diversity, and we stress the current and future limitations of race and ethnicity as proxies for pharmacogenomic variation. Full article
(This article belongs to the Special Issue Advances in Clinical Pharmacogenetics)
Show Figures

Figure 1

19 pages, 2458 KiB  
Article
A Comprehensive Analysis of 21 Actionable Pharmacogenes in the Spanish Population: From Genetic Characterisation to Clinical Impact
by Rocio Nunez-Torres, Guillermo Pita, María Peña-Chilet, Daniel López-López, Jorge Zamora, Gema Roldán, Belén Herráez, Nuria Álvarez, María Rosario Alonso, Joaquín Dopazo and Anna Gonzalez-Neira
Pharmaceutics 2023, 15(4), 1286; https://doi.org/10.3390/pharmaceutics15041286 - 19 Apr 2023
Cited by 1 | Viewed by 2252
Abstract
The implementation of pharmacogenetics (PGx) is a main milestones of precision medicine nowadays in order to achieve safer and more effective therapies. Nevertheless, the implementation of PGx diagnostics is extremely slow and unequal worldwide, in part due to a lack of ethnic PGx [...] Read more.
The implementation of pharmacogenetics (PGx) is a main milestones of precision medicine nowadays in order to achieve safer and more effective therapies. Nevertheless, the implementation of PGx diagnostics is extremely slow and unequal worldwide, in part due to a lack of ethnic PGx information. We analysed genetic data from 3006 Spanish individuals obtained by different high-throughput (HT) techniques. Allele frequencies were determined in our population for the main 21 actionable PGx genes associated with therapeutical changes. We found that 98% of the Spanish population harbours at least one allele associated with a therapeutical change and, thus, there would be a need for a therapeutical change in a mean of 3.31 of the 64 associated drugs. We also identified 326 putative deleterious variants that were not previously related with PGx in 18 out of the 21 main PGx genes evaluated and a total of 7122 putative deleterious variants for the 1045 PGx genes described. Additionally, we performed a comparison of the main HT diagnostic techniques, revealing that after whole genome sequencing, genotyping with the PGx HT array is the most suitable solution for PGx diagnostics. Finally, all this information was integrated in the Collaborative Spanish Variant Server to be available to and updated by the scientific community. Full article
(This article belongs to the Special Issue Advances in Clinical Pharmacogenetics)
Show Figures

