Module 3

Module 3

Course description

This course focuses on the available analytical and bioinformatics high-throughput platforms, and its application to pharmacogenomics. In brief, the candidate will learn the analytical basis of omic procedures employed in pharmacogenomics, to process and mine the analytical data, and to transfer it to clinicians and pharmacists to personalize patient treatments. During the course candidates will also learn to design new omic analyses in drug discovery environments, and to apply state-of-the-art methodologies to phenotype patients under several pathologies.

Intended Learning outcomes (ILOs)

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

  1. Understand the analytical basis of most of the high-throughput methodologies employed in pharmacogenomics.
  2. Differentiate the different analytical platforms employed in omics assay.
  3. Choose the appropriate analytical platforms for specific pharmacogenomics procedures (genotyping, splicing variants, enzymatic assays, pharmacokinetic assays, …)
  4. Process raw data coming from transcriptomics, genomics, proteomics or metabolomics platforms.
  5. Design custom pharmacogenomic analytical procedures.
  6. Employ omics data to evaluate and personalize patient treatments.
  7. Realize the role of sex and ethnicity on drug-response and its importance when applying pharmacogenomic algorithms
  8. Create new links between hospital pharmacy and laboratory to provide precision treatments for individual patients

Course description

This course provides an overview of somatic and germline Pharmacogenetics in cancer patients. It illustrates pharmacogenomic-based diagnostics in oncology and pharmacogenomic factors that are associated with inter-individual differences in toxicity and therapeutic response of many chemotherapy agents. The course introduces different types of biomarkers that are associated with the response and toxicities of chemotherapeutics. Patient cases and clinical recommendations are also presented.

Intended Learning outcomes (ILOs)

By the end of the course, the student will be able to:

  1. Describe the impact of pharmacogenomics on optimizing cancer chemotherapy and reducing adverse drug effects and drug toxicities to improve therapeutic outcomes in cancer patients.
  2. Apply pharmacogenomics concepts to cancer therapy to solve relevant problems in healthcare.
  3. Discuss case studies reporting clinical consequences of pharmacogenomics chemotherapeutic agents efficacy.
  4. Evaluate relevant literature with respect to pharmacogenomics in genetic testing for inherited cancer susceptibility and its impact on patient care.
  5. Encourage the healthcare team collaboration regarding the dissemination of Pharmacogenomics concepts, patient education, and collaboration between healthcare providers.

 

Course description

This course offers a broad understanding of current knowledge on rare and complex diseases through a genomics lens. This clear understanding of the latest molecular and genomic technologies used to elucidate human disorders brings readers closer to unravelling many more that remain undiscovered. The challenges associated with performing such analysis and the opportunities to understand disease architecture and pathophysiology better. Specific areas are to be discussed including large scale sequencing project in rare diseases, comparative genomics, epigenome-wide analysis and proteomics.

Intended Learning outcomes (ILOs)

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

  1. Understand the importance of comparative genome sequencing projects to reveal evolution processes and help interpret regions in the human genome.
  2. Understand how next-generation sequencing and chromosomal microarray can be used to detect copy number variation in genetic testing of fetuses.
  3. Familiarize with the application of human epigenomic methods to understand the Health and Disease States.
  4. Highlight how the molecular diagnosis of Cystic fibrosis is being made
  5. Outline the different mechanisms by which mutations can affect human health.
  6. Discuss the genetic heterogeneity seen in inherited retinal diseases.
  7. Familiarize with several important diseases with genetic components, and knowing which ones can be treated and for which one’s lifestyle adjustments can reduce the danger inherent in genetic risk factors.
  8. Understand the fundamental chemical structure of proteins and common post-translational modifications.
  9. Understand polyacrylamide gel electrophoresis (PAGE) to separate proteins.
  10. Understand the technique and uses of mass spectrometry.
  11. Identify the molecular causes of the yet-to-be-discovered genetic disorders.
  12. Understand the pathophysiology of the myriad of rare diseases.
  13. Utilize the knowledge to provide better molecular diagnostics.
  14. Gain insight into the different types of Inherited Cardiac Conditions.
  15. Identify the key genetic determinants of Hypertrophic Cardiomyopathy.
  16. Explain the value of genetic testing and its implications on the patient and family members.
  17. Understand basic concepts of epigenetics and their relation to CVDs & their pharmaco-epigenomic treatment.
  18. Underline the advantages and disadvantages of the iPSCs model.
  19. Describe the translational applications of the iPSCs-derived cardiomyocytes.
  20. Examine the different epigenomic methods currently used.
  21. Discover the importance of comparative genomics in revealing conserved elements that are likely to have exciting functions.
  22. Interpret the Impact of CFTR Variants and the Challenge of Penetrance.
  23. Outline the coordination of changes in genotype and phenotype.
  24. Summarize the complexity of human biology.
  25. Develop the sense of the discipline of systems biology as an integrative approach to all the ‘omics’ disciplines.
  26. analyse how next-generation sequencing technologies have helped with the genetic heterogeneity, cost, and poorly defined genotype–phenotype correlation challenges that practitioners (especially physicians) face.
  27. Analyse how transcriptomic analyses help elucidating the mechanisms underlying human diseases that remained elusive through DNA analysis alone.
  28. Understand the practical procedure of karyotyping using Giemsa Stain.
  29. Solve the human karyotype though identifying the type of aneuploidy.
  30. Demonstrate professional behavior and teamwork (when needed).
  31. Participate in the activities of individual and group learning.

Course description

This course discusses the potential application of genomics and pharmacogenomics in disease monitoring, treatment, and control in individuals and populations by improving diagnostic accuracy. In addition, the course discusses the use of genomic technologies such as the genome-wide association studies and next-generation sequencing in infectious diseases.

Intended Learning outcomes (ILOs)

  1. Describe how the principles of human genetics are employed to optimize antimicrobial therapy and patient care.
  2. Recognize the impact of Pharmacogenomics in Antimicrobial treatment and how it improves various problems in drug therapy optimizations and patient care.
  3. Recognize the principles of genetic associations with susceptibility to infectious diseases.
  4. Describe bacterial pathogenicity and the development of antibiotics.
  5. Validate the use of metagenomic next-generation sequencing for diagnosing of infectious diseases.
  6. Examine how scientists craft an equitable and just ethical framework and a sustainable environment for effective policies.
  7. Investigate the genome-wide association results for the major infectious diseases.
  8. Examine challenges in pharmacogenomics professional practice towards infectious diseases and making relevant decisions in the light of guidelines and supplied information.
  9. Apply pharmacogenomics concepts to a particular antimicrobial therapy to solve relevant problems in healthcare.
  10. Determine the impact of genetic variation on susceptibility to infectious diseases.
  11. Show proficiency in applying basic pharmacogenomics and molecular genetic techniques in infectious diseases.
  12. Critically evaluate relevant literature with respect to pharmacogenomics in genetic testing for infectious diseases and its impact on patient care.
  13. Encourage the healthcare team collaboration regarding the dissemination of Pharmacogenomics concepts, patient education and collaboration between healthcare providers.

Course description

The course will study the molecular basis of drug action, protein structure-activity relationships, receptor-ligand interactions, signal transduction, different types of receptors from molecular point of view and their signalling mechanisms, quantitative aspects of ligand binding, receptor antagonism, functional selectivity, biologicals, mechanisms of drug resistance in cancer, consequences of genetic and epigenetic alteration in cancer, role of miRNA and its applications.

Intended Learning outcomes (ILOs)

At the end of this study-unit, students will be able to:

  1. Define pharmacological terms and concepts and outline the modes of action of drug at the cellular level.
  2. Differentiate between the primary characteristics of the principal superfamilies of drug targets (ligand-gated ion channels, growth factor receptors, steroid receptors and their signalling mechanisms)
  3. Summarise the development of biological and biotechnological drugs.
  4. Describe mechanisms of drug resistance in cancer and consequences of genetic and epigenetic alteration in cancer
  5. Detect the role of miRNA and possibilities of diagnostics and therapeutic applications
  6. Analyze methods of radio-ligand binding techniques and their application in the study of the receptor molecular pharmacology.
  7. Comprehend the efficacy of monoclonal antibodies and molecular target drugs
  8. Assess the mechanisms of action of clinically important drugs on receptors and enzymes
  9. Analyze data and solve problems related to drug resistance in cancer
  10. Predict potential new sites of drug action in the relevant cell signalling pathways.
  11. Develop contemporary application of scientific advances in the discipline of molecular pharmacology
  12. Develop skills critical for the application of radio-ligand binding techniques
  13. Correlate genetic and epigenetic alterations with cancer development
  14. Correlate microRNA with possible application in early cancer diagnosis and therapy
  15. Apply professional skills in the field of molecular pharmacology
  16. Self-assessment and self- and continuous learning.
  17. Use of different sources to obtain information and knowledge.
  18. Independently manage time.
  19. Communicate the molecular pharmacology data obtained from different resources to other colleagues