Our pharmacogenomics platform will focus on population pharmacogenomics for safety and dose optimization. Through sequencing and advanced algorithms, the dosage of drugs for patients is optimized to enhance drug safety. By analyzing the genomics of different populations, we can precisely determine biomarker-based administration guidelines and potential risks of adverse reactions.
Population pharmacogenomics is becoming a key discipline in today's rapidly developing medical field. Link genomics with drug safety to understand the responses of different groups to drugs and specify medication methods that align with efficacy and safety.
One major focus is to utilize genetic data from different populations to enhance drug safety and optimize dosage. By applying advanced sequencing and algorithms, researchers can discover complex gene-drug interactions and predict patients' responses to treatment.
A significant part of this work involves identifying genetic factors that increase the risk of adverse drug reactions (ADRs)—a serious healthcare issue contributing to illness, mortality, and high medical costs. Variations in genes related to drug metabolism, transport, or targets can alter how a drug behaves in the body, sometimes leading to severe side effects. By analyzing genetic data alongside real-world drug response records, we can pinpoint these risks and develop practical prevention strategies, such as pre-treatment genetic screening.
When it comes to optimizing drug doses, population pharmacogenomics helps determine the right dose for each patient based on their genetic profile. By using genetic data in pharmacokinetic and pharmacodynamic models, we can predict how a person will process and respond to a drug. This allows us to adjust the dose accordingly—maximizing the drug's benefits while reducing risks, especially for medications with a narrow therapeutic window.
Our Population Pharmacogenomics Service for Safety & Dosing Optimization Elevates Your Research with:
Population pharmacogenomics studies genetic variations among populations to predict drug responses. Through genomics of different populations, identify the population's response to drugs and optimize the dosage by taking genetic factors into account. This approach enhances the therapeutic effect, minimizes risks to the greatest extent, and ensures safer and more targeted drug treatment.
1. Whole Genome Sequencing (WGS)
2. Targeted Panel Sequencing
2. Polygenic Risk Scores (PRS) for Personalized Drug Therapy
3. Pathway and Network Analysis
Figure 1: How We Deliver This Solution: Population Pharmacogenomics for Safety & Dosing Optimization
We have established our key advantage - a comprehensive database that integrates global genetic data from different races, ages and regions. Integrate this information onto one platform to make our data analysis more efficient. For instance, we can obtain drug metabolism data by analyzing the genomics of Asian and European populations with the same disease. This can provide everyone with more precise dosage recommendations and safer drug use.
We partner with leading academic and pharmaceutical institutions worldwide. These collaborations give us access to a wealth of resources-from specialized labs and top-tier research talent to diverse patient groups. By working together on joint projects, we combine our strengths to speed up the discovery of new pharmacogenomic markers and create more effective dosing guidelines.
Traditional drug safety risk assessment relies on clinical evaluation and has individual differences. Population pharmacogenomics services employ advanced sequencing analysis and statistical models to evaluate large-scale genomic data of different populations. In cardiology, we can identify individuals at high risk of arrhythmia when taking certain medications based on specific genetic markers.
Patient responses to drugs can change over time due to various factors, including genetic evolution, disease progression, and environmental influences. Our service incorporates dynamic monitoring of genetic and clinical data. By continuously collecting and analyzing samples from patients during treatment, we can detect changes in genetic expression or the emergence of new genetic variants that may impact drug response. Based on these real - time insights, we can recommend adaptive dosing strategies. For example, in the treatment of chronic viral infections, we can adjust the dose of antiviral drugs according to the changes in the viral genetic sequence and the patient's immune - related genetic markers, ensuring sustained treatment effectiveness.
Our approach brings together multiple layers of biological data—not just genetics, but also gene expression (transcriptomics), protein levels (proteomics), and metabolic profiles (metabolomics). This helps us fill in the gaps between a person's genes and their actual response to a drug. For example, in the study of neurodegenerative diseases, integrating genomic and proteomic data can help us identify novel biomarkers and therapeutic targets related to drug efficacy and safety. This multi-omics approach provides a more accurate and in - depth basis for optimizing drug dosing and ensuring patient safety.
Figure 2: Relative predicted PGx medication exposure for underrepresented racial/ethnic populations with chronic health conditions compared to Whites, 2014–2021. (Saulsberry, 2025)
Frequency and Implications of High-Risk Pharmacogenomic Phenotypes Identified in a Diverse Australian Pediatric Oncology Cohort.
Journal:Clin Transl Sci.
Published:2025
Pediatric oncology patients have seen improved survival outcomes, with 5-year overall survival rates exceeding 80%. However, treatment-related toxicities remain severe, debilitating, and even life-threatening. Although pharmacogenomics (PGx) can help predict, prevent, and treat these toxicities, as well as improve medication efficacy, it is widely underutilized in clinical pediatric oncology settings. Despite the existence of international, evidence-based PGx guidelines from organizations like CPIC and DPWG, only one guideline (TPMT/NUDT15 for thiopurines in lymphoblastic leukemia) has been incorporated into pediatric standard of care in Australia. Other available guidelines for commonly used medications in pediatric oncology supportive care have not been implemented.
This study aims to determine the prevalence of high-risk PGx phenotypes in an Australian pediatric oncology cohort of 180 patients. By identifying the most common high-risk phenotypes, the research provides insights into which genes are most important to assess and which medications are most frequently affected. This information can guide personalized treatment strategies in Australia, helping to prioritize PGx testing and implement relevant guidelines to improve both the safety and efficacy of medications used in pediatric oncology.
The study found that the highest frequency of high-risk phenotypes was for CYP2C19, with nearly a third of patients being poor/intermediate metabolizers and another third being rapid/ultrarapid metabolizers. UGT1A1 had approximately 40% of patients as intermediate metabolizers, while almost half of the patients had a high-risk phenotype for CYP2D6 (mostly intermediate/poor metabolizers). Reduced ABCG2 function was identified in a quarter of the cohort, and no high-risk phenotypes were found for MT-RNR1 or G6PD. Overall, more than 90% of patients had at least one high-risk phenotype, with 20% carrying four or more and only 6.7% having none. These findings highlight the potential of PGx to significantly improve treatment outcomes in pediatric oncology by enabling personalized medication prescribing based on genetic information.
Figure 3: Proportion of high-risk phenotypes by gene (A) and total number of high-risk phenotypes (B).
A: By analyzing genetic data from large populations, we can identify specific genetic markers that are associated with an increased risk of adverse drug reactions. For example, certain genetic variants in drug - metabolizing enzymes can lead to the accumulation of toxic drug metabolites in the body. Once these markers are identified, healthcare providers can screen patients before prescribing medications. Patients with high - risk genetic profiles can be given alternative drugs or have their dosages adjusted to prevent potential safety issues, thus significantly reducing the incidence of adverse drug events.
References