Graphical abstract

11 pages, 280 KiB  
Article
Abigene, a Prospective, Multicentric Study of Abiraterone Acetate Pharmacogenetics in Metastatic Castration-Resistant Prostate Cancer
by Jean-Marc Ferrero, Hakim Mahammedi, Gwenaelle Gravis, Guilhem Roubaud, Philippe Beuzeboc, Remi Largillier, Delphine Borchiellini, Claude Linassier, Nathalie Ebran, Tanguy Pace-Loscos, Marie-Christine Etienne-Grimaldi, Renaud Schiappa, Jocelyn Gal and Gérard Milano
Pharmaceutics 2023, 15(2), 651; https://doi.org/10.3390/pharmaceutics15020651 - 15 Feb 2023
Cited by 1 | Viewed by 1298
Abstract
Abiraterone acetate (AA) is the first-in-class of drugs belonging to the second-generation of agents inhibiting androgen neosynthesis in advanced prostate cancer. A cumulative experience attests that germinal gene polymorphisms may play a role in the prediction of anticancer agent pharmacodynamics variability. In the [...] Read more.
Abiraterone acetate (AA) is the first-in-class of drugs belonging to the second-generation of agents inhibiting androgen neosynthesis in advanced prostate cancer. A cumulative experience attests that germinal gene polymorphisms may play a role in the prediction of anticancer agent pharmacodynamics variability. In the present prospective, multicentric study, gene polymorphisms of CYP17A1 (AA direct target) and the androgen transporter genes SLCO2B1 and SLCO1B3 (potential modulators of AA activity) were confronted with AA pharmacodynamics (treatment response and toxicity) in a group of 137 advanced prostate cancer patients treated in the first line by AA. The median follow-up was 56.3 months (95% CI [52.5–61]). From multivariate analysis, rs2486758 C/C (CYP17A1) and PSA (≥10 ng/mL) were associated with a shorter 3-year biological PFS (HR = 4.05, IC95% [1.46–11.22]; p = 0.007 and HR = 2.08, IC95% [1.31–3.30]; p = 0.002, respectively). From a multivariate analysis, the rs743572 (CYP17A1) and performance status were independently associated with significant toxicity (OR = 3.78 (IC95% [1.42–9.75]; p = 0.006 and OR = 4.54; IC95% [1.46–13.61]; p = 0.007, respectively). Host genome characteristics may help to predict AA treatment efficacy and identify patients at risk for toxicity. Full article
(This article belongs to the Special Issue Advances in Clinical Pharmacogenetics)
14 pages, 1338 KiB  
Article
Relevance of CYP2D6 Gene Variants in Population Genetic Differentiation
by Anita Stojanović Marković, Matea Zajc Petranović, Tatjana Škarić-Jurić, Željka Celinšćak, Maja Šetinc, Željka Tomas and Marijana Peričić Salihović
Pharmaceutics 2022, 14(11), 2481; https://doi.org/10.3390/pharmaceutics14112481 - 16 Nov 2022
Cited by 1 | Viewed by 1474
Abstract
A significant portion of the variability in complex features, such as drug response, is likely caused by human genetic diversity. One of the highly polymorphic pharmacogenes is CYP2D6, encoding an enzyme involved in the metabolism of about 25% of commonly prescribed drugs. [...] Read more.
A significant portion of the variability in complex features, such as drug response, is likely caused by human genetic diversity. One of the highly polymorphic pharmacogenes is CYP2D6, encoding an enzyme involved in the metabolism of about 25% of commonly prescribed drugs. In a directed search of the 1000 Genomes Phase III variation data, 86 single nucleotide polymorphisms (SNPs) in the CYP2D6 gene were extracted from the genotypes of 2504 individuals from 26 populations, and then used to reconstruct haplotypes. Analyses were performed using Haploview, Phase, and Arlequin softwares. Haplotype and nucleotide diversity were high in all populations, but highest in populations of African ancestry. Pairwise FST showed significant results for eleven SNPs, six of which were characteristic of African populations, while four SNPs were most common in East Asian populations. A principal component analysis of CYP2D6 haplotypes showed that African populations form one cluster, Asian populations form another cluster with East and South Asian populations separated, while European populations form the third cluster. Linkage disequilibrium showed that all African populations have three or more haplotype blocks within the CYP2D6 gene, while other world populations have one, except for Chinese Dai and Punjabi in Pakistan populations, which have two. Full article
(This article belongs to the Special Issue Advances in Clinical Pharmacogenetics)
Show Figures

Figure 1

12 pages, 1105 KiB  
Article
An Investigation of O-Demethyl Tramadol/Tramadol Ratio for Cytochrome P450 2D6 Phenotyping: The CYTRAM Study
by Blandine De La Gastine, Soizic Percevault, Laurent Varin, Nicolas Richard, Fabienne Fobe, Benoît Plaud, Georges Daccache, Vincent Compere, Jean-Jacques Parienti, Antoine Coquerel, Magalie Loilier, Nathalie Bleyzac, Laurent Bourguignon, Sylvain Goutelle and Véronique Lelong-Boulouard
Pharmaceutics 2022, 14(10), 2177; https://doi.org/10.3390/pharmaceutics14102177 - 12 Oct 2022
Viewed by 1335
Abstract
Cytochrome P450 2D6 (CYP2D6) gene polymorphisms influence the exposure to tramadol (T) and its pharmacologically active metabolite, O-demethyl tramadol (O-dT). Tramadol has been considered as a candidate probe drug for CYP2D6 phenotyping. The objective of the CYTRAM study was to investigate [...] Read more.
Cytochrome P450 2D6 (CYP2D6) gene polymorphisms influence the exposure to tramadol (T) and its pharmacologically active metabolite, O-demethyl tramadol (O-dT). Tramadol has been considered as a candidate probe drug for CYP2D6 phenotyping. The objective of the CYTRAM study was to investigate the value of plasma O-dT/T ratio for CYP2D6 phenotyping. European adult patients who received IV tramadol after surgery were included. CYP2D6 genotyping was performed and subjects were classified as extensive (EM), intermediate (IM), poor (PM), or ultra-rapid (UM) CYP2D6 metabolizers. Plasma concentrations of tramadol and O-dT were determined at 24 h and 48 h. The relationship between O-dT/T ratio and CYP2D6 phenotype was examined in both a learning and a validation group. Genotype data were obtained in 301 patients, including 23 PM (8%), 117 IM (39%), 154 EM (51%), and 7 UM (2%). Tramadol trough concentrations at 24 h were available in 297 patients. Mean value of O-dT/T ratio was significantly lower in PM than in non-PM individuals (0.061 ± 0.031 versus 0.178 ± 0.09, p < 0.01). However, large overlap was observed in the distributions of O-dT/T ratio between groups. Statistical models based on O-dT/T ratio failed to identify CYP2D6 phenotype with acceptable sensitivity and specificity. Those results suggest that tramadol is not an adequate probe drug for CYP2D6 phenotyping. Full article
(This article belongs to the Special Issue Advances in Clinical Pharmacogenetics)
Show Figures

Graphical abstract

8 pages, 262 KiB  
Article
Genotyping of UGT1A1*80 as an Alternative to UGT1A1*28 Genotyping in Spain
by Adrián Bravo-Gómez, Sara Salvador-Martín, Paula Zapata-Cobo, María Sanjurjo-Sáez and Luis Andrés López-Fernández
Pharmaceutics 2022, 14(10), 2082; https://doi.org/10.3390/pharmaceutics14102082 - 29 Sep 2022
Cited by 2 | Viewed by 1878
Abstract
Background: The variant rs34983651 (UGT1A1*28) and its genotyping are used to prevent irinotecan-induced toxicity. Several variants are in close linkage disequilibrium. Our objective was to evaluate the potential correlation of genotyping UGT1A1*80 instead of UGT1A1*28 in different populations. Methods: [...] Read more.
Background: The variant rs34983651 (UGT1A1*28) and its genotyping are used to prevent irinotecan-induced toxicity. Several variants are in close linkage disequilibrium. Our objective was to evaluate the potential correlation of genotyping UGT1A1*80 instead of UGT1A1*28 in different populations. Methods: We studied SNPs in linkage disequilibrium with UGT1A1*28 in several populations and selected rs887829 to develop an inexpensive and rapid genotyping method and compare it with the one we currently use for UGT1A1*28 genotyping. Samples from cancer patients (n = 701) already tested using PCR and electrophoresis prior to treatment with irinotecan for rs34983651 (UGT1A1*28) in a Spanish hospital were genotyped for rs887829 (UGT1A1*80) using real-time PCR with a TaqMan probe. Results: We observed a complete match for both genotypes, except in one sample. This method was 100% efficient in correctly genotyping *28/*28 patients, 99.68% efficient for *1/*28, and 100% efficient for *1/*1. Linkage disequilibrium between populations showed the Iberian population to be the most suitable for the clinical use of UGT1A1*80. This method is less expensive and the time to decision is shorter. Conclusion: Genotyping of rs887829 using the proposed method may be used to substitute genotyping of rs34983651 as a pharmacogenetics test in cancer patients prior to starting irinotecan-based treatments, mainly in the Iberian population. In addition, it is less expensive than other conventional methods and easy to implement, with a shorter time to decision than UGT1A1*28. Full article
(This article belongs to the Special Issue Advances in Clinical Pharmacogenetics)
Show Figures

Graphical abstract

12 pages, 500 KiB  
Article
Patient Perceptions and Potential Utility of Pharmacogenetic Testing in Chronic Pain Management and Opioid Use Disorder in the Camden Opioid Research Initiative
by Dara Kusic, Jessica Heil, Stefan Zajic, Andrew Brangan, Oluseun Dairo, Gretchen Smith, Diego Morales-Scheihing, Russell J. Buono, Thomas N. Ferraro, Rachel Haroz, Matthew Salzman, Kaitlan Baston, Elliot Bodofsky, Michael Sabia, Alissa Resch and Laura B. Scheinfeldt
Pharmaceutics 2022, 14(9), 1863; https://doi.org/10.3390/pharmaceutics14091863 - 03 Sep 2022
Cited by 1 | Viewed by 1792
Abstract
Pharmacogenetics (PGx) has the potential to improve opioid medication management. Here, we present patient perception data, pharmacogenetic data and medication management trends in patients with chronic pain (arm 1) and opioid use disorder (arm 2) treated at Cooper University Health Care in Camden [...] Read more.
Pharmacogenetics (PGx) has the potential to improve opioid medication management. Here, we present patient perception data, pharmacogenetic data and medication management trends in patients with chronic pain (arm 1) and opioid use disorder (arm 2) treated at Cooper University Health Care in Camden City, NJ. Our results demonstrate that the majority of patients in both arms of the study (55% and 65%, respectively) are open to pharmacogenetic testing, and most (66% and 69%, respectively) believe that genetic testing has the potential to improve their medical care. Our results further support the potential for CYP2D6 PGx testing to inform chronic pain medication management for poor metabolizers (PMs) and ultrarapid metabolizers (UMs). Future efforts to implement PGx testing in chronic pain management, however, must address patient concerns about genetic test result access and genetic discrimination. Full article
(This article belongs to the Special Issue Advances in Clinical Pharmacogenetics)
Show Figures

Figure 1

15 pages, 454 KiB  
Article
Genetic Polymorphisms in VEGFR Coding Genes (FLT1/KDR) on Ranibizumab Response in High Myopia and Choroidal Neovascularization Patients
by David Blánquez-Martínez, Xando Díaz-Villamarín, Sonia García-Rodríguez, Alba Antúnez-Rodríguez, Ana Pozo-Agundo, Luis Javier Martínez-González, José Ignacio Muñoz-Ávila and Cristina Lucía Dávila-Fajardo
Pharmaceutics 2022, 14(8), 1555; https://doi.org/10.3390/pharmaceutics14081555 - 26 Jul 2022
Cited by 2 | Viewed by 1764
Abstract
A severe form of myopia defined as pathologic/high myopia is the main cause of visual impairment and one of the most frequent causes of blindness worldwide. It is characterized by at least 6 diopters or axial length (AL) of eyeball > 26 mm [...] Read more.
A severe form of myopia defined as pathologic/high myopia is the main cause of visual impairment and one of the most frequent causes of blindness worldwide. It is characterized by at least 6 diopters or axial length (AL) of eyeball > 26 mm and choroidal neovascularization (CNV) in 5 to 10% of cases. Ranibizumab is a humanized recombinant monoclonal antibody fragment targeted against human vascular endothelial growth factor A (VEGF-A) used in the treatment of CNV. It acts by preventing VEGF-A from interacting with its receptors (VEGFR-1 and -2) encoded by the FLT1 and KDR genes. Several studies found that the KDR and FLT1 genotypes may represent predictive determinants of efficacy in ranibizumab-treated neovascular age-related macular degeneration (nAMD) patients. We performed a retrospective study to evaluate the association of single nucleotide polymorphisms (SNPs) in VEGFR coding genes with the response rate to ranibizumab in patients with high myopia and CNV. In the association study of genotypes in FLT1 with the response to ranibizumab, we found a significant association between two FLT1 variants (rs9582036, rs7993418) with ranibizumab efficacy at the 12-month follow-up. About the KDR gene, we found that two KDR variants (rs2305948, rs2071559) are associated with best-corrected visual acuity (BCVA) improvement and KDR (rs2239702) is associated with lower rates of BCVA worsening considering a 12-month follow-up period. Full article
(This article belongs to the Special Issue Advances in Clinical Pharmacogenetics)
Show Figures

Figure 1

Review

Jump to: Research

23 pages, 569 KiB  
Review
Clinical Utility and Implementation of Pharmacogenomics for the Personalisation of Antipsychotic Treatments
by Marta Hernandez, Natalia Cullell, Marc Cendros, Alexandre Serra-Llovich and Maria J. Arranz
Pharmaceutics 2024, 16(2), 244; https://doi.org/10.3390/pharmaceutics16020244 - 07 Feb 2024
Viewed by 994
Abstract
Decades of pharmacogenetic research have revealed genetic biomarkers of clinical response to antipsychotics. Genetic variants in antipsychotic targets, dopamine and serotonin receptors in particular, and in metabolic enzymes have been associated with the efficacy and toxicity of antipsychotic treatments. However, genetic prediction of [...] Read more.
Decades of pharmacogenetic research have revealed genetic biomarkers of clinical response to antipsychotics. Genetic variants in antipsychotic targets, dopamine and serotonin receptors in particular, and in metabolic enzymes have been associated with the efficacy and toxicity of antipsychotic treatments. However, genetic prediction of antipsychotic response based on these biomarkers is far from accurate. Despite the clinical validity of these findings, the clinical utility remains unclear. Nevertheless, genetic information on CYP metabolic enzymes responsible for the biotransformation of most commercially available antipsychotics has proven to be effective for the personalisation of clinical dosing, resulting in a reduction of induced side effects and in an increase in efficacy. However, pharmacogenetic information is rarely used in psychiatric settings as a prescription aid. Lack of studies on cost-effectiveness, absence of clinical guidelines based on pharmacogenetic biomarkers for several commonly used antipsychotics, the cost of genetic testing and the delay in results delivery hamper the implementation of pharmacogenetic interventions in clinical settings. This narrative review will comment on the existing pharmacogenetic information, the clinical utility of pharmacogenetic findings, and their current and future implementations. Full article
(This article belongs to the Special Issue Advances in Clinical Pharmacogenetics)
Show Figures

Figure 1

20 pages, 2195 KiB  
Review
New Drug Development and Clinical Trial Design by Applying Genomic Information Management
by Young Kyung Ko and Jeong-An Gim
Pharmaceutics 2022, 14(8), 1539; https://doi.org/10.3390/pharmaceutics14081539 - 24 Jul 2022
Cited by 3 | Viewed by 2125
Abstract
Depending on the patients’ genotype, the same drug may have different efficacies or side effects. With the cost of genomic analysis decreasing and reliability of analysis methods improving, vast amount of genomic information has been made available. Several studies in pharmacology have been [...] Read more.
Depending on the patients’ genotype, the same drug may have different efficacies or side effects. With the cost of genomic analysis decreasing and reliability of analysis methods improving, vast amount of genomic information has been made available. Several studies in pharmacology have been based on genomic information to select the optimal drug, determine the dose, predict efficacy, and prevent side effects. This paper reviews the tissue specificity and genomic information of cancer. If the tissue specificity of cancer is low, cancer is induced in various organs based on a single gene mutation. Basket trials can be performed for carcinomas with low tissue specificity, confirming the efficacy of one drug for a single gene mutation in various carcinomas. Conversely, if the tissue specificity of cancer is high, cancer is induced in only one organ based on a single gene mutation. An umbrella trial can be performed for carcinomas with a high tissue specificity. Some drugs are effective for patients with a specific genotype. A companion diagnostic strategy that prescribes a specific drug for patients selected with a specific genotype is also reviewed. Genomic information is used in pharmacometrics to identify the relationship among pharmacokinetics, pharmacodynamics, and biomarkers of disease treatment effects. Utilizing genomic information, sophisticated clinical trials can be designed that will be better suited to the patients of specific genotypes. Genomic information also provides prospects for innovative drug development. Through proper genomic information management, factors relating to drug response and effects can be determined by selecting the appropriate data for analysis and by understanding the structure of the data. Selecting pre-processing and appropriate machine-learning libraries for use as machine-learning input features is also necessary. Professional curation of the output result is also required. Personalized medicine can be realized using a genome-based customized clinical trial design. Full article
(This article belongs to the Special Issue Advances in Clinical Pharmacogenetics)
Show Figures

Figure 1

Back to TopTop