Module 2

Module 2

Course description

This course will introduce to learners the concepts of pharmacogenetics and pharmacogenomics, and provide a brief history of pharmacogenomics and personalized medicine. It will cover examples of pharmacogenomic polymorphisms relevant to drug metabolism, drug transport, drug targets and immune complexes, and differentiate between somatic and germline variants in diseases such as cancer. Methods and ethical issues in pharmacogenomics will be discussed in addition to the pharmacogenomics of special diseases such as cystic fibrosis. The course will help learners understand pharmacogenomics data to potentially choose the appropriate drug or dose for a certain patient based on pharmacogenomic testing.

Intended Learning outcomes (ILOs)

By the end of the course the learner will be able to:

1.  Recognize how the principles of human genetics are employed to optimize drug therapy and patient care.

2.  Identify the genetic basis of variability in drug response and how it contributes to drug efficacy and toxicity, adverse drug reactions, and drug-drug interaction.

3.  Explain how gene variants can affect the pharmacokinetics and/or pharmacodynamics of drugs.

4. Analyse pharmacogenomic test results from different genotyping methods.

5. Explain pharmacogenomic concepts to a particular drug therapy to solve relevant problems in healthcare.

6. Illustrate legal and ethical issues of pharmacogenomic testing.

7. Outline social and economic issues of implementation of pharmacogenetic in health care

8. Identify the key components and steps required for successful implementation of pharmacogenomics in clinical practice.

9. Recognize emerging advances in the pharmacogenomics field.

 

Course description

This course covers the principles of individualization of drug therapy in different clinical setting, drug use in renal patients, special populations and during idiosyncratic reactions. Pharmacogenetics, drug-drug interactions and adverse drug reactions are covered in this context.

Intended Learning outcomes (ILOs)

At the end of this course the student will be able to:

  1. Outline clinically relevant diversity in age, sex and genetic related variations that affects drug response.
  2. Recognize basic principles of dose adjustment and factors contributing to individual variability in different clinical situations
  3. Describe different types of drug-drug interactions and their impacts on patient safety issues
  4. Describe the rationale for proper drug choice in kidney failure
  5. Outline important drugs -induced hypersensitivity situations and the impact of pharmacogenetic testing
  6. Define pharmacovigilance and the importance of drug information centers
  7. Underline Pharmacogenetic issues related to drugs according to CPIC recommendations
  8. Analyze data and problems related to genetic variability and its impact on drug dosing and side effects.
  9. Explore special population-related drug use adjustment.
  10. Explain the importance of pharmacogenomics for individual variation in adverse drug reactions.
  11. Explain types of adverse drug reactions, why they occur, and their association with poor medication compliance
  12. Analyze inter-individual differences or changes in pharmacokinetics parameters due to genetics, sex and cardiac, renal or hepatic function
  13. Categorize the seriousness, expectedness, and causality of adverse events
  14. Apply pharmacovigilance concepts and procedures in the drug information centers
  15. Distinguish various types of drugs-induced hypersensitivity and the importance of pharmacogenetic testing
  16. Evaluate research articles in adverse drug reactions and interactions issues
  17. Correlate population diversity data with altered drug response and adjust the dosage properly.
  18. Apply appropriate terms to describe different types of adverse effect.
  19. Discuss the history, principles, and regulatory framework for clinical drug safety
  20. Evaluate risk factors and possible dose adjustment of drugs-induced pro-arrhythmias
  21. Effectively communicate their understanding of research through presentation
  22. Develop professional documents pertinent to organizational needs (e.g., monographs, policy documents).
  23. Apply the information technology to remain updated with the recent advances in professional practices

Course description

This course focuses on the application of pharmacogenomics in clinical practice, and enhances the student’s capabilities to apply and implement the concepts of pharmacogenomics in real life settings. The candidates will learn to interpret pharmacogenomics tests and use the data to choose the appropriate drug or dose for a certain patient using clinically-based case scenarios.

Intended Learning outcomes (ILOs)

By the end of this course, the learners should be able to:

  1. Explain the relevance of analytical validity, clinical validity and clinical utility in pharmacogenomics.
  2. Find drug-specific pharmacogenomic clinical and prescription guidelines.
  3. Evaluate drug-specific pharmacogenomic clinical and prescription guidelines.
  4. Choose the appropriate drug or dose for a certain patient based on pharmacogenomic test results.
  5. Realize the role of ethnicity on applying pharmacogenomic algorithms.
  6. List examples of pharmacogenomic programs and tests that are applied clinically.
  7. Interpret pharmacogenomic laboratory tests, reports and clinical decision supports.
  8. Differentiate different genotyping platforms used for pharmacogenomic testing.
  9. Compare preemptive to reactive point of care genotyping.
  10. Recognize barriers and challenges of implementing pharmacogenomics in patient care.

 

Course description

The course describes the principles of human genetics. Thus, first, we will explain the essential components of inheritance, all the genetic components of the human genome, and human diseases from a genetic point of view.

Intended Learning outcomes (ILOs)

By the end of this course, the learners should be able to:

  1. Describe the basics of human genetics from DNA, chromosomes to genes.
  2. Outline the Human Genome structure and its functional elements.
  3. Recognize the principles of genetic associations with disease susceptibility.
  4. Explain some of the current genomics sequencing technologies and illustrate how these can be used in personalized medicine.
  5. Understand the relevance of applying genomic testing to the patients' health.
  6. Recognize how the chromosomes behave during meiosis and mitosis in terms of numbers and inheritance.
  7. Provide examples of drug therapy recommendations for specific human diseases.
  8. Categorize genetic variations according to their frequency and size
  9. Discuss the genome structure and function
  10. Discuss methods to determine the functional relevance of genetic variants
  11. Explain examples of high penetrance genes for genetic diseases
  12. Discuss examples of genetic disease risk factors
  13. Discuss examples of gene-environment interactions
  14. Explain ethical and legal issues of genetic testing.
  15. Use proper online tools to study the DNA structure and identify its shape.
  16. Demonstrate professional behavior and teamwork (when needed).
  17. Participate in the activities of individual and group learning