Clinical Omics

A.Y. 2023/2024
6
Max ECTS
48
Overall hours
SSD
MED/06 MED/11 MED/15
Language
English
Learning objectives
Precision medicine is rapidly evolving within cancer medicine, cardiovascular and neurological diseases and has brought many new concepts and terminologies that are often poorly defined when first introduced. We defined specific learning objectives for any specific learning module:
a) Omics in Oncology
Our learning objectives will be to
1. To acquire an omic "glossary" for clinical oncology (focus on solid tumors)
2. To understand the clinical utility and clinical validity of genomic testing in cancer medicine.
3. To assess risk of relapse according to omics data
4. To increase knowledge on risk assessment and patient stratification in the context of clinical trials
5. To assess minimal residual disease and to define the clinical utility and clinical validity of available tests.
b) Omics in Hematological Malignancies
1. To acquire an omic "glossary" for hematology-oncology (focus on hematological malignancies)
2. To understand the clinical utility and clinical validity of genomic testing in hematology.
3. To assess risk of relapse according to omics data
4. To increase knowledge on risk assessment and patient stratification in the context of clinical trials
5. To assess minimal residual disease and to define the clinical utility and clinical validity of available tests.
c) Cardio-genomics
1. To acquire an omic "glossary" for cardiology
2. To understand the clinical utility and clinical validity of genomic and proteomic testing in cardiology
3. To assess cardiac risk of relapse according to omics data
d) Neuro-genomics
1. To acquire an omic "glossary" for neurology
2. To understand the clinical utility and clinical validity of genomic and proteomic testing in neuroogical diseases
3. To assess risk of neurodegenerative disease according to omics data
Expected learning outcomes
We expect a better knowledge on the role of
1) Precision medicine in cancer, cardiology and neurology.
An healthcare approach with the primary aim of identifying which interventions are likely to be of most benefit to which patients based upon the features of the individual and their disease. In cancer, the term usually refers to the use of therapeutics that are expected to confer benefit to a subset of patients whose cancer displays specific molecular or cellular features (most commonly genomic changes and gene or protein expression patterns). Nevertheless, the term also includes the use of prognostic markers, predictors of toxicities and any parameter such as environmental and lifestyle factors that leads to treatment tailoring. Characterisation approaches in the future are expected to encompass a wider range of technologies such as functional imaging.
2) Pharmacogenomics.
A component of precision medicine—the study of how genomic variation within the individual or their disease (including gene expression, epigenetics, germline and somatic mutations) influences his/her response to drugs. In pharmacogenomics genomic variation is correlated with pharmacodynamics and pharmacokinetics. The aim of pharmacogenomics is to optimise drug therapy by maximising therapeutic effect and minimising adverse effects.
3) Stratified medicine
The use of a molecular assay to define subpopulations, rather than individuals, who are likely to benefit from a treatment intervention.
4) Molecular tumour board
A molecular tumour board is a specific type of multidisciplinary tumour board. In common with a classical multidisciplinary board it aims at providing clinical recommendations. A molecular tumour board, however, deals not only with the classical radiological, clinical and standard biological data of the patient, but also with modern molecular diagnostic tests. Its composition therefhours goes beyond that of multidisciplinary boards, encompassing molecular biologists, geneticists and bioinformaticians.
Single course

This course can be attended as a single course.

Course syllabus and organization

Single session

Course syllabus
2 CFU: Omics in Oncology (16 hours)
Clinical applications of omics technologies:- 4 hours
· Omics in cancer medicine
· Gene signature
· Prognostic and predictive gene signatures
The molecular tumor board:- 3 hours
· Role of the molecular tumor board
· Structure
· Reporting
· Data integration
Patient stratification and enrichement strategies in clinical trials- 4 hours
· Use of omic to stratify patients in clinical trials
· Umbrella trials vs basket trials
Clinical trial design and disease risk-assessment- 2 hours
· Enrichment trials
· Genomics signature in context of clinical trials and application in clinical practice
New tools for disease monitoring: the role of liquid biopsy :- 2 hours
· Prognostic role of circulating tumor DNA
· Predictive role of liquid biopsy

2 CFU: Omics in Hematological Malignancies (16 hours)
Clinical applications of omics technologies in Hematology:- 4 hours
· What is a biomarker? Types of biomarkers.
· Molecular diagnostic testing.
· Driver vs. passenger mutations
· Actionable molecular alterations
· .
· Genomic online resources
NGS and clinical decision making in Hematology:- 4 hours
Patient stratification and enrichement strategies in clinical trials:- 4 hours
· Use of omic to stratify patients in clinical trials
· Umbrella trials vs basket trials
· Disease and clonal evolution
Clinical trial design and disease risk-assessment:- 2 hours
· Enrichment trials
· Genomics signature in context of clinical trials and application in clinical practice
New tools for disease monitoring Alessandro - 2 hours
· Liquid biopsy and circulating biomarkers
1 CFU: Cardio-Genomics (8 hours)
Biomarkers in patient risk assessment: - 3 hours
Omic biomarkers for cardiovascular risk assessment
Patient risk stratification: - 3 hours
· Integration of omics to predict the risk of cardiovascular disease
Clinical trial design and cardio-toxicity risk-assessment: - 2 hours
Quality and outcome indicators
· Integration of big data and omics to predict cardio-toxicity
1 CFU: Neurogenomics (8 hours)
Neorogenomics in clinical setting:- 3 hours
· NGS approach to study Mitochondrial DNA
· Genes and neurological disease: Genotype and Phenotype correlation
Omics-based biomarkers for early diagnosis of neurodegenerative disorders - 3 hours
· Genomics of neurodegenerative disorders
Clinical trial design and risk-assessment for brain tumors: - 2 hours
How to design an clinical study in rare neurological diseases
· Emerging omics data in brain tumors
Prerequisites for admission
No prior knowledge is required.
Teaching methods
Lectures will be accompanied by practicals, working gropu and time for reflection and collective discussion.
Teaching Resources
Slides and scientific material provided during lectures
Assessment methods and Criteria
The examination is based on a multiple-choice test. We will ask 30 multiple choice questions and a minimum of 18 correct answers will be required to pass the examination
Attendance is required to be allowed to take the exam. Unexcused absence is tolerated up to 30% of the course activities
MED/06 - MEDICAL ONCOLOGY
MED/11 - CARDIOVASCULAR DISEASES
MED/15 - BLOOD DISEASES
Lessons: 48 hours
Educational website(s)
Professor(s)
Reception:
Please request an appointment via e-mail
Reception:
by telephone appointment
via L. Temolo 4, 20126 Milano - Lab of Neurogenetics and mitochondrial disorders
Reception:
To be arranged by email
ASST Grande Ospedale Metropolitano Niguarda, Blocco Sud