L5, Genomics and Health II Flashcards

1
Q

Trends in effect size/penetrance and allele frequency of gene variants:

A
  • Highly penetrant mutations (large effect size) tend to have lower frequency (e.g. CF)
  • Conversely, common variants typically have small effects (e.g. LMTK2 in Prostate cancer)
  • Outliers include APOE4 (Alzheimer’s)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

GWAS: What is it? How is association established?

A
  • Comparing two populations to see if frequency of genotypes is different
  • SNP array -> generating genotypes for individuals with and without trait
  • Higher incidence of a SNP allele in individuals with the trait compared to those without the trait = association (significance depends on degree of association and sample size)
  • Nearby candidate genes can be identified and tested
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Why are SNPs useful in GWAS? What are tag and lead SNPs?

A
  • SNP arrays use tag SNPs to represent SNPs nearby (haplotype), reducing the need to study every SNP
  • LD clumping allows computational determination of index (lead SNP) within LD blocks -> reduce amount of information, can thus combine results from multiple studies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Manhattan plots: What are they for?

A
  • Used to visualise the p-values for investigated SNPs across the genome locations in GWAS
  • Significant p-values show as peaks (uses -log^10(P) value)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Limitations of GWAS:

A
  • Cost millions
  • Low predictive value (statistical association only with odds ratios less than 1.5)
  • Around 90% of associated SNPs in non-coding DNA
  • Difficult to study combinations of genes as disease cause -> Future prospect: polygenic risk scores
  • Can’t correct for environmental factors (logistically difficult)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Precision medicine:

A
  • Therapy, instead of being generalised and responding to symptoms only, is tailored through DNA/RNA profiling
  • Targeted therapy avoids the different effects of individuals to drugs
  • Alternatively, DNA and RNA profiling can be used in determining how genetics influence drug response (pharmacogenetics)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Application of transcriptomics to breast cancer treatment:

A
  • Approximately 70% of all breast cancer patients present with ER+, HER2- breast cancer
  • Adjuvant chemotherapy improves prognoses, but is exposing a large percentage needlessly to radiotherapy
  • Oncotype DX (reverse transcriptase PCR-based), Mammaprint (micro-array based) are examples of transcriptomic methods for informing treatment via biomarker discovery
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Pharmacogenetics through genotyping arrays:

A
  • Using DNA as inputs, custom microarrays and amplicon sequencing can be used to investigate gene variants for specific gene targets after PCR amplification -> more appropriate drug dosing
  • e.g. CYP450 test (amplichip)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Amplichip CYP450 test:

A
  • Cytochrome P450 superfamily is a large and diverse group of enzymes that form the major system for metabolising lipids, hormones, toxins and drugs-> 12 of these are implicated in metabolism of most commonly used drugs
  • Amplichip tests for CYP2D6 and CYP2C19 alleles -> ascertaining which metabolizer subgroup an individual belongs to
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Use of genetic testing for rare and inherited disorders and cancers:

A
  • NHS currently tests a wide range of genetic disorders
  • Both whole genome and whole exome techniques (among others)
  • Useful to use a subset of genes in a panel - exon capture or exon amplification -> all possible alleles can be identified in one test (e.g. 450 gene panel for eye disorders)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Filtering criteria for candidates to test for:

A
  • Mutation must be likely to cause a change in gene expression or protein structure (e.g. nonsense, strong missense, splice site changes, frameshifts)
  • Mutation should not be found in SNP databases or control genome sequences
  • Same gene mutated in affected, unrelated individuals (and no unaffected individuals with the putative disease-causing genotype)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What issues have caused prior clinical tests to fail?

A
  • Causative variants in a gene not include in tested gene panel for amplicon sequencing / microarray
  • Intron variants and exon deletions are difficult to detect with exome sequencing
  • Non-coding RNA are not covered by exome sequencing
  • Poor coverage in initial genome sequencing
  • Novel triplet expansion disorder cannot be accurately estimated by short-read sequencing
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What might cause an NGS dead zone? How may they be recovered?

A
  • Genes with high homology or repetitive regions
  • Can sometimes be recovered using long-read NGS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

How can methylation status be used to detect cancers?

A
  • Circulating cell-free DNA is released form dying cells; in healthy individuals, mainly released from hematopoietic cells whereas in cancers, high rate of apoptosis/necrosis
  • Methylation patterns in cell-free DNA can indicate their tissue of origin (e.g. Galleri test -> enrichment through hybridisation capture and bisulfite DNA conversion and sequencing)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